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:

AAV-mediated direct in vivo CRISPR screen identifies functional suppressors in glioblastoma

This article has been updated

Abstract

A causative understanding of genetic factors that regulate glioblastoma pathogenesis is of central importance. Here we developed an adeno-associated virus–mediated, autochthonous genetic CRISPR screen in glioblastoma. Stereotaxic delivery of a virus library targeting genes commonly mutated in human cancers into the brains of conditional-Cas9 mice resulted in tumors that recapitulate human glioblastoma. Capture sequencing revealed diverse mutational profiles across tumors. The mutation frequencies in mice correlated with those in two independent patient cohorts. Co-mutation analysis identified co-occurring driver combinations such as B2mNf1, Mll3Nf1 and Zc3h13Rb1, which were subsequently validated using AAV minipools. Distinct from Nf1-mutant tumors, Rb1-mutant tumors are undifferentiated and aberrantly express homeobox gene clusters. The addition of Zc3h13 or Pten mutations altered the gene expression profiles of Rb1 mutants, rendering them more resistant to temozolomide. Our study provides a functional landscape of gliomagenesis suppressors in vivo.

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

Figure 1: Autochthonous brain tumorigenesis induced by an AAV-mediated CRISPR library.
Figure 2: AAV-mTSG-induced brain tumors recapitulate pathological features of GBM.
Figure 3: Targeted-capture sequencing of sgRNA sites in AAV-mTSG-induced mouse GBM.
Figure 4: Integrative analysis of functional mutations in driving tumorigenesis.
Figure 5: Co-mutation analysis uncovers synergistic gene pairs in GBM.
Figure 6: Validation of driver combinations.
Figure 7: Transcriptional profiling of mouse GBM driver combinations.
Figure 8: Transcriptional profiling of mouse GBM driver combinations in the presence and absence of a chemotherapeutic agent.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

Change history

  • 21 August 2017

    In the PDF version of this article initially published online, one of the Online Methods headings read “vRNA-seq differential expression analysis”; this has been changed to “RNA-seq differential expression analysis.” The error has been corrected in the print and PDF versions of this article.

References

  1. Sturm, D. et al. Paediatric and adult glioblastoma: multiform (epi)genomic culprits emerge. Nat. Rev. Cancer 14, 92–107 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Louis, D.N. et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114, 97–109 (2007).

    PubMed  PubMed Central  Google Scholar 

  3. Krex, D. et al. Long-term survival with glioblastoma multiforme. Brain 130, 2596–2606 (2007).

    PubMed  Google Scholar 

  4. American Brain Tumor Association. Glioblastoma and Malignant Astrocytoma http://www.abta.org/secure/glioblastoma-brochure.pdf (2016).

  5. Claus, E.B. & Black, P.M. Survival rates and patterns of care for patients diagnosed with supratentorial low-grade gliomas: data from the SEER program, 1973-2001. Cancer 106, 1358–1363 (2006).

    PubMed  Google Scholar 

  6. Stupp, R. et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 352, 987–996 (2005).

    CAS  PubMed  Google Scholar 

  7. Stupp, R. et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10, 459–466 (2009).

    CAS  PubMed  Google Scholar 

  8. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

  9. Brennan, C.W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Verhaak, R.G. et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17, 98–110 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Bai, H. et al. Integrated genomic characterization of IDH1-mutant glioma malignant progression. Nat. Genet. 48, 59–66 (2016).

    CAS  PubMed  Google Scholar 

  12. Ceccarelli, M. et al. Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell 164, 550–563 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Parsons, D.W. et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Hanahan, D. & Weinberg, R.A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    CAS  PubMed  Google Scholar 

  15. Hainaut, P. & Plymoth, A. Targeting the hallmarks of cancer: towards a rational approach to next-generation cancer therapy. Curr. Opin. Oncol. 25, 50–51 (2013).

    PubMed  Google Scholar 

  16. Cairncross, J.G. et al. Specific genetic predictors of chemotherapeutic response and survival in patients with anaplastic oligodendrogliomas. J. Natl. Cancer Inst. 90, 1473–1479 (1998).

    CAS  PubMed  Google Scholar 

  17. Napoli, M. & Flores, E.R. The p53 family orchestrates the regulation of metabolism: physiological regulation and implications for cancer therapy. Br. J. Cancer 116, 149–155 (2017).

    CAS  PubMed  Google Scholar 

  18. Muller, P.A. & Vousden, K.H. p53 mutations in cancer. Nat. Cell Biol. 15, 2–8 (2013).

    CAS  PubMed  Google Scholar 

  19. Feng, Z., Hu, W., Rajagopal, G. & Levine, A.J. The tumor suppressor p53: cancer and aging. Cell Cycle 7, 842–847 (2008).

    CAS  PubMed  Google Scholar 

  20. Levine, A.J. p53, the cellular gatekeeper for growth and division. Cell 88, 323–331 (1997).

    CAS  PubMed  Google Scholar 

  21. Berns, A. Cancer: the blind spot of p53. Nature 468, 519–520 (2010).

    CAS  PubMed  Google Scholar 

  22. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Lawrence, M.S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495–501 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Lawrence, M.S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Davoli, T. et al. Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948–962 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Guo, F., Li, Y., Liu, Y., Wang, J. & Li, G. ARL6IP1 mediates cisplatin-induced apoptosis in CaSki cervical cancer cells. Oncol. Rep. 23, 1449–1455 (2010).

    CAS  PubMed  Google Scholar 

  27. Ribas, A. Tumor immunotherapy directed at PD-1. N. Engl. J. Med. 366, 2517–2519 (2012).

    CAS  PubMed  Google Scholar 

  28. Frese, K.K. & Tuveson, D.A. Maximizing mouse cancer models. Nat. Rev. Cancer 7, 645–658 (2007).

    CAS  PubMed  Google Scholar 

  29. Holland, E.C. Gliomagenesis: genetic alterations and mouse models. Nat. Rev. Genet. 2, 120–129 (2001).

    CAS  PubMed  Google Scholar 

  30. Huse, J.T. & Holland, E.C. Genetically engineered mouse models of brain cancer and the promise of preclinical testing. Brain Pathol. 19, 132–143 (2009).

    CAS  PubMed  Google Scholar 

  31. Alcantara Llaguno, S. et al. Malignant astrocytomas originate from neural stem/progenitor cells in a somatic tumor suppressor mouse model. Cancer Cell 15, 45–56 (2009).

    PubMed  PubMed Central  Google Scholar 

  32. Friedmann-Morvinski, D. et al. Dedifferentiation of neurons and astrocytes by oncogenes can induce gliomas in mice. Science 338, 1080–1084 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Marumoto, T. et al. Development of a novel mouse glioma model using lentiviral vectors. Nat. Med. 15, 110–116 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Perry, A. et al. Malignant gliomas with primitive neuroectodermal tumor-like components: a clinicopathologic and genetic study of 53 cases. Brain Pathol. 19, 81–90 (2009).

    PubMed  Google Scholar 

  35. Reilly, K.M., Loisel, D.A., Bronson, R.T., McLaughlin, M.E. & Jacks, T. Nf1;Trp53 mutant mice develop glioblastoma with evidence of strain-specific effects. Nat. Genet. 26, 109–113 (2000).

    CAS  PubMed  Google Scholar 

  36. Schmid, R.S., Vitucci, M. & Miller, C.R. Genetically engineered mouse models of diffuse gliomas. Brain Res. Bull. 88, 72–79 (2012).

    CAS  PubMed  Google Scholar 

  37. Qazi, M. et al. Generation of murine xenograft models of brain tumors from primary human tissue for in vivo analysis of the brain tumor-initiating cell. Methods Mol. Biol. 1210, 37–49 (2014).

    CAS  PubMed  Google Scholar 

  38. Agemy, L. et al. Targeted nanoparticle enhanced proapoptotic peptide as potential therapy for glioblastoma. Proc. Natl. Acad. Sci. USA 108, 17450–17455 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Chow, L.M. et al. Cooperativity within and among Pten, p53, and Rb pathways induces high-grade astrocytoma in adult brain. Cancer Cell 19, 305–316 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Chen, J. et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 488, 522–526 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Sánchez-Rivera, F.J. & Jacks, T. Applications of the CRISPR-Cas9 system in cancer biology. Nat. Rev. Cancer 15, 387–395 (2015).

    PubMed  PubMed Central  Google Scholar 

  45. Weitzman, M.D., Kyöstiö, S.R.M., Kotin, R.M. & Owens, R.A. Adeno-associated virus (AAV) Rep proteins mediate complex formation between AAV DNA and its integration site in human DNA. Proc. Natl. Acad. Sci. USA 91, 5808–5812 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Yeung, J.T. et al. LOH in the HLA class I region at 6p21 is associated with shorter survival in newly diagnosed adult glioblastoma. Clin. Cancer Res. 19, 1816–1826 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Patil, V., Pal, J. & Somasundaram, K. Elucidating the cancer-specific genetic alteration spectrum of glioblastoma derived cell lines from whole exome and RNA sequencing. Oncotarget 6, 43452–43471 (2015).

    PubMed  PubMed Central  Google Scholar 

  48. Bale, T.A. et al. Genomic characterization of recurrent high-grade astroblastoma. Cancer Genet. 209, 321–330 (2016).

    CAS  PubMed  Google Scholar 

  49. Aithal, M.G. & Rajeswari, N. Validation of housekeeping genes for gene expression analysis in glioblastoma using quantitative real-time polymerase chain reaction. Brain Tumor Res. Treat. 3, 24–29 (2015).

    PubMed  PubMed Central  Google Scholar 

  50. Friedman, A.A., Letai, A., Fisher, D.E. & Flaherty, K.T. Precision medicine for cancer with next-generation functional diagnostics. Nat. Rev. Cancer 15, 747–756 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  53. 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).

    CAS  PubMed  Google Scholar 

  54. Platt, R.J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Chen, S. et al. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell 160, 1246–1260 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Franklin, K.B.J. & Paxinos, G. The Mouse Brain in Stereotaxic Coordinates (Academic Press, 2013).

  57. Fedorov, A. et al. 3D Slicer as an image computing platform for the Quantitative Imaging Network. Magn. Reson. Imaging 30, 1323–1341 (2012).

    PubMed  PubMed Central  Google Scholar 

  58. Schindelin, J., Rueden, C.T., Hiner, M.C. & Eliceiri, K.W. The ImageJ ecosystem: an open platform for biomedical image analysis. Mol. Reprod. Dev. 82, 518–529 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Chen, S. et al. Global microRNA depletion suppresses tumor angiogenesis. Genes Dev. 28, 1054–1067 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Quinlan, A.R. & Hall, I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Barnett, D.W., Garrison, E.K., Quinlan, A.R., Strömberg, M.P. & Marth, G.T. BamTools: a C. API and toolkit for analyzing and managing BAM files. Bioinformatics 27, 1691–1692 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

  64. Koboldt, D.C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    PubMed  PubMed Central  Google Scholar 

  66. Bray, N.L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    CAS  PubMed  Google Scholar 

  67. Robinson, M.D., McCarthy, D.J. & Smyth, G.K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  68. Huang, W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank all members of the Chen, Sharp, Zhang and Platt laboratories, as well as our colleagues in the Yale Department of Genetics, Systems Biology Institute, Yale Cancer Center and Stem Cell Center, Koch Institute and Broad Institute at MIT for assistance and/or discussions. We thank the Center for Genome Analysis, Center for Molecular Discovery, High Performance Computing Center, West Campus Analytical Chemistry Core and West Campus Imaging Core and Keck Biotechnology Resource Laboratory at Yale, as well as Swanson Biotechnology Center at MIT, for technical support. S.C. is supported by Yale SBI/Genetics Startup Fund, Damon Runyon (DRG-2117-12; DFS-13-15), Melanoma Research Alliance (412806, 16-003524), St. Baldrick's Foundation (426685), American Cancer Society (IRG 58-012-54), Breast Cancer Alliance, Cancer Research Institute (CLIP), AACR (499395), DoD (W81XWH-17-1-0235) and NIH/NCI (1U54CA209992, 5P50CA196530-A10805, 4P50CA121974-A08306). R.J.P. is supported by NCCRMSE and ETH Zurich, the McGovern Institute and NSF (1122374). P.A.S. is supported by NIH (R01-CA133404, R01-GM034277, CCNE), Skoltech Center and the Casimir-Lambert Fund. F.Z. is supported by the NIH/NIMH (5DP1-MH100706 and 1R01-MH110049), NSF, NY Stem Cell Foundation, HHMI, Poitras, Simons, Paul G. Allen Family, Vallee Foundations, D.R. Cheng and B. Metcalfe. C.D.G. and P.R. are supported by an NIH Graduate Training Grant (T32GM007499). R.D.C., M.B.D. and M.W.Y. are supported by an NIH MSTP training grant (T32GM007205). F.S. is supported by NCCRMSE and ETH Zurich. G.W. is supported by RJ Anderson and CRI Irvington Postdoctoral Fellowships.

Author information

Authors and Affiliations

Authors

Contributions

S.C. and R.J.P. conceived the study, designed the study and performed the initial set of experiments. R.D.C. developed the algorithms and performed integrative analyses of all the data. C.D.G. performed validation, performed histology and established primary cell lines. G.W. performed exome-capture, mutant cell line generation, drug treatment and RNA-seq. F.S. performed AAV production. S.C. performed MRI. M.W.Y. contributed to data analysis. L.Y., Y.E., M.B.D., M.A.M., S.Z. and P.R. contributed to experiments including mouse breeding, genotyping, cloning, cell culture, virus prep, injection, necropsy and sample prep. K.B. assisted in captured and exome sequencing. M.G. provided clinical insights. P.A.S., F.Z., R.J.P. and S.C. jointly supervised the work. R.D.C. and S.C. wrote the manuscript with inputs from all authors.

Corresponding authors

Correspondence to Randall J Platt or Sidi Chen.

Ethics declarations

Competing interests

F.Z. is a cofounder of Editas Medicine and a scientific advisor for Editas Medicine and Horizon Discovery. A patent application has been filed on the methods pertaining to this work.

Integrated supplementary information

Supplementary Figure 1 Additional data for massively parallel GBM suppressor analysis by AAV-CRISPR library-mediated pooled mutagenesis

(a) Schematic of the AAV vector used in the study. The vector contains a cassette expressing Cre recombinase under a GFAP promoter, a p53 sgRNA under U6 promoter, and an empty cassette for expression of custom cloned sgRNA(s).

(b) Plasmid library representation of the AAV-CRISPR mTSG library (n = 2 plasmid library replicates, averaged).

(c) A representative AAV-mTSG injected mouse showing macrocephaly.

(d) Dissected whole brains from PBS, AAV-vector and AAV-mTSG injected mice (left) and sections (right) visualized under a fluorescent stereoscope.

(e) Full-spectrum MRI series of representative mouse brains in PBS, vector and mTSG group. Mice under anesthesia were imaged with a small animal MRI imaging system. 20 MRI sections are shown for each condition. Brain tumors were found in AAV-mTSG injected mice but not in matched PBS or AAV-vector injected mice.

Supplementary Figure 2 Full-scan histology images for special staining of mouse brain sections in vector and mTSG groups

Panels from top to bottom: Luxol fast blue Cresyl violet (LFB/CV) staining, Wight Giemsa staining, Masson staining and Alcian blue Periodic acidSchiff (AB/PAS) staining of representative mouse brain sections in vector and mTSG groups. Scale bar = 1 mm.

Supplementary Figure 3 Representative histopathology images of human GBM

(a) Representative images of H&E stained brain sections from human GBM patient samples from Yale Glioma tissue bank. Images from the three rows represent GBM with significant mutations in NF1, PTEN and RB1, respectively. Pathological features such as giant aneuploid cells with pleomorphic nuclei, angiogenesis, necrosis and hemorrhage were evident in these tumors. Scale bar = 0.5 mm.

(b) Representative images of anti-GFAP stained brain sections from human GBM patient samples from Yale Glioma tissue bank. Images from the two rows represent GBM with significant mutations in PTEN and RB1, respectively. PTEN tumors were mostly GFAP-positive. RB1 tumors have mixtures of GFAP-positive and GFAP-negative cells. NF1 tumors were not shown due to availability of GFAP staining sections. Scale bar = 0.5 mm.

Supplementary Figure 4 Early time point analysis of sgRNA cutting efficiency by molecular inversion probe sequencing

(a) Heatmap of sum variant frequencies for each sgRNA across the 3 in vivo infection replicates. Each row denotes one gene, while each column corresponds to a specific sgRNA and replicate. Variant frequencies are square-rooted to improve visibility.

(b) Dissected whole brain from an AAV-mTSG injected mouse for early time point analysis, visualized under a fluorescent stereoscope. GFP (green) is shown as an overlay on the brightfield image.

(c) Venn diagram detailing the overlap between cutting sgRNAs identified in early-stage mutagenesis and late-stage GBMs. Differences in the identified cutting sgRNAs were likely due to differential selection pressures, insufficient time for CRISPR mutagenesis to occur in early time point brains, and/or allele frequencies below detection limit of capture sequencing.

Supplementary Figure 5 Mutational signatures of all GBM mice induced with AAV-CRISPR mTSG library

Waterfall plots of significantly mutated sgRNA sites across all mTSG brain samples, sorted by sum variant frequency. Two samples (mTSG brain 1, mTSG brain 7) are not shown, as these samples were not found to have any significantly mutated sgRNA sites per the stringent variant calling strategy. The extensive mutational landscape in these samples shows strong positive selection for loss-of-function mutations in mTSGs during gliomagenesis.

Supplementary Figure 6 Additional analysis of mutational signatures

(a) Scatterplots of the number of samples with an SMS call per sgRNA (left) or SMG call per gene (right), using two different thresholds for calling SMSs. In conjunction with the FDR approach, the use of either a flat 5% or 10% variant frequency cutoff did not affect the results at either the sgRNA or gene level. Spearman correlation coefficients and associated p-values are shown on the plots.

(b) Gaussian kernel density estimate of variant frequencies within each mTSG brain sample. The number of peaks in the kernel density estimate is an approximation for the clonality of each sample. From this analysis, most (20/22) samples appeared to be composed of multiple clones, with only two (mTSG brain 15, mTSG brain 20) monoclonal samples. Of note, 3/25 sequenced mTSG brain samples did not have sufficient high-frequency variants for clustering analysis.

Supplementary Figure 7 Additional analysis of comutated pairs and exome sequencing

(a) Scatterplot of the co-occurrence rate of a given mutation pair, plotted against -log10 p-values. All pairs involving Trp53 were excluded from this analysis.

(b) Scatterplot of pairwise Spearman correlations plotted against -log10 p-values. All pairs involving Trp53 were excluded from this analysis.

(c) Scatterplot of the co-occurrence rate of a given mutation pair in the TCGA human GBM dataset, plotted against -log10 p-values.

(d) Venn diagram of co-occurring pairs identified in mouse GBM (Benjamini-Hochberg adjusted p < 0.05, either co-occurrence or Spearman correlation analysis) and/or in human GBM (p < 0.05). 7 gene pairs were found to be significant in both mouse and human GBM. The overlap between the two datasets was significant (hypergeometric test, p = 0.001).

(e) Whole-exome analysis of possible off-target mutations generated by AAV-CRISPR mTSG (n = 7). Chromosomal map of potential off-targets in AAV-CRISPR mTSG brain samples. Indels in mTSG genes are marked in red, while possible off-target mutations and AAV insertions are marked in blue.

Supplementary Figure 8 GFAP immunohistochemical characterization of brain sections from mice treated with AAV sgRNA minipools

GFAP immunohistochemistry of brain sections from mice treated with various AVV minipools. Brain tumors in Nf1, Nf1;Pten, and Nf1;B2m mice were strongly positive for GFAP, while tumors in Nf1;Mll3 mice were positive at an intermediate level. Brain tumors in Rb1, Rb1;Pten, and Rb1;Zc3h13 mice contained a mixture of GFAP positive and negative cells, similar to the GFAP staining pattern with human patient GBM samples. Brain tumors in Mll2 mice were variably GFAP positive. Scale bar = 0.5 mm.

Supplementary Figure 9 Additional supplemental data related to the study

(a) Kaplan-Meier overall survival curves for mice injected with control (n = 9), B2m (n = 4), Nf1 (n = 8), and Nf1;B2m (n = 4) AAV minipools. All control and B2m mice were tumor-free and survived the entire duration of the experiment; control and B2m curves are offset for visibility. Mice treated with Nf1;B2m AAVs had significantly worse survival compared to mice treated with Nf1 or B2m AAVs alone (Log-rank (LR) test, p = 0.0067).

(b) T7E1 nuclease assay to confirm mutagenesis by CRISPR/Cas9 at the indicated target genes. Indel frequencies are indicated.

(c-d) LentiCRISPR mTSG direct in vivo GBM screen

(c) IVIS imaging of mice injected with lenti-vector or lenti-mTSG library, showing luminescence in the brains of a fraction of lenti-mTSG injected mice, but not in vector injected mice. Mice were imaged at 6.5 months post injection (mpi), where 4/18 mice imaged were luciferase positive (10 were shown). These 4 mice were sacrificed as they developed poor body conditions and brain tumors, before the end of 10 mpi. Mice were imaged again at 11 mpi, where 6/14 mice imaged luciferase positive, which were subsequently sacrificed as they developed poor body conditions and brain tumors.

(d) Kaplan-Meier curves for overall survival (OS) of mice injected with PBS (n = 2), lenti-vector (n = 5) or lenti-mTSG library (n = 18). OS for PBS and vector groups were both 100%, where the curves are dashed and slightly offset for visibility. LR test, p < 0.0239, mTSG vs. vector or PBS; LR test, p = 1, vector vs. PBS.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 (PDF 3540 kb)

Life Sciences Reporting Summary (PDF 129 kb)

Supplementary Tables

Supplementary Tables 1–33 (XLSX 10772 kb)

Supplementary Software

Code used for analysis (ZIP 610 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chow, R., Guzman, C., Wang, G. et al. AAV-mediated direct in vivo CRISPR screen identifies functional suppressors in glioblastoma. Nat Neurosci 20, 1329–1341 (2017). https://doi.org/10.1038/nn.4620

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4620

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer