411
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Identification of oncogenic driver mutations by genome-wide CRISPR-Cas9 dropout screening

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Genome-wide CRISPR-Cas9 dropout screens can identify genes whose knockout affects cell viability. Recent CRISPR screens detected thousands of essential genes required for cellular survival and key cellular processes; however discovering novel lineage-specific genetic dependencies from the many hits still remains a challenge.

          Results

          To assess whether CRISPR-Cas9 dropout screens can help identify cancer dependencies, we screened two human cancer cell lines carrying known and distinct oncogenic mutations using a genome-wide sgRNA library. We found that the gRNA targeting the driver mutation EGFR was one of the highest-ranking candidates in the EGFR-mutant HCC-827 lung adenocarcinoma cell line. Likewise, sgRNAs for NRAS and MAP2K1 (MEK1), a downstream kinase of mutant NRAS, were identified among the top hits in the NRAS-mutant neuroblastoma cell line CHP-212. Depletion of these genes targeted by the sgRNAs strongly correlated with the sensitivity to specific kinase inhibitors of the EGFR or RAS pathway in cell viability assays. In addition, we describe other dependencies such as TBK1 in HCC-827 cells and TRIB2 in CHP-212 cells which merit further investigation.

          Conclusions

          We show that genome-wide CRISPR dropout screens are suitable for the identification of oncogenic drivers and other essential genes.

          Electronic supplementary material

          The online version of this article (doi:10.1186/s12864-016-3042-2) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references12

          • Record: found
          • Abstract: found
          • Article: not found

          The protein kinase complement of the human genome.

          G. Manning (2002)
          We have catalogued the protein kinase complement of the human genome (the "kinome") using public and proprietary genomic, complementary DNA, and expressed sequence tag (EST) sequences. This provides a starting point for comprehensive analysis of protein phosphorylation in normal and disease states, as well as a detailed view of the current state of human genome analysis through a focus on one large gene family. We identify 518 putative protein kinase genes, of which 71 have not previously been reported or described as kinases, and we extend or correct the protein sequences of 56 more kinases. New genes include members of well-studied families as well as previously unidentified families, some of which are conserved in model organisms. Classification and comparison with model organism kinomes identified orthologous groups and highlighted expansions specific to human and other lineages. We also identified 106 protein kinase pseudogenes. Chromosomal mapping revealed several small clusters of kinase genes and revealed that 244 kinases map to disease loci or cancer amplicons.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library.

            Identification of genes influencing a phenotype of interest is frequently achieved through genetic screening by RNA interference (RNAi) or knockouts. However, RNAi may only achieve partial depletion of gene activity, and knockout-based screens are difficult in diploid mammalian cells. Here we took advantage of the efficiency and high throughput of genome editing based on type II, clustered, regularly interspaced, short palindromic repeats (CRISPR)-CRISPR-associated (Cas) systems to introduce genome-wide targeted mutations in mouse embryonic stem cells (ESCs). We designed 87,897 guide RNAs (gRNAs) targeting 19,150 mouse protein-coding genes and used a lentiviral vector to express these gRNAs in ESCs that constitutively express Cas9. Screening the resulting ESC mutant libraries for resistance to either Clostridium septicum alpha-toxin or 6-thioguanine identified 27 known and 4 previously unknown genes implicated in these phenotypes. Our results demonstrate the potential for efficient loss-of-function screening using the CRISPR-Cas9 system.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis.

              Genetic screens are powerful tools for identifying genes responsible for diverse phenotypes. Here we describe a genome-wide CRISPR/Cas9-mediated loss-of-function screen in tumor growth and metastasis. We mutagenized a non-metastatic mouse cancer cell line using a genome-scale library with 67,405 single-guide RNAs (sgRNAs). The mutant cell pool rapidly generates metastases when transplanted into immunocompromised mice. Enriched sgRNAs in lung metastases and late-stage primary tumors were found to target a small set of genes, suggesting that specific loss-of-function mutations drive tumor growth and metastasis. Individual sgRNAs and a small pool of 624 sgRNAs targeting the top-scoring genes from the primary screen dramatically accelerate metastasis. In all of these experiments, the effect of mutations on primary tumor growth positively correlates with the development of metastases. Our study demonstrates Cas9-based screening as a robust method to systematically assay gene phenotypes in cancer evolution in vivo.
                Bookmark

                Author and article information

                Contributors
                michael.kiessling@usz.ch
                sven.schuierer@novartis.com
                stertz.silke@virology.uzh.ch
                martin.beibel@novartis.com
                sebastian.bergling@novartis.com
                judith.knehr@novartis.com
                walter.carbone@novartis.com
                Cheryl.DeValliere@usz.ch
                Joelle.Tchinda@kispi.uzh.ch
                tewis.bouwmeester@novartis.com
                klaus.seuwen@novartis.com
                Gerhard.rogler@usz.ch
                Guglielmo.roma@novartis.com
                Journal
                BMC Genomics
                BMC Genomics
                BMC Genomics
                BioMed Central (London )
                1471-2164
                9 September 2016
                9 September 2016
                2016
                : 17
                : 1
                : 723
                Affiliations
                [1 ]Department of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
                [2 ]Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
                [3 ]Institute of Medical Virology, University of Zürich, Zürich, Switzerland
                [4 ]Department of Oncology, Children University Hospital Zürich, Zürich, Switzerland
                Article
                3042
                10.1186/s12864-016-3042-2
                5016932
                27613601
                bb6994d8-2833-4042-83a4-f70f4deffe24
                © The Author(s). 2016

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 21 August 2016
                : 24 August 2016
                Categories
                Methodology Article
                Custom metadata
                © The Author(s) 2016

                Genetics
                whole genome crispr screen,dropout,negative selection,driver mutations,egfr,nras,kinase
                Genetics
                whole genome crispr screen, dropout, negative selection, driver mutations, egfr, nras, kinase

                Comments

                Comment on this article