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.

  • Article
  • Published:

Genome-wide CRISPR screens reveal synthetic lethality of RNASEH2 deficiency and ATR inhibition

Abstract

Ataxia telangiectasia mutated and RAD3 related (ATR) protein kinase plays critical roles in ensuring DNA replication, DNA repair, and cell cycle control in response to replication stress, making ATR inhibition a promising therapeutic strategy for cancer treatment. To identify genes whose loss makes tumor cells hypersensitive to ATR inhibition, we performed CRISPR/Cas9-based whole-genome screens in 3 independent cell lines treated with a highly selective ATR inhibitor, AZD6738. These screens uncovered a comprehensive genome-wide profile of ATR inhibitor sensitivity. From the candidate genes, we demonstrated that RNASEH2 deficiency is synthetic lethal with ATR inhibition both in vitro and in vivo. RNASEH2-deficient cells exhibited elevated levels of DNA damage and, when treated with AZD6738, underwent apoptosis (short-time treated) or senescence (long-time treated). Notably, RNASEH2 deficiency is frequently found in prostate adenocarcinoma; we found decreased RNASEH2B protein levels in prostate adenocarcinoma patient-derived xenograft (PDX) samples. Our findings suggest that ATR inhibition may be beneficial for cancer patients with reduced levels of RNASEH2 and that RNASEH2 merits further exploration as a potential biomarker for ATR inhibitor-based therapy.

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

Access options

Buy this article

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

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Data availability

All relevant data not presented in the main figures or Supplementary Data are available from the authors.

References

  1. Saldivar JC, Cortez D, Cimprich KA. The essential kinase ATR: ensuring faithful duplication of a challenging genome. Nat Rev Mol Cell Biol. 2017;18:622–36.

    Article  CAS  Google Scholar 

  2. Keszthelyi A, Minchell NE, Baxter J. The causes and consequences of topological stress during DNA replication. Genes (Basel). 2016;7:pii: E134.

    Article  Google Scholar 

  3. Zeman MK, Cimprich KA. Causes and consequences of replication stress. Nat Cell Biol. 2014;16:2–9.

    Article  CAS  Google Scholar 

  4. Techer H, Koundrioukoff S, Nicolas A, Debatisse M. The impact of replication stress on replication dynamics and DNA damage in vertebrate cells. Nat Rev Genet. 2017;18:535–50.

    Article  CAS  Google Scholar 

  5. Saldivar JC, Cortez D, Cimprich KA. The essential kinase ATR: ensuring faithful duplication of a challenging genome. Nat Rev Mol Cell Biol. 2017;18:622–36.

    Article  CAS  Google Scholar 

  6. de Klein A, Muijtjens M, van Os R, Verhoeven Y, Smit B, Carr AM. et al. Targeted disruption of the cell-cycle checkpoint gene ATR leads to early embryonic lethality in mice. Curr Biol. 2000;10:479–82.

    Article  CAS  Google Scholar 

  7. Brown EJ, Baltimore D. ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev. 2000;14:397–402.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Wright JA, et al. Protein kinase mutants of human ATR increase sensitivity to UV and ionizing radiation and abrogate cell cycle checkpoint control. Proc Natl Acad Sci USA. 1998;95:7445–50.

    Article  CAS  Google Scholar 

  9. Rundle S, Bradbury A, Drew Y, Curtin NJ. Targeting the ATR-CHK1 axis in cancer therapy. Cancers (Basel). 2017;9:pii: E41.

    Article  Google Scholar 

  10. Charrier JD. et al. Discovery of potent and selective inhibitors of Ataxia telangiectasia mutated and Rad3 related (ATR) protein kinase as potential anticancer agents. J Med Chem. 2011;54:2320–30.

    Article  CAS  Google Scholar 

  11. Hall AB, et al. Potentiation of tumor responses to DNA damaging therapy by the selective ATR inhibitor VX-970. Oncotarget. 2014;5:5674–85.

    Article  Google Scholar 

  12. Mohni KN, Kavanaugh GM, Cortez D. ATR pathway inhibition is synthetically lethal in cancer cells with ERCC1 deficiency. Cancer Res. 2014;74:2835–45.

    Article  CAS  Google Scholar 

  13. Williamson CT. et al. ATR inhibitors as a synthetic lethal therapy for tumours deficient in ARID1A. Nat Commun. 2016;7:13837.

    Article  Google Scholar 

  14. Kwok M, Davies N, Agathanggelou A. et al. ATR inhibition induces synthetic lethality and overcomes chemoresistance in TP53- or ATM-defective chronic lymphocytic leukemia cells. Blood. 2016;127:582–95.

    Article  CAS  Google Scholar 

  15. Reaper PM, et al. Selective killing of ATM- or p53-deficient cancer cells through inhibition of ATR. Nat Chem Biol. 2011;7:428–30.

    Article  CAS  Google Scholar 

  16. Prevo R, et al. The novel ATR inhibitor VE-821 increases sensitivity of pancreatic cancer cells to radiation and chemotherapy. Cancer Biol Ther. 2012;13:1072–81.

    Article  CAS  Google Scholar 

  17. Fokas E. et al. Targeting ATR in vivo using the novel inhibitor VE-822 results in selective sensitization of pancreatic tumors to radiation. Cell Death Dis. 2012;3:e441.

    Article  Google Scholar 

  18. Zhu S, Zhou Y, Wei W. Genome-wide CRISPR/Cas9 screening for high-throughput functional genomics in human cells. Methods Mol Biol. 2017;1656:175–81.

    Article  CAS  Google Scholar 

  19. Steinhart Z, et al. Genome-wide CRISPR screens reveal a Wnt-FZD5 signaling circuit as a druggable vulnerability of RNF43-mutant pancreatic tumors. Nat Med. 2017;23:60–8.

    Article  CAS  Google Scholar 

  20. Sidik SM, et al. A genome-wide CRISPR screen in toxoplasma identifies essential apicomplexan genes. Cell. 2016;166:1423–35 e1412.

    Article  CAS  Google Scholar 

  21. Ruiz S, et al. A genome-wide CRISPR screen identifies CDC25A as a determinant of sensitivity to ATR inhibitors. Mol Cell. 2016;62:307–13.

    Article  CAS  Google Scholar 

  22. Hart T. et al. High-resolution CRISPR screens reveal fitness genes and genotype-specific cancer liabilities. Cell. 2015;163:1515–26.

    Article  CAS  Google Scholar 

  23. Hart T, Brown KR, Sircoulomb F, Rottapel R, Moffat J. Measuring error rates in genomic perturbation screens: gold standards for human functional genomics. Mol Syst Biol. 2014;10:733.

    Article  Google Scholar 

  24. Hart T, Moffat J. BAGEL: a computational framework for identifying essential genes from pooled library screens. BMC Bioinformatics. 2016;17:164.

    Article  Google Scholar 

  25. Checkley S. et al. Bridging the gap between in vitro and in vivo: Dose and schedule predictions for the ATR inhibitor AZD6738. Sci Rep. 2015;5:13545.

    Article  Google Scholar 

  26. Guichard SM, Brown E, Odedra R, Hughes A, Heathcote D, Barnes J, et al. The pre-clinical in vitro and in vivo activity of AZD6738: a potent and selective inhibitor of ATR kinase. Cancer Res. 73, https://doi.org/10.1158/1538-7445.Am2013-3343 (2013).

  27. Hiller B, et al. Mammalian RNase H2 removes ribonucleotides from DNA to maintain genome integrity. J Exp Med. 2012;209:1419–26.

    Article  CAS  Google Scholar 

  28. Perrino FW, Harvey S, Shaban NM, Hollis T. RNaseH2 mutants that cause Aicardi-Goutieres syndrome are active nucleases. J Mol Med. 2009;87:25–30.

    Article  CAS  Google Scholar 

  29. Pendergraft WF 3rd, Means TK. AGS, SLE, and RNASEH2 mutations: translating insights into therapeutic advances. J Clin Invest. 2015;125:102–4.

    Article  Google Scholar 

  30. Crow YJ, et al. Mutations in genes encoding ribonuclease H2 subunits cause Aicardi-Goutieres syndrome and mimic congenital viral brain infection. Nat Genet. 2006;38:910–6.

    Article  CAS  Google Scholar 

  31. Mackenzie KJ, et al. Ribonuclease H2 mutations induce a cGAS/STING-dependent innate immune response. EMBO J. 2016;35:831–44.

    Article  CAS  Google Scholar 

  32. Pokatayev V, et al. RNase H2 catalytic core Aicardi-Goutieres syndrome-related mutant invokes cGAS-STING innate immune-sensing pathway in mice. J Exp Med. 2016;213:329–36.

    Article  CAS  Google Scholar 

  33. Williams JS, Gehle DB, Kunkel TA. The role of RNase H2 in processing ribonucleotides incorporated during DNA replication. DNA Repair (Amst). 2017;53:52–8.

    Article  CAS  Google Scholar 

  34. Pizzi S, et al. Reduction of hRNase H2 activity in Aicardi-Goutieres syndrome cells leads to replication stress and genome instability. Hum Mol Genet. 2015;24:649–58.

    Article  CAS  Google Scholar 

  35. Chon H, et al. RNase H2 roles in genome integrity revealed by unlinking its activities. Nucleic Acids Res. 2013;41:3130–43.

    Article  CAS  Google Scholar 

  36. McElhinny SAN, et al. Genome instability due to ribonucleotide incorporation into DNA. Nat Chem Biol. 2010;6:774–81.

    Article  Google Scholar 

  37. Reijns MAM. et al. Enzymatic removal of ribonucleotides from DNA is essential for mammalian genome integrity and development. Cell. 2012;149:1008–22.

    Article  CAS  Google Scholar 

  38. Zimmermann M, et al. CRISPR screens identify genomic ribonucleotides as a source of PARP-trapping lesions. Nature. 2018;559:285–9.

    Article  CAS  Google Scholar 

  39. Hart T, et al. Evaluation and Design of Genome-Wide CRISPR/SpCas9 Knockout Screens. G3-Genes Genom Genet. 2017;7:2719–27.

    CAS  Google Scholar 

  40. Kim H, George E, Brown E, Zhang RG, Krepler C, Tanyi J, et al. Targeting the ATR/CHK1 axis in BRCA1/2 mutant ovarian cancer using an orthotopic patient-derived xenograft (PDX) model. Clin Cancer Res. 2016;22 (2 Suppl).

  41. Kim H, et al. Targeting the ATR/CHK1 axis with PARP inhibition results in tumor regression in BRCA-mutant ovarian cancer models. Clin Cancer Res. 2017;23:3097–108.

    Article  CAS  Google Scholar 

  42. Huang S, et al. MED12 controls the response to multiple cancer drugs through regulation of TGF-beta receptor signaling. Cell. 2012;151:937–50.

    Article  CAS  Google Scholar 

  43. He T, et al. Methylation of SLFN11 is a marker of poor prognosis and cisplatin resistance in colorectal cancer. Epigenomics. 2017;9:849–62.

    Article  CAS  Google Scholar 

  44. Mu Y, et al. SLFN11 inhibits checkpoint maintenance and homologous recombination repair. EMBO Rep. 2016;17:94–109.

    Article  CAS  Google Scholar 

  45. Bartsch K, et al. Absence of RNase H2 triggers generation of immunogenic micronuclei removed by autophagy. Hum Mol Genet. 2017;26:3960–72.

    Article  CAS  Google Scholar 

  46. Rice GI, et al. Synonymous mutations in RNASEH2A create cryptic splice sites impairing RNase H2 enzyme function in Aicardi-Goutieres syndrome. Hum Mutat. 2013;34:1066–70.

    Article  CAS  Google Scholar 

  47. Grasso CS, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature. 2012;487:239–43.

    Article  CAS  Google Scholar 

  48. Robinson D, et al. Integrative clinical genomics of advanced prostate. Cancer Cell. 2015;162:454.

    Google Scholar 

  49. Kumar A, et al. Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer. Nat Med. 2016;22:369.

    Article  CAS  Google Scholar 

  50. Abeshouse A, et al. The molecular taxonomy of primary prostate. Cancer Cell. 2015;163:1011–25.

    CAS  Google Scholar 

  51. Taylor BS, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22.

    Article  CAS  Google Scholar 

  52. Uhlen M, et al. Proteomics. Tissue-based map of the human proteome. Science. 2015;347:1260419.

    Article  Google Scholar 

  53. Gao J, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:pl1.

    Article  Google Scholar 

  54. Li T, Chen ZJ. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J Exp Med. 2018;215:1287–99.

    Article  CAS  Google Scholar 

  55. Yang H, Wang H, Ren J, Chen Q, Chen ZJ. cGAS is essential for cellular senescence. Proc Natl Acad Sci USA. 2017;114:E4612–20.

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the members of Dr. Junjie Chen’s lab for their kind help and Dr. Lei Li for his suggestions regarding the experimental design. We also thank Amy Ninetto from the Department of Scientific Publications at MD Anderson for editing the manuscript. This work was supported in part by CPRIT (RP160667) and NIH grants (CA157448, CA193124, CA210929, CA216911, and CA216437) to JC and MD Anderson’s NIH Cancer Center Support Grant (CA016672).

Author contributions

CW and JC conceived the project. CW, JZ, XF, MT, ZC, and MS performed the experiments. MEM provided technical support for the screen work. GW and GTH analyzed the deep-sequencing results. PS and NMN provided and analyzed the PDX samples. CW and JC wrote the manuscript with input from all authors.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Junjie Chen.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, C., Wang, G., Feng, X. et al. Genome-wide CRISPR screens reveal synthetic lethality of RNASEH2 deficiency and ATR inhibition. Oncogene 38, 2451–2463 (2019). https://doi.org/10.1038/s41388-018-0606-4

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-018-0606-4

This article is cited by

Search

Quick links