146 results on '"Yusa, Kosuke"'
Search Results
102. Hyperactive piggyBac Gene Transfer in Human Cells and In Vivo
- Author
-
Doherty, Joseph E., primary, Huye, Leslie E., additional, Yusa, Kosuke, additional, Zhou, Liqin, additional, Craig, Nancy L., additional, and Wilson, Matthew H., additional
- Published
- 2012
- Full Text
- View/download PDF
103. A homozygous mutant embryonic stem cell bank applicable for phenotype-driven genetic screening
- Author
-
Horie, Kyoji, primary, Kokubu, Chikara, additional, Yoshida, Junko, additional, Akagi, Keiko, additional, Isotani, Ayako, additional, Oshitani, Akiko, additional, Yusa, Kosuke, additional, Ikeda, Ryuji, additional, Huang, Yue, additional, Bradley, Allan, additional, and Takeda, Junji, additional
- Published
- 2011
- Full Text
- View/download PDF
104. Towards Cell Based Therapy For Alpha1-Antitrypsin Deficiency Through Bi-Allelic Gene Correction In Human IPS Cells
- Author
-
Lomas, David A., primary, Rashid, S Tamir, additional, Yusa, Kosuke, additional, Marciniak, Stefan J., additional, Miranda, Elena, additional, Ordonez, Adriana, additional, Hannan, Nick, additional, Alexander, Graeme, additional, Bradley, Allan, additional, and Vallier, Ludovic, additional
- Published
- 2011
- Full Text
- View/download PDF
105. Butyrate Greatly Enhances Derivation of Human Induced Pluripotent Stem Cells by Promoting Epigenetic Remodeling and the Expression of Pluripotency-Associated Genes
- Author
-
Mali, Prashant, primary, Chou, Bin-Kuan, additional, Yen, Jonathan, additional, Ye, Zhaohui, additional, Zou, Jizhong, additional, Dowey, Sarah, additional, Brodsky, Robert A., additional, Ohm, Joyce E., additional, Yu, Wayne, additional, Baylin, Stephen B., additional, Yusa, Kosuke, additional, Bradley, Allan, additional, Meyers, David J., additional, Mukherjee, Chandrani, additional, Cole, Philip A., additional, and Cheng, Linzhao, additional
- Published
- 2010
- Full Text
- View/download PDF
106. Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon
- Author
-
Yusa, Kosuke, primary, Rad, Roland, additional, Takeda, Junji, additional, and Bradley, Allan, additional
- Published
- 2009
- Full Text
- View/download PDF
107. Sleeping Beauty Transposase Has an Affinity for Heterochromatin Conformation
- Author
-
Ikeda, Ryuji, primary, Kokubu, Chikara, additional, Yusa, Kosuke, additional, Keng, Vincent W., additional, Horie, Kyoji, additional, and Takeda, Junji, additional
- Published
- 2007
- Full Text
- View/download PDF
108. Off-target assessment of CRISPR- Cas9 guiding RNAs in human i PS and mouse ES cells.
- Author
-
Tan, E-Pien, Li, Yilong, Del Castillo Velasco-Herrera, Martin, Yusa, Kosuke, and Bradley, Allan
- Published
- 2015
- Full Text
- View/download PDF
109. Sleeping Beauty Transposon-Based Phenotypic Analysis of Mice: Lack of Arpc3 Results in Defective Trophoblast Outgrowth
- Author
-
Yae, Kojiro, primary, Keng, Vincent W., additional, Koike, Masato, additional, Yusa, Kosuke, additional, Kouno, Michiyoshi, additional, Uno, Yoshihiro, additional, Kondoh, Gen, additional, Gotow, Takahiro, additional, Uchiyama, Yasuo, additional, Horie, Kyoji, additional, and Takeda, Junji, additional
- Published
- 2006
- Full Text
- View/download PDF
110. Bloom's syndrome gene-deficient phenotype in mouse primary cells induced by a modified tetracycline-controlled trans-silencer
- Author
-
Hayakawa, Tomoko, primary, Yusa, Kosuke, additional, Kouno, Michiyoshi, additional, Takeda, Junji, additional, and Horie, Kyoji, additional
- Published
- 2006
- Full Text
- View/download PDF
111. Region-specific saturation germline mutagenesis in mice using the Sleeping Beauty transposon system
- Author
-
Keng, Vincent W, primary, Yae, Kojiro, additional, Hayakawa, Tomoko, additional, Mizuno, Sumi, additional, Uno, Yoshihiro, additional, Yusa, Kosuke, additional, Kokubu, Chikara, additional, Kinoshita, Taroh, additional, Akagi, Keiko, additional, Jenkins, Nancy A, additional, Copeland, Neal G, additional, Horie, Kyoji, additional, and Takeda, Junji, additional
- Published
- 2005
- Full Text
- View/download PDF
112. Enhancement of Sleeping Beauty Transposition by CpG Methylation: Possible Role of Heterochromatin Formation
- Author
-
Yusa, Kosuke, primary, Takeda, Junji, additional, and Horie, Kyoji, additional
- Published
- 2004
- Full Text
- View/download PDF
113. Characterization of Sleeping Beauty Transposition and Its Application to Genetic Screening in Mice
- Author
-
Horie, Kyoji, primary, Yusa, Kosuke, additional, Yae, Kojiro, additional, Odajima, Junko, additional, Fischer, Sylvia E. J., additional, Keng, Vincent W., additional, Hayakawa, Tomoko, additional, Mizuno, Sumi, additional, Kondoh, Gen, additional, Ijiri, Takashi, additional, Matsuda, Yoichi, additional, Plasterk, Ronald H. A., additional, and Takeda, Junji, additional
- Published
- 2003
- Full Text
- View/download PDF
114. Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library.
- Author
-
Koike-Yusa, Hiroko, Li, Yilong, Tan, E-Pien, Velasco-Herrera, Martin Del Castillo, and Yusa, Kosuke
- Subjects
GENETIC testing ,RNA interference ,PALINDROMIC DNA ,LENTIVIRUSES ,GENETIC mutation ,EMBRYONIC stem cells ,EXONS (Genetics) - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
115. Hyperactive piggyBacGene Transfer in Human Cells and In Vivo.
- Author
-
Doherty, Joseph E., Huye, Leslie E., Yusa, Kosuke, Zhou, Liqin, Craig, Nancy L., and Wilson, Matthew H.
- Published
- 2012
- Full Text
- View/download PDF
116. Targeted gene correction of ?1-antitrypsin deficiency in induced pluripotent stem cells.
- Author
-
Yusa, Kosuke, Rashid, S. Tamir, Strick-Marchand, Helene, Varela, Ignacio, Liu, Pei-Qi, Paschon, David E., Miranda, Elena, Ordóñez, Adriana, Hannan, Nicholas R. F., Rouhani, Foad J., Darche, Sylvie, Alexander, Graeme, Marciniak, Stefan J., Fusaki, Noemi, Hasegawa, Mamoru, Holmes, Michael C., Di Santo, James P., Lomas, David A., Bradley, Allan, and Vallier, Ludovic
- Subjects
GENE targeting ,GENETIC recombination ,PLURIPOTENT stem cells ,REGENERATIVE medicine ,TRYPSIN inhibitors ,MEDICAL genetics - Abstract
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 ?
1 -antitrypsin (A1AT, also known as SERPINA1) gene that is responsible for ?1 -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. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
117. Butyrate Greatly Enhances Derivation of Human Induced Pluripotent Stem Cells by Promoting Epigenetic Remodeling and the Expression of Pluripotency-Associated Genes.
- Author
-
MALL, PRASHANT, BIN-KUAN CHOU, YEN, JONATHAN, ZHAOHUI YE, JIZHONG ZOU, DOWEY, SARAH, BRODSKY, ROBERT A., OHM, JOYCE E., WAYNE YU, BAYLIN, STEPHEN B., YUSA, KOSUKE, BRADLEY, ALLAN, EYERS, DAVID J., MUKHERJEE, CHANDRANI, COLE, PHILIP A., and LINZHAO CHENG
- Subjects
STEM cells ,SCOLYTIDAE ,CELL culture ,SICKLE cell anemia ,CHROMOSOME abnormalities - Abstract
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 transpuson 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. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
118. The CADM1 tumor suppressor gene is a major candidate gene in MDS with deletion of the long arm of chromosome 11
- Author
-
Lafage-Pochitaloff, Marina, Gerby, Bastien, Baccini, Véronique, Largeaud, Laetitia, Fregona, Vincent, Prade, Naïs, Juvin, Pierre-Yves, Jamrog, Laura, Bories, Pierre, Hébrard, Sylvie, Lagarde, Stéphanie, Mansat-De Mas, Véronique, Dovey, Oliver M, Yusa, Kosuke, Vassiliou, George S, Jansen, Joop H, Tekath, Tobias, Rombaut, David, Ameye, Geneviève, Barin, Carole, Bidet, Audrey, Boudjarane, John, Collonge-Rame, Marie-Agnès, Gervais, Carine, Ittel, Antoine, Lefebvre, Christine, Luquet, Isabelle, Michaux, Lucienne, Nadal, Nathalie, Poirel, Hélène A, Radford-Weiss, Isabelle, Ribourtout, Bénédicte, Richebourg, Steven, Struski, Stéphanie, Terré, Christine, Tigaud, Isabelle, Penther, Dominique, Eclache, Virginie, Fontenay, Michaela, Broccardo, Cyril, and Delabesse, Eric
- Subjects
Leukemia, Myeloid, Acute ,Mice ,hemic and lymphatic diseases ,Chromosomes, Human, Pair 11 ,Myelodysplastic Syndromes ,Cell Adhesion Molecule-1 ,Animals ,Humans ,Female ,Genes, Tumor Suppressor ,Chromosome Deletion ,3. Good health - Abstract
Myelodysplastic syndromes (MDS) represent a heterogeneous group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis leading to peripheral cytopenias and in a substantial proportion of cases to acute myeloid leukemia. The deletion of the long arm of chromosome 11, del(11q), is a rare but recurrent clonal event in MDS. Here, we detail the largest series of 113 cases of MDS and myelodysplastic syndromes/myeloproliferative neoplasms (MDS/MPN) harboring a del(11q) analyzed at clinical, cytological, cytogenetic, and molecular levels. Female predominance, a survival prognosis similar to other MDS, a low monocyte count, and dysmegakaryopoiesis were the specific clinical and cytological features of del(11q) MDS. In most cases, del(11q) was isolated, primary and interstitial encompassing the 11q22-23 region containing ATM, KMT2A, and CBL genes. The common deleted region at 11q23.2 is centered on an intergenic region between CADM1 (also known as Tumor Suppressor in Lung Cancer 1) and NXPE2. CADM1 was expressed in all myeloid cells analyzed in contrast to NXPE2. At the functional level, the deletion of Cadm1 in murine Lineage-Sca1+Kit+ cells modifies the lymphoid-to-myeloid ratio in bone marrow, although not altering their multilineage hematopoietic reconstitution potential after syngenic transplantation. Together with the frequent simultaneous deletions of KMT2A, ATM, and CBL and mutations of ASXL1, SF3B1, and CBL, we show that CADM1 may be important in the physiopathology of the del(11q) MDS, extending its role as tumor-suppressor gene from solid tumors to hematopoietic malignancies.
119. ATM orchestrates the DNA-damage response to counter toxic non-homologous end-joining at broken replication forks
- Author
-
Balmus, Gabriel, Pilger, Domenic, Coates, Julia, Demir, Mukerrem, Sczaniecka-Clift, Matylda, Barros, Ana C, Woods, Michael, Fu, Beiyuan, Yang, Fengtang, Chen, Elisabeth, Ostermaier, Matthias, Stankovic, Tatjana, Ponstingl, Hannes, Herzog, Mareike, Yusa, Kosuke, Martinez, Francisco Munoz, Durant, Stephen T, Galanty, Yaron, Beli, Petra, Adams, David J, Bradley, Allan, Metzakopian, Emmanouil, Forment, Josep V, and Jackson, Stephen P
- Subjects
DNA Replication ,Mice, Knockout ,DNA End-Joining Repair ,endocrine system diseases ,BRCA1 Protein ,Cell Survival ,Antineoplastic Agents ,Mouse Embryonic Stem Cells ,Ataxia Telangiectasia Mutated Proteins ,Neoplasms, Experimental ,Piperazines ,3. Good health ,DNA Ligase ATP ,Mice ,Drug Resistance, Neoplasm ,Mice, Inbred NOD ,Cell Line, Tumor ,Mutation ,Animals ,Humans ,Phthalazines ,DNA Breaks, Double-Stranded ,Female ,CRISPR-Cas Systems ,Topotecan - Abstract
Mutations in the ATM tumor suppressor gene confer hypersensitivity to DNA-damaging chemotherapeutic agents. To explore genetic resistance mechanisms, we performed genome-wide CRISPR-Cas9 screens in cells treated with the DNA topoisomerase I inhibitor topotecan. Thus, we here establish that inactivating terminal components of the non-homologous end-joining (NHEJ) machinery or of the BRCA1-A complex specifically confer topotecan resistance to ATM-deficient cells. We show that hypersensitivity of ATM-mutant cells to topotecan or the poly-(ADP-ribose) polymerase (PARP) inhibitor olaparib reflects delayed engagement of homologous recombination at DNA-replication-fork associated single-ended double-strand breaks (DSBs), allowing some to be subject to toxic NHEJ. Preventing DSB ligation by NHEJ, or enhancing homologous recombination by BRCA1-A complex disruption, suppresses this toxicity, highlighting a crucial role for ATM in preventing toxic LIG4-mediated chromosome fusions. Notably, suppressor mutations in ATM-mutant backgrounds are different to those in BRCA1-mutant scenarios, suggesting new opportunities for patient stratification and additional therapeutic vulnerabilities for clinical exploitation.
120. A CRISPR Dropout Screen Identifies Genetic Vulnerabilities and Therapeutic Targets in Acute Myeloid Leukemia
- Author
-
Tzelepis, Konstantinos, Koike-Yusa, Hiroko, De Braekeleer, Etienne, Li, Yilong, Metzakopian, Emmanouil, Dovey, Oliver M., Mupo, Annalisa, Grinkevich, Vera, Li, Meng, Mazan, Milena, Gozdecka, Malgorzata, Ohnishi, Shuhei, Cooper, Jonathan, Patel, Miten, McKerrell, Thomas, Chen, Bin, Domingues, Ana Filipa, Gallipoli, Paolo, Teichmann, Sarah, Ponstingl, Hannes, McDermott, Ultan, Saez-Rodriguez, Julio, Huntly, Brian J P., Iorio, Francesco, Pina, Cristina, Vassiliou, George S., and Yusa, Kosuke
- Subjects
3. Good health - Abstract
Cell reports 17(4), 1193-1205 (2016). doi:10.1016/j.celrep.2016.09.079, Published by Cell Press, Maryland Heights, MO
121. Additional file 1: of Pooled extracellular receptor-ligand interaction screening using CRISPR activation
- Author
-
Zheng-Shan Chong, Ohnishi, Shuhei, Yusa, Kosuke, and Wright, Gavin
- Subjects
3. Good health - Abstract
Table S1. A table listing the gRNAs sequences targeting the promoter regions for the named genes. The gene symbol, accession number of the target transcript and chromosomal location are provided. Table S2. A table detailing the sequences of the synthesized DNA fragments and PCR primers used for plasmid construction and sequencing, primers used for q-RT-PCR, and primers for gRNA library preparation and amplification. Table S3. A table providing the sources, and where appropriate, clone names of the primary monoclonal and conjugated secondary antibodies used in this study. (PDF 553 kb)
122. Additional file 2: of Pooled extracellular receptor-ligand interaction screening using CRISPR activation
- Author
-
Zheng-Shan Chong, Ohnishi, Shuhei, Yusa, Kosuke, and Wright, Gavin
- Subjects
3. Good health - Abstract
Figure S1. CRISPR activation enables rapid and stable upregulation of cell surface proteins. Figure S2. A CRISPR activation gRNA library targeting membrane-associated proteins. Figure S3. Enrichment of gRNAs targeting known receptors in cells selected using their corresponding ligand. Figure S4. ADGRB1 directly interacts with all three members of the RTN4R family. (PDF 489 kb)
123. ARID1A influences HDAC1/BRD4 activity, intrinsic proliferative capacity and breast cancer treatment response
- Author
-
Nagarajan, Sankari, Rao, Shalini V, Sutton, Joseph, Cheeseman, Danya, Dunn, Shanade, Papachristou, Evangelia K, Prada, Jose-Enrique Gonzalez, Couturier, Dominique-Laurent, Kumar, Sanjeev, Kishore, Kamal, Chilamakuri, Chandra Sekhar Reddy, Glont, Silvia-Elena, Archer Goode, Emily, Brodie, Cara, Guppy, Naomi, Natrajan, Rachael, Bruna, Alejandra, Caldas, Carlos, Russell, Alasdair, Siersbæk, Rasmus, Yusa, Kosuke, Chernukhin, Igor, and Carroll, Jason S
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Estrogen Receptor alpha ,Acetylation ,Breast Neoplasms ,Cell Cycle Proteins ,Histone Deacetylase 1 ,Xenograft Model Antitumor Assays ,3. Good health ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Drug Resistance, Neoplasm ,Mice, Inbred NOD ,MCF-7 Cells ,Animals ,Humans ,Clustered Regularly Interspaced Short Palindromic Repeats ,Female ,Cell Proliferation ,Transcription Factors - Abstract
Using genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) screens to understand endocrine drug resistance, we discovered ARID1A and other SWI/SNF complex components as the factors most critically required for response to two classes of estrogen receptor-alpha (ER) antagonists. In this context, SWI/SNF-specific gene deletion resulted in drug resistance. Unexpectedly, ARID1A was also the top candidate in regard to response to the bromodomain and extraterminal domain inhibitor JQ1, but in the opposite direction, with loss of ARID1A sensitizing breast cancer cells to bromodomain and extraterminal domain inhibition. We show that ARID1A is a repressor that binds chromatin at ER cis-regulatory elements. However, ARID1A elicits repressive activity in an enhancer-specific, but forkhead box A1-dependent and active, ER-independent manner. Deletion of ARID1A resulted in loss of histone deacetylase 1 binding, increased histone 4 lysine acetylation and subsequent BRD4-driven transcription and growth. ARID1A mutations are more frequent in treatment-resistant disease, and our findings provide mechanistic insight into this process while revealing rational treatment strategies for these patients.
124. Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon
- Author
-
Yusa, Kosuke, Rad, Roland, Takeda, Junji, and Bradley, Allan
- Subjects
Pluripotent Stem Cells ,Chromosomal Proteins, Non-Histone ,Genetic Vectors ,Kruppel-Like Transcription Factors ,Gene Expression ,Transposases ,Moths ,Transfection ,Proto-Oncogene Proteins c-myc ,Kruppel-Like Factor 4 ,Mice ,Animals ,Humans ,Transgenes ,Homeodomain Proteins ,Chimera ,SOXB1 Transcription Factors ,Valproic Acid ,fungi ,Teratoma ,RNA-Binding Proteins ,Cell Differentiation ,Nanog Homeobox Protein ,Cell Dedifferentiation ,Fibroblasts ,Alkaline Phosphatase ,Embryo, Mammalian ,3. Good health ,Repressor Proteins ,DNA Transposable Elements ,Octamer Transcription Factor-3 - Abstract
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.
125. Additional file 1: of Pooled extracellular receptor-ligand interaction screening using CRISPR activation
- Author
-
Zheng-Shan Chong, Ohnishi, Shuhei, Yusa, Kosuke, and Wright, Gavin
- Subjects
3. Good health - Abstract
Table S1. A table listing the gRNAs sequences targeting the promoter regions for the named genes. The gene symbol, accession number of the target transcript and chromosomal location are provided. Table S2. A table detailing the sequences of the synthesized DNA fragments and PCR primers used for plasmid construction and sequencing, primers used for q-RT-PCR, and primers for gRNA library preparation and amplification. Table S3. A table providing the sources, and where appropriate, clone names of the primary monoclonal and conjugated secondary antibodies used in this study. (PDF 553 kb)
126. KAT7 is a genetic vulnerability of acute myeloid leukemias driven by MLL rearrangements
- Author
-
Au, Yan Zi, Gu, Muxin, De Braekeleer, Etienne, Gozdecka, Malgorzata, Aspris, Demetrios, Tarumoto, Yusuke, Cooper, Jonathan, Yu, Jason, Ong, Swee Hoe, Chen, Xi, Tzelepis, Konstantinos, Huntly, Brian JP, Vassiliou, George, and Yusa, Kosuke
- Subjects
Gene Rearrangement ,Oncogene Proteins, Fusion ,Disease Management ,Apoptosis ,Cell Differentiation ,Histone-Lysine N-Methyltransferase ,3. Good health ,Epigenesis, Genetic ,Histones ,Gene Knockout Techniques ,Leukemia, Myeloid, Acute ,hemic and lymphatic diseases ,Cell Line, Tumor ,Biomarkers, Tumor ,Humans ,Genetic Predisposition to Disease ,Myeloid Cells ,Promoter Regions, Genetic ,neoplasms ,Genetic Association Studies ,Myeloid-Lymphoid Leukemia Protein ,Histone Acetyltransferases ,Protein Binding - Abstract
Histone acetyltransferases (HATs) catalyze the transfer of an acetyl group from acetyl-CoA to lysine residues of histones and play a central role in transcriptional regulation in diverse biological processes. Dysregulation of HAT activity can lead to human diseases including developmental disorders and cancer. Through genome-wide CRISPR-Cas9 screens, we identified several HATs of the MYST family as fitness genes for acute myeloid leukemia (AML). Here we investigate the essentiality of lysine acetyltransferase KAT7 in AMLs driven by the MLL-X gene fusions. We found that KAT7 loss leads to a rapid and complete loss of both H3K14ac and H4K12ac marks, in association with reduced proliferation, increased apoptosis, and differentiation of AML cells. Acetyltransferase activity of KAT7 is essential for the proliferation of these cells. Mechanistically, our data propose that acetylated histones provide a platform for the recruitment of MLL-fusion-associated adaptor proteins such as BRD4 and AF4 to gene promoters. Upon KAT7 loss, these factors together with RNA polymerase II rapidly dissociate from several MLL-fusion target genes that are essential for AML cell proliferation, including MEIS1, PBX3, and SENP6. Our findings reveal that KAT7 is a plausible therapeutic target for this poor prognosis AML subtype.
127. Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells
- Author
-
Yusa, Kosuke, Rashid, S Tamir, Strick-Marchand, Helene, Varela, Ignacio, Liu, Pei-Qi, Paschon, David E, Miranda, Elena, Ordóñez, Adriana, Hannan, Nicholas RF, Rouhani, Foad J, Darche, Sylvie, Alexander, Graeme, Marciniak, Stefan J, Fusaki, Noemi, Hasegawa, Mamoru, Holmes, Michael C, Di Santo, James P, Lomas, David A, Bradley, Allan, and Vallier, Ludovic
- Subjects
Time Factors ,Induced Pluripotent Stem Cells ,Serum Albumin, Human ,3. Good health ,Cell Line ,Mice ,Liver ,alpha 1-Antitrypsin Deficiency ,alpha 1-Antitrypsin ,Hepatocytes ,DNA Transposable Elements ,Animals ,Humans ,Serum Albumin ,Targeted Gene Repair - Abstract
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 α(1)-antitrypsin (A1AT, also known as SERPINA1) gene that is responsible for α(1)-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.
128. Additional file 2: of Pooled extracellular receptor-ligand interaction screening using CRISPR activation
- Author
-
Zheng-Shan Chong, Ohnishi, Shuhei, Yusa, Kosuke, and Wright, Gavin
- Subjects
3. Good health - Abstract
Figure S1. CRISPR activation enables rapid and stable upregulation of cell surface proteins. Figure S2. A CRISPR activation gRNA library targeting membrane-associated proteins. Figure S3. Enrichment of gRNAs targeting known receptors in cells selected using their corresponding ligand. Figure S4. ADGRB1 directly interacts with all three members of the RTN4R family. (PDF 489 kb)
129. A genetic progression model of Braf(V600E)-induced intestinal tumorigenesis reveals targets for therapeutic intervention
- Author
-
Rad, Roland, Cadiñanos, Juan, Rad, Lena, Varela, Ignacio, Strong, Alexander, Kriegl, Lydia, Constantino-Casas, Fernando, Eser, Stefan, Hieber, Maren, Seidler, Barbara, Price, Stacey, Fraga, Mario F, Calvanese, Vincenzo, Hoffman, Gary, Ponstingl, Hannes, Schneider, Günter, Yusa, Kosuke, Grove, Carolyn, Schmid, Roland M, Wang, Wei, Vassiliou, George, Kirchner, Thomas, McDermott, Ultan, Liu, Pentao, Saur, Dieter, and Bradley, Allan
- Subjects
Proto-Oncogene Proteins B-raf ,MAP Kinase Signaling System ,digestive system diseases ,3. Good health ,Neoplasm Proteins ,Mice ,Cell Transformation, Neoplastic ,Mutation ,Animals ,Microsatellite Instability ,Neoplasm Invasiveness ,Drug Screening Assays, Antitumor ,Tumor Suppressor Protein p53 ,Colorectal Neoplasms ,Wnt Signaling Pathway ,Cyclin-Dependent Kinase Inhibitor p16 ,Phosphoinositide-3 Kinase Inhibitors - Abstract
We show that BRAF(V600E) 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 Braf(V600E) 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.
130. SRPK1 maintains acute myeloid leukemia through effects on isoform usage of epigenetic regulators including BRD4
- Author
-
Tzelepis, Konstantinos, De Braekeleer, Etienne, Aspris, Demetrios, Barbieri, Isaia, Vijayabaskar, MS, Liu, Wen-Hsin, Gozdecka, Malgorzata, Metzakopian, Emmanouil, Toop, Hamish D, Dudek, Monika, Robson, Samuel C, Hermida-Prado, Francisco, Yang, Yu Hsuen, Babaei-Jadidi, Roya, Garyfallos, Dimitrios A, Ponstingl, Hannes, Dias, Joao ML, Gallipoli, Paolo, Seiler, Michael, Buonamici, Silvia, Vick, Binje, Bannister, Andrew J, Rad, Roland, Prinjha, Rab K, Marioni, John C, Huntly, Brian, Batson, Jennifer, Morris, Jonathan C, Pina, Cristina, Bradley, Allan, Jeremias, Irmela, Bates, David O, Yusa, Kosuke, Kouzarides, Tony, and Vassiliou, George S
- Subjects
RNA Splicing ,Nuclear Proteins ,Cell Cycle Proteins ,Cell Differentiation ,HL-60 Cells ,Cell Cycle Checkpoints ,Protein Serine-Threonine Kinases ,Chromatin ,3. Good health ,Epigenesis, Genetic ,Hematopoiesis ,Leukemia, Myeloid, Acute ,hemic and lymphatic diseases ,Humans ,Protein Isoforms ,K562 Cells ,Transcription Factors - Abstract
We recently identified the splicing kinase gene SRPK1 as a genetic vulnerability of acute myeloid leukemia (AML). Here, we show that genetic or pharmacological inhibition of SRPK1 leads to cell cycle arrest, leukemic cell differentiation and prolonged survival of mice transplanted with MLL-rearranged AML. RNA-seq analysis demonstrates that SRPK1 inhibition leads to altered isoform levels of many genes including several with established roles in leukemogenesis such as MYB, BRD4 and MED24. We focus on BRD4 as its main isoforms have distinct molecular properties and find that SRPK1 inhibition produces a significant switch from the short to the long isoform at the mRNA and protein levels. This was associated with BRD4 eviction from genomic loci involved in leukemogenesis including BCL2 and MYC. We go on to show that this switch mediates at least part of the anti-leukemic effects of SRPK1 inhibition. Our findings reveal that SRPK1 represents a plausible new therapeutic target against AML.
131. Agreement between two large pan-cancer CRISPR-Cas9 gene dependency data sets
- Author
-
Dempster, Joshua M, Pacini, Clare, Pantel, Sasha, Behan, Fiona M, Green, Thomas, Krill-Burger, John, Beaver, Charlotte M, Younger, Scott T, Zhivich, Victor, Najgebauer, Hanna, Allen, Felicity, Gonçalves, Emanuel, Shepherd, Rebecca, Doench, John G, Yusa, Kosuke, Vazquez, Francisca, Parts, Leopold, Boehm, Jesse S, Golub, Todd R, Hahn, William C, Root, David E, Garnett, Mathew J, Tsherniak, Aviad, and Iorio, Francesco
- Subjects
Genes, Essential ,Gene Expression Profiling ,Datasets as Topic ,Reproducibility of Results ,Antineoplastic Agents ,Genomics ,Oncogenes ,3. Good health ,Small Molecule Libraries ,Cell Line, Tumor ,Neoplasms ,Biomarkers, Tumor ,Humans ,Molecular Targeted Therapy ,CRISPR-Cas Systems ,Drug Screening Assays, Antitumor ,Precision Medicine - Abstract
Genome-scale CRISPR-Cas9 viability screens performed in cancer cell lines provide a systematic approach to identify cancer dependencies and new therapeutic targets. As multiple large-scale screens become available, a formal assessment of the reproducibility of these experiments becomes necessary. We analyze data from recently published pan-cancer CRISPR-Cas9 screens performed at the Broad and Sanger Institutes. Despite significant differences in experimental protocols and reagents, we find that the screen results are highly concordant across multiple metrics with both common and specific dependencies jointly identified across the two studies. Furthermore, robust biomarkers of gene dependency found in one data set are recovered in the other. Through further analysis and replication experiments at each institute, we show that batch effects are driven principally by two key experimental parameters: the reagent library and the assay length. These results indicate that the Broad and Sanger CRISPR-Cas9 viability screens yield robust and reproducible findings.
132. Genetic Vulnerabilities of DNMT3AR882Hin Myeloid Malignancies
- Author
-
Gozdecka, Malgorzata, Dudek, Monika, Tzelepis, Konstantinos, Damaskou, Aristi, Baskar, Vijay, Wright, Penny, Duddy, Graham, Meduri, Eshwar, Mazan, Milena, Yusa, Kosuke, Huntly, Brian J.P., and Vassiliou, George S.
- Abstract
Mutations affecting the gene for the de novoDNA Methyltransferase 3A (DNMT3A) are the most common drivers of clonal hematopoiesis (CH) and amongst the most common somatic events in acute myeloid leukaemia (AML). Approximately two thirds of AML-associated DNMT3Amutations are heterozygous substitutions affecting codon R882, located within the methytransferase domain and are correlated with global hypomethylation. DNMT3Amutations are initiating events in leukemogenesis and leukemic progression relies on the acquisition of additional mutations in genes such as NPM1, TET2, IDH1/2, FLT3 and NRAS. AMLs harboring DNMT3A-R882mutations display increased resistance to chemotherapy and carry a worse prognosis for patients, but the molecular basis of this is not well understood.
- Published
- 2019
- Full Text
- View/download PDF
133. Enhancement of microhomology-mediated genomic rearrangements by transient loss of mouse Bloom syndrome helicase.
- Author
-
Yamanishi, Ayako, Yusa, Kosuke, Horie, Kyoji, Tokunaga, Masahiro, Kusano, Kohji, Kokubu, Chikara, and Takeda, Junji
- Subjects
- *
EMBRYONIC stem cells , *HUMAN cloning , *BLOOM syndrome , *COMPARATIVE genomic hybridization , *NUCLEIC acids - Abstract
Bloom syndrome, an autosomal recessive disorder of the BLM gene, confers predisposition to a broad spectrum of early-onset cancers in multiple tissue types. Loss of genomic integrity is a primary hallmark of such human malignancies, but many studies using disease-affected specimens are limited in that they are retrospective and devoid of an appropriate experimental control. To overcome this, we devised an experimental system to recapitulate the early molecular events in genetically engineered mouse embryonic stem cells, in which cells undergoing loss of heterozygosity (LOH) can be enriched after inducible down-regulation of Blm expression, with or without site-directed DNA double-strand break (DSB) induction. Transient loss of BLM increased the rate of LOH, whose breakpoints were distributed along the chromosome. Combined with site-directed DSB induction, loss of BLM synergistically increased the rate of LOH and concentrated the breakpoints around the targeted chromosomal region. We characterized the LOH events using specifically tailored genomic tools, such as high-resolution array comparative genomic hybridization and high-density single nucleotide polymorphism genotyping, revealing that the combination of BLM suppression and DSB induction enhanced genomic rearrangements, including deletions and insertions, whose breakpoints were clustered in genomic inverted repeats and associated with junctional microhomologies. Our experimental approach successfully uncovered the detailed molecular mechanisms of as-yet-uncharacterized loss of heterozygosities and reveals the significant contribution of microhomology-mediated genomic rearrangements, which could be widely applicable to the early steps of cancer formation in general. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
134. Genetic dissection of the exit of pluripotency in mouse embryonic stem cells by CRISPR-based screening
- Author
-
Li, Meng and Yusa, Kosuke
- Subjects
571.8 ,Pluripotency ,CRISPR ,Genetic screen - Abstract
The ground state naive pluripotency is established in the epiblast of the blastocyst and can be captured by culturing mouse embryonic stem cells (mESCs) with MEK and GSK3 inhibitors (2i). The transcription network that maintains pluripotency has been extensively studied with the indispensable core factors being Oct4, Sox2 and Nanog, together with other ancillary factors reinforcing the network. However, how this network is dissolved at the onset of differentiation is still not fully understood. To identify genes required for differentiation in an unbiased fashion, I conducted a genome-wide CRISPR-Cas9-mediated screen in Rex1GFPd2 mESCs. This cell line expresses GFP specifically in the naive state and rapidly down-regulate upon differentiation. I differentiated mutagenised mESCs for two days and sorted mutants that kept higher GFP expression. gRNA representation was subsequently analysed by sequencing. I identified 563 and 8 genes whose mutants showed delayed and accelerated differentiation, respectively, at a false discovery rate (FDR) cutoff of 10%. The majority of the previously known genes were identified in my screen, suggesting faithful representation of genes regulating differentiation. Detailed screening result analysis revealed a comprehensive picture of pathways involved in the dissolution of naive pluripotency. Amongst the genes identified are 19 mTORC1 regulators and components of the mTORC2 complex. Deficiency in the TSC and GATOR complexes resulted in mTORC1 upregulation in consistent with previous studies. However, they showed opposite phenotype during ESC differentiation: TSC complex knockout cells showed delayed differentiation, whereas GATOR1 deficiency accelerated differentiation I found that the pattern of GSK3b phosphorylation is highly correlated with differentiation phenotype. I conclude that mTORC1 is involved in pluripotency maintenance and differentiation through cross-talk with the Wnt signalling pathway. My screen has demonstrated the power of CRISPR-Cas9-mediated screen and provided further insights in biological pathways involved in regulating differentiation. It would be interesting to explore the remaining unstudied genes for better understanding of the mechanisms underlying mESC differentiation.
- Published
- 2018
- Full Text
- View/download PDF
135. Genome-wide screening identifies Polycomb repressive complex 1.3 as an essential regulator of human naïve pluripotent cell reprogramming
- Author
-
Amanda J. Collier, Adam Bendall, Charlene Fabian, Andrew A. Malcolm, Katarzyna Tilgner, Claudia I. Semprich, Katarzyna Wojdyla, Paola Serena Nisi, Kamal Kishore, Valar Nila Roamio Franklin, Bahar Mirshekar-Syahkal, Clive D’Santos, Kathrin Plath, Kosuke Yusa, Peter J. Rugg-Gunn, Collier, Amanda J [0000-0003-1137-6874], Bendall, Adam [0000-0002-6865-2625], Malcolm, Andrew A [0000-0001-6240-7701], Semprich, Claudia I [0000-0002-2446-9028], Wojdyla, Katarzyna [0000-0003-4509-1818], Kishore, Kamal [0000-0002-4650-8745], Mirshekar-Syahkal, Bahar [0000-0002-2337-9442], D'Santos, Clive [0000-0003-3425-7288], Plath, Kathrin [0000-0001-7796-3372], Yusa, Kosuke [0000-0002-3442-021X], Rugg-Gunn, Peter J [0000-0002-9601-5949], and Apollo - University of Cambridge Repository
- Subjects
Pluripotent Stem Cells ,Polycomb Repressive Complex 1 ,Multidisciplinary ,1.1 Normal biological development and functioning ,Human Genome ,Cell Differentiation ,Stem Cell Research ,Regenerative Medicine ,Cellular Reprogramming ,Gene Expression Regulation ,Underpinning research ,Genetics ,Humans ,Generic health relevance ,Stem Cell Research - Embryonic - Human - Abstract
Uncovering the mechanisms that establish naïve pluripotency in humans is crucial for the future applications of pluripotent stem cells including the production of human blastoids. However, the regulatory pathways that control the establishment of naïve pluripotency by reprogramming are largely unknown. Here, we use genome-wide screening to identify essential regulators as well as major impediments of human primed to naïve pluripotent stem cell reprogramming. We discover that factors essential for cell state change do not typically undergo changes at the level of gene expression but rather are repurposed with new functions. Mechanistically, we establish that the variant Polycomb complex PRC1.3 and PRDM14 jointly repress developmental and gene regulatory factors to ensure naïve cell reprogramming. In addition, small-molecule inhibitors of reprogramming impediments improve naïve cell reprogramming beyond current methods. Collectively, this work defines the principles controlling the establishment of human naïve pluripotency and also provides new insights into mechanisms that destabilize and reconfigure cell identity during cell state transitions.
- Published
- 2022
136. The CADM1 tumor suppressor gene is a major candidate gene in MDS with deletion of the long arm of chromosome 11
- Author
-
Virginie Eclache, John Boudjarane, Naïs Prade, Stéphanie Struski, Laetitia Largeaud, Cyril Broccardo, Joop H. Jansen, Christine Terré, Marie-Agnès Collonge-Rame, Pierre-Yves Juvin, Dominique Penther, Stéphanie Lagarde, Antoine Ittel, Véronique Mansat-De Mas, G Ameye, Isabelle Luquet, Marina Lafage-Pochitaloff, Carole Barin, David Rombaut, Bastien Gerby, Carine Gervais, Steven Richebourg, Oliver M. Dovey, Pierre Bories, Christine Lefebvre, Isabelle Radford-Weiss, Audrey Bidet, Isabelle Tigaud, George S. Vassiliou, Benedicte Ribourtout, Tobias Tekath, Michaela Fontenay, Lucienne Michaux, Sylvie Hébrard, Hélène Antoine-Poirel, Laura Jamrog, Vincent Fregona, Nathalie Nadal, Eric Delabesse, Véronique Baccini, Kosuke Yusa, Gerby, Bastien [0000-0002-2657-4200], Baccini, Véronique [0000-0003-3913-7664], Largeaud, Laetitia [0000-0001-5341-5427], Fregona, Vincent [0000-0003-4857-1737], Prade, Naïs [0000-0003-4718-7848], Jamrog, Laura [0000-0003-2288-0806], Mansat-De Mas, Véronique [0000-0003-1878-9129], Dovey, Oliver M [0000-0003-3586-4813], Yusa, Kosuke [0000-0002-3442-021X], Vassiliou, George S [0000-0003-4337-8022], Jansen, Joop H [0000-0001-9459-568X], Tekath, Tobias [0000-0002-9315-5452], Rombaut, David [0000-0001-8910-0945], Ameye, Geneviève [0000-0002-5838-2879], Ittel, Antoine [0000-0001-5067-575X], Michaux, Lucienne [0000-0002-8357-7942], Poirel, Hélène A [0000-0002-0712-5127], Struski, Stéphanie [0000-0002-2282-4364], Fontenay, Michaela [0000-0002-5492-6349], Broccardo, Cyril [0000-0003-3016-6549], Delabesse, Eric [0000-0002-0928-0753], Apollo - University of Cambridge Repository, Centre de Recherches en Cancérologie de Toulouse (CRCT), Université Toulouse III - Paul Sabatier (UT3), and Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
INVOLVEMENT ,Candidate gene ,Myeloid ,Tumor suppressor gene ,SCORING SYSTEM ,[SDV]Life Sciences [q-bio] ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Biology ,CLASSIFICATION ,03 medical and health sciences ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,TSLC1/IGSF4 ,030304 developmental biology ,MYELODYSPLASTIC SYNDROME ,0303 health sciences ,Science & Technology ,Myeloid Neoplasia ,Myelodysplastic syndromes ,MALE-INFERTILITY ,Chromosomes, Human, Pair 11 ,TSLC1 ,Cell Adhesion Molecule-1 ,Myeloid leukemia ,KARYOTYPE ,Hematology ,medicine.disease ,3. Good health ,Transplantation ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,KMT2A ,030220 oncology & carcinogenesis ,CELL-ADHESION MOLECULE ,Myelodysplastic Syndromes ,Cancer research ,biology.protein ,Female ,Bone marrow ,Chromosome Deletion ,Life Sciences & Biomedicine ,LEUKEMIA - Abstract
Key Points We detail at clinical, cytological, cytogenetic, and molecular levels 113 cases of MDS and MDS/MPN with del(11q), a rare recurrent event.CADM1, a tumor suppressor gene identified initially in solid tumors, ATM, CBL, and KMT2A are deleted and/or mutated in del(11q)., Visual Abstract, Myelodysplastic syndromes (MDS) represent a heterogeneous group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis leading to peripheral cytopenias and in a substantial proportion of cases to acute myeloid leukemia. The deletion of the long arm of chromosome 11, del(11q), is a rare but recurrent clonal event in MDS. Here, we detail the largest series of 113 cases of MDS and myelodysplastic syndromes/myeloproliferative neoplasms (MDS/MPN) harboring a del(11q) analyzed at clinical, cytological, cytogenetic, and molecular levels. Female predominance, a survival prognosis similar to other MDS, a low monocyte count, and dysmegakaryopoiesis were the specific clinical and cytological features of del(11q) MDS. In most cases, del(11q) was isolated, primary and interstitial encompassing the 11q22-23 region containing ATM, KMT2A, and CBL genes. The common deleted region at 11q23.2 is centered on an intergenic region between CADM1 (also known as Tumor Suppressor in Lung Cancer 1) and NXPE2. CADM1 was expressed in all myeloid cells analyzed in contrast to NXPE2. At the functional level, the deletion of Cadm1 in murine Lineage-Sca1+Kit+ cells modifies the lymphoid-to-myeloid ratio in bone marrow, although not altering their multilineage hematopoietic reconstitution potential after syngenic transplantation. Together with the frequent simultaneous deletions of KMT2A, ATM, and CBL and mutations of ASXL1, SF3B1, and CBL, we show that CADM1 may be important in the physiopathology of the del(11q) MDS, extending its role as tumor-suppressor gene from solid tumors to hematopoietic malignancies.
- Published
- 2022
- Full Text
- View/download PDF
137. Low rates of mutation in clinical grade human pluripotent stem cells under different culture conditions
- Author
-
John Alexander, Ivana Barbaric, Zoe Hewitt, Sian Gregory, Wolf Reik, Peter W. Andrews, Richard Weightman, Harry Moore, Simon Andrews, Oliver Thompson, Serena Nik-Zainal, Andrew Wood, Felix Krueger, Kosuke Yusa, Paul J. Gokhale, Ferdinand von Meyenn, Marta Milo, von Meyenn, Ferdinand [0000-0001-9920-3075], Hewitt, Zoe [0000-0001-7519-7029], Alexander, John [0000-0003-3973-6501], Wood, Andrew [0000-0002-5973-2092], Weightman, Richard [0000-0002-2593-8631], Gregory, Sian [0000-0002-3014-2603], Krueger, Felix [0000-0002-5513-3324], Gokhale, Paul J. [0000-0001-7225-4403], Milo, Marta [0000-0002-6996-6431], Nik-Zainal, Serena [0000-0001-5054-1727], Yusa, Kosuke [0000-0002-3442-021X], Andrews, Peter W. [0000-0001-7215-4410], Apollo - University of Cambridge Repository, Gokhale, Paul J [0000-0001-7225-4403], and Andrews, Peter W [0000-0001-7215-4410]
- Subjects
0301 basic medicine ,Mutation rate ,49/47 ,DNA Mutational Analysis ,631/532/2064/2117 ,Cell Culture Techniques ,General Physics and Astronomy ,medicine.disease_cause ,38/70 ,Epigenesis, Genetic ,0302 clinical medicine ,Mutation Rate ,38/23 ,Induced pluripotent stem cell ,lcsh:Science ,Mutation ,Multidisciplinary ,article ,Chromatin ,Cell biology ,DNA methylation ,DNA, Intergenic ,Pluripotent Stem Cells ,Embryonic stem cells ,Science ,631/114/2403 ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,631/208/726/649 ,03 medical and health sciences ,13/100 ,medicine ,Humans ,Genetic variation ,Epigenetics ,Chromosomes, Human, X ,Whole Genome Sequencing ,Sequence Analysis, RNA ,631/208/176/1988 ,General Chemistry ,DNA Methylation ,Embryonic stem cell ,Culture Media ,Oxygen ,Oxidative Stress ,030104 developmental biology ,Cell culture ,Gene ontology ,lcsh:Q ,030217 neurology & neurosurgery - Abstract
The occurrence of repetitive genomic changes that provide a selective growth advantage in pluripotent stem cells is of concern for their clinical application. However, the effect of different culture conditions on the underlying mutation rate is unknown. Here we show that the mutation rate in two human embryonic stem cell lines derived and banked for clinical application is low and not substantially affected by culture with Rho Kinase inhibitor, commonly used in their routine maintenance. However, the mutation rate is reduced by >50% in cells cultured under 5% oxygen, when we also found alterations in imprint methylation and reversible DNA hypomethylation. Mutations are evenly distributed across the chromosomes, except for a slight increase on the X-chromosome, and an elevation in intergenic regions suggesting that chromatin structure may affect mutation rate. Overall the results suggest that pluripotent stem cells are not subject to unusually high rates of genetic or epigenetic alterations., Nature Communications, 11 (1), ISSN:2041-1723
- Published
- 2020
138. piggyBac Transposon.
- Author
-
Yusa K
- Subjects
- Animals, DNA genetics, DNA metabolism, Models, Biological, Recombination, Genetic, DNA Transposable Elements, Insecta genetics, Mammals genetics
- Abstract
The piggyBac transposon was originally isolated from the cabbage looper moth, Trichoplusia ni, in the 1980s. Despite its early discovery and dissimilarity to the other DNA transposon families, the piggyBac transposon was not recognized as a member of a large transposon superfamily for a long time. Initially, the piggyBac transposon was thought to be a rare transposon. This view, however, has now been completely revised as a number of fully sequenced genomes have revealed the presence of piggyBac-like repetitive elements. The isolation of active copies of the piggyBac-like elements from several distinct species further supported this revision. This includes the first isolation of an active mammalian DNA transposon identified in the bat genome. To date, the piggyBac transposon has been deeply characterized and it represents a number of unique characteristics. In general, all members of the piggyBac superfamily use TTAA as their integration target sites. In addition, the piggyBac transposon shows precise excision, i.e., restoring the sequence to its preintegration state, and can transpose in a variety of organisms such as yeasts, malaria parasites, insects, mammals, and even in plants. Biochemical analysis of the chemical steps of transposition revealed that piggyBac does not require DNA synthesis during the actual transposition event. The broad host range has attracted researchers from many different fields, and the piggyBac transposon is currently the most widely used transposon system for genetic manipulations.
- Published
- 2015
- Full Text
- View/download PDF
139. Off-target assessment of CRISPR-Cas9 guiding RNAs in human iPS and mouse ES cells.
- Author
-
Tan EP, Li Y, Velasco-Herrera Mdel C, Yusa K, and Bradley A
- Subjects
- Animals, Bacterial Proteins genetics, Base Sequence, CRISPR-Associated Protein 9, Clustered Regularly Interspaced Short Palindromic Repeats, DNA Cleavage, DNA Repair, Endonucleases genetics, Gene Targeting, HEK293 Cells, Humans, Male, Mice, Embryonic Stem Cells physiology, Genetic Engineering, Induced Pluripotent Stem Cells physiology, RNA, Guide, CRISPR-Cas Systems genetics
- Abstract
The CRISPR-Cas9 system consists of a site-specific, targetable DNA nuclease that holds great potential in gene editing and genome-wide screening applications. To apply the CRISPR-Cas9 system to these assays successfully, the rate at which Cas9 induces DNA breaks at undesired loci must be understood. We characterized the rate of Cas9 off-target activity in typical Cas9 experiments in two human and one mouse cell lines. We analyzed the Cas9 cutting activity of 12 gRNAs in both their targeted sites and ∼90 predicted off-target sites per gRNA. In a Cas9-based knockout experiment, gRNAs induced detectable Cas9 cutting activity in all on-target sites and in only a few off-target sites genome-wide in human 293FT, human-induced pluripotent stem (hiPS) cells, and mouse embryonic stem (ES) cells. Both the cutting rates and DNA repair patterns were highly correlated between the two human cell lines in both on-target and off-target sites. In clonal Cas9 cutting analysis in mouse ES cells, biallelic Cas9 cutting was observed with low off-target activity. Our results show that off-target activity of Cas9 is low and predictable by the degree of sequence identity between the gRNA and a potential off-target site. Off-target Cas9 activity can be minimized by selecting gRNAs with few off-target sites of near complementarity., (© 2014 The Authors. Genesis Published by Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
140. piggyBac transposon-based insertional mutagenesis in mouse haploid embryonic stem cells.
- Author
-
Pettitt SJ, Tan EP, and Yusa K
- Subjects
- Animals, Cell Culture Techniques, Cell Separation methods, Immunohistochemistry, Mice, DNA Transposable Elements, Embryonic Stem Cells metabolism, Haploidy, Mutagenesis, Insertional
- Abstract
Forward genetic screening is a powerful non-hypothesis-driven approach to unveil the molecular mechanisms and pathways underlying phenotypes of interest. In this approach, a genome-wide mutant library is first generated and then screened for a phenotype of interest. Subsequently, genes responsible for the phenotype are identified. There have been a number of successful screens in yeasts, Caenorhabditis elegans and Drosophila. These model organisms all allow loss-of-function mutants to be generated easily on a genome-wide scale: yeasts have a haploid stage in their reproductive cycles and the latter two organisms have short generation times, allowing mutations to be systematically bred to homozygosity. However, in mammals, the diploid genome and long generation time have always hampered rapid and efficient production of homozygous mutant cells and animals. The recent discovery of several haploid mammalian cell lines promises to revolutionize recessive genetic screens in mammalian cells. In this protocol, we describe an overview of insertional mutagenesis, focusing on DNA transposons, and provide a method for an efficient generation of genome-wide mutant libraries using mouse haploid embryonic stem cells.
- Published
- 2015
- Full Text
- View/download PDF
141. The critical role of histone H2A-deubiquitinase Mysm1 in hematopoiesis and lymphocyte differentiation.
- Author
-
Nijnik A, Clare S, Hale C, Raisen C, McIntyre RE, Yusa K, Everitt AR, Mottram L, Podrini C, Lucas M, Estabel J, Goulding D, Adams N, Ramirez-Solis R, White JK, Adams DJ, Hancock RE, and Dougan G
- Subjects
- Animals, Blood Cell Count, Blotting, Western, Endopeptidases metabolism, Female, Flow Cytometry, Gene Expression Profiling, Genotype, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Histones metabolism, Lymphocytes cytology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Oligonucleotide Array Sequence Analysis, Phenotype, Reactive Oxygen Species metabolism, Reverse Transcriptase Polymerase Chain Reaction, Trans-Activators, Tumor Suppressor Protein p53 metabolism, Ubiquitin-Specific Proteases, Cell Differentiation genetics, Endopeptidases genetics, Hematopoiesis genetics, Lymphocytes metabolism
- Abstract
Stem cell differentiation and lineage specification depend on coordinated programs of gene expression, but our knowledge of the chromatin-modifying factors regulating these events remains incomplete. Ubiquitination of histone H2A (H2A-K119u) is a common chromatin modification associated with gene silencing, and controlled by the ubiquitin-ligase polycomb repressor complex 1 (PRC1) and H2A-deubiquitinating enzymes (H2A-DUBs). The roles of H2A-DUBs in mammalian development, stem cells, and hematopoiesis have not been addressed. Here we characterized an H2A-DUB targeted mouse line Mysm1(tm1a/tm1a) and demonstrated defects in BM hematopoiesis, resulting in lymphopenia, anemia, and thrombocytosis. Development of lymphocytes was impaired from the earliest stages of their differentiation, and there was also a depletion of erythroid cells and a defect in erythroid progenitor function. These phenotypes resulted from a cell-intrinsic requirement for Mysm1 in the BM. Importantly, Mysm1(tm1a/tm1a) HSCs were functionally impaired, and this was associated with elevated levels of reactive oxygen species, γH2AX DNA damage marker, and p53 protein in the hematopoietic progenitors. Overall, these data establish a role for Mysm1 in the maintenance of BM stem cell function, in the control of oxidative stress and genetic stability in hematopoietic progenitors, and in the development of lymphoid and erythroid lineages.
- Published
- 2012
- Full Text
- View/download PDF
142. Interhomolog recombination and loss of heterozygosity in wild-type and Bloom syndrome helicase (BLM)-deficient mammalian cells.
- Author
-
LaRocque JR, Stark JM, Oh J, Bojilova E, Yusa K, Horie K, Takeda J, and Jasin M
- Subjects
- Animals, Base Sequence, Blotting, Western, Cell Line, Crossing Over, Genetic genetics, Cytogenetic Analysis, DNA Primers genetics, Electroporation, Genetic Vectors genetics, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Polymorphism, Single Nucleotide genetics, Sequence Analysis, DNA, Chromosomes, Mammalian genetics, DNA Breaks, Double-Stranded, Gene Conversion genetics, Loss of Heterozygosity genetics, RecQ Helicases genetics
- Abstract
Genomic integrity often is compromised in tumor cells, as illustrated by genetic alterations leading to loss of heterozygosity (LOH). One mechanism of LOH is mitotic crossover recombination between homologous chromosomes, potentially initiated by a double-strand break (DSB). To examine LOH associated with DSB-induced interhomolog recombination, we analyzed recombination events using a reporter in mouse embryonic stem cells derived from F1 hybrid embryos. In this study, we were able to identify LOH events although they occur only rarely in wild-type cells (≤2.5%). The low frequency of LOH during interhomolog recombination suggests that crossing over is rare in wild-type cells. Candidate factors that may suppress crossovers include the RecQ helicase deficient in Bloom syndrome cells (BLM), which is part of a complex that dissolves recombination intermediates. We analyzed interhomolog recombination in BLM-deficient cells and found that, although interhomolog recombination is slightly decreased in the absence of BLM, LOH is increased by fivefold or more, implying significantly increased interhomolog crossing over. These events frequently are associated with a second homologous recombination event, which may be related to the mitotic bivalent structure and/or the cell-cycle stage at which the initiating DSB occurs.
- Published
- 2011
- Full Text
- View/download PDF
143. A hyperactive piggyBac transposase for mammalian applications.
- Author
-
Yusa K, Zhou L, Li MA, Bradley A, and Craig NL
- Subjects
- Alternative Splicing genetics, Animals, Cells, Cultured, Comparative Genomic Hybridization, Embryo, Mammalian cytology, Embryonic Stem Cells cytology, Fibroblasts cytology, Fibroblasts metabolism, Genome genetics, HEK293 Cells, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Mice, Models, Genetic, Moths genetics, Mutation, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Transposases genetics, DNA Transposable Elements genetics, Embryonic Stem Cells metabolism, Mutagenesis, Insertional, Transposases metabolism
- Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
144. PiggyBac transposon mutagenesis: a tool for cancer gene discovery in mice.
- Author
-
Rad R, Rad L, Wang W, Cadinanos J, Vassiliou G, Rice S, Campos LS, Yusa K, Banerjee R, Li MA, de la Rosa J, Strong A, Lu D, Ellis P, Conte N, Yang FT, Liu P, and Bradley A
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neoplasms genetics, Oncogenes, Promoter Regions, Genetic, DNA Transposable Elements, Genes, Neoplasm, Genetic Testing methods, Mutagenesis, Insertional
- Abstract
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.
- Published
- 2010
- Full Text
- View/download PDF
145. [Recent progress in mouse transposon research].
- Author
-
Horie K, Yusa K, and Takeda J
- Subjects
- Animals, DNA Methylation, Evolution, Molecular, Genes, Intracisternal A-Particle genetics, Genes, Intracisternal A-Particle physiology, Green Fluorescent Proteins genetics, Long Interspersed Nucleotide Elements genetics, Long Interspersed Nucleotide Elements physiology, Mice, RNA Interference, Retroelements genetics, Retroelements physiology, Transposases, DNA Transposable Elements genetics, DNA Transposable Elements physiology, Genome, Mice, Mutant Strains genetics
- Published
- 2004
146. [New perspectives on genetically engineered mice in post-genomic era].
- Author
-
Yusa K, Horie K, and Takeda J
- Subjects
- Animals, DNA Transposable Elements, Embryo, Mammalian cytology, Ethylnitrosourea, Mutagens, RNA Interference, Animals, Genetically Modified, Genomics methods, Mice genetics
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.