Publications by ISTT members
This list contains selected publications by ISTT members as of 2017. For a general overview of transgenic technology publications, see the extensive collections of our members Thom Saunders and Lluís Montoliu.
To import this collection into your preferred reference management software, please contact the webmaster.
2024
Elrick, H., Peterson, K. A., Willis, B. J., Lanza, D. G., Acar, E. F., Ryder, E. J., Teboul, L., Kasparek, P., Birling, M. C., Adams, D. J., Bradley, A., Braun, R. E., Brown, S. D., Caulder, A., Codner, G. F., Demayo, F. J., Dickinson, M. E., Doe, B., Duddy, G., Gertsenstein, M., Goodwin, L. O., Hérault, Y., Lintott, L. G., Lloyd, K. C. K., Lorenzo, I., Mackenzie, M., Mallon, A. M., Mckerlie, C., Parkinson, H., Ramirez-Solis, R., Seavitt, J. R., Sedlacek, R., Skarnes, W. C., Smedley, D., Wells, S., White, J. K., Wood, J. A., Murray, S. A., Heaney, J. D. & Nutter, L. M. J. 2024. Impact of essential genes on the success of genome editing experiments generating 3313 new genetically engineered mouse lines. Sci Rep, 14, 22626, https://doi.org/10.1038/s41598-024-72418-8
Mccabe, C. V., Price, P. D., Codner, G. F., Allan, A. J., Caulder, A., Christou, S., Loeffler, J., Mackenzie, M., Malzer, E., Mianné, J., Nowicki, K. J., O'neill, E. J., Pike, F. J., Hutchison, M., Petit-Demoulière, B., Stewart, M. E., Gates, H., Wells, S., Sanderson, N. D. & Teboul, L. 2024. Long-read sequencing for fast and robust identification of correct genome-edited alleles: PCR-based and Cas9 capture methods. PLoS Genet, 20, e1011187, https://doi.org/10.1371/journal.pgen.1011187
Yamaga, K., Nakao, S., Mikoda, N., Sztein, J. M., Nakagata, N. & Takeo, T. 2024. High-concentration bovine serum albumin enhances fertilization ability of cold-stored rat sperm. J Reprod Dev, https://doi.org/10.1262/jrd.2023-085
Muhammad Waqas, L., Piroon, J., Jessica, S., Nikhil, S., Mitchell, C., Pattaraporn, N., Lars, M. I., Thidathip, W. & Fabien, D. 2024. Long Read Sequencing reveals transgene concatemerization and backbone integration following AAV-driven electroporation of CRISPR RNP complexes in mouse zygotes. bioRxiv, 2024.02.18.580906, http://www.doi.org/10.1101/2024.02.18.580906
Chen, J. Y., Zhang, L., Yang, M., Hughes, E. D., Freeman, Z. T., Saunders, T. L. & Lin, F. 2024. Development of a C3 Humanized Rat as a New Model for Evaluating Novel C3 Inhibitors. J Innate Immun, 16, 56-65, https://www.doi.org/10.1159/000534963
Kim, J. K., Villa-Diaz, L. G., Saunders, T. L., Saul, R. P., Timilsina, S., Liu, F., Mishina, Y. & Krebsbach, P. H. 2024. Selective Inhibition of mTORC1 Signaling Supports the Development and Maintenance of Pluripotency. Stem Cells, 42, 13-28, https://www.doi.org/10.1093/stmcls/sxad079
Eghbalsaied, S., Lawler, C., Petersen, B., Hajiyev, R. A., Bischoff, S. R. & Frankenberg, S. 2024. CRISPR/Cas9-mediated base editors and their prospects for mitochondrial genome engineering. Gene Ther, https://doi.org/10.1038/s41434-023-00434-w
Kim-Yip, R. P., Mcnulty, R., Joyce, B., Mollica, A., Chen, P. J., Ravisankar, P., Law, B. K., Liu, D. R., Toettcher, J. E., Ivakine, E. A., Posfai, E. & Adamson, B. 2024. Efficient prime editing in two-cell mouse embryos using PEmbryo. Nat Biotechnol, https://www.doi.org/10.1038/s41587-023-02106-x.com
2023
Bossert, J. M., Mejias-Aponte, C. A., Saunders, T., Altidor, L., Emery, M., Fredriksson, I., Batista, A., Claypool, S. M., Caldwell, K. E., Reiner, D. J., Chow, J. J., Foltz, M., Kumar, V., Seasholtz, A., Hughes, E., Filipiak, W., Harvey, B. K., Richie, C. T., Vautier, F., Gomez, J. L., Michaelides, M., Kieffer, B. L., Watson, S. J., Akil, H. & Shaham, Y. 2023. Effect of Selective Lesions of Nucleus Accumbens µ-Opioid Receptor-Expressing Cells on Heroin Self-Administration in Male and Female Rats: A Study with Novel Oprm1-Cre Knock-in Rats. J Neurosci, 43, 1692-1713, https://www.doi.org/10.1523/jneurosci.2049-22.2023
Laubreton, D., Djebali, S., Angleraux, C., Chain, B., Dubois, M., Henry, F., Leverrier, Y., Teixeira, M., Markossian, S. & Marvel, J. 2023. Generation of a C57BL/6J mouse strain expressing the CD45.1 epitope to improve hematopoietic stem cell engraftment and adoptive cell transfer experiments. Lab Anim (NY), 52, 324-331, https://www.doi.org/10.1038/s41684-023-01275-1
Lee, J., Wang, J., Ally, R., Trzaska, S., Hickey, J., Mujica, A., Miloscio, L., Mastaitis, J., Morse, B., Smith, J., Atanasio, A., Chiao, E., Chen, H., Latuszek, A., Hu, Y., Valenzuela, D., Romano, C., Zambrowicz, B. & Auerbach, W. 2023. Production of large, defined genome modifications in rats by targeting rat embryonic stem cells. Stem Cell Reports, 18, 394-409, https://www.doi.org/10.1016/j.stemcr.2022.11.012
Matsushita, Y., Liu, J., Chu, A. K. Y., Tsutsumi-Arai, C., Nagata, M., Arai, Y., Ono, W., Yamamoto, K., Saunders, T. L., Welch, J. D. & Ono, N. 2023. Bone marrow endosteal stem cells dictate active osteogenesis and aggressive tumorigenesis. Nat Commun, 14, 2383, https://www.doi.org/10.1038/s41467-023-38034-2
Strepay, D., Olszewski, R. T., Nixon, S., Korrapati, S., Adadey, S., Griffith, A. J., Su, Y., Liu, J., Vishwasrao, H., Gu, S., Saunders, T., Roux, I. & Hoa, M. 2023. Transgenic Tg(Kcnj10-ZsGreen) Fluorescent Reporter Mice Allow Visualization of Intermediate Cells in the Stria Vascularis. Res Sq, https://www.doi.org/10.21203/rs.3.rs-3393161/v1
Vohnoutka, R. B., Kuppa, A., Hegde, Y., Chen, Y., Pant, A., Tohme, M. E., Choi, E. K., Mccarty, S. M., Bagchi, D. P., Du, X., Chen, Y., Chen, V. L., Mori, H., Bielak, L. F., Maguire, L. H., Handelman, S. K., Sexton, J. Z., Saunders, T. L., Halligan, B. D. & Speliotes, E. K. 2023. Knockout of murine Lyplal1 confers sex-specific protection against diet-induced obesity. J Mol Endocrinol, 70, https://www.doi.org/10.1530/jme-22-0131
Zhao, Y., Zhao, G., Chang, Z., Zhu, T., Zhao, Y., Lu, H., Xue, C., Saunders, T. L., Guo, Y., Chang, L., Chen, Y. E. & Zhang, J. 2023. Generating endogenous Myh11-driven Cre mice for sex-independent gene deletion in smooth muscle cells. JCI Insight, 8, https://www.doi.org/10.1172/jci.insight.171661
Eghbalsaied, S. & Kues, W. A. 2023. CRISPR/Cas9-mediated targeted knock-in of large constructs using nocodazole and RNase HII. Sci Rep, 13, 2690, https://doi.org/10.1038/s41598-023-29789-1
Lee, J., Wang, J., Ally, R., Trzaska, S., Hickey, J., Mujica, A., Miloscio, L., Mastaitis, J., Morse, B., Smith, J., Atanasio, A., Chiao, E., Chen, H., Latuszek, A., Hu, Y., Valenzuela, D., Romano, C., Zambrowicz, B. & Auerbach, W. 2023. Production of large, defined genome modifications in rats by targeting rat embryonic stem cells. Stem Cell Reports, 18, 394-409, https://doi.org/10.1016/j.stemcr.2022.11.012
Papaioannou, V. E. & Behringer, R. 2023. Mouse phenotypes : generation and analysis of mutants. Second edition. ed. Cold Spring Harbor, New York: Cold Spring Harbor Laboratory Press, https://cshlpress.com
Boura-Halfon, S., Haffner-Krausz, R., Ben-Dor, S., Kim, J. S. & Jung, S. 2024. Tackling Tissue Macrophage Heterogeneity by SplitCre Transgenesis. Methods Mol Biol, 2713, 481-503, https://doi.org/10.1007/978-1-0716-3437-0_32
Davis, D. J., Mcnew, J. F., Maresca-Fichter, H., Chen, K., Telugu, B. P. & Bryda, E. C. 2023. Efficient DNA knock-in using AAV-mediated delivery with 2-cell embryo CRISPR-Cas9 electroporation. Frontiers in Genome Editing, 5, https://www.doi.org/10.3389/fgeed.2023.1256451
Wigger, M., Schneider, M., Feldmann, A., Assenmacher, S., Zevnik, B. & Tröder, S. E. 2023. Successful use of HTF as a basal fertilization medium during SEcuRe mouse in vitro fertilization. BMC Res Notes, 16, 184, https://doi.org/10.1186/s13104-023-06452-6
Trotman, J. B., Braceros, A. K., Bischoff, S. R., Murvin, M. M., Boyson, S. P., Cherney, R. E., Eberhard, Q. E., Abrash, E. W., Cowley, D. O. & Calabrese, J. M. 2023. Ectopically expressed Airn lncRNA deposits Polycomb with a potency that rivals Xist. bioRxiv, 2023.05.09.539960, https://www.doi.org/10.1101/2023.05.09.539960
Peterson, K. A., Khalouei, S., Hanafi, N., Wood, J. A., Lanza, D. G., Lintott, L. G., Willis, B. J., Seavitt, J. R., Braun, R. E., Dickinson, M. E., White, J. K., Lloyd, K. C. K., Heaney, J. D., Murray, S. A., Ramani, A. & Nutter, L. M. J. 2023. Whole genome analysis for 163 gRNAs in Cas9-edited mice reveals minimal off-target activity. Commun Biol, 6, 626, https://www.doi.org/10.1038/s42003-023-04974-0
Lintott, L. G. & Nutter, L. M. J. 2023. Genetic and Molecular Quality Control of Genetically Engineered Mice. Methods Mol Biol, 2631, 53-101, https://www.doi.org/10.1007/978-1-0716-2990-1_3
Mcbeath, E., Fujiwara, K. & Hofmann, M. C. 2023. Evidence-Based Guide to Using Artificial Introns for Tissue-Specific Knockout in Mice. Int J Mol Sci, 24, https://www.doi.org/10.3390/ijms241210258
Nakao, S., Ito, K., Sugahara, C., Watanabe, H., Kondoh, G., Nakagata, N. & Takeo, T. 2023. Synchronization of the ovulation and copulation timings increased the number of in vivo fertilized oocytes in superovulated female mice. PLoS One, 18, e0281330, https://www.doi.org/10.1371/journal.pone.0281330
Zhong, L., Gordillo, M., Wang, X., Qin, Y., Huang, Y., Soshnev, A., Kumar, R., Nanjangud, G., James, D., David Allis, C., Evans, T., Carey, B. & Wen, D. 2023. Dual role of lipids for genome stability and pluripotency facilitates full potency of mouse embryonic stem cells. Protein Cell, https://www.doi.org/10.1093/procel/pwad008
Nakao, S., Ito, K., Sakoh, K., Takemoto, K., Watanabe, H., Kondoh, G., Irie, T., Nakagata, N. & Takeo, T. 2023. Dimethyl-α-cyclodextrin induces capacitation by removing phospholipids from the plasma membrane of mouse sperm†. Biol Reprod, 108, 671-681, https://www.doi.org/10.1093/biolre/ioad013
Nakagata, N., Nakao, S., Mikoda, N., Yamaga, K. & Takeo, T. 2023. Observation of the in vitro fertilization process in living oocytes using frozen-thawed sperm in rats. Theriogenology, 199, 69-76, https://www.doi.org/10.1016/j.theriogenology.2023.01.016
Montoro-Gámez, C., Nolte, H., Molinié, T., Evangelista, G., Tröder, S., Barth, E., Popovic, M., Trifunovic, A., Zevnik, B., Langer, T. & Rugarli, E. I. 2023. SARM1 deletion delays cerebellar but not spinal cord degeneration in an enhanced mouse model of SPG7 deficiency. Brain, https://www.doi.org/10.1093/brain/awad136
Mathew, S. M. 2023. Strategies for generation of mice via CRISPR/HDR-mediated knock-in. Mol Biol Rep, 50, 3189-3204, https://www.doi.org/10.1007/s11033-023-08278-8
Maino, E., Scott, O., Rizvi, S. Z., Visuvanathan, S., Zablah, Y. B., Li, H., Sengar, A. S., Salter, M. W., Jia, Z., Rossant, J., Cohn, R. D., Gu, B. & Ivakine, E. A. 2023. A Cas9-fusion proximity-based approach generates an Irak1-Mecp2 tandem duplication mouse model for the study of MeCP2 duplication syndrome. bioRxiv, 2023.02.07.527511, https://www.doi.org/10.1101/2023.02.07.527511
Lee, J., Wang, J., Ally, R., Trzaska, S., Hickey, J., Mujica, A., Miloscio, L., Mastaitis, J., Morse, B., Smith, J., Atanasio, A., Chiao, E., Chen, H., Latuszek, A., Hu, Y., Valenzuela, D., Romano, C., Zambrowicz, B. & Auerbach, W. 2023. Production of large, defined genome modifications in rats by targeting rat embryonic stem cells. Stem Cell Reports, 18, 394-409, https://www.doi.org/10.1016/j.stemcr.2022.11.012
Erbs, V., Lorentz, R., Eisenman, B., Schaeffer, L., Luppi, L., Lindner, L., Hérault, Y., Pavlovic, G., Wattenhofer-Donzé, M. & Birling, M. C. 2023. Increased On-Target Rate and Risk of Concatemerization after CRISPR-Enhanced Targeting in ES Cells. Genes (Basel), 14, https://www.doi.org/10.3390/genes14020401
Chenouard, V., Leray, I., Tesson, L., Remy, S., Allan, A., Archer, D., Caulder, A., Fortun, A., Bernardeau, K., Cherifi, Y., Teboul, L., David, L. & Anegon, I. 2023. Excess of guide RNA reduces knockin efficiency and drastically increases on-target large deletions. iScience, 26, 106399, https://www.doi.org/10.1016/j.isci.2023.106399
Buch, T., Jerchow, B. & Zevnik, B. 2023. Practical Application of the 3Rs in Rodent Transgenesis. Methods Mol Biol, 2631, 33-51, https://www.doi.org/10.1007/978-1-0716-2990-1_2
Bryant, W. B., Yang, A., Griffin, S. H., Zhang, W., Rafiq, A. M., Han, W., Deak, F., Mills, M. K., Long, X. & Miano, J. M. 2023. CRISPR-Cas9 Long-Read Sequencing for Mapping Transgenes in the Mouse Genome. Crispr j, 6, 163-175, https://www.doi.org/10.1089/crispr.2022.0099
2022
Chen, D., Hughes, E. D., Saunders, T. L., Wu, J., Vasquez, M. N. H., Makinen, T. & King, P. D. 2022. Angiogenesis depends upon EPHB4-mediated export of collagen IV from vascular endothelial cells. JCI Insight, 7, https://www.doi.org/10.1172/jci.insight.156928
Liang, T., Wang, S. K., Smith, C., Zhang, H., Hu, Y., Seymen, F., Koruyucu, M., Kasimoglu, Y., Kim, J. W., Zhang, C., Saunders, T. L., Simmer, J. P. & Hu, J. C. 2022. Enamel defects in Acp4(R110C/R110C) mice and human ACP4 mutations. Sci Rep, 12, 16477, https://www.doi.org/10.1038/s41598-022-20684-9
Marinelli, I., Parekh, V., Fletcher, P., Thompson, B., Ren, J., Tang, X., Saunders, T. L., Ha, J., Sherman, A., Bertram, R. & Satin, L. S. 2022. Slow oscillations persist in pancreatic beta cells lacking phosphofructokinase M. Biophys J, 121, 692-704, https://www.doi.org/10.1016/j.bpj.2022.01.027
Tsou, P. S., Lu, C., Gurrea-Rubio, M., Muraoka, S., Campbell, P. L., Wu, Q., Model, E. N., Lind, M. E., Vichaikul, S., Mattichak, M. N., Brodie, W. D., Hervoso, J. L., Ory, S., Amarista, C. I., Pervez, R., Junginger, L., Ali, M., Hodish, G., O'mara, M. M., Ruth, J. H., Robida, A. M., Alt, A. J., Zhang, C., Urquhart, A. G., Lawton, J. N., Chung, K. C., Maerz, T., Saunders, T. L., Groppi, V. E., Fox, D. A. & Amin, M. A. 2022. Soluble CD13 induces inflammatory arthritis by activating the bradykinin receptor B1. J Clin Invest, 132, https://www.doi.org/10.1172/jci151827
Verhaegen, M. E., Harms, P. W., Van Goor, J. J., Arche, J., Patrick, M. T., Wilbert, D., Zabawa, H., Grachtchouk, M., Liu, C. J., Hu, K., Kelly, M. C., Chen, P., Saunders, T. L., Weidinger, S., Syu, L. J., Runge, J. S., Gudjonsson, J. E., Wong, S. Y., Brownell, I., Cieslik, M., Udager, A. M., Chinnaiyan, A. M., Tsoi, L. C. & Dlugosz, A. A. 2022. Direct cellular reprogramming enables development of viral T antigen-driven Merkel cell carcinoma in mice. J Clin Invest, 132, https://www.doi.org/10.1172/jci152069
Zvick, J., Tarnowska-Sengül, M., Ghosh, A., Bundschuh, N., Gjonlleshaj, P., Hinte, L. C., Trautmann, C. L., Noé, F., Qabrati, X., Domenig, S. A., Kim, I., Hennek, T., Von Meyenn, F. & Bar-Nur, O. 2022. Exclusive generation of rat spermatozoa in sterile mice utilizing blastocyst complementation with pluripotent stem cells. Stem Cell Reports, 17, 1942-1958, https://www.doi.org/10.1016/j.stemcr.2022.07.005
Zevnik, B., Jerchow, B. & Buch, T. 2022. 3R measures in facilities for the production of genetically modified rodents. Lab Animal, 51, 162-177, https://www.doi.org/10.1038/s41684-022-00978-1
Tanaka, M., Yokoyama, K., Hayashi, H., Isaki, S., Kitatani, K., Wang, T., Kawata, H., Matsuzawa, H., Gurumurthy, C. B., Miura, H. & Ohtsuka, M. 2022. CRISPR-KRISPR: a method to identify on-target and random insertion of donor DNAs and their characterization in knock-in mice. Genome Biol, 23, 228, https://www.doi.org/10.1186/s13059-022-02779-8
Takeo, T., Nakao, S., Mikoda, N., Yamaga, K., Maeda, R., Tsuchiyama, S., Nakatsukasa, E. & Nakagata, N. 2022. Optimized protocols for sperm cryopreservation and in vitro fertilization in the rat. Lab Anim (NY), 51, 256-274, https://www.doi.org/10.1038/s41684-022-01053-5
Qin, Y., Geng, F. & Wen, D. 2022. Generation of sex-reversed female clonal mice via CRISPR/Cas9-mediated Y chromosome deletion in male embryonic stem cells. Methods Cell Biol, 170, 203-210, https://www.doi.org/10.1016/bs.mcb.2022.02.015
Montoliu, L. 2022. Historical DNA Manipulation Overview. Methods Mol Biol, 2495, 3-28, https://www.doi.org/10.1007/978-1-0716-2301-5_1
Low, B. E., Hosur, V., Lesbirel, S. & Wiles, M. V. 2022. Efficient targeted transgenesis of large donor DNA into multiple mouse genetic backgrounds using bacteriophage Bxb1 integrase. Sci Rep, 12, 5424, https://www.doi.org/10.1038/s41598-022-09445-w
Jiao, H., Wachsmuth, L., Wolf, S., Lohmann, J., Nagata, M., Kaya, G. G., Oikonomou, N., Kondylis, V., Rogg, M., Diebold, M., Tröder, S. E., Zevnik, B., Prinz, M., Schell, C., Young, G. R., Kassiotis, G. & Pasparakis, M. 2022. ADAR1 averts fatal type I interferon induction by ZBP1. Nature, https://www.doi.org/10.1038/s41586-022-04878-9
Desjardins, J., Cowan, M. & Yamanaka, Y. 2022. Designing Genetically Engineered Mouse Models (GEMMs) Using CRISPR Mediated Genome Editing. Methods Mol Biol, 2429, 515-531, https://www.doi.org/10.1007/978-1-0716-1979-7_36
Delerue, F. & Ittner, L. M. 2022. Microinjection of Zygotes for CRISPR/Cas9-Mediated Insertion of Transgenes into the Murine Rosa26 Safe Harbor. Methods Mol Biol, 2495, 115-128, https://www.doi.org/10.1007/978-1-0716-2301-5_7
Chin, H. J., Dobbie, M. S., Gao, X., Hennessy, J. E., Nam, K. H., Seong, J. K., Shiroishi, T., Takeo, T., Yoshiki, A., Zao, J. & Wang, C. L. 2022. Asian Mouse Mutagenesis Resource Association (AMMRA): mouse genetics and laboratory animal resources in the Asia Pacific. Mamm Genome, 33, 192-202, https://www.doi.org/10.1007/s00335-021-09912-1
2021
Gertsenstein, M. & Nutter, L. M. J. 2021. Production of knockout mouse lines with Cas9. Methods, 191, 32-43, https://doi.org/10.1016/j.ymeth.2021.01.005
Birling, M. C., Yoshiki, A., Adams, D. J., Ayabe, S., Beaudet, A. L., Bottomley, J., Bradley, A., Brown, S. D. M., Bürger, A., Bushell, W., Chiani, F., Chin, H. G., Christou, S., Codner, G. F., Demayo, F. J., Dickinson, M. E., Doe, B., Donahue, L. R., Fray, M. D., Gambadoro, A., Gao, X., Gertsenstein, M., Gomez-Segura, A., Goodwin, L. O., Heaney, J. D., Hérault, Y., De Angelis, M. H., Jiang, S. T., Justice, M. J., Kasparek, P., King, R. E., Kühn, R., Lee, H., Lee, Y. J., Liu, Z., Lloyd, K. C. K., Lorenzo, I., Mallon, A. M., Mckerlie, C., Meehan, T. F., Fuentes, V. M., Newman, S., Nutter, L. M. J., Oh, G. T., Pavlovic, G., Ramirez-Solis, R., Rosen, B., Ryder, E. J., Santos, L. A., Schick, J., Seavitt, J. R., Sedlacek, R., Seisenberger, C., Seong, J. K., Skarnes, W. C., Sorg, T., Steel, K. P., Tamura, M., Tocchini-Valentini, G. P., Wang, C. L., Wardle-Jones, H., Wattenhofer-Donzé, M., Wells, S., Wiles, M. V., Willis, B. J., Wood, J. A., Wurst, W., Xu, Y., Teboul, L. & Murray, S. A. 2021. A resource of targeted mutant mouse lines for 5,061 genes. Nat Genet, 53, 416-419, https://www.doi.org/10.1038/s41588-021-00825-y
Yamaga, K., Nakao, S., Mikoda, N., Yoshimoto, H., Nakatsukasa, E., Nakagata, N. & Takeo, T. 2021. Quercetin-treated rat sperm enables refrigerated transport with motility and fertility for five days. Sci Rep, 11, 22641, https://www.doi.org/10.1038/s41598-021-02166-6
Xu, C. C. Y., Ramsay, C., Cowan, M., Dehghani, M., Lasko, P. & Barrett, R. D. H. 2021. Transgenes of genetically modified animals detected non-invasively via environmental DNA. PLoS One, 16, e0249439, https://www.doi.org/10.1371/journal.pone.0249439
Wigger, M., Tröder, S. E. & Zevnik, B. 2021. A simple and economic protocol for efficient in vitro fertilization using cryopreserved mouse sperm. PLoS One, 16, e0259202, https://www.doi.org/10.1371/journal.pone.0259202
Tröder, S. E. & Zevnik, B. 2021. History of genome editing: From meganucleases to CRISPR. Lab Anim, 23677221994613, https://www.doi.org/10.1177/0023677221994613
Miura, H., Imafuku, J., Kurosaki, A., Sato, M., Ma, Y., Zhang, G., Mizutani, A., Kamimura, K., Gurumurthy, C. B., Liu, D. & Ohtsuka, M. 2021. Novel reporter mouse models useful for evaluating in vivo gene editing and for optimization of methods of delivering genome editing tools. Mol Ther Nucleic Acids, 24, 325-336, https://www.doi.org/10.1016/j.omtn.2021.03.003
Gurumurthy, C. B., Saunders, T. L. & Ohtsuka, M. 2021. Designing and generating a mouse model: frequently asked questions. J Biomed Res, 35, 76-90, https://www.doi.org/10.7555/JBR.35.20200197
Goto, M., Takeo, T., Takahashi, R. & Nakagata, N. 2021. Efficient production of immunodeficient non-obese diabetic/Shi-scid IL2rγ(null) mice via the superovulation technique using inhibin antiserum and gonadotropin. Lab Anim, 55, 13-20, https://www.doi.org/10.1177/0023677220928091
Bunton-Stasyshyn, R. K., Codner, G. F. & Teboul, L. 2021. Screening and validation of genome-edited animals. Lab Anim, 236772211016922, https://www.doi.org/10.1177/00236772211016922
2020
Trotman, J. B., Lee, D. M., Cherney, R. E., Kim, S. O., Inoue, K., Schertzer, M. D., Bischoff, S. R., Cowley, D. O. & Calabrese, J. M. 2020. Elements at the 5' end of Xist harbor SPEN-independent transcriptional antiterminator activity. Nucleic Acids Res, 48, 10500-10517, https://www.doi.org/10.1093/nar/gkaa789
Mccann, J. V., Bischoff, S. R., Zhang, Y., Cowley, D. O., Sanchez-Gonzalez, V., Daaboul, G. D. & Dudley, A. C. 2020. Reporter mice for isolating and auditing cell type-specific extracellular vesicles in vivo. Genesis, 58, e23369, https://www.doi.org/10.1002/dvg.23369
Yoshimoto, H., Takeo, T. & Nakagata, N. 2020. Simple Transportation of Genetically Engineered Mice via Cold Storage Techniques. Methods Mol Biol, 2066, 211-216, https://www.doi.org/10.1007/978-1-4939-9837-1_17
Takeo, T., Nakao, S., Nakagawa, Y., Sztein, J. M. & Nakagata, N. 2020. Cryopreservation of mouse resources. Lab Anim Res, 36, 33, https://www.doi.org/10.1186/s42826-020-00066-w
Takeo, T. & Nakagata, N. 2020. Cryobanking and Recovery of Genetically Modified Mice. Methods Mol Biol, 2066, 195-209, https://www.doi.org/10.1007/978-1-4939-9837-1_16
Pradas-Juni, M., Hansmeier, N. R., Link, J. C., Schmidt, E., Larsen, B. D., Klemm, P., Meola, N., Topel, H., Loureiro, R., Dhaouadi, I., Kiefer, C. A., Schwarzer, R., Khani, S., Oliverio, M., Awazawa, M., Frommolt, P., Heeren, J., Scheja, L., Heine, M., Dieterich, C., Buning, H., Yang, L., Cao, H., Jesus, D. F., Kulkarni, R. N., Zevnik, B., Troder, S. E., Knippschild, U., Edwards, P. A., Lee, R. G., Yamamoto, M., Ulitsky, I., Fernandez-Rebollo, E., Vallim, T. Q. A. & Kornfeld, J. W. 2020. A MAFG-lncRNA axis links systemic nutrient abundance to hepatic glucose metabolism. Nat Commun, 11, 644, https://www.doi.org/10.1038/s41467-020-14323-y
Nakao, S., Takeo, T., Watanabe, H., Kondoh, G. & Nakagata, N. 2020. Successful selection of mouse sperm with high viability and fertility using microfluidics chip cell sorter. Sci Rep, 10, 8862, https://www.doi.org/10.1038/s41598-020-65931-z
Nakagata, N., Mikoda, N., Nakao, S., Nakatsukasa, E. & Takeo, T. 2020. Establishment of sperm cryopreservation and in vitro fertilisation protocols for rats. Sci Rep, 10, 93, https://www.doi.org/10.1038/s41598-019-57090-7
Muñoz-Santos, D., Montoliu, L. & Fernández, A. 2020. Generation of Genetically Modified Mice Using CRISPR/Cas9. In: COSTA, C. (ed.) Xenotransplantation: Methods and Protocols. New York, NY: Springer US, https://www.doi.org/10.1007/978-1-0716-0255-3_9
Mukunoki, A., Takeo, T., Nakao, S., Tamura, K., Horikoshi, Y. & Nakagata, N. 2020. Simple transport and cryopreservation of cold-stored mouse embryos. Exp Anim, 69, 423-429, https://www.doi.org/10.1538/expanim.20-0042
Mcbeath, E., Parker-Thornburg, J., Fujii, Y., Aryal, N., Smith, C., Hofmann, M. C., Abe, J. I. & Fujiwara, K. 2020. Rapid Evaluation of CRISPR Guides and Donors for Engineering Mice. Genes (Basel), 11, https://www.doi.org/10.3390/genes11060628
Larson, M. A., Gibson, K. A. & Vivian, J. L. 2020. In Vivo Validation of CRISPR Reagents in Preimplantation Mouse Embryos. Methods Mol Biol, 2066, 47-57, https://www.doi.org/10.1007/978-1-4939-9837-1_4
Gurumurthy, C. B., Quadros, R. M., Richardson, G. P., Poluektova, L. Y., Mansour, S. L. & Ohtsuka, M. 2020. Genetically modified mouse models to help fight COVID-19. Nat Protoc, 15, 3777-3787, https://www.doi.org/10.1038/s41596-020-00403-2
Gu, B., Posfai, E., Gertsenstein, M. & Rossant, J. 2020. Efficient Generation of Large-Fragment Knock-In Mouse Models Using 2-Cell (2C)-Homologous Recombination (HR)-CRISPR. Curr Protoc Mouse Biol, 10, e67, https://www.doi.org/10.1002/cpmo.67
Gu, B., Gertsenstein, M. & Posfai, E. 2020. Generation of Large Fragment Knock-In Mouse Models by Microinjecting into 2-Cell Stage Embryos. Methods Mol Biol, 2066, 89-100, https://www.doi.org/10.1007/978-1-4939-9837-1_7
Fernandez, A., Morin, M., Munoz-Santos, D., Josa, S., Montero, A., Rubio-Fernandez, M., Cantero, M., Fernandez, J., Del Hierro, M. J., Castrillo, M., Moreno-Pelayo, M. A. & Montoliu, L. 2020. Simple Protocol for Generating and Genotyping Genome-Edited Mice With CRISPR-Cas9 Reagents. Curr Protoc Mouse Biol, 10, e69, https://www.doi.org/10.1002/cpmo.69
Burgio, G. & Teboul, L. 2020. Anticipating and Identifying Collateral Damage in Genome Editing. Trends Genet, 36, 905-914, https://www.doi.org/10.1016/j.tig.2020.09.011
2019
Schertzer, M. D., Braceros, K. C. A., Starmer, J., Cherney, R. E., Lee, D. M., Salazar, G., Justice, M., Bischoff, S. R., Cowley, D. O., Ariel, P., Zylka, M. J., Dowen, J. M., Magnuson, T. & Calabrese, J. M. 2019. lncRNA-Induced Spread of Polycomb Controlled by Genome Architecture, RNA Abundance, and CpG Island DNA. Mol Cell, 75, 523-537.e10, https://www.doi.org/10.1016/j.molcel.2019.05.028
Takeo, T., Sztein, J. & Nakagata, N. 2019. The CARD Method for Mouse Sperm Cryopreservation and In Vitro Fertilization Using Frozen-Thawed Sperm. Methods Mol Biol, 1874, 243-256, https://www.doi.org/10.1007/978-1-4939-8831-0_14
Takeo, T., Mukunoki, A. & Nakagata, N. 2019. Ovulation of juvenile, mature, and aged female C57BL/6 mice following coadministration of inhibin antiserum and equine chorionic gonadotropin. Theriogenology, 135, 1-6, https://www.doi.org/10.1016/j.theriogenology.2019.05.028
Pineault, K. M., Novoa, A., Lozovska, A., Wellik, D. M. & Mallo, M. 2019. Two CRISPR/Cas9-mediated methods for targeting complex insertions, deletions, or replacements in mouse. MethodsX, 6, 2088-2100, https://www.doi.org/10.1016/j.mex.2019.09.003
Nakagata, N. & Takeo, T. 2019. Basic mouse reproductive techniques developed and modified at the Center for Animal Resources and Development (CARD), Kumamoto University. Exp Anim, 68, 391-395, https://www.doi.org/10.1538/expanim.19-0070
Nakagata, N., Sztein, J. & Takeo, T. 2019. The CARD Method for Simple Vitrification of Mouse Oocytes: Advantages and Applications. Methods Mol Biol, 1874, 229-242, https://www.doi.org/10.1007/978-1-4939-8831-0_13
Mukunoki, A., Takeo, T. & Nakagata, N. 2019. N-acetyl cysteine restores the fertility of vitrified-warmed mouse oocytes derived through ultrasuperovulation. PLoS One, 14, e0224087, https://www.doi.org/10.1371/journal.pone.0224087
Gurumurthy, C. B., Sato, M., Nakamura, A., Inui, M., Kawano, N., Islam, M. A., Ogiwara, S., Takabayashi, S., Matsuyama, M., Nakagawa, S., Miura, H. & Ohtsuka, M. 2019. Creation of CRISPR-based germline-genome-engineered mice without ex vivo handling of zygotes by i-GONAD. Nat Protoc, https://www.doi.org/10.1038/s41596-019-0187-x
Gurumurthy, C. B., O'brien, A. R., Quadros, R. M., Adams, J., Jr., Alcaide, P., Ayabe, S., Ballard, J., Batra, S. K., Beauchamp, M. C., Becker, K. A., Bernas, G., Brough, D., Carrillo-Salinas, F., Chan, W., Chen, H., Dawson, R., Demambro, V., D'hont, J., Dibb, K. M., Eudy, J. D., Gan, L., Gao, J., Gonzales, A., Guntur, A. R., Guo, H., Harms, D. W., Harrington, A., Hentges, K. E., Humphreys, N., Imai, S., Ishii, H., Iwama, M., Jonasch, E., Karolak, M., Keavney, B., Khin, N. C., Konno, M., Kotani, Y., Kunihiro, Y., Lakshmanan, I., Larochelle, C., Lawrence, C. B., Li, L., Lindner, V., Liu, X. D., Lopez-Castejon, G., Loudon, A., Lowe, J., Jerome-Majewska, L. A., Matsusaka, T., Miura, H., Miyasaka, Y., Morpurgo, B., Motyl, K., Nabeshima, Y. I., Nakade, K., Nakashiba, T., Nakashima, K., Obata, Y., Ogiwara, S., Ouellet, M., Oxburgh, L., Piltz, S., Pinz, I., Ponnusamy, M. P., Ray, D., Redder, R. J., Rosen, C. J., Ross, N., Ruhe, M. T., Ryzhova, L., Salvador, A. M., Alam, S. S., Sedlacek, R., Sharma, K., Smith, C., Staes, K., Starrs, L., Sugiyama, F., Takahashi, S., Tanaka, T., Trafford, A. W., Uno, Y., Vanhoutte, L., Vanrockeghem, F., Willis, B. J., Wright, C. S., Yamauchi, Y., Yi, X., Yoshimi, K., Zhang, X., Zhang, Y., Ohtsuka, M., Das, S., Garry, D. J., Hochepied, T., Thomas, P., Parker-Thornburg, J., Adamson, A. D., Yoshiki, A., et al. 2019. Reproducibility of CRISPR-Cas9 methods for generation of conditional mouse alleles: a multi-center evaluation. Genome Biol, 20, 171, https://www.doi.org/10.1186/s13059-019-1776-2
2018
Nutter, L. M. J., Heaney, J. D., Lloyd, K. C. K., Murray, S. A., Seavitt, J. R., Skarnes, W. C., Teboul, L., Brown, S. D. M. & Moore, M. 2018. Response to "Unexpected mutations after CRISPR-Cas9 editing in vivo". Nat Methods, 15, 235-236, https://www.doi.org/10.1038/nmeth.4559
Teixeira, M., Py, B. F., Bosc, C., Laubreton, D., Moutin, M. J., Marvel, J., Flamant, F. & Markossian, S. 2018. Electroporation of mice zygotes with dual guide RNA/Cas9 complexes for simple and efficient cloning-free genome editing. Sci Rep, 8, 474, https://www.doi.org/10.1038/s41598-017-18826-5
Tröder, S. E., Ebert, L. K., Butt, L., Assenmacher, S., Schermer, B. & Zevnik, B. 2018. An optimized electroporation approach for efficient CRISPR/Cas9 genome editing in murine zygotes. PLoS One, 13, e0196891, https://www.doi.org/10.1371/journal.pone.0196891
Takeo, T. & Nakagata, N. 2018. In Vitro Fertilization in Mice. Cold Spring Harb Protoc, 2018, https://www.doi.org/10.1101/pdb.prot094524
Takeo, T. & Nakagata, N. 2018. Mouse Sperm Cryopreservation Using Cryoprotectant Containing l-Glutamine. Cold Spring Harb Protoc, 2018, https://www.doi.org/10.1101/pdb.prot094516
Sztein, J. M., Takeo, T. & Nakagata, N. 2018. History of cryobiology, with special emphasis in evolution of mouse sperm cryopreservation. Cryobiology, 82, 57-63, https://www.doi.org/10.1016/j.cryobiol.2018.04.008
Sato, M., Ohtsuka, M., Nakamura, S., Sakurai, T., Watanabe, S. & Gurumurthy, C. B. 2018. In vivo genome editing targeted towards the female reproductive system. Arch Pharm Res, https://www.doi.org/10.1007/s12272-018-1053-z
Roth, T. L., Puig-Saus, C., Yu, R., Shifrut, E., Carnevale, J., Li, P. J., Hiatt, J., Saco, J., Krystofinski, P., Li, H., Tobin, V., Nguyen, D. N., Lee, M. R., Putnam, A. L., Ferris, A. L., Chen, J. W., Schickel, J. N., Pellerin, L., Carmody, D., Alkorta-Aranburu, G., Del Gaudio, D., Matsumoto, H., Morell, M., Mao, Y., Cho, M., Quadros, R. M., Gurumurthy, C. B., Smith, B., Haugwitz, M., Hughes, S. H., Weissman, J. S., Schumann, K., Esensten, J. H., May, A. P., Ashworth, A., Kupfer, G. M., Greeley, S. a. W., Bacchetta, R., Meffre, E., Roncarolo, M. G., Romberg, N., Herold, K. C., Ribas, A., Leonetti, M. D. & Marson, A. 2018. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature, 559, 405-409, https://www.doi.org/10.1038/s41586-018-0326-5
Raspa, M., Fray, M., Paoletti, R., Montoliu, L., Giuliani, A. & Scavizzi, F. 2018. A new, simple and efficient liquid nitrogen free method to cryopreserve mouse spermatozoa at -80 °C. Theriogenology, 119, 52-59, https://www.doi.org/10.1016/j.theriogenology.2018.06.020
Ohtsuka, M., Sato, M., Miura, H., Takabayashi, S., Matsuyama, M., Koyano, T., Arifin, N., Nakamura, S., Wada, K. & Gurumurthy, C. B. 2018. i-GONAD: a robust method for in situ germline genome engineering using CRISPR nucleases. Genome Biol, 19, 25, https://www.doi.org/10.1186/s13059-018-1400-x
Nakagawa, Y., Sakuma, T., Takeo, T., Nakagata, N. & Yamamoto, T. 2018. Electroporation-mediated genome editing in vitrified/warmed mouse zygotes created by IVF via ultra-superovulation. Exp Anim, 67, 535-543, https://www.doi.org/10.1538/expanim.18-0062
Miyasaka, Y., Uno, Y., Yoshimi, K., Kunihiro, Y., Yoshimura, T., Tanaka, T., Ishikubo, H., Hiraoka, Y., Takemoto, N., Tanaka, T., Ooguchi, Y., Skehel, P., Aida, T., Takeda, J. & Mashimo, T. 2018. CLICK: one-step generation of conditional knockout mice. BMC Genomics, 19, 318, https://www.doi.org/10.1186/s12864-018-4713-y
Miura, H., Quadros, R. M., Gurumurthy, C. B. & Ohtsuka, M. 2018. Easi-CRISPR for creating knock-in and conditional knockout mouse models using long ssDNA donors. Nat Protoc, 13, 195-215, https://www.doi.org/10.1038/nprot.2017.153
Mahabir, E., Volland, R., Landsberger, A., Manz, S., Na, E., Urban, I. & Michel, G. 2018. Reproductive Performance after Unilateral or Bilateral Oviduct Transfer of 2-Cell Embryos in Mice. J Am Assoc Lab Anim Sci, 57, 110-114, https://www.doi.org/
Kobayashi, T., Namba, M., Koyano, T., Fukushima, M., Sato, M., Ohtsuka, M. & Matsuyama, M. 2018. Successful production of genome-edited rats by the rGONAD method. BMC Biotechnol, 18, 19, https://www.doi.org/10.1186/s12896-018-0430-5
Heykants, M., Scherb, H., Michel, G. & Mahabir, E. 2018. Influence of polygamous versus monogamous mating on embryo production in four different strains of mice after superovulatory treatment. Theriogenology, 114, 85-94, https://www.doi.org/10.1016/j.theriogenology.2018.03.023
Gu, B., Posfai, E. & Rossant, J. 2018. Efficient generation of targeted large insertions by microinjection into two-cell-stage mouse embryos. Nat Biotechnol, 36, 632-637, https://www.doi.org/10.1038/nbt.4166
Gertsenstein, M. & Nutter, L. M. J. 2018. Engineering Point Mutant and Epitope-Tagged Alleles in Mice Using Cas9 RNA-Guided Nuclease. Curr Protoc Mouse Biol, 8, 28-53, https://www.doi.org/10.1002/cpmo.40
Charpentier, M., Khedher, A. H. Y., Menoret, S., Brion, A., Lamribet, K., Dardillac, E., Boix, C., Perrouault, L., Tesson, L., Geny, S., De Cian, A., Itier, J. M., Anegon, I., Lopez, B., Giovannangeli, C. & Concordet, J. P. 2018. CtIP fusion to Cas9 enhances transgene integration by homology-dependent repair. Nat Commun, 9, 1133, https://www.doi.org/10.1038/s41467-018-03475-7
Burgio, G. 2018. Redefining mouse transgenesis with CRISPR/Cas9 genome editing technology. Genome Biol, 19, 27, https://www.doi.org/10.1186/s13059-018-1409-1
2017
Yoshimoto, H., Takeo, T., Irie, T. & Nakagata, N. 2017. Fertility of cold-stored mouse sperm is recovered by promoting acrosome reaction and hyperactivation after cholesterol efflux by methyl-beta-cyclodextrin. Biol Reprod, 96, 446-455, https://www.doi.org/10.1095/biolreprod.116.142901
Remy, S., Chenouard, V., Tesson, L., Usal, C., Ménoret, S., Brusselle, L., Heslan, J. M., Nguyen, T. H., Bellien, J., Merot, J., De Cian, A., Giovannangeli, C., Concordet, J. P. & Anegon, I. 2017. Generation of gene-edited rats by delivery of CRISPR/Cas9 protein and donor DNA into intact zygotes using electroporation. Sci Rep, 7, 16554, https://www.doi.org/10.1038/s41598-017-16328-y
Quadros, R. M., Miura, H., Harms, D. W., Akatsuka, H., Sato, T., Aida, T., Redder, R., Richardson, G. P., Inagaki, Y., Sakai, D., Buckley, S. M., Seshacharyulu, P., Batra, S. K., Behlke, M. A., Zeiner, S. A., Jacobi, A. M., Izu, Y., Thoreson, W. B., Urness, L. D., Mansour, S. L., Ohtsuka, M. & Gurumurthy, C. B. 2017. Easi-CRISPR: a robust method for one-step generation of mice carrying conditional and insertion alleles using long ssDNA donors and CRISPR ribonucleoproteins. Genome Biol, 18, 92, https://www.doi.org/10.1186/s13059-017-1220-4
Nakagawa, Y., Sakuma, T., Nishimichi, N., Yokosaki, Y., Takeo, T., Nakagata, N. & Yamamoto, T. 2017. Culture time of vitrified/warmed zygotes before microinjection affects the production efficiency of CRISPR-Cas9-mediated knock-in mice. Biol Open, 6, 706-713, https://www.doi.org/10.1242/bio.025122
Mianne, J., Codner, G. F., Caulder, A., Fell, R., Hutchison, M., King, R., Stewart, M. E., Wells, S. & Teboul, L. 2017. Analysing the outcome of CRISPR-aided genome editing in embryos: Screening, genotyping and quality control. Methods, https://www.doi.org/10.1016/j.ymeth.2017.03.016
Meehan, T. F., Conte, N., West, D. B., Jacobsen, J. O., Mason, J., Warren, J., Chen, C. K., Tudose, I., Relac, M., Matthews, P., Karp, N., Santos, L., Fiegel, T., Ring, N., Westerberg, H., Greenaway, S., Sneddon, D., Morgan, H., Codner, G. F., Stewart, M. E., Brown, J., Horner, N., International Mouse Phenotyping, C., Haendel, M., Washington, N., Mungall, C. J., Reynolds, C. L., Gallegos, J., Gailus-Durner, V., Sorg, T., Pavlovic, G., Bower, L. R., Moore, M., Morse, I., Gao, X., Tocchini-Valentini, G. P., Obata, Y., Cho, S. Y., Seong, J. K., Seavitt, J., Beaudet, A. L., Dickinson, M. E., Herault, Y., Wurst, W., De Angelis, M. H., Lloyd, K. C. K., Flenniken, A. M., Nutter, L. M. J., Newbigging, S., Mckerlie, C., Justice, M. J., Murray, S. A., Svenson, K. L., Braun, R. E., White, J. K., Bradley, A., Flicek, P., Wells, S., Skarnes, W. C., Adams, D. J., Parkinson, H., Mallon, A. M., Brown, S. D. M. & Smedley, D. 2017. Disease model discovery from 3,328 gene knockouts by The International Mouse Phenotyping Consortium. Nat Genet, 49, 1231-1238, https://www.doi.org/10.1038/ng.3901
Jacobi, A. M., Rettig, G. R., Turk, R., Collingwood, M. A., Zeiner, S. A., Quadros, R. M., Harms, D. W., Bonthuis, P. J., Gregg, C., Ohtsuka, M., Gurumurthy, C. B. & Behlke, M. A. 2017. Simplified CRISPR tools for efficient genome editing and streamlined protocols for their delivery into mammalian cells and mouse zygotes. Methods, 121-122, 16-28, https://www.doi.org/10.1016/j.ymeth.2017.03.021
Hosur, V., Low, B. E., Avery, C., Shultz, L. D. & Wiles, M. V. 2017. Development of Humanized Mice in the Age of Genome Editing. J Cell Biochem, https://www.doi.org/10.1002/jcb.26002
Fernandez, A., Josa, S. & Montoliu, L. 2017. A history of genome editing in mammals. Mamm Genome, 28, 237-246, https://www.doi.org/10.1007/s00335-017-9699-2