Skip to main navigation menu Skip to main content Skip to site footer

Review article: Biomedical intelligence

Vol. 145 No. 4344 (2015)

Astonishing advances in mouse genetic tools for biomedical research

  • Walker Scot Jackson
  • Lech Kaczmarczyk
DOI
https://doi.org/10.4414/smw.2015.14186
Cite this as:
Swiss Med Wkly. 2015;145:w14186
Published
18.10.2015

Summary

The humble house mouse has long been a workhorse model system in biomedical research. The technology for introducing site-specific genome modifications led to Nobel Prizes for its pioneers and opened a new era of mouse genetics. However, this technology was very time-consuming and technically demanding. As a result, many investigators continued to employ easier genome manipulation methods, though resulting models can suffer from overlooked or underestimated consequences. Another breakthrough, invaluable for the molecular dissection of disease mechanisms, was the invention of high-throughput methods to measure the expression of a plethora of genes in parallel. However, the use of samples containing material from multiple cell types could obfuscate data, and thus interpretations. In this review we highlight some important issues in experimental approaches using mouse models for biomedical research. We then discuss recent technological advances in mouse genetics that are revolutionising human disease research. Mouse genomes are now easily manipulated at precise locations thanks to guided endonucleases, such as transcription activator-like effector nucleases (TALENs) or the CRISPR/Cas9 system, both also having the potential to turn the dream of human gene therapy into reality. Newly developed methods of cell type-specific isolation of transcriptomes from crude tissue homogenates, followed by detection with next generation sequencing (NGS), are vastly improving gene regulation studies. Taken together, these amazing tools simplify the creation of much more accurate mouse models of human disease, and enable the extraction of hitherto unobtainable data.

References

  1. Fahlstrom A, Yu Q, Ulfhake B. Behavioral changes in aging female C57BL/6 mice. Neurobiol Aging. 2011;32(10):1868–80.
  2. Aasum E, Hafstad AD, Severson DL, Larsen TS. Age-dependent changes in metabolism, contractile function, and ischemic sensitivity in hearts from db/db mice. Diabetes. 2003;52(2):434–41.
  3. Houtkooper RH, Argmann C, Houten SM, Canto C, Jeninga EH, Andreux PA, et al. The metabolic footprint of aging in mice. Sci Rep. 2011;1:134.
  4. Lister R, Mukamel EA, Nery JR, Urich M, Puddifoot CA, Johnson ND, et al. Global epigenomic reconfiguration during mammalian brain development. Science. 2013;341(6146):1237905. Epub 2013/07/06.
  5. Al-Mahdawi S, Virmouni SA, Pook MA. The emerging role of 5-hydroxymethylcytosine in neurodegenerative diseases. Front Neurosci. 2014;8:397. Epub 2014/12/30.
  6. Cheng Y, Ma Z, Kim BH, Wu W, Cayting P, Boyle AP, et al. Principles of regulatory information conservation between mouse and human. Nature. 2014;515(7527):371–5. Epub 2014/11/21.
  7. Pope BD, Ryba T, Dileep V, Yue F, Wu W, Denas O, et al. Topologically associating domains are stable units of replication-timing regulation. Nature. 2014;515(7527):402–5. Epub 2014/11/21.
  8. Stergachis AB, Neph S, Sandstrom R, Haugen E, Reynolds AP, Zhang M, et al. Conservation of trans-acting circuitry during mammalian regulatory evolution. Nature. 2014;515(7527):365–70. Epub 2014/11/21.
  9. Yue F, Cheng Y, Breschi A, Vierstra J, Wu W, Ryba T, et al. A comparative encyclopedia of DNA elements in the mouse genome. Nature. 2014;515(7527):355–64. Epub 2014/11/21.
  10. Wu LC, Sun CW, Ryan TM, Pawlik KM, Ren J, Townes TM. Correction of sickle cell disease by homologous recombination in embryonic stem cells. Blood. 2006;108(4):1183–8. Epub 2006/04/28.
  11. Hanna J, Wernig M, Markoulaki S, Sun CW, Meissner A, Cassady JP, et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science. 2007;318(5858):1920–3. Epub 2007/12/08.
  12. Heyer J, Kwong LN, Lowe SW, Chin L. Non-germline genetically engineered mouse models for translational cancer research. Nat Rev Cancer. 2010;10(7):470–80. Epub 2010/06/25.
  13. Walrath JC, Hawes JJ, Van Dyke T, Reilly KM. Genetically engineered mouse models in cancer research. Adv Cancer Res. 2010;106:113–64. Epub 2010/04/20.
  14. Philips T, Rothstein JD, Pouladi MA. Preclinical models: needed in translation? A Pro/Con debate. Mov Disord. 2014;29(11):1391–6. Epub 2014/09/13.
  15. Jaenisch R, Mintz B. Simian virus 40 DNA sequences in DNA of healthy adult mice derived from preimplantation blastocysts injected with viral DNA. Proc Natl Acad Sci U S A. 1974;71(4):1250–4. Epub 1974/04/01.
  16. Brinster RL, Chen HY, Trumbauer M, Senear AW, Warren R, Palmiter RD. Somatic expression of herpes thymidine kinase in mice following injection of a fusion gene into eggs. Cell. 1981;27(1 Pt 2):223–31. Epub 1981/11/01.
  17. Costantini F, Lacy E. Introduction of a rabbit beta-globin gene into the mouse germ line. Nature. 1981;294(5836):92–4. Epub 1981/11/05.
  18. Wurtele H, Little KC, Chartrand P. Illegitimate DNA integration in mammalian cells. Gene Ther. 2003;10(21):1791–9. Epub 2003/09/10.
  19. Smithies O, Gregg RG, Boggs SS, Koralewski MA, Kucherlapati RS. Insertion of DNA sequences into the human chromosomal beta-globin locus by homologous recombination. Nature. 1985;317(6034):230–4.
  20. Thomas KR, Capecchi MR. Site-directed mutagenesis by gene targeting in mouse embryo-derived stem cells. Cell. 1987;51(3):503–12.
  21. Robertson E, Bradley A, Kuehn M, Evans M. Germ-line transmission of genes introduced into cultured pluripotential cells by retroviral vector. Nature. 1986;323(6087):445–8. Epub 1986/10/02.
  22. Hooper M, Hardy K, Handyside A, Hunter S, Monk M. HPRT-deficient (Lesch-Nyhan) mouse embryos derived from germline colonization by cultured cells. Nature. 1987;326(6110):292–5. Epub 1987/03/19.
  23. Kuehn MR, Bradley A, Robertson EJ, Evans MJ. A potential animal model for Lesch-Nyhan syndrome through introduction of HPRT mutations into mice. Nature. 1987;326(6110):295–8. Epub 1987/03/19.
  24. Koller BH, Hagemann LJ, Doetschman T, Hagaman JR, Huang S, Williams PJ, et al. Germ-line transmission of a planned alteration made in a hypoxanthine phosphoribosyltransferase gene by homologous recombination in embryonic stem cells. Proc Natl Acad Sci U S A. 1989;86(22):8927–31. Epub 1989/11/01.
  25. Thompson S, Clarke AR, Pow AM, Hooper ML, Melton DW. Germ line transmission and expression of a corrected HPRT gene produced by gene targeting in embryonic stem cells. Cell. 1989;56(2):313–21. Epub 1989/01/27.
  26. Zijlstra M, Li E, Sajjadi F, Subramani S, Jaenisch R. Germ-line transmission of a disrupted beta 2-microglobulin gene produced by homologous recombination in embryonic stem cells. Nature. 1989;342(6248):435–8. Epub 1989/11/23.
  27. Thomas KR, Capecchi MR. Targeted disruption of the murine int-1 proto-oncogene resulting in severe abnormalities in midbrain and cerebellar development. Nature. 1990;346(6287):847–50. Epub 1990/08/30.
  28. Smithies O, Maeda N. Gene targeting approaches to complex genetic diseases: atherosclerosis and essential hypertension. Proc Natl Acad Sci U S A. 1995;92(12):5266–72. Epub 1995/06/06.
  29. Melton DW. Gene targeting in the mouse. Bioessays. 1994;16(9):633–8.
  30. Detloff PJ, Lewis J, John SW, Shehee WR, Langenbach R, Maeda N, et al. Deletion and replacement of the mouse adult beta-globin genes by a “plug and socket” repeated targeting strategy. Mol Cell Biol. 1994;14(10):6936–43. Epub 1994/10/01.
  31. Stacey A, Schnieke A, McWhir J, Cooper J, Colman A, Melton DW. Use of double-replacement gene targeting to replace the murine alpha-lactalbumin gene with its human counterpart in embryonic stem cells and mice. Mol Cell Biol. 1994;14(2):1009–16. Epub 1994/02/01.
  32. Moore RC, Redhead NJ, Selfridge J, Hope J, Manson JC, Melton DW. Double replacement gene targeting for the production of a series of mouse strains with different prion protein gene alterations. Biotechnology (N Y). 1995;13(9):999–1004.
  33. Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev M, Ragoczy T, Telling A, et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science. 2009;326(5950):289–93. Epub 2009/10/10.
  34. Gheldof N, Smith EM, Tabuchi TM, Koch CM, Dunham I, Stamatoyannopoulos JA, et al. Cell-type-specific long-range looping interactions identify distant regulatory elements of the CFTR gene. Nucleic Acids Res. 2010;38(13):4325–36. Epub 2010/04/03.
  35. Gibcus JH, Dekker J. The hierarchy of the 3D genome. Mol Cell. 2013;49(5):773–82. Epub 2013/03/12.
  36. Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-Andre V, Sigova AA, et al. Super-enhancers in the control of cell identity and disease. Cell. 2013;155(4):934–47. Epub 2013/10/15.
  37. Dowen JM, Fan ZP, Hnisz D, Ren G, Abraham BJ, Zhang LN, et al. Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell. 2014;159(2):374–87. Epub 2014/10/11.
  38. Burright EN, Clark HB, Servadio A, Matilla T, Feddersen RM, Yunis WS, et al. SCA1 transgenic mice: a model for neurodegeneration caused by an expanded CAG trinucleotide repeat. Cell. 1995;82(6):937–48.
  39. Caroni P. Overexpression of growth-associated proteins in the neurons of adult transgenic mice. J Neurosci Methods. 1997;71(1):3–9. Epub 1997/01/01.
  40. Feng G, Mellor RH, Bernstein M, Keller-Peck C, Nguyen QT, Wallace M, et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron. 2000;28(1):41–51.
  41. Livet J, Weissman TA, Kang H, Draft RW, Lu J, Bennis RA, et al. Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system. Nature. 2007;450(7166):56–62. Epub 2007/11/02.
  42. Borchelt DR, Davis J, Fischer M, Lee MK, Slunt HH, Ratovitsky T, et al. A vector for expressing foreign genes in the brains and hearts of transgenic mice. Genet Anal. 1996;13(6):159–63.
  43. Fischer M, Rulicke T, Raeber A, Sailer A, Moser M, Oesch B, et al. Prion protein (PrP) with amino-proximal deletions restoring susceptibility of PrP knockout mice to scrapie. Embo J. 1996;15(6):1255–64.
  44. Faas H, Jackson WS, Borkowski AW, Wang X, Ma J, Lindquist S, et al. Context-dependent perturbation of neural systems in transgenic mice expressing a cytosolic prion protein. Neuroimage. 2010;49(3):2607–17. Epub 2009/10/20.
  45. Karapetyan YE, Saa P, Mahal SP, Sferrazza GF, Sherman A, Sales N, et al. Prion strain discrimination based on rapid in vivo amplification and analysis by the cell panel assay. PLoS One. 2009;4(5):e5730. Epub 2009/05/30.
  46. Gong S, Zheng C, Doughty ML, Losos K, Didkovsky N, Schambra UB, et al. A gene expression atlas of the central nervous system based on bacterial artificial chromosomes. Nature. 2003;425(6961):917–25. Epub 2003/10/31.
  47. Zabel M, Greenwood C, Thackray AM, Pulford B, Rens W, Bujdoso R. Perturbation of T-cell development by insertional mutation of a PrP transgene. Immunology. 2009;127(2):226–36. Epub 2009/01/16.
  48. Akhtar W, de Jong J, Pindyurin AV, Pagie L, Meuleman W, de Ridder J, et al. Chromatin position effects assayed by thousands of reporters integrated in parallel. Cell. 2013;154(4):914–27. Epub 2013/08/21.
  49. Lewis J, Yang B, Kim R, Sierakowska H, Kole R, Smithies O, et al. A common human beta globin splicing mutation modeled in mice. Blood. 1998;91(6):2152–6. Epub 1998/04/16.
  50. Lin CH, Tallaksen-Greene S, Chien WM, Cearley JA, Jackson WS, Crouse AB, et al. Neurological abnormalities in a knock-in mouse model of Huntington’s disease. Hum Mol Genet. 2001;10(2):137–44. Epub 2001/01/12.
  51. Watase K, Weeber EJ, Xu B, Antalffy B, Yuva-Paylor L, Hashimoto K, et al. A long CAG repeat in the mouse Sca1 locus replicates SCA1 features and reveals the impact of protein solubility on selective neurodegeneration. Neuron. 2002;34(6):905–19. Epub 2002/06/28.
  52. Jackson WS, Borkowski AW, Faas H, Steele AD, King OD, Watson N, et al. Spontaneous generation of prion infectivity in fatal familial insomnia knockin mice. Neuron. 2009;63(4):438–50.
  53. Jackson WS. Selective vulnerability to neurodegenerative disease: the curious case of Prion Protein. Dis Model Mech. 2014;7(1):21–9. Epub 2014/01/08.
  54. Lieber MR. The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu Rev Biochem. 2010;79:181–211. Epub 2010/03/03.
  55. van den Bosch M, Lohman PH, Pastink A. DNA double-strand break repair by homologous recombination. Biol Chem. 2002;383(6):873–92. Epub 2002/09/12.
  56. Szostak JW, Orr-Weaver TL, Rothstein RJ, Stahl FW. The double-strand-break repair model for recombination. Cell. 1983;33(1):25–35. Epub 1983/05/01.
  57. Aymard F, Bugler B, Schmidt CK, Guillou E, Caron P, Briois S, et al. Transcriptionally active chromatin recruits homologous recombination at DNA double-strand breaks. Nat Struct Mol Biol. 2014;21(4):366–74. Epub 2014/03/25.
  58. Weissmann C, Bueler H. A mouse to remember. Cell. 2004;116(2 Suppl):S111–3, 2 p following S3. Epub 2004/04/02.
  59. Miranda A, Pericuesta E, Ramirez MA, Gutierrez-Adan A. Prion protein expression regulates embryonic stem cell pluripotency and differentiation. PLoS One. 2011;6(4):e18422. Epub 2011/04/13.
  60. Palais G, Nguyen Dinh Cat A, Friedman H, Panek-Huet N, Millet A, Tronche F, et al. Targeted transgenesis at the HPRT locus: an efficient strategy to achieve tightly controlled in vivo conditional expression with the tet system. Physiol Genomics. 2009;37(2):140–6. Epub 2009/01/15.
  61. Soriano P. Generalized lacZ expression with the ROSA26 Cre reporter strain. Nat Genet. 1999;21(1):70–1. Epub 1999/01/23.
  62. Madisen L, Zwingman TA, Sunkin SM, Oh SW, Zariwala HA, Gu H, et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci. 2010;13(1):133–40. Epub 2009/12/22.
  63. Zambrowicz BP, Imamoto A, Fiering S, Herzenberg LA, Kerr WG, Soriano P. Disruption of overlapping transcripts in the ROSA beta geo 26 gene trap strain leads to widespread expression of beta-galactosidase in mouse embryos and hematopoietic cells. Proc Natl Acad Sci U S A. 1997;94(8):3789–94. Epub 1997/04/15.
  64. Zeng H, Horie K, Madisen L, Pavlova MN, Gragerova G, Rohde AD, et al. An inducible and reversible mouse genetic rescue system. PLoS Genet. 2008;4(5):e1000069. Epub 2008/05/10.
  65. Madisen L, Garner AR, Shimaoka D, Chuong AS, Klapoetke NC, Li L, et al. Transgenic mice for intersectional targeting of neural sensors and effectors with high specificity and performance. Neuron. 2015;85(5):942–58. Epub 2015/03/06.
  66. Kim YG, Cha J, Chandrasegaran S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc Natl Acad Sci U S A. 1996;93(3):1156–60. Epub 1996/02/06.
  67. Bibikova M, Beumer K, Trautman JK, Carroll D. Enhancing gene targeting with designed zinc finger nucleases. Science. 2003;300(5620):764. Epub 2003/05/06.
  68. Bibikova M, Golic M, Golic KG, Carroll D. Targeted chromosomal cleavage and mutagenesis in Drosophila using zinc-finger nucleases. Genetics. 2002;161(3):1169–75. Epub 2002/07/24.
  69. Carbery ID, Ji D, Harrington A, Brown V, Weinstein EJ, Liaw L, et al. Targeted genome modification in mice using zinc-finger nucleases. Genetics. 2010;186(2):451–9. Epub 2010/07/16.
  70. Kimberlin RH, Walker CA. Pathogenesis of mouse scrapie: effect of route of inoculation on infectivity titres and dose-response curves. J Comp Pathol. 1978;88(1):39–47. Epub 1978/01/01.
  71. Porteus MH, Baltimore D. Chimeric nucleases stimulate gene targeting in human cells. Science. 2003;300(5620):763. Epub 2003/05/06.
  72. Segal DJ, Meckler JF. Genome engineering at the dawn of the golden age. Annu Rev Genomics Hum Genet. 2013;14:135–58. Epub 2013/05/25.
  73. Rahman SH, Maeder ML, Joung JK, Cathomen T. Zinc-finger nucleases for somatic gene therapy: the next frontier. Hum Gene Ther. 2011;22(8):925–33. Epub 2011/06/03.
  74. Kim HJ, Lee HJ, Kim H, Cho SW, Kim JS. Targeted genome editing in human cells with zinc finger nucleases constructed via modular assembly. Genome Res. 2009;19(7):1279–88. Epub 2009/05/28.
  75. Maeder ML, Thibodeau-Beganny S, Sander JD, Voytas DF, Joung JK. Oligomerized pool engineering (OPEN): an “open-source” protocol for making customized zinc-finger arrays. Nat Protoc. 2009;4(10):1471–501. Epub 2009/10/03.
  76. Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. Genome editing with engineered zinc finger nucleases. Nat Rev Genet. 2010;11(9):636–46. Epub 2010/08/19.
  77. Ferguson C, McKay M, Harris RA, Homanics GE. Toll-like receptor 4 (Tlr4) knockout rats produced by transcriptional activator-like effector nuclease (TALEN)-mediated gene inactivation. Alcohol. 2013;47(8):595–9. Epub 2013/11/10.
  78. Sung YH, Baek IJ, Kim DH, Jeon J, Lee J, Lee K, et al. Knockout mice created by TALEN-mediated gene targeting. Nat Biotechnol. 2013;31(1):23–4. Epub 2013/01/11.
  79. Sung YH, Kim JM, Kim HT, Lee J, Jeon J, Jin Y, et al. Highly efficient gene knockout in mice and zebrafish with RNA-guided endonucleases. Genome Res. 2014;24(1):125–31. Epub 2013/11/21.
  80. Tesson L, Usal C, Menoret S, Leung E, Niles BJ, Remy S, et al. Knockout rats generated by embryo microinjection of TALENs. Nat Biotechnol. 2011;29(8):695–6. Epub 2011/08/09.
  81. Wefers B, Meyer M, Ortiz O, Hrabe de Angelis M, Hansen J, Wurst W, et al. Direct production of mouse disease models by embryo microinjection of TALENs and oligodeoxynucleotides. Proc Natl Acad Sci U S A. 2013;110(10):3782–7. Epub 2013/02/22.
  82. Gilles AF, Averof M. Functional genetics for all: engineered nucleases, CRISPR and the gene editing revolution. Evodevo. 2014;5:43. Epub 2015/02/24.
  83. Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014;346(6213):1258096. Epub 2014/11/29.
  84. Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for genome engineering. Cell. 2014;157(6):1262–78. Epub 2014/06/07.
  85. Gaj T, Gersbach CA, Barbas CF, 3rd. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 2013;31(7):397–405. Epub 2013/05/15.
  86. Pu J, Frescas D, Zhang B, Feng J. Utilization of TALEN and CRISPR/Cas9 technologies for gene targeting and modification. Exp Biol Med (Maywood). 2015. Epub 2015/05/10.
  87. Cho SW, Kim S, Kim Y, Kweon J, Kim HS, Bae S, et al. Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res. 2014;24(1):132–41. Epub 2013/11/21.
  88. Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung JK, et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol. 2013;31(9):822–6. Epub 2013/06/25.
  89. Ran FA, Hsu PD, Lin CY, Gootenberg JS, Konermann S, Trevino AE, et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell. 2013;154(6):1380–9. Epub 2013/09/03.
  90. Guilinger JP, Thompson DB, Liu DR. Fusion of catalytically inactive Cas9 to FokI nuclease improves the specificity of genome modification. Nat Biotechnol. 2014;32(6):577–82. Epub 2014/04/29.
  91. Sommer D, Peters A, Wirtz T, Mai M, Ackermann J, Thabet Y, et al. Efficient genome engineering by targeted homologous recombination in mouse embryos using transcription activator-like effector nucleases. Nat Commun. 2014;5:3045. Epub 2014/01/15.
  92. Briggs AW, Rios X, Chari R, Yang L, Zhang F, Mali P, et al. Iterative capped assembly: rapid and scalable synthesis of repeat-module DNA such as TAL effectors from individual monomers. Nucleic Acids Res. 2012;40(15):e117. Epub 2012/06/29.
  93. Reyon D, Tsai SQ, Khayter C, Foden JA, Sander JD, Joung JK. FLASH assembly of TALENs for high-throughput genome editing. Nat Biotechnol. 2012;30(5):460–5. Epub 2012/04/10.
  94. Cermak T, Doyle EL, Christian M, Wang L, Zhang Y, Schmidt C, et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 2011;39(12):e82. Epub 2011/04/16.
  95. Schmid-Burgk JL, Schmidt T, Kaiser V, Honing K, Hornung V. A ligation-independent cloning technique for high-throughput assembly of transcription activator-like effector genes. Nat Biotechnol. 2013;31(1):76–81. Epub 2012/12/18.
  96. Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW, Zhang F, et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell. 2013;153(4):910–8. Epub 2013/05/07.
  97. Platt RJ, Chen S, Zhou Y, Yim MJ, Swiech L, Kempton HR, et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell. 2014;159(2):440–55. Epub 2014/09/30.
  98. Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature. 2015;520(7546):186–91. Epub 2015/04/02.
  99. Ousterout DG, Kabadi AM, Thakore PI, Majoros WH, Reddy TE, Gersbach CA. Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy. Nat Commun. 2015;6:6244. Epub 2015/02/19.
  100. Liao HK, Gu Y, Diaz A, Marlett J, Takahashi Y, Li M, et al. Use of the CRISPR/Cas9 system as an intracellular defense against HIV-1 infection in human cells. Nat Commun. 2015;6:6413. Epub 2015/03/11.
  101. Osborn MJ, Gabriel R, Webber BR, DeFeo AP, McElroy AN, Jarjour J, et al. Fanconi anemia gene editing by the CRISPR/Cas9 system. Hum Gene Ther. 2015;26(2):114–26. Epub 2014/12/30.
  102. Heckl D, Kowalczyk MS, Yudovich D, Belizaire R, Puram RV, McConkey ME, et al. Generation of mouse models of myeloid malignancy with combinatorial genetic lesions using CRISPR-Cas9 genome editing. Nat Biotechnol. 2014;32(9):941–6. Epub 2014/06/24.
  103. Lee HJ, Kweon J, Kim E, Kim S, Kim JS. Targeted chromosomal duplications and inversions in the human genome using zinc finger nucleases. Genome Res. 2012;22(3):539–48. Epub 2011/12/21.
  104. Renouf B, Piganeau M, Ghezraoui H, Jasin M, Brunet E. Creating cancer translocations in human cells using Cas9 DSBs and nCas9 paired nicks. Methods Enzymol. 2014;546:251–71. Epub 2014/11/16.
  105. Maddalo D, Manchado E, Concepcion CP, Bonetti C, Vidigal JA, Han YC, et al. In vivo engineering of oncogenic chromosomal rearrangements with the CRISPR/Cas9 system. Nature. 2014;516(7531):423–7. Epub 2014/10/23.
  106. Mou H, Kennedy Z, Anderson DG, Yin H, Xue W. Precision cancer mouse models through genome editing with CRISPR-Cas9. Genome Med. 2015;7(1):53. Epub 2015/06/11.
  107. Qin W, Dion SL, Kutny PM, Zhang Y, Cheng A, Jillette NL, et al. Efficient CRISPR/Cas9-Mediated Genome Editing in Mice by Zygote Electroporation of Nuclease. Genetics. 2015. Epub 2015/03/31.
  108. Maruyama T, Dougan SK, Truttmann MC, Bilate AM, Ingram JR, Ploegh HL. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat Biotechnol. 2015;33(5):538–42. Epub 2015/03/24.
  109. Cheng AW, Wang H, Yang H, Shi L, Katz Y, Theunissen TW, et al. Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res. 2013;23(10):1163–71. Epub 2013/08/28.
  110. Konermann S, Brigham MD, Trevino AE, Joung J, Abudayyeh OO, Barcena C, et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature. 2015;517(7536):583–8. Epub 2014/12/11.
  111. Konermann S, Brigham MD, Trevino AE, Hsu PD, Heidenreich M, Cong L, et al. Optical control of mammalian endogenous transcription and epigenetic states. Nature. 2013;500(7463):472–6. Epub 2013/07/24.
  112. Polstein LR, Gersbach CA. A light-inducible CRISPR-Cas9 system for control of endogenous gene activation. Nat Chem Biol. 2015;11(3):198–200. Epub 2015/02/11.
  113. Heller EA, Cates HM, Pena CJ, Sun H, Shao N, Feng J, et al. Locus-specific epigenetic remodeling controls addiction- and depression-related behaviors. Nat Neurosci. 2014;17(12):1720–7. Epub 2014/10/28.
  114. Hilton IB, D’Ippolito AM, Vockley CM, Thakore PI, Crawford GE, Reddy TE, et al. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat Biotechnol. 2015. Epub 2015/04/08.
  115. Polstein L, Perez-Pinera P, Kocak D, Vockley C, Bledsoe P, Song L, et al. Genome-wide specificity of DNA-binding, gene regulation, and chromatin remodeling by TALE- and CRISPR/Cas9-based transcriptional activators. Genome Res. 2015. Epub 2015/05/31.
  116. Bueler H, Fischer M, Lang Y, Bluethmann H, Lipp HP, DeArmond SJ, et al. Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature. 1992;356(6370):577–82.
  117. Manson JC, Clarke AR, Hooper ML, Aitchison L, McConnell I, Hope J. 129/Ola mice carrying a null mutation in PrP that abolishes mRNA production are developmentally normal. Mol Neurobiol. 1994;8(2-3):121–7.
  118. Sakaguchi S, Katamine S, Shigematsu K, Nakatani A, Moriuchi R, Nishida N, et al. Accumulation of proteinase K-resistant prion protein (PrP) is restricted by the expression level of normal PrP in mice inoculated with a mouse-adapted strain of the Creutzfeldt-Jakob disease agent. J Virol. 1995;69(12):7586–92. Epub 1995/12/01.
  119. Jackson WS, Krost C, Borkowski AW, Kaczmarczyk L. Translation of the Prion Protein mRNA Is Robust in Astrocytes but Does Not Amplify during Reactive Astrocytosis in the Mouse Brain. PLoS One. 2014;9(4):e95958. Epub 2014/04/23.
  120. Nuvolone M, Kana V, Hutter G, Sakata D, Mortin-Toth SM, Russo G, et al. SIRPalpha polymorphisms, but not the prion protein, control phagocytosis of apoptotic cells. J Exp Med. 2013;210(12):2539–52. Epub 2013/10/23.
  121. de Almeida CJ, Chiarini LB, da Silva JP, PM ES, Martins MA, Linden R. The cellular prion protein modulates phagocytosis and inflammatory response. J Leukoc Biol. 2005;77(2):238–46. Epub 2004/11/13.
  122. Gerlai R. Gene-targeting studies of mammalian behavior: is it the mutation or the background genotype? Trends Neurosci. 1996;19(5):177–81. Epub 1996/05/01.
  123. Crusio WE. Flanking gene and genetic background problems in genetically manipulated mice. Biol Psychiatry. 2004;56(6):381–5. Epub 2004/09/15.
  124. Striebel JF, Race B, Pathmajeyan M, Rangel A, Chesebro B. Lack of influence of prion protein gene expression on kainate-induced seizures in mice: studies using congenic, coisogenic and transgenic strains. Neuroscience. 2013;238:11–8. Epub 2013/02/19.
  125. Ordway JM, Tallaksen-Greene S, Gutekunst CA, Bernstein EM, Cearley JA, Wiener HW, et al. Ectopically expressed CAG repeats cause intranuclear inclusions and a progressive late onset neurological phenotype in the mouse. Cell. 1997;91(6):753–63. Epub 1997/12/31.
  126. Jackson WS, Tallaksen-Greene SJ, Albin RL, Detloff PJ. Nucleocytoplasmic transport signals affect the age at onset of abnormalities in knock-in mice expressing polyglutamine within an ectopic protein context. Hum Mol Genet. 2003;12(13):1621–9.
  127. Jackson WS, Borkowski AW, Watson NE, King OD, Faas H, Jasanoff A, et al. Profoundly different prion diseases in knock-in mice carrying single PrP codon substitutions associated with human diseases. Proc Natl Acad Sci U S A. 2013;110(36):14759–64. Epub 2013/08/21.
  128. Diedrich J, Wietgrefe S, Zupancic M, Staskus K, Retzel E, Haase AT, et al. The molecular pathogenesis of astrogliosis in scrapie and Alzheimer's disease. Microb Pathog. 1987;2(6):435–42. Epub 1987/06/01.
  129. Lashkari DA, DeRisi JL, McCusker JH, Namath AF, Gentile C, Hwang SY, et al. Yeast microarrays for genome wide parallel genetic and gene expression analysis. Proc Natl Acad Sci U S A. 1997;94(24):13057–62. Epub 1997/12/16.
  130. Schena M, Shalon D, Davis RW, Brown PO. Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science. 1995;270(5235):467–70. Epub 1995/10/20.
  131. Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet. 2009;10(1):57–63. Epub 2008/11/19.
  132. Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci U S A. 2013;110(9):3507–12. Epub 2013/02/13.
  133. Takao K, Miyakawa T. Genomic responses in mouse models greatly mimic human inflammatory diseases. Proc Natl Acad Sci U S A. 2015;112(4):1167–72. Epub 2014/08/06.
  134. Kuhn A, Goldstein DR, Hodges A, Strand AD, Sengstag T, Kooperberg C, et al. Mutant huntingtin’s effects on striatal gene expression in mice recapitulate changes observed in human Huntington’s disease brain and do not differ with mutant huntingtin length or wild-type huntingtin dosage. Hum Mol Genet. 2007;16(15):1845–61. Epub 2007/05/24.
  135. Pereson S, Wils H, Kleinberger G, McGowan E, Vandewoestyne M, Van Broeck B, et al. Progranulin expression correlates with dense-core amyloid plaque burden in Alzheimer disease mouse models. J Pathol. 2009;219(2):173–81. Epub 2009/06/27.
  136. Neueder A, Bates GP. A common gene expression signature in Huntington inverted question marks disease patient brain regions. BMC Med Genomics. 2014;7(1):60. Epub 2014/11/02.
  137. Adiconis X, Borges-Rivera D, Satija R, DeLuca DS, Busby MA, Berlin AM, et al. Comparative analysis of RNA sequencing methods for degraded or low-input samples. Nat Methods. 2013;10(7):623–9. Epub 2013/05/21.
  138. Islam S, Zeisel A, Joost S, La Manno G, Zajac P, Kasper M, et al. Quantitative single-cell RNA-seq with unique molecular identifiers. Nat Methods. 2014;11(2):163–6. Epub 2013/12/24.
  139. Bhargava V, Head SR, Ordoukhanian P, Mercola M, Subramaniam S. Technical variations in low-input RNA-seq methodologies. Scientific reports. 2014;4:3678. Epub 2014/01/15.
  140. Shanker S, Paulson A, Edenberg HJ, Peak A, Perera A, Alekseyev YO, et al. Evaluation of commercially available RNA amplification kits for RNA sequencing using very low input amounts of total RNA. J Biomol Tech. 2015;26(1):4–18. Epub 2015/02/05.
  141. Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson KS, et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J Neurosci. 2008;28(1):264–78. Epub 2008/01/04.
  142. Foo LC, Allen NJ, Bushong EA, Ventura PB, Chung WS, Zhou L, et al. Development of a method for the purification and culture of rodent astrocytes. Neuron. 2011;71(5):799–811. Epub 2011/09/10.
  143. Emmert-Buck MR, Bonner RF, Smith PD, Chuaqui RF, Zhuang Z, Goldstein SR, et al. Laser capture microdissection. Science. 1996;274(5289):998–1001. Epub 1996/11/08.
  144. Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature. 2007;445(7124):168–76.
  145. Majer A, Medina SJ, Niu Y, Abrenica B, Manguiat KJ, Frost KL, et al. Early mechanisms of pathobiology are revealed by transcriptional temporal dynamics in hippocampal CA1 neurons of prion infected mice. PLoS Pathog. 2012;8(11):e1003002. Epub 2012/11/13.
  146. Gu H, Marth JD, Orban PC, Mossmann H, Rajewsky K. Deletion of a DNA polymerase beta gene segment in T cells using cell type-specific gene targeting. Science. 1994;265(5168):103–6. Epub 1994/07/01.
  147. Taniguchi H, He M, Wu P, Kim S, Paik R, Sugino K, et al. A resource of Cre driver lines for genetic targeting of GABAergic neurons in cerebral cortex. Neuron. 2011;71(6):995–1013. Epub 2011/09/29.
  148. Vong L, Ye C, Yang Z, Choi B, Chua S, Jr., Lowell BB. Leptin action on GABAergic neurons prevents obesity and reduces inhibitory tone to POMC neurons. Neuron. 2011;71(1):142–54. Epub 2011/07/13.
  149. Guenthner CJ, Miyamichi K, Yang HH, Heller HC, Luo L. Permanent genetic access to transiently active neurons via TRAP: targeted recombination in active populations. Neuron. 2013;78(5):773–84. Epub 2013/06/15.
  150. Gay L, Miller MR, Ventura PB, Devasthali V, Vue Z, Thompson HL, et al. Mouse TU tagging: a chemical/genetic intersectional method for purifying cell type-specific nascent RNA. Genes Dev. 2013;27(1):98–115. Epub 2013/01/12.
  151. Gay L, Karfilis KV, Miller MR, Doe CQ, Stankunas K. Applying thiouracil tagging to mouse transcriptome analysis. Nat Protoc. 2014;9(2):410–20. Epub 2014/01/25.
  152. Guo H, Ingolia NT, Weissman JS, Bartel DP. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 2010;466(7308):835–40. Epub 2010/08/13.
  153. Hammond SM, Boettcher S, Caudy AA, Kobayashi R, Hannon GJ. Argonaute2, a link between genetic and biochemical analyses of RNAi. Science. 2001;293(5532):1146–50.
  154. He M, Liu Y, Wang X, Zhang MQ, Hannon GJ, Huang ZJ. Cell-type-based analysis of microRNA profiles in the mouse brain. Neuron. 2012;73(1):35–48. Epub 2012/01/17.
  155. Sanz E, Yang L, Su T, Morris DR, McKnight GS, Amieux PS. Cell-type-specific isolation of ribosome-associated mRNA from complex tissues. Proc Natl Acad Sci U S A. 2009;106(33):13939–44. Epub 2009/08/12.
  156. Doyle JP, Dougherty JD, Heiman M, Schmidt EF, Stevens TR, Ma G, et al. Application of a translational profiling approach for the comparative analysis of CNS cell types. Cell. 2008;135(4):749–62. Epub 2008/11/18.
  157. Heiman M, Schaefer A, Gong S, Peterson JD, Day M, Ramsey KE, et al. A translational profiling approach for the molecular characterization of CNS cell types. Cell. 2008;135(4):738–48. Epub 2008/11/18.
  158. Battle A, Khan Z, Wang SH, Mitrano A, Ford MJ, Pritchard JK, et al. Genomic variation. Impact of regulatory variation from RNA to protein. Science. 2015;347(6222):664–7. Epub 2015/02/07.
  159. Achour M, Le Gras S, Keime C, Parmentier F, Lejeune FX, Boutillier AL, et al. Neuronal Identity Genes Regulated by Super-Enhancers Are Preferentially Down-Regulated in the Striatum of Huntington’s Disease Mice. Hum Mol Genet. 2015. Epub 2015/03/19.
  160. Gu H, Zou YR, Rajewsky K. Independent control of immunoglobulin switch recombination at individual switch regions evidenced through Cre-loxP-mediated gene targeting. Cell. 1993;73(6):1155–64. Epub 1993/06/18.
  161. Danielian PS, Muccino D, Rowitch DH, Michael SK, McMahon AP. Modification of gene activity in mouse embryos in utero by a tamoxifen-inducible form of Cre recombinase. Curr Biol. 1998;8(24):1323–6. Epub 1998/12/09.
  162. Pabo CO, Peisach E, Grant RA. Design and selection of novel Cys2His2 zinc finger proteins. Annu Rev Biochem. 2001;70:313–40. Epub 2001/06/08.
  163. Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S, et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science. 2009;326(5959):1509–12. Epub 2009/11/26.
  164. Kim JS, Lee HJ, Carroll D. Genome editing with modularly assembled zinc-finger nucleases. Nat Methods. 2010;7(2):91; author reply -2. Epub 2010/01/30.
  165. Morbitzer R, Elsaesser J, Hausner J, Lahaye T. Assembly of custom TALE-type DNA binding domains by modular cloning. Nucleic Acids Res. 2011;39(13):5790–9. Epub 2011/03/23.
  166. Zhang F, Cong L, Lodato S, Kosuri S, Church GM, Arlotta P. Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription. Nat Biotechnol. 2011;29(2):149–53. Epub 2011/01/21.
  167. Sanjana NE, Cong L, Zhou Y, Cunniff MM, Feng G, Zhang F. A transcription activator-like effector toolbox for genome engineering. Nat Protoc. 2012;7(1):171–92. Epub 2012/01/10.
  168. Reyon D, Maeder ML, Khayter C, Tsai SQ, Foley JE, Sander JD, et al. Engineering customized TALE nucleases (TALENs) and TALE transcription factors by fast ligation-based automatable solid-phase high-throughput (FLASH) assembly. Curr Protoc Mol Biol. 2013;Chapter 12:Unit 12 6. Epub 2013/07/04.
  169. Cornu TI, Thibodeau-Beganny S, Guhl E, Alwin S, Eichtinger M, Joung JK, et al. DNA-binding specificity is a major determinant of the activity and toxicity of zinc-finger nucleases. Mol Ther. 2008;16(2):352–8. Epub 2007/11/21.
  170. Ramirez CL, Foley JE, Wright DA, Muller-Lerch F, Rahman SH, Cornu TI, et al. Unexpected failure rates for modular assembly of engineered zinc fingers. Nat Methods. 2008;5(5):374–5. Epub 2008/05/01.