Identification of potential drug targets for tuberous sclerosis complex by synthetic screens combining CRISPR-based knockouts with RNAi

Combining targeted CRISPR-mediated gene editing with an RNAi-mediated screen identifies candidate drug targets. Double screening for drug targets The tumor suppressors TSC1 and TSC2 form a complex that limits the activity of mTORC1, a multiprotein complex that couples nutrient availability to cell proliferation and growth. Individuals with mutations in either of the TSC-encoding genes develop tumors in various organs. Housden et al. used CRISPR to generate Drosophila cell lines that lacked either TSC1 or TSC2 and performed RNAi screens directed against kinase- and phosphatase-encoding genes in these cell lines to identify those genes that limited the growth of cells lacking either TSC1 or TSC2. Candidate genes that, when knocked down, suppressed the growth of both of the TSC1- and TSC2-deficient Drosophila cell lines (but not that of normal cells) were validated in TSC2-deficient human cells. Because these candidates are evolutionarily conserved, they are more likely to be bona fide drug targets, and this combination of techniques and multispecies screening could be used to identify drug targets for other diseases. The tuberous sclerosis complex (TSC) family of tumor suppressors, TSC1 and TSC2, function together in an evolutionarily conserved protein complex that is a point of convergence for major cell signaling pathways that regulate mTOR complex 1 (mTORC1). Mutation or aberrant inhibition of the TSC complex is common in various human tumor syndromes and cancers. The discovery of novel therapeutic strategies to selectively target cells with functional loss of this complex is therefore of clinical relevance to patients with nonmalignant TSC and those with sporadic cancers. We developed a CRISPR-based method to generate homogeneous mutant Drosophila cell lines. By combining TSC1 or TSC2 mutant cell lines with RNAi screens against all kinases and phosphatases, we identified synthetic interactions with TSC1 and TSC2. Individual knockdown of three candidate genes (mRNA-cap, Pitslre, and CycT; orthologs of RNGTT, CDK11, and CCNT1 in humans) reduced the population growth rate of Drosophila cells lacking either TSC1 or TSC2 but not that of wild-type cells. Moreover, individual knockdown of these three genes had similar growth-inhibiting effects in mammalian TSC2-deficient cell lines, including human tumor-derived cells, illustrating the power of this cross-species screening strategy to identify potential drug targets.

[1]  W. Talbot,et al.  The drosophila EcR gene encodes an ecdysone receptor, a new member of the steroid receptor superfamily , 1991, Cell.

[2]  L. Cherbas,et al.  Transformation techniques for Drosophila cell lines. , 1994, Methods in cell biology.

[3]  J. Lis,et al.  P-TEFb kinase recruitment and function at heat shock loci. , 2000, Genes & development.

[4]  B. Dickson,et al.  The Drosophila Tuberous Sclerosis Complex Gene Homologs Restrict Cell Growth and Cell Proliferation , 2001, Cell.

[5]  Gary D Bader,et al.  Systematic Genetic Analysis with Ordered Arrays of Yeast Deletion Mutants , 2001, Science.

[6]  A. Coulson,et al.  A functional genomic analysis of cell morphology using RNA interference , 2003, Journal of biology.

[7]  Hongbing Zhang,et al.  Loss of Tsc1/Tsc2 activates mTOR and disrupts PI3K-Akt signaling through downregulation of PDGFR. , 2003, The Journal of clinical investigation.

[8]  Frank B. Golley,et al.  Paradigm shift , 1989, Landscape Ecology.

[9]  G. Thomas,et al.  The mTOR/S6K signalling pathway: the role of the TSC1/2 tumour suppressor complex and the proto-oncogene Rheb. , 2004, Novartis Foundation symposium.

[10]  D. Kwiatkowski,et al.  Tuberous sclerosis: a GAP at the crossroads of multiple signaling pathways. , 2005, Human molecular genetics.

[11]  Steven P. Gygi,et al.  mTOR and S6K1 Mediate Assembly of the Translation Preinitiation Complex through Dynamic Protein Interchange and Ordered Phosphorylation Events , 2005, Cell.

[12]  C. Proud Regulation of protein synthesis by insulin. , 2006, Biochemical Society transactions.

[13]  P. Crino,et al.  The tuberous sclerosis complex. , 2006, The New England journal of medicine.

[14]  D. Guertin,et al.  Functional Genomics Identifies TOR-Regulated Genes that Control Growth and Division , 2006, Current Biology.

[15]  A. Tong,et al.  Synthetic genetic array analysis in Saccharomyces cerevisiae. , 2006, Methods in molecular biology.

[16]  Seungbok Lee,et al.  The F-actin-microtubule crosslinker Shot is a platform for Krasavietz-mediated translational regulation of midline axon repulsion , 2007, Development.

[17]  N. Perrimon,et al.  GFP reporters detect the activation of the Drosophila JAK/STAT pathway in vivo. , 2007, Gene expression patterns : GEP.

[18]  B. Manning,et al.  The TSC1-TSC2 complex: a molecular switchboard controlling cell growth. , 2008, The Biochemical journal.

[19]  Vincent J Schmithorst,et al.  Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis. , 2008, The New England journal of medicine.

[20]  B. Housden,et al.  Direct Response to Notch Activation: Signaling Crosstalk and Incoherent Logic , 2009, Science Signaling.

[21]  B. Berger,et al.  Inference of RhoGAP/GTPase regulation using single-cell morphological data from a combinatorial RNAi screen. , 2010, Genome research.

[22]  R. Jensen,et al.  Rapamycin-insensitive up-regulation of MMP2 and other genes in tuberous sclerosis complex 2-deficient lymphangioleiomyomatosis-like cells. , 2010, American journal of respiratory cell and molecular biology.

[23]  Wolfgang Huber,et al.  Mapping of signaling networks through synthetic genetic interaction analysis by RNAi , 2011, Nature Methods.

[24]  J. Krischer,et al.  Efficacy and safety of sirolimus in lymphangioleiomyomatosis. , 2011, The New England journal of medicine.

[25]  Julian Mintseris,et al.  A Protein Complex Network of Drosophila melanogaster , 2011, Cell.

[26]  Anne E Carpenter,et al.  Genome-scale RNAi on living-cell microarrays identifies novel regulators of Drosophila melanogaster TORC1-S6K pathway signaling. , 2011, Genome research.

[27]  K. Ryan,et al.  The cyclin-dependent kinase PITSLRE/CDK11 is required for successful autophagy , 2011, Autophagy.

[28]  P. Finan,et al.  TBC1D7 is a third subunit of the TSC1-TSC2 complex upstream of mTORC1. , 2012, Molecular cell.

[29]  J. Stockman,et al.  Everolimus for Subependymal Giant-Cell Astrocytomas in Tuberous Sclerosis , 2012 .

[30]  Daniel F. Voytas,et al.  Efficient TALEN-mediated gene knockout in livestock , 2012, Proceedings of the National Academy of Sciences.

[31]  J. Doudna,et al.  A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity , 2012, Science.

[32]  N. Perrimon,et al.  Stringent Analysis of Gene Function and Protein–Protein Interactions Using Fluorescently Tagged Genes , 2012, Genetics.

[33]  W. Kaelin,et al.  Use and Abuse of RNAi to Study Mammalian Gene Function , 2012, Science.

[34]  Eli J. Fine,et al.  DNA targeting specificity of RNA-guided Cas9 nucleases , 2013, Nature Biotechnology.

[35]  David R. Liu,et al.  High-throughput profiling of off-target DNA cleavage reveals RNA-programmed Cas9 nuclease specificity , 2013, Nature Biotechnology.

[36]  A. Choi,et al.  Autophagy-Dependent Metabolic Reprogramming Sensitizes TSC2-Deficient Cells to the Antimetabolite 6-Aminonicotinamide , 2013, Molecular Cancer Research.

[37]  Koji Sugiura,et al.  Efficient generation of large-scale genome-modified mice using gRNA and CAS9 endonuclease , 2013, Nucleic acids research.

[38]  Le Cong,et al.  Multiplex Genome Engineering Using CRISPR/Cas Systems , 2013, Science.

[39]  Susan R. Wente,et al.  Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system , 2013, Proceedings of the National Academy of Sciences.

[40]  Jianzhong Xi,et al.  Optimized gene editing technology for Drosophila melanogaster using germ line-specific Cas9 , 2013, Proceedings of the National Academy of Sciences.

[41]  Dong-Yeon Cho,et al.  DNA copy number evolution in Drosophila cell lines , 2014, Genome Biology.

[42]  Chris P. Ponting,et al.  Highly Efficient Targeted Mutagenesis of Drosophila with the CRISPR/Cas9 System , 2013, Cell reports.

[43]  Andrew R. Bassett,et al.  Mutagenesis and homologous recombination in Drosophila cell lines using CRISPR/Cas9 , 2013, Biology Open.

[44]  G. Church,et al.  CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering , 2013, Nature Biotechnology.

[45]  Melissa M. Harrison,et al.  Genome Engineering of Drosophila with the CRISPR RNA-Guided Cas9 Nuclease , 2013, Genetics.

[46]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[47]  Bo Zhang,et al.  Highly Efficient Genome Modifications Mediated by CRISPR/Cas9 in Drosophila , 2013, Genetics.

[48]  Robert L Sutherland,et al.  ZNF300P1 Encodes a lincRNA that regulates cell polarity and is epigenetically silenced in type II epithelial ovarian cancer , 2014, Molecular Cancer.

[49]  J. Keith Joung,et al.  High frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells , 2013, Nature Biotechnology.

[50]  B. Dujon,et al.  Highly Specific Contractions of a Single CAG/CTG Trinucleotide Repeat by TALEN in Yeast , 2014, PloS one.

[51]  Gang Wang,et al.  Optimization of Genome Engineering Approaches with the CRISPR/Cas9 System , 2014, PloS one.

[52]  Norbert Perrimon,et al.  RNAi screening comes of age: improved techniques and complementary approaches , 2014, Nature Reviews Molecular Cell Biology.

[53]  Andrew R. Jones,et al.  ProteomeXchange provides globally co-ordinated proteomics data submission and dissemination , 2014, Nature Biotechnology.

[54]  U. Gaul,et al.  Efficient chromosomal gene modification with CRISPR/cas9 and PCR-based homologous recombination donors in cultured Drosophila cells , 2014, Nucleic acids research.

[55]  Sangsu Bae,et al.  Microhomology-based choice of Cas9 nuclease target sites , 2014, Nature Methods.

[56]  Jing Li,et al.  Estradiol and mTORC2 cooperate to enhance prostaglandin biosynthesis and tumorigenesis in TSC2-deficient LAM cells , 2014, The Journal of experimental medicine.

[57]  E. Lander,et al.  Genetic Screens in Human Cells Using the CRISPR-Cas9 System , 2013, Science.

[58]  Neville E. Sanjana,et al.  Genome-Scale CRISPR-Cas9 Knockout Screening in Human Cells , 2014, Science.

[59]  Ying Peng,et al.  A simplified and efficient germline-specific CRISPR/Cas9 system for Drosophila genomic engineering , 2013, Fly.

[60]  N. Perrimon,et al.  Combining genetic perturbations and proteomics to examine kinase-phosphatase networks in Drosophila embryos. , 2014, Developmental cell.

[61]  Chris P Ponting,et al.  Highly Efficient Targeted Mutagenesis of Drosophila with the CRISPR/Cas9 System , 2014, Cell reports.

[62]  Meagan E. Sullender,et al.  Rational design of highly active sgRNAs for CRISPR-Cas9–mediated gene inactivation , 2014, Nature Biotechnology.

[63]  E. Henske,et al.  Therapeutic Targeting of Cellular Metabolism in Cells with Hyperactive mTORC1: A Paradigm Shift , 2014, Molecular Cancer Research.

[64]  Yanhui Hu,et al.  Enhanced specificity and efficiency of the CRISPR/Cas9 system with optimized sgRNA parameters in Drosophila. , 2014, Cell reports.

[65]  E. Roach,et al.  Tuberous sclerosis complex. , 2015, Handbook of clinical neurology.

[66]  V. Cowling,et al.  Myc and mRNA capping☆ , 2015, Biochimica et biophysica acta.