1932

Abstract

High-order interactions among components of interconnected genetic networks regulate complex functions in biological systems, but deciphering these interactions is challenging. New strategies are emerging to decode these combinatorial genetic interactions across a wide range of organisms. Here, we review advances in multiplexed and combinatorial genetic perturbation technologies and high-throughput profiling platforms that are enabling the systematic dissection of complex genetic networks. These rapidly evolving technologies are being harnessed to probe combinatorial gene functions in functional genomics studies and have the potential to advance our understanding of how genetic networks regulate sophisticated biological phenotypes, to generate novel therapeutic strategies, and to enable the engineering of complex artificial gene networks.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-genet-120215-034902
2016-11-23
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/genet/50/1/annurev-genet-120215-034902.html?itemId=/content/journals/10.1146/annurev-genet-120215-034902&mimeType=html&fmt=ahah

Literature Cited

  1. Abujarour R, Efe J, Ding S. 1.  2010. Genome-wide gain-of-function screen identifies novel regulators of pluripotency. Stem Cells 28:91487–97 [Google Scholar]
  2. Amendola M, Venneri MA, Biffi A, Vigna E, Naldini L. 2.  2005. Coordinate dual-gene transgenesis by lentiviral vectors carrying synthetic bidirectional promoters. Nat. Biotechnol. 23:1108–16 [Google Scholar]
  3. Askou AL, Aagaard L, Kostic C, Arsenijevic Y, Hollensen AK. 3.  et al. 2015. Multigenic lentiviral vectors for combined and tissue-specific expression of miRNA- and protein-based antiangiogenic factors. Mol. Ther. Methods Clin. Dev. 2:14064 [Google Scholar]
  4. Atsumi S, Higashide W, Liao JC. 4.  2009. Direct photosynthetic recycling of carbon dioxide to isobutyraldehyde. Nat. Biotechnol. 27:121177–80 [Google Scholar]
  5. Bartel DP. 5.  2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–97 [Google Scholar]
  6. Baryshnikova A, Costanzo M, Myers CL, Andrews B, Boone C. 6.  2013. Genetic interaction networks: toward an understanding of heritability. Annu. Rev. Genom. Hum. Genet. 14:111–33 [Google Scholar]
  7. Bassik MC, Kampmann M, Lebbink RJ, Wang S, Hein MY. 7.  et al. 2013. A systematic mammalian genetic interaction map reveals pathways underlying ricin susceptibility. Cell 152:909–22 [Google Scholar]
  8. Beerli RR, Segal DJ, Dreier B, Barbas CF III. 8.  1998. Toward controlling gene expression at will: specific regulation of the erbB-2/HER-2 promoter by using polydactyl zinc finger proteins constructed from modular building blocks. PNAS 95:14628–33 [Google Scholar]
  9. Bendall SC, Simonds EF, Qiu P, Amir ED, Krutzik PO. 9.  et al. 2011. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 332:6030687–96 [Google Scholar]
  10. Bernstein DL, Le Lay JE, Ruano EG, Kaestner KH. 10.  2015. TALE-mediated epigenetic suppression of CDKN2A increases replication in human fibroblasts. J. Clin. Investig. 125:51998–2006 [Google Scholar]
  11. Beronj S, Janki P, Heller E, Lien W-H, Keyes B. 11.  et al. 2013. RNAi screens in mice identify physiological regulators of oncogenic growth. Nature 501:185–90 [Google Scholar]
  12. Boutros M, Ahringer J. 12.  2008. The art and design of genetic screens: RNA interference. Nat. Rev. Genet. 9:7554–66 [Google Scholar]
  13. Broccoli V, Caiazzo M, Dell'Anno MT. 13.  2011. Setting a highway for converting skin into neurons. J. Mol. Cell Biol. 3:322–23 [Google Scholar]
  14. Buehler E, Chen YC, Martin S. 14.  2012. C911: a bench-level control for sequence specific siRNA off-target effects. PLOS ONE 7:12e51942 [Google Scholar]
  15. Busino L, Chiesa M, Draetta GF, Donzelli M. 15.  2004. Cdc25A phosphatase: combinatorial phosphorylation, ubiquitylation and proteolysis. Oncogene 23:112050–56 [Google Scholar]
  16. Butland G, Babu M, Díaz-Mejía JJ, Bohdana F, Phanse S. 16.  et al. 2008. eSGA: E. coli synthetic genetic array analysis. Nat. Methods 5:789–95 [Google Scholar]
  17. Cahan P, Li H, Morris SA, Lummertz da Rocha E, Daley GQ. 17.  et al. 2014. CellNet: network biology applied to stem cell engineering. Cell 158:4903–15 [Google Scholar]
  18. Calnan DR, Brunet A. 18.  2008. The FoxO code. Oncogene 27:162276–88 [Google Scholar]
  19. Canver MC, Smith EC, Sher F, Pinello L, Sanjana NE. 19.  et al. 2015. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature 527:7577192–97 [Google Scholar]
  20. Carpenter AE, Sabatini DM. 20.  2004. Systematic genome-wide screens of gene function. Nat. Rev. Genet. 5:111–22 [Google Scholar]
  21. Carroll D. 21.  2014. Genome engineering with targetable nucleases. Annu. Rev. Biochem. 83:409–39 [Google Scholar]
  22. Chavez A, Scheiman J, Vora S, Pruitt BW, Tuttle M. 22.  et al. 2015. Highly efficient Cas9-mediated transcriptional programming. Nat. Methods 12:4326–28 [Google Scholar]
  23. Cheng AA, Ding H, Lu TK. 23.  2014. Enhanced killing of antibiotic-resistant bacteria enabled by massively parallel combinatorial genetics. PNAS 111:12462–67 [Google Scholar]
  24. Cheng AA, Lu TK. 24.  2012. Synthetic biology: an emerging engineering discipline. Annu. Rev. Biomed. Eng. 14:155–78 [Google Scholar]
  25. Cheng AW, Wang H, Yang H, Shi L, Katz Y. 25.  et al. 2013. Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res. 23:1163–71 [Google Scholar]
  26. Coba MP, Pocklington AJ, Collins MO, Kopanitsa MV, Uren RT. 26.  et al. 2009. Neurotransmitters drive combinatorial multistate postsynaptic density networks. Sci. Signal. 2:68ra19 [Google Scholar]
  27. Cong L, Zhou R, Kuo Y, Cunniff M, Zhang F. 27.  2012. Comprehensive interrogation of natural TALE DNA-binding modules and transcriptional repressor domains. Nat. Commun. 3:968 [Google Scholar]
  28. Cordell HJ. 28.  2009. Detecting gene-gene interactions that underlie human diseases. Nat. Rev. Genet. 10:6392–404 [Google Scholar]
  29. Dahlman JE, Abudayyeh OO, Joung J, Gootenberg JS, Zhang F, Konermann S. 29.  2015. Orthogonal gene knockout and activation with a catalytically active Cas9 nuclease. Nat. Biotechnol. 33:111159–61 [Google Scholar]
  30. Delay C, Calon F, Mathews P, Hebert S. 30.  2011. Alzheimer-specific variants in the 3′ UTR of Amyloid precursor protein affect microRNA function. Mol. Neurodegener. 6:70 [Google Scholar]
  31. De Silva S, Mastrangelo MA, Lotta LT, Burris CA, Federoff HJ, Bowers WJ. 31.  2010. Extending the transposable payload limit of Sleeping Beauty (SB) using the Herpes Simplex Virus (HSV)/SB amplicon-vector platform. Gene Ther. 17:3424–31 [Google Scholar]
  32. Dietzl G, Chen D, Schnorrer F, Su K-C, Barinova Y. 32.  et al. 2007. A genome-wide transgenic RNAi library for conditional gene inactivation in Drosophila. Nature 448:7150151–56 [Google Scholar]
  33. Dixon SJ, Costanzo M, Baryshnikova A, Andrews B, Boone C. 33.  2009. Systematic mapping of genetic interaction networks. Annu. Rev. Genet. 43:601–25 [Google Scholar]
  34. Doench JG, Fusi N, Sullender M, Hegde M, Vaimberg EW. 34.  et al. 2016. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34:184–91 [Google Scholar]
  35. Doench JG, Hartenian E, Graham DB, Tothova Z, Hegde M. 35.  et al. 2014. Rational design of highly active sgRNAs for CRISPR-Cas9-mediated gene inactivation. Nat. Biotechnol. 32:121–13 [Google Scholar]
  36. Doudna JA, Charpentier E. 36.  2014. The new frontier of genome engineering with CRISPR-Cas9. Science 346:62131258096 [Google Scholar]
  37. Drees BL, Thorsson V, Carter GW, Rives AW, Raymond MZ. 37.  et al. 2005. Derivation of genetic interaction networks from quantitative phenotype data. Genome Biol. 6:R38 [Google Scholar]
  38. Duportet X, Wroblewska L, Guye P, Li Y, Eyquem J. 38.  et al. 2014. A platform for rapid prototyping of synthetic gene networks in mammalian cells. Nucleic Acids Res. 42:2113440–51 [Google Scholar]
  39. Ebert MS, Neilson JR, Sharp PA. 39.  2007. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat. Methods 4:721–26 [Google Scholar]
  40. Echeverri CJ, Perrimon N. 40.  2006. High-throughput RNAi screening in cultured cells: a user's guide. Nat. Rev. Genet. 7:5373–84 [Google Scholar]
  41. Esvelt KM, Mali P, Braff JL, Moosburner M, Yaung SJ, Church GM. 41.  2013. Orthogonal Cas9 proteins for RNA-guided gene regulation and editing. Nat. Methods 10:111116–21 [Google Scholar]
  42. Farzadfard F, Lu TK. 42.  2014. Genomically encoded analog memory with precise in vivo DNA writing in living cell populations. Science 346:62111256272 [Google Scholar]
  43. Farzadfard F, Perli SD, Lu TK. 43.  2013. Tunable and multifunctional eukaryotic transcription factors based on CRISPR/Cas. ACS Synth. Biol. 2:604–13 [Google Scholar]
  44. Félix M-A, Barkoulas M. 44.  2015. Pervasive robustness in biological systems. Nat. Rev. Genet. 16:8483–96 [Google Scholar]
  45. Fellmann C, Zuber J, McJunkin K, Chang K, Malone CD. 45.  et al. 2011. Functional identification of optimized RNAi triggers using a massively parallel sensor assay. Mol. Cell 41:6733–46 [Google Scholar]
  46. Ferraris L, Stewart AP, Kang J, DeSimone AM, Gemberling M. 46.  et al. 2011. Combinatorial binding of transcription factors in the pluripotency control regions of the genome. Genome Res. 21:71055–64 [Google Scholar]
  47. Findlay GM, Boyle EA, Hause RJ, Klein JC, Shendure J. 47.  2014. Saturation editing of genomic regions by multiplex homology-directed repair. Nature 513:7516120–23 [Google Scholar]
  48. Gaj T, Gersbach CA, Barbas CF. 48.  2013. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 31:7397–405 [Google Scholar]
  49. Garg A, Lohmueller JJ, Silver PA, Armel TZ. 49.  2012. Engineering synthetic TAL effectors with orthogonal target sites. Nucleic Acids Res. 40:157584–95 [Google Scholar]
  50. Gilbert LA, Horlbeck MA, Adamson B, Villalta JE, Chen Y. 50.  et al. 2014. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159:647–61 [Google Scholar]
  51. Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA. 51.  et al. 2013. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154:442–51 [Google Scholar]
  52. Golding MC, Mann MRW. 52.  2011. A bidirectional promoter architecture enhances lentiviral transgenesis in embryonic and extraembryonic stem cells. Gene Ther. 18:8817–26 [Google Scholar]
  53. Gompel N, Prud'homme B, Wittkopp PJ, Kassner VA, Carrol SB. 53.  2005. Chance caught on the wing: cis-regulatory evolution and the origin of pigment patterns in Drosophila. Nature 433:481–87 [Google Scholar]
  54. González F, Zhu Z, Shi ZD, Lelli K, Verma N. 54.  et al. 2014. An iCRISPR platform for rapid, multiplexable, and inducible genome editing in human pluripotent stem cells. Cell Stem Cell 15:2215–26 [Google Scholar]
  55. Greber D, Fussenegger M. 55.  2007. Multi-gene engineering: simultaneous expression and knockdown of six genes off a single platform. Biotechnol. Bioeng. 96:5821–34 [Google Scholar]
  56. Grindley NDF, Whiteson KL, Rice PA. 56.  2006. Mechanisms of site-specific recombination. Annu. Rev. Biochem. 75:567–605 [Google Scholar]
  57. Guttman M, Donaghey J, Carey BW, Garber M, Grenier JK. 57.  et al. 2011. LincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 477:295–300 [Google Scholar]
  58. Haber JE, Braberg H, Wu Q, Alexander R, Haase J. 58.  et al. 2013. Systematic triple-mutant analysis uncovers functional connectivity between pathways involved in chromosome regulation. Cell Rep. 3:62168–78 [Google Scholar]
  59. Haimovich AD, Muir P, Isaacs FJ. 59.  2015. Genomes by design. Nat. Rev. Genet. 16:9501–16 [Google Scholar]
  60. Hausser J, Zavolan M. 60.  2014. Identification and consequences of miRNA-target interactions: beyond repression of gene expression. Nat. Rev. Genet. 15:9599–612 [Google Scholar]
  61. Heckl D, Kowalczyk MS, Yudovich D, Belizaire R, Puram RV. 61.  et al. 2014. Generation of mouse models of myeloid malignancy with combinatorial genetic lesions using CRISPR-Cas9 genome editing. Nat. Biotechnol. 32:9941–46 [Google Scholar]
  62. Hilton I, D'Ippolito AM, Vockley CM, Thakore PI, Crawford GE. 62.  et al. 2015. Epigenome editing by a CRISPR-Cas9–based acetyltransferase activates genes from promoters and enhancers. Nat. Biotechnol. 33:5510–17 [Google Scholar]
  63. Hon LS, Zhang Z. 63.  2007. The roles of binding site arrangement and combinatorial targeting in microRNA repression of gene expression. Genome Biol. 8:8R166 [Google Scholar]
  64. Hotta A, Yamanaka S. 64.  2015. From genomics to gene therapy: induced pluripotent stem cells meet genome editing. Annu. Rev. Genet. 49:47–70 [Google Scholar]
  65. Hsu PD, Lander ES, Zhang F. 65.  2014. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157:1262–78 [Google Scholar]
  66. Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S. 66.  et al. 2013. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31:827–32 [Google Scholar]
  67. Huang P, Zhang L, Gao Y, He Z, Yao D. 67.  et al. 2014. Direct reprogramming of human fibroblasts to functional and expandable hepatocytes. Cell Stem Cell 14:3370–84 [Google Scholar]
  68. Hutchison CA III, Chuang R-Y, Noskov VN, Assad-Garcia N, Deerinck TJ. 68.  et al. 2016. Design and synthesis of a minimal bacterial genome. Science 351:6280aad6253 [Google Scholar]
  69. Ibrahimi A, Vande Velde G, Reumers V, Toelen J, Thiry J. 69.  et al. 2009. Highly efficient multicistronic lentiviral vectors with peptide 2A sequences. Hum. Gene Ther. 20:8845–60 [Google Scholar]
  70. Ivey KN, Srivastava D. 70.  2010. MicroRNAs as regulators of differentiation and cell fate decisions. Cell Stem Cell 7:36–41 [Google Scholar]
  71. Jao L-E, Wente SR, Chen W. 71.  2013. Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system. PNAS 110:3413904–9 [Google Scholar]
  72. Jiang W, Bikard D, Cox D, Zhang F, Marraffini L. 72.  2013. CRISPR-assisted editing of bacterial genomes. Nat. Biotechnol. 31:3233–39 [Google Scholar]
  73. Jolma A, Kivioja T, Toivonen J, Cheng L, Wei G. 73.  et al. 2010. Multiplexed massively parallel SELEX for characterization of human transcription factor binding specificities. Genome Res. 20:6861–73 [Google Scholar]
  74. Kamath RS, Fraser AG, Dong Y, Poulin G, Durbin R. 74.  et al. 2003. Systematic functional analysis of the Caenorhabditis elegans genome using RNAi. Nature 421:6920231–37 [Google Scholar]
  75. Kampmann M, Horlbeck MA, Chen Y, Tsai JC, Bassik MC. 75.  et al. 2015. Next-generation libraries for robust RNA interference-based genome-wide screens. PNAS 112:26E3384–91 [Google Scholar]
  76. Kearns N, Pham H, Tabak B, Genga R, Silverstein N. 76.  et al. 2015. Functional annotation of native enhancers with a Cas9-histone demethylase fusion. Nat. Methods 12:5401–3 [Google Scholar]
  77. Khalil AS, Collins JJ. 77.  2010. Synthetic biology: applications come of age. Nat. Rev. Genet. 11:5367–79 [Google Scholar]
  78. Khalil AS, Lu TK, Bashor CJ, Ramirez CL, Pyenson NC. 78.  et al. 2012. A synthetic biology framework for programming eukaryotic transcription functions. Cell 150:3647–58 [Google Scholar]
  79. Kiani S, Chavez A, Tuttle M, Hall RN, Chari R. 79.  et al. 2015. Cas9 gRNA engineering for genome editing, activation and repression. Nat. Methods 12:111051–54 [Google Scholar]
  80. Kim H, Kim J-S. 80.  2014. A guide to genome engineering with programmable nucleases. Nat. Rev. Genet. 15:5321–34 [Google Scholar]
  81. Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT. 81.  et al. 2016. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529:490–95 [Google Scholar]
  82. Kleinstiver BP, Prew MS, Tsai SQ, Nguyen NT, Topkar VV. 82.  et al. 2015. Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition. Nat. Biotechnol. 33:121293–98 [Google Scholar]
  83. Kleinstiver BP, Prew MS, Tsai SQ, Topkar VV, Nguyen NT. 83.  et al. 2015. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 523:7561481–85 [Google Scholar]
  84. Kling J. 84.  2015. Cytometry: measure for measure. Nature 518:439–43 [Google Scholar]
  85. Knott SRV, Maceli AR, Erard N, Chang K, Marran K. 85.  et al. 2014. A computational algorithm to predict shRNA potency. Mol. Cell 56:6796–807 [Google Scholar]
  86. Konermann S, Brigham MD, Trevino AE, Joung J, Abudayyeh OO. 86.  et al. 2014. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517:7536583–88 [Google Scholar]
  87. Korkmaz G, Lopes R, Ugalde AP, Nevedomskaya E, Han R. 87.  et al. 2016. Functional genetic screens for enhancer elements in the human genome using CRISPR-Cas9. Nat. Biotechnol. 34:2192–98 [Google Scholar]
  88. Krek A, Grün D, Poy MN, Wolf R, Rosenberg L. 88.  et al. 2005. Combinatorial microRNA target predictions. Nat. Genet. 37:495–500 [Google Scholar]
  89. Kwok R. 89.  2010. Five hard truths for synthetic biology. Nature 463:7279288–90 [Google Scholar]
  90. Lander ES. 90.  2011. Initial impact of the sequencing of the human genome. Nature 470:187–97 [Google Scholar]
  91. Laufer C, Fischer B, Billmann M, Huber W, Boutros M. 91.  2013. Mapping genetic interactions in human cancer cells with RNAi and multiparametric phenotyping. Nat. Methods 10:5427–31 [Google Scholar]
  92. Lewis NE, Nagarajan H, Palsson . 92.  2012. Constraining the metabolic genotype-phenotype relationship using a phylogeny of in silico methods. Nat. Rev. Microbiol. 10:4291–305 [Google Scholar]
  93. Liberali P, Snijder B, Pelkmans L. 93.  2014. Single-cell and multivariate approaches in genetic perturbation screens. Nat. Rev. Genet. 16:118–32 [Google Scholar]
  94. Lim C, Allada R. 94.  2013. Emerging roles for post-transcriptional regulation in circadian clocks. Nat. Neurosci. 16:111544–50 [Google Scholar]
  95. Lin Y, Sohn CH, Dalal CK, Cai L, Elowitz MB. 95.  2015. Combinatorial gene regulation by modulation of relative pulse timing. Nature 527:757654–58 [Google Scholar]
  96. Liu W, Ma Q, Wong K, Li W, Ohgi K. 96.  et al. 2013. Brd4 and JMJD6-associated anti-pause enhancers in regulation of transcriptional pause release. Cell 155:71581–95 [Google Scholar]
  97. Londin E, Lohera P, Telonis AG, Quann K, Clark P. 97.  et al. 2015. Analysis of 13 cell types reveals evidence for the expression of numerous novel primate- and tissue-specific microRNAs. PNAS 112:10E1106–15 [Google Scholar]
  98. Lu R, Neff NF, Quake SR, Weissman IL. 98.  2011. Tracking single hematopoietic stem cells in vivo using high-throughput sequencing in conjunction with viral genetic barcoding. Nat. Biotechnol. 29:10928–33 [Google Scholar]
  99. Macneil LT, Walhout AJM. 99.  2011. Gene regulatory networks and the role of robustness and stochasticity in the control of gene expression. Genome Res. 21:5645–57 [Google Scholar]
  100. Maeder ML, Linder SJ, Cascio VM, Fu Y, Ho QH, Joung JK. 100.  2013. CRISPR RNA-guided activation of endogenous human genes. Nat. Methods 10:977–79 [Google Scholar]
  101. Maeder ML, Linder SJ, Reyon D, Angstman JF, Fu Y. 101.  et al. 2013. Robust, synergistic regulation of human gene expression using TALE activators. Nat. Methods 10:3243–45 [Google Scholar]
  102. Mali P, Aach J, Stranges PB, Esvelt KM, Moosburner M. 102.  et al. 2013. Cas9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat. Biotechnol. 31:833–38 [Google Scholar]
  103. Marder E, Goaillard J-M. 103.  2006. Variability, compensation and homeostasis in neuron and network function. Nat. Rev. Neurosci. 7:563–74 [Google Scholar]
  104. Maresca M, Lin VG, Guo N, Yang Y. 104.  2013. Obligate ligation-gated recombination (obligare): custom-designed nuclease-mediated targeted integration through nonhomologous end joining. Genome Res. 23:3539–46 [Google Scholar]
  105. Maston GA, Evans SK, Green MR. 105.  2006. Transcriptional regulatory elements in the human genome. Annu. Rev. Genom. Hum. Genet. 7:29–59 [Google Scholar]
  106. Matano M, Date S, Shimokawa M, Takano A, Fujii M. 106.  et al. 2015. Modeling colorectal cancer using CRISPR-Cas9–mediated engineering of human intestinal organoids. Nat. Med. 21:3256–62 [Google Scholar]
  107. Medema MH, van Raaphorst R, Takano E, Breitling R. 107.  2012. Computational tools for the synthetic design of biochemical pathways. Nat. Rev. Microbiol. 10:3191–202 [Google Scholar]
  108. Moehle EA, Rock JM, Lee Y-L, Jouvenot Y, DeKelver RC. 108.  et al. 2007. Targeted gene addition into a specified location in the human genome using designed zinc finger nucleases. PNAS 104:93055–60 [Google Scholar]
  109. Mohr S, Bakal C, Perrimon N. 109.  2010. Genomic screening with RNAi: results and challenges. Annu. Rev. Biochem. 79:37–64 [Google Scholar]
  110. Mohr SE, Smith JA, Shamu CE, Neumüller RA. 110.  2014. RNAi screening comes of age: improved techniques and complementary approaches. Nat. Rev. Mol. Cell Biol. 15:591–600 [Google Scholar]
  111. Mullokandov G, Baccarini A, Ruzo A, Jayaprakash AD, Tung N. 111.  et al. 2012. High-throughput assessment of microRNA activity and function using microRNA sensor and decoy libraries. Nat. Methods 9:8840–46 [Google Scholar]
  112. Nussinov R, Tsai CJ, Xin F, Radivojac P. 112.  2012. Allosteric post-translational modification codes. Trends Biochem. Sci. 37:10447–55 [Google Scholar]
  113. Pasquinelli AE. 113.  2012. MicroRNAs and their targets: recognition, regulation and an emerging reciprocal relationship. Nat. Rev. Genet. 13:4271–82 [Google Scholar]
  114. Perez-Pinera P, Kocak DD, Vockley CM, Adler AF, Kabadi AM. 114.  et al. 2013. RNA-guided gene activation by CRISPR-Cas9–based transcription factors. Nat. Methods 10:973–76 [Google Scholar]
  115. Perli SD, Cui CH, Lu TK. 115.  2016. Continuous genetic recording with self-targeting CRISPR-Cas in human cells. Science doi: 10.1126/science.aag0511
  116. Phillips PC. 116.  2008. Epistasis: the essential role of gene interactions in the structure and evolution of genetic systems. Nat. Rev. Genet. 9:11855–67 [Google Scholar]
  117. Pilpel Y, Sudarsanam P, Church GM. 117.  2001. Identifying regulatory networks by combinatorial analysis of promoter elements. Nat. Genet. 29:153–59 [Google Scholar]
  118. Pritsker M, Ford NR, Jenq HT, Lemischka IR. 118.  2006. Genomewide gain-of-function genetic screen identifies functionally active genes in mouse embryonic stem cells. PNAS 103:186946–51 [Google Scholar]
  119. Purcell O, Lu TK. 119.  2014. Synthetic analog and digital circuits for cellular computation and memory. Curr. Opin. Biotechnol. 29:146–55 [Google Scholar]
  120. Purnick PEM, Weiss R. 120.  2009. The second wave of synthetic biology: from modules to systems. Nat. Rev. Mol. Cell Biol. 10:6410–22 [Google Scholar]
  121. Qian L, Srivastava D. 121.  2013. Direct cardiac reprogramming: from developmental biology to cardiac regeneration. Circ. Res. 113:7915–21 [Google Scholar]
  122. Ramirez CL, Foley JE, Wright DA, Müller-Lerch F, Rahman SH. 122.  et al. 2008. Unexpected failure rates for modular assembly of engineered zinc fingers. Nat. Methods 5:374–75 [Google Scholar]
  123. Rackham OJ, Firas J, Fang H, Oates ME, Holmes ML. 123.  et al. 2016. A predictive computational framework for direct reprogramming between human cell types. Nat. Genet. 48:331–35 [Google Scholar]
  124. Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS. 124.  et al. 2015. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520:7546186–90 [Google Scholar]
  125. Ravasi T, Suzuki H, Cannistraci CV, Katayama S, Bajic VB. 125.  et al. 2010. An atlas of combinatorial transcriptional regulation in mouse and man. Cell 140:5744–52 [Google Scholar]
  126. Roguev A, Talbot D, Negri GL, Shales M, Cagney G. 126.  et al. 2013. Quantitative genetic-interaction mapping in mammalian cells. Nat. Methods 10:5432–37 [Google Scholar]
  127. Ronda C, Pedersen LE, Sommer MOA, Nielsen AT. 127.  2016. CRMAGE: CRISPR optimized mage recombineering. Sci. Rep. 6:19452 [Google Scholar]
  128. Roquet N, Soleimany AP, Ferris AC, Aaronson S, Lu TK. 128.  2016. Synthetic recombinase-based state machines in living cells. Science 353:6297aad8559 [Google Scholar]
  129. Sakuma T, Barry MA, Ikeda Y. 129.  2012. Lentiviral vectors: basic to translational. Biochem. J. 443:3603–18 [Google Scholar]
  130. Sancho-Martinez I, Baek SH, Izpisua Belmonte JC. 130.  2012. Lineage conversion methodologies meet the reprogramming toolbox. Nat. Cell Biol. 14:9892–99 [Google Scholar]
  131. Sandoval NR, Kim JYH, Glebes TY, Reeder PJ, Aucoin HR. 131.  et al. 2012. Strategy for directing combinatorial genome engineering in Escherichia coli. PNAS 109:2610540–45 [Google Scholar]
  132. Sebban S, Buganim Y. 132.  2016. Nuclear reprogramming by defined factors: quantity versus quality. Trends Cell Biol. 26:65–75 [Google Scholar]
  133. Shalem O, Sanjana NE, Zhang F. 133.  2015. High-throughput functional genomics using CRISPR-Cas9. Nat. Rev. Genet. 16:299–311 [Google Scholar]
  134. Sharon E, Kalma Y, Sharp A, Raveh-Sadka T, Levo M. 134.  et al. 2012. Inferring gene regulatory logic from high-throughput measurements of thousands of systematically designed promoters. Nat. Biotechnol. 30:6521–30 [Google Scholar]
  135. Shechner DM, Hacisuleyman E, Younger ST, Rinn JL. 135.  2015. Multiplexable, locus-specific targeting of long RNAs with CRISPR-display. Nat. Methods 12:7664–70 [Google Scholar]
  136. Skarnes WC, Rosen B, West AP, Koutsourakis M, Bushell W. 136.  et al. 2011. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474:7351337–42 [Google Scholar]
  137. Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, Zhang F. 137.  2015. Rationally engineered Cas9 nucleases with improved specificity. Science 351:626884–88 [Google Scholar]
  138. Slusarczyk AL, Lin A, Weiss R. 138.  2012. Foundations for the design and implementation of synthetic genetic circuits. Nat. Rev. Genet. 13:6406–20 [Google Scholar]
  139. Smanski MJ, Bhatia S, Zhao D, Park Y, Woodruff LBA. 139.  et al. 2014. Functional optimization of gene clusters by combinatorial design and assembly. Nat. Biotechnol. 32:121241–49 [Google Scholar]
  140. Sopko R, Huang D, Preston N, Chua G, Papp B. 140.  et al. 2006. Mapping pathways and phenotypes by systematic gene overexpression. Mol. Cell 21:3319–30 [Google Scholar]
  141. Spitz F, Furlong EEM. 141.  2012. Transcription factors: from enhancer binding to developmental control. Nat. Rev. Genet. 13:9613–26 [Google Scholar]
  142. Stampfel G, Kazmar T, Frank O, Wienerroither S, Reiter F, Stark A. 142.  2015. Transcriptional regulators form diverse groups with context-dependent regulatory functions. Nature 528:7580147–51 [Google Scholar]
  143. Takahashi K, Yamanaka S. 143.  2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:4663–76 [Google Scholar]
  144. Tian J, Andreadis ST. 144.  2009. Independent and high-level dual-gene expression in adult stem-progenitor cells from a single lentiviral vector. Gene Ther. 16:7874–84 [Google Scholar]
  145. Tong AH, Evangelista M, Parsons AB, Xu H, Bader GD. 145.  et al. 2001. Systematic genetic analysis with ordered arrays of yeast deletion mutants. Science 294:55502364–68 [Google Scholar]
  146. Tong AHY, Lesage G, Bader GD, Ding H, Xu H. 146.  et al. 2004. Global mapping of the yeast genetic interaction network. Science 303:808–13 [Google Scholar]
  147. Typas A, Nichols RJ, Siegele DA, Shales M, Collins SR. 147.  et al. 2008. High-throughput, quantitative analyses of genetic interactions in E. coli. Nat. Methods 5:9781–87 [Google Scholar]
  148. Vaquerizas JM, Kummerfeld SK, Teichmann SA, Luscombe NM. 148.  2009. A census of human transcription factors: function, expression and evolution. Nat. Rev. Genet. 10:4252–63 [Google Scholar]
  149. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Südhof TC. 149.  et al. 2010. Direct conversion of fibroblasts to functional neurons by defined factors. Nature 463:1035–41 [Google Scholar]
  150. Vierbuchen T, Wernig M. 150.  2012. Molecular roadblocks for cellular reprogramming. Mol. Cell 47:827–38 [Google Scholar]
  151. Vizeacoumar FJ, Arnold R, Vizeacoumar FS, Chandrashekhar M, Buzina A. 151.  et al. 2013. A negative genetic interaction map in isogenic cancer cell lines reveals cancer cell vulnerabilities. Mol. Syst. Biol. 9:696 [Google Scholar]
  152. Voorhoeve PM, Le Sage C, Schrier M, Gilis AJM, Stoop H. 152.  et al. 2007. A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell 131:102–14 [Google Scholar]
  153. Wang B, Li K, Wang A, Reiser M, Saunders T. 153.  et al. 2015. Highly efficient CRISPR/HDR-mediated knock-in for mouse embryonic stem cells and zygotes. Biotechniques 59:4201–8 [Google Scholar]
  154. Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW. 154.  et al. 2013. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153:4910–18 [Google Scholar]
  155. Wang HH, Isaacs FJ, Carr PA, Sun ZZ, Xu G. 155.  et al. 2009. Programming cells by multiplex genome engineering and accelerated evolution. Nature 460:7257894–98 [Google Scholar]
  156. Wirth D, Gama-Norton L, Riemer P, Sandhu U, Schucht R, Hauser H. 156.  2007. Road to precision: recombinase-based targeting technologies for genome engineering. Curr. Opin. Biotechnol. 18:5411–19 [Google Scholar]
  157. Witzgall R, O'Leary E, Leaf A, Onaldi D, Bonventre JV. 157.  1994. The Krüppel-associated box-A (KRAB-A) domain of zinc finger proteins mediates transcriptional repression. PNAS 91:104514–18 [Google Scholar]
  158. Wolfe D, Craft AM, Cohen JB, Glorioso JC. 158.  2010. A herpes simplex virus vector system for expression of complex cellular cDNA libraries. J. Virol. 84:147360–68 [Google Scholar]
  159. Wong ASL, Choi GCG, Cheng AA, Purcell O, Lu TK. 159.  2015. Massively parallel high-order combinatorial genetics in human cells. Nat. Biotechnol. 33:952–61 [Google Scholar]
  160. Wong ASL, Choi GCG, Cui CH, Pregernig G, Milani P. 160.  et al. 2016. Multiplexed barcoded CRISPR-Cas9 screening enabled by CombiGEM. PNAS 113:92544–49 [Google Scholar]
  161. Yang X, Boehm JS, Yang X, Salehi-Ashtiani K, Hao T. 161.  et al. 2011. A public genome-scale lentiviral expression library of human ORFs. Nat. Methods 8:8659–61 [Google Scholar]
  162. Yim H, Haselbeck R, Niu W, Pujol-Baxley C, Burgard A. 162.  et al. 2011. Metabolic engineering of Escherichia coli for direct production of 1,4-butanediol. Nat. Chem. Biol. 7:7445–52 [Google Scholar]
  163. Yoo AS, Sun AX, Li L, Shcheglovitov A, Portman T. 163.  et al. 2011. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 476:228–31 [Google Scholar]
  164. Yu H, Tardivo L, Tam S, Weiner E, Gebreab F. 164.  et al. 2011. Next-generation sequencing to generate interactome datasets. Nat. Methods 8:478–80 [Google Scholar]
  165. Zalatan JG, Lee ME, Almeida R, Gilbert LA, Whitehead EH. 165.  et al. 2015. Engineering complex synthetic transcriptional programs with CRISPR RNA scaffolds. Cell 160:1–2339–50 [Google Scholar]
  166. Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS. 166.  et al. 2015. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163:3759–71 [Google Scholar]
  167. Zhang F, Cong L, Lodato S, Kosuri S, Church GM, Arlotta P. 167.  2011. Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription. Nat. Biotechnol. 29:2149–53 [Google Scholar]
  168. Zhu F, Gamboa M, Farruggio AP, Hippenmeyer S, Tasic B. 168.  et al. 2013. DICE, an efficient system for iterative genomic editing in human pluripotent stem cells. Nucleic Acids Res 42:e34 [Google Scholar]
  169. Zhu LJ. 169.  2015. Overview of guide RNA design tools for CRISPR-Cas9 genome editing technology. Front. Biol. 10:4289–96 [Google Scholar]
/content/journals/10.1146/annurev-genet-120215-034902
Loading
/content/journals/10.1146/annurev-genet-120215-034902
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error