Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Am I ready for CRISPR? A user's guide to genetic screens

Key Points

  • Pooled screens are a cost-effective approach to investigating phenotypes at the genome scale and build on technological innovations with lentivirus, oligonucleotide synthesis and massively parallel sequencing.

  • CRISPR technology is extremely powerful, with many modalities for perturbing gene function. A successful genetic screen, however, requires not only a good library of perturbations but also a relevant model and optimized assay.

  • When executing a screen, maintaining representation of the library is essential to good quantification. All steps in the process should be investigated experimentally in advance of the screen to ensure efficiency.

  • Designing a library of single-guide RNAs (sgRNAs) requires genomic information from multiple sources, which will change over time as the genome is better annotated and may vary depending on the type of cell under investigation.

  • Defining a systematic follow-up path, both analytically and experimentally, should be thought through before conducting a screen. Secondary pooled screens with customized libraries can be very powerful at this stage.

Abstract

Exciting new technologies are often self-limiting in their rollout, as access to state-of-the-art instrumentation or the need for years of hands-on experience, for better or worse, ensures slow adoption by the community. CRISPR technology, however, presents the opposite dilemma, where the simplicity of the system enabled the parallel development of many applications, improvements and derivatives, and new users are now presented with an almost paralyzing abundance of choices. This Review intends to guide users through the process of applying CRISPR technology to their biological problems of interest, especially in the context of discovering gene function at scale.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 2: Diversity of activities with Cas9.
Figure 1: Maintaining representation in pooled screens.
Figure 3: Design considerations for CRISPR-based knockout.

Similar content being viewed by others

References

  1. Genomes Project Consortium et al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

  2. Plenge, R. M. Disciplined approach to drug discovery and early development. Sci. Transl Med. 8, 349ps15 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Berns, K. et al. A large-scale RNAi screen in human cells identifies new components of the p53 pathway. Nature 428, 431–437 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Westbrook, T. F. et al. A genetic screen for candidate tumor suppressors identifies REST. Cell 121, 837–848 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Wang, T., Wei, J. J., Sabatini, D. M. & Lander, E. S. Genetic screens in human cells using the CRISPR-Cas9 system. Science 343, 80–84 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Koike-Yusa, H., Li, Y., Tan, E.-P., Velasco-Herrera, M. D. C. & Yusa, K. Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library. Nat. Biotechnol. 32, 267–273 (2014). References 5–7 provide the first examples of the use of CRISPR technology for large-scale screens in mammalian cells.

    Article  CAS  PubMed  Google Scholar 

  8. Luo, B. et al. Highly parallel identification of essential genes in cancer cells. Proc. Natl Acad. Sci. USA 105, 20380–20385 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Wang, T., Lander, E. S. & Sabatini, D. M. Viral packaging and cell culture for CRISPR-based screens. Cold Spring Harb. Protoc. 2016, db.prot090811 (2016).

    Article  Google Scholar 

  10. Hartenian, E. & Doench, J. G. Genetic screens and functional genomics using CRISPR/Cas9 technology. FEBS J. 282, 1383–1393 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Wang, T., Lander, E. S. & Sabatini, D. M. Single guide RNA library design and construction. Cold Spring Harb. Protoc. 2016, db.prot090803 (2016).

    Article  Google Scholar 

  12. Tan, J. & Martin, S. E. Validation of synthetic CRISPR reagents as a tool for arrayed functional genomic screening. PLoS ONE 11, e0168968 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Esvelt, K. M. et al. Orthogonal Cas9 proteins for RNA-guided gene regulation and editing. Nat. Methods 10, 1116–1121 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hou, Z. et al. Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis. Proc. Natl Acad. Sci. USA 110, 15644–15649 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shmakov, S. et al. Discovery and functional characterization of diverse class 2 CRISPR-Cas systems. Mol. Cell 60, 385–397 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ran, F. A. et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520, 186–191 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zetsche, B. et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163, 759–771 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mohanraju, P. et al. Diverse evolutionary roots and mechanistic variations of the CRISPR-Cas systems. Science 353, aad5147 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Doench, J. G. et al. Rational design of highly active sgRNAs for CRISPR-Cas9-mediated gene inactivation. Nat. Biotechnol. 32, 1262–1267 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cheung, H. W. et al. Systematic investigation of genetic vulnerabilities across cancer cell lines reveals lineage-specific dependencies in ovarian cancer. Proc. Natl Acad. Sci. USA 108, 12372–12377 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Wang, T. et al. Gene essentiality profiling reveals gene networks and synthetic lethal interactions with oncogenic Ras. Cell 168, 890–903.e15 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tzelepis, K. et al. A CRISPR dropout screen identifies genetic vulnerabilities and therapeutic targets in acute myeloid leukemia. Cell Rep. 17, 1193–1205 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kryukov, G. V. et al. MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells. Science 351, 1214–1218 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Tsherniak, A. et al. Defining a cancer dependency map. Cell 170, 564–576.e16 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. McDonald, E. R. et al. Project DRIVE: a compendium of cancer dependencies and synthetic lethal relationships uncovered by large-scale, deep RNAi screening. Cell 170, 577–592.e10 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Hart, T. et al. High-resolution CRISPR screens reveal fitness genes and genotype-specific cancer liabilities. Cell 163, 1515–1526 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Arroyo, J. D. et al. A genome-wide CRISPR death screen identifies genes essential for oxidative phosphorylation. Cell Metab. 24, 875–885 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zwang, Y. et al. Synergistic interactions with PI3K inhibition that induce apoptosis. eLife 6, e24523 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Rotem, A. et al. Alternative to the soft-agar assay that permits high-throughput drug and genetic screens for cellular transformation. Proc. Natl Acad. Sci. USA 112, 5708–5713 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Jain, I. H. et al. Hypoxia as a therapy for mitochondrial disease. Science 352, 54–61 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, B. et al. ATXN1L, CIC, and ETS transcription factors modulate sensitivity to MAPK pathway inhibition. Cell Rep. 18, 1543–1557 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Krall, E. B. et al. KEAP1 loss modulates sensitivity to kinase targeted therapy in lung cancer. eLife 6, e18970 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Vecchione, L. et al. A vulnerability of a subset of colon cancers with potential clinical utility. Cell 165, 317–330 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Johannessen, C. M. et al. A melanocyte lineage program confers resistance to MAP kinase pathway inhibition. Nature 504, 138–142 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Orchard, R. C. et al. Discovery of a proteinaceous cellular receptor for a norovirus. Science 353, 933–936 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ma, H. et al. A CRISPR-based screen identifies genes essential for West-Nile-virus-induced cell death. Cell Rep. 12, 673–683 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Blondel, C. J. et al. CRISPR/Cas9 screens reveal requirements for host cell sulfation and fucosylation in bacterial type III secretion system-mediated cytotoxicity. Cell Host Microbe 20, 226–237 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang, R. et al. A CRISPR screen defines a signal peptide processing pathway required by flaviviruses. Nature 535, 164–168 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Marceau, C. D. et al. Genetic dissection of Flaviviridae host factors through genome-scale CRISPR screens. Nature 535, 159–163 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Deans, R. M. et al. Parallel shRNA and CRISPR-Cas9 screens enable antiviral drug target identification. Nat. Chem. Biol. 12, 361–366 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ma, Y. et al. CRISPR/Cas9 screens reveal Epstein-Barr virus-transformed B cell host dependency factors. Cell Host Microbe 21, 580–591.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Parnas, O. et al. A genome-wide CRISPR screen in primary immune cells to dissect regulatory networks. Cell 162, 675–686 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Brockmann, M. et al. Genetic wiring maps of single-cell protein states reveal an off-switch for GPCR signalling. Nature 546, 307–311 (2017).

    Article  CAS  PubMed  Google Scholar 

  44. Park, R. J. et al. A genome-wide CRISPR screen identifies a restricted set of HIV host dependency factors. Nat. Genet. 49, 193–203 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Braun, C. J. et al. Versatile in vivo regulation of tumor phenotypes by dCas9-mediated transcriptional perturbation. Proc. Natl Acad. Sci. USA 113, E3892–E3900 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Beronja, S. et al. RNAi screens in mice identify physiological regulators of oncogenic growth. Nature 501, 185–190 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Schramek, D. et al. Direct in vivo RNAi screen unveils myosin IIa as a tumor suppressor of squamous cell carcinomas. Science 343, 309–313 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Heckl, D. et al. Generation of mouse models of myeloid malignancy with combinatorial genetic lesions using CRISPR-Cas9 genome editing. Nat. Biotechnol. 32, 941–946 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Manguso, R. T. et al. In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature 547, 413–418 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Quintana, E. et al. Efficient tumour formation by single human melanoma cells. Nature 456, 593–598 (2008). This study provides a demonstration of the complex interplay between cell type and mouse background in the determination of xenograft efficiency.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Godec, J. et al. Inducible RNAi in vivo reveals that the transcription factor BATF is required to initiate but not maintain CD8+ T-cell effector differentiation. Proc. Natl Acad. Sci. USA 112, 512–517 (2015).

    Article  CAS  PubMed  Google Scholar 

  52. Bhang, H.-E. C. et al. Studying clonal dynamics in response to cancer therapy using high-complexity barcoding. Nat. Med. 21, 440–448 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Dixit, A. et al. Perturb-Seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell 167, 1853–1866.e17 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Adamson, B. et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell 167, 1867–1882.e21 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Datlinger, P. et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat. Methods 14, 297–301 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Jaitin, D. A. et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell 167, 1883–1896.e15 (2016). References 53–56 combine CRISPR screens with single-cell RNA sequencing readouts.

    Article  CAS  PubMed  Google Scholar 

  57. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Jinek, M. et al. RNA-programmed genome editing in human cells. eLife 2, e00471 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Mali, P. et al. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat. Biotechnol. 31, 833–838 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gilbert, L. A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014). This study presents the first use of dCas9 for genetic screens in mammalian cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2015).

    Article  CAS  PubMed  Google Scholar 

  65. Rajagopal, N. et al. High-throughput mapping of regulatory DNA. Nat. Biotechnol. 34, 167–174 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Vojta, A. et al. Repurposing the CRISPR-Cas9 system for targeted DNA methylation. Nucleic Acids Res. 44, 5615–5628 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Choudhury, S. R., Cui, Y., Lubecka, K., Stefanska, B. & Irudayaraj, J. CRISPR-dCas9 mediated TET1 targeting for selective DNA demethylation at BRCA1 promoter. Oncotarget 7, 46545–46556 (2016).

    PubMed  PubMed Central  Google Scholar 

  68. Hilton, I. B. et al. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat. Biotechnol. 33, 510–517 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kearns, N. A. et al. Functional annotation of native enhancers with a Cas9–histone demethylase fusion. Nat. Methods 12, 401–403 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Kwon, D. Y., Zhao, Y.-T., Lamonica, J. M. & Zhou, Z. Locus-specific histone deacetylation using a synthetic CRISPR-Cas9-based HDAC. Nat. Commun. 8, 15315 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Polstein, L. R. et al. Genome-wide specificity of DNA binding, gene regulation, and chromatin remodeling by TALE- and CRISPR/Cas9-based transcriptional activators. Genome Res. 25, 1158–1169 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Thakore, P. I. et al. Highly specific epigenome editing by CRISPR-Cas9 repressors for silencing of distal regulatory elements. Nat. Methods 12, 1143–1149 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016). This study presents the development of 'Base Editor' Cas9, which enables specific nucleotide changes without the need for double-stranded DNA breaks and homology-directed repair.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Hess, G. T. et al. Directed evolution using dCas9-targeted somatic hypermutation in mammalian cells. Nat. Methods 13, 1036–1042 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ma, Y. et al. Targeted AID-mediated mutagenesis (TAM) enables efficient genomic diversification in mammalian cells. Nat. Methods 13, 1029–1035 (2016).

    Article  CAS  PubMed  Google Scholar 

  76. Kampmann, M. et al. Next-generation libraries for robust RNA interference-based genome-wide screens. Proc. Natl Acad. Sci. USA 112, E3384–E3391 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Smith, I. et al. Evaluation of RNAi and CRISPR technologies by large-scale gene expression profiling in the connectivity map. Cold Spring Harb. Lab. http://dx.doi.org/10.1101/147504 (2017).

  78. Morgens, D. W., Deans, R. M., Li, A. & Bassik, M. C. Systematic comparison of CRISPR/Cas9 and RNAi screens for essential genes. Nat. Biotechnol. 34, 634–636 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Evers, B. et al. CRISPR knockout screening outperforms shRNA and CRISPRi in identifying essential genes. Nat. Biotechnol. 34, 631–633 (2016).

    Article  CAS  PubMed  Google Scholar 

  80. Anderson, E. M. et al. Experimental validation of the importance of seed complement frequency to siRNA specificity. RNA 14, 853–861 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Singh, S. et al. Morphological profiles of RNAi-induced gene knockdown are highly reproducible but dominated by seed effects. PLoS ONE 10, e0131370 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Berger, A. H. et al. High-throughput phenotyping of lung cancer somatic mutations. Cancer Cell 30, 214–228 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Majithia, A. R. et al. Prospective functional classification of all possible missense variants in PPARG. Nat. Genet. 48, 1570–1575 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191 (2016). This study applies machine learning to sgRNA design.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Wang, T. et al. Identification and characterization of essential genes in the human genome. Science 350, 1096–1101 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Horlbeck, M. A. et al. Nucleosomes impede Cas9 access to DNA in vivo and in vitro. eLife 5, e12677 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Horlbeck, M. A. et al. Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation. eLife 5, e19760 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Read, A., Gao, S., Batchelor, E. & Luo, J. Flexible CRISPR library construction using parallel oligonucleotide retrieval. Nucleic Acids Res. 45, e101 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Hough, S. H. et al. Guide Picker is a comprehensive design tool for visualizing and selecting guides for CRISPR experiments. BMC Bioinformatics 18, 167 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Shi, J. et al. Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains. Nat. Biotechnol. 33, 661–667 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Munoz, D. M. et al. CRISPR screens provide a comprehensive assessment of cancer vulnerabilities but generate false-positive hits for highly amplified genomic regions. Cancer Discov. 6, 900–913 (2016).

    Article  CAS  PubMed  Google Scholar 

  92. Rodriguez, J. M. et al. APPRIS: annotation of principal and alternative splice isoforms. Nucleic Acids Res. 41, D110–D117 (2013).

    Article  CAS  PubMed  Google Scholar 

  93. Xu, H. et al. Sequence determinants of improved CRISPR sgRNA design. Genome Res. 25, 1147–1157 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Haeussler, M. et al. Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR. Genome Biol. 17, 148 (2016). This study presents a systematic comparison of on-target algorithms and off-target algorithms for designing sgRNAs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. van Overbeek, M. et al. DNA repair profiling reveals nonrandom outcomes at Cas9-mediated breaks. Mol. Cell 63, 633–646 (2016).

    Article  CAS  PubMed  Google Scholar 

  96. Bae, S., Kweon, J., Kim, H. S. & Kim, J.-S. Microhomology-based choice of Cas9 nuclease target sites. Nat. Methods 11, 705–706 (2014).

    Article  CAS  PubMed  Google Scholar 

  97. Tsai, S. Q. & Joung, J. K. Defining and improving the genome-wide specificities of CRISPR-Cas9 nucleases. Nat. Rev. Genet. 17, 300–312 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Listgarten, J. et al. Predicting off-target effects for end-to-end CRISPR guide design. Cold Spring Harb. Lab. http://dx.doi.org/10.1101/078253 (2017).

  99. Radzisheuskaya, A., Shlyueva, D., Müller, I. & Helin, K. Optimizing sgRNA position markedly improves the efficiency of CRISPR/dCas9-mediated transcriptional repression. Nucleic Acids Res. 44, e141 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Lizio, M. et al. Update of the FANTOM web resource: high resolution transcriptome of diverse cell types in mammals. Nucleic Acids Res. 45, D737–D743 (2017).

    Article  CAS  PubMed  Google Scholar 

  101. Graham, D. B. & Root, D. E. Resources for the design of CRISPR gene editing experiments. Genome Biol. 16, 260 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Heigwer, F., Kerr, G. & Boutros, M. E-CRISP: fast CRISPR target site identification. Nat. Methods 11, 122–123 (2014).

    Article  CAS  PubMed  Google Scholar 

  103. Meier, J. A., Zhang, F. & Sanjana, N. E. GUIDES: sgRNA design for loss-of-function screens. Nat. Methods 14, 831–832 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Bae, S., Park, J. & Kim, J.-S. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics 30, 1473–1475 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Fulco, C. P. et al. Systematic mapping of functional enhancer-promoter connections with CRISPR interference. Science 354, 769–773 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Canver, M. C. et al. Variant-aware saturating mutagenesis using multiple Cas9 nucleases identifies regulatory elements at trait-associated loci. Nat. Genet. 49, 625–634 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Canver, M. C. et al. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature 527, 192–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Liu, S. J. et al. CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells. Science 355, eaah7111 (2017).

    Article  CAS  Google Scholar 

  109. Winter, J. et al. caRpools: an R package for exploratory data analysis and documentation of pooled CRISPR/Cas9 screens. Bioinformatics 32, 632–634 (2016).

    Article  CAS  PubMed  Google Scholar 

  110. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Hart, T. et al. Evaluation and design of genome-wide CRISPR/SpCas9 knockout screens. G3 7, 2719–2727 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Hart, T., Brown, K. R., Sircoulomb, F., Rottapel, R. & Moffat, J. Measuring error rates in genomic perturbation screens: gold standards for human functional genomics. Mol. Syst. Biol. 10, 733 (2014). This study presents a systematic catalogue of essential and nonessential genes for benchmarking screen performance.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Donovan, K. F. et al. Creation of novel protein variants with CRISPR/Cas9-mediated mutagenesis: turning a screening by-product into a discovery tool. PLoS ONE 12, e0170445 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Ipsaro, J. J. et al. Rapid generation of drug-resistance alleles at endogenous loci using CRISPR-Cas9 indel mutagenesis. PLoS ONE 12, e0172177 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Aguirre, A. J. et al. Genomic copy number dictates a gene-independent cell response to CRISPR/Cas9 targeting. Cancer Discov. 6, 914–929 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Rosenbluh, J. et al. Complementary information derived from CRISPR Cas9 mediated gene deletion and suppression. Nat. Commun. 8, 15403 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Brinkman, E. K., Chen, T., Amendola, M. & van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Caskey, C. T. & Kruh, G. D. The HPRT locus. Cell 16, 1–9 (1979).

    Article  CAS  PubMed  Google Scholar 

  119. Sagi, I. et al. Derivation and differentiation of haploid human embryonic stem cells. Nature 532, 107–111 (2016).

    Article  CAS  PubMed  Google Scholar 

  120. Rauscher, B., Heigwer, F., Breinig, M., Winter, J. & Boutros, M. GenomeCRISPR — a database for high-throughput CRISPR/Cas9 screens. Nucleic Acids Res. 45, D679–D686 (2017).

    Article  CAS  PubMed  Google Scholar 

  121. Han, K. et al. Synergistic drug combinations for cancer identified in a CRISPR screen for pairwise genetic interactions. Nat. Biotechnol. 35, 463–474 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Wong, A. S. L. et al. Multiplexed barcoded CRISPR-Cas9 screening enabled by CombiGEM. Proc. Natl Acad. Sci. USA 113, 2544–2549 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Shen, J. P. et al. Combinatorial CRISPR–Cas9 screens for de novo mapping of genetic interactions. Nat. Methods 17, 10–19 (2017).

    Google Scholar 

  124. Bassik, M. C. et al. A systematic mammalian genetic interaction map reveals pathways underlying ricin susceptibility. Cell 152, 909–922 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Costanzo, M. et al. A global genetic interaction network maps a wiring diagram of cellular function. Science 353, aaf1420 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. van Leeuwen, J. et al. Exploring genetic suppression interactions on a global scale. Science 354, aag0839 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Lu, Q. et al. Applications of CRISPR genome editing technology in drug target identification and validation. Expert Opin. Drug Discov. 12, 541–552 (2017).

    Article  CAS  PubMed  Google Scholar 

  128. Schenone, M., Dancík, V., Wagner, B. K. & Clemons, P. A. Target identification and mechanism of action in chemical biology and drug discovery. Nat. Chem. Biol. 9, 232–240 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Chavez, A. et al. Comparison of Cas9 activators in multiple species. Nat. Methods 13, 563–567 (2016). This study systematically compares numerous CRISPRa approaches.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Liu, X. S. et al. Editing DNA methylation in the mammalian genome. Cell 167, 233–247.e17 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Laufer, B. I. & Singh, S. M. Strategies for precision modulation of gene expression by epigenome editing: an overview. Epigenet. Chromatin 8, 34 (2015).

    Article  CAS  Google Scholar 

  132. Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187–197 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The author thanks K. Donovan for assistance in manuscript preparation, M. Hegde for assistance with analysis and the entire Genetic Perturbation Platform (GPP) at the Broad Institute. For insightful discussions, the author thanks J. Arroyo, O. Parnas and Z. Tothova (Broad Institute); J. Listgarten and N. Fusi (Microsoft Research); C.Wilen and R. Orchard (Washington University); J. Klappenbach (Merck); L. Brody (Desktop Genetics); and M. Fan (Addgene). This work is dedicated to Francis Edward Sheehan.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to John G. Doench.

Ethics declarations

Competing interests

J.G.D. is a consultant for Tango Therapeutics.

Related links

PowerPoint slides

Supplementary information

Supplementary information S1 (box)

Description of Simulations from Figure 1 (DOC 29 kb)

Glossary

Confirmation bias

The tendency to focus on information that confirms a pre-existing belief to the exclusion of contradictory information. In genetic screens, this can manifest in choosing to follow up a gene that scores with marginal statistical significance in the primary screen, rather than focusing on the experimentally identified top hits.

Single-guide RNAs

(sgRNAs). The first CRISPR systems characterized in prokaryotes required two RNAs to program the Cas9 protein: a CRISPR RNA (crRNA) and a transactivating crRNA (tracrRNA). To simplify the system, these two independent RNAs can instead be merged into a single transcript, the sgRNA, which has practical benefits especially for ease of expression in mammalian cells.

Titre

The titre of a lentivirus is the number of infectious particles per unit of volume, and the ratio of lentiviral integrants to cells is the multiplicity of infection (MOI). Importantly, cells differ in their inherent infectivity, and thus the volume of virus that is sufficient to achieve a given infection efficiency in cell type A is not necessarily the same in cell type B.

Xenograft

The transplantation of cells from one species to another. Often, it involves introducing human cancer cells into a mouse model to study their behaviour in complex microenvironments that are difficult to model in cell culture. Mice with an active immune system will recognize foreign cells and clear them out; thus, such experiments must be performed in immunodeficient mice.

Paralogues

Two genes that are produced by a gene duplication event and that, owing to their shared sequence, may have the same or similar functions. Thus, loss of one of them is often insufficient to manifest a phenotype, as the other paralogue can compensate.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Doench, J. Am I ready for CRISPR? A user's guide to genetic screens. Nat Rev Genet 19, 67–80 (2018). https://doi.org/10.1038/nrg.2017.97

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg.2017.97

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research