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:

A CRISPR activation screen identifies FBXO22 supporting targeted protein degradation

Abstract

Targeted protein degradation (TPD) represents a potent chemical biology paradigm that leverages the cellular degradation machinery to pharmacologically eliminate specific proteins of interest. Although multiple E3 ligases have been discovered to facilitate TPD, there exists a compelling requirement to diversify the pool of E3 ligases available for such applications. Here we describe a clustered regularly interspaced short palindromic repeats (CRISPR)-based transcriptional activation screen focused on human E3 ligases, with the goal of identifying E3 ligases that can facilitate heterobifunctional compound-mediated target degradation. Through this approach, we identified a candidate proteolysis-targeting chimera (PROTAC), 22-SLF, that induces the degradation of FK506-binding protein 12 when the transcription of FBXO22 gene is activated. Subsequent mechanistic investigations revealed that 22-SLF interacts with C227 and/or C228 in F-box protein 22 (FBXO22) to achieve target degradation. Lastly, we demonstrated the versatility of FBXO22-based PROTACs by effectively degrading additional endogenous proteins, including bromodomain-containing protein 4 and the echinoderm microtubule-associated protein-like 4–anaplastic lymphoma kinase fusion protein.

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: An E3 ligase-focused CRISPR–Cas9 transcriptional activation screen identifies FBXO22 that supports 22-SLF-induced reduction in FKBP12-EGFP expression levels.
Fig. 2: 22-SLF promotes FBXO22-dependent proteasomal degradation of FKBP12.
Fig. 3: FBXO22 C227 and C228 are involved in 22-SLF-mediated degradation of FKBP12.
Fig. 4: 22-SLF induces the formation of a ternary complex involving 22-SLF, FKBP12 and FBXO22.
Fig. 5: Harnessing FBXO22 for the degradation of BRD4 and EML4–ALK.

Similar content being viewed by others

Data availability

The mass spectrometry proteomics data were deposited to the ProteomeXchange Consortium through the PRIDE39 partner repository with the dataset identifier PXD050270. Protein sequences were retrieved from UniProt (https://www.uniprot.org) with the following accession codes: FBXO22 (human), Q8NEZ5; FBXO22 (mouse), Q78JE5. The predicted protein structure was retrieved from the AlphaFold Protein Structure Database (https://alphafold.ebi.ac.uk) under accession code FBXO22 (human), AF-Q8NEZ5-F1. The data supporting the findings of this study are available within the article and Supplementary Information. Source data are provided with this paper.

References

  1. Lai, A. C. & Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discov. 16, 101–114 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Nalawansha, D. A. & Crews, C. M. PROTACs: an emerging therapeutic modality in precision medicine. Cell Chem. Biol. 27, 998–1014 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kronke, J. et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, 301–305 (2014).

    Article  PubMed  Google Scholar 

  4. Lu, G. et al. The myeloma drug lenalidomide promotes the Cereblon-dependent destruction of Ikaros proteins. Science 343, 305–309 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Kannt, A. & Dikic, I. Expanding the arsenal of E3 ubiquitin ligases for proximity-induced protein degradation. Cell Chem. Biol. 28, 1014–1031 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Belcher, B. P., Ward, C. C. & Nomura, D. K. Ligandability of E3 ligases for targeted protein degradation applications. Biochemistry 62, 588–600 (2023).

    Article  CAS  PubMed  Google Scholar 

  7. Buckley, D. L. et al. Targeting the Von Hippel–Lindau E3 ubiquitin ligase using small molecules to disrupt the VHL/HIF-1α interaction. J. Am. Chem. Soc. 134, 4465–4468 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ito, T. et al. Identification of a primary target of thalidomide teratogenicity. Science 327, 1345–1350 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. Spradlin, J. N. et al. Harnessing the anti-cancer natural product nimbolide for targeted protein degradation. Nat. Chem. Biol. 15, 747–755 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tao, Y. et al. Targeted protein degradation by electrophilic PROTACs that stereoselectively and site-specifically engage DCAF1. J. Am. Chem. Soc. 144, 18688–18699 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang, X. et al. DCAF11 supports targeted protein degradation by electrophilic proteolysis-targeting chimeras. J. Am. Chem. Soc. 143, 5141–5149 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, X., Crowley, V. M., Wucherpfennig, T. G., Dix, M. M. & Cravatt, B. F. Electrophilic PROTACs that degrade nuclear proteins by engaging DCAF16. Nat. Chem. Biol. 15, 737–746 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ghandi, M. et al. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 569, 503–508 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Nusinow, D. P. et al. Quantitative proteomics of the Cancer Cell Line Encyclopedia. Cell 180, 387–402 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Khan, S. et al. A selective BCL-XL PROTAC degrader achieves safe and potent antitumor activity. Nat. Med. 25, 1938–1947 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Slabicki, M. et al. The CDK inhibitor CR8 acts as a molecular glue degrader that depletes cyclin K. Nature 585, 293–297 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Slabicki, M. et al. Small-molecule-induced polymerization triggers degradation of BCL6. Nature 588, 164–168 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Winter, G. E. et al. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 348, 1376–1381 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Nabet, B. et al. The dTAG system for immediate and target-specific protein degradation. Nat. Chem. Biol. 14, 431–441 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR–Cas9 complex. Nature 517, 583–588 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Tempest, P. A. Recent advances in heterocycle generation using the efficient Ugi multiple-component condensation reaction. Curr. Opin. Drug Discov. Devel. 8, 776–788 (2005).

    CAS  PubMed  Google Scholar 

  22. Soucy, T. A. et al. An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature 458, 732–736 (2009).

    Article  CAS  PubMed  Google Scholar 

  23. Skaar, J. R., Pagan, J. K. & Pagano, M. SCF ubiquitin ligase-targeted therapies. Nat. Rev. Drug Discov. 13, 889–903 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cheng, J. et al. Emerging role of FBXO22 in carcinogenesis. Cell Death Discov. 6, 66 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Tan, M. K., Lim, H. J. & Harper, J. W. SCFFBXO22 regulates histone H3 lysine 9 and 36 methylation levels by targeting histone demethylase KDM4A for ubiquitin-mediated proteasomal degradation. Mol. Cell. Biol. 31, 3687–3699 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Johmura, Y. et al. Fbxo22-mediated KDM4B degradation determines selective estrogen receptor modulator activity in breast cancer. J. Clin. Invest. 128, 5603–5619 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Johmura, Y. et al. SCFFbxo22-KDM4A targets methylated p53 for degradation and regulates senescence. Nat. Commun. 7, 10574 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang, L. et al. FBXO22 promotes the development of hepatocellular carcinoma by regulating the ubiquitination and degradation of p21. J. Exp. Clin. Cancer Res. 38, 101 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Ge, M. K. et al. FBXO22 degrades nuclear PTEN to promote tumorigenesis. Nat. Commun. 11, 1720 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tian, X. et al. F-box protein FBXO22 mediates polyubiquitination and degradation of KLF4 to promote hepatocellular carcinoma progression. Oncotarget 6, 22767–22775 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Zhu, X. N. et al. FBXO22 mediates polyubiquitination and inactivation of LKB1 to promote lung cancer cell growth. Cell Death Discov. 10, 486 (2019).

    Article  Google Scholar 

  32. Liang, D. et al. Protein CutA undergoes an unusual transfer into the secretory pathway and affects the folding, oligomerization, and secretion of acetylcholinesterase. J. Biol. Chem. 284, 5195–5207 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Vinogradova, E. V. et al. An activity-guided map of electrophile–cysteine interactions in primary human T cells. Cell 182, 1009–1026 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Backus, K. M. et al. Proteome-wide covalent ligand discovery in native biological systems. Nature 534, 570–574 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Casement, R., Bond, A., Craigon, C. & Ciulli, A. Mechanistic and structural features of PROTAC ternary complexes. Methods Mol. Biol. 2365, 79–113 (2021).

    Article  CAS  PubMed  Google Scholar 

  36. Hines, J., Lartigue, S., Dong, H., Qian, Y. & Crews, C. M. MDM2-recruiting PROTAC offers superior, synergistic antiproliferative activity via simultaneous degradation of BRD4 and stabilization of p53. Cancer Res. 79, 251–262 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Galkin, A. V. et al. Identification of NVP-TAE684, a potent, selective, and efficacious inhibitor of NPM-ALK. Proc. Natl Acad. Sci. USA 104, 270–275 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Koivunen, J. P. et al. EML4–ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin. Cancer Res. 14, 4275–4283 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Perez-Riverol, Y. et al. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 50, D543–D552 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge the support of the National Institutes of Health (NIH) R00 CA248715 (X.Z.), NIH T32 GM105538 (A.A.B.), NIH T32 GM149439 (A.M. and M.A.C.), National Science Foundation Graduate Research Fellowship Program (I.A.R.), Damon Runyon Cancer Research Foundation DFS-53-22 (X.Z.) and Illumina Pilot Project Program (X.Z.). We thank the Robert H. Lurie Comprehensive Cancer Center of Northwestern University for the use of the Flow Cytometry Core Facility. We thank H. Li for the helpful discussions regarding CRISPR library cloning and CRISPR screens.

Author information

Authors and Affiliations

Authors

Contributions

A.A.B. designed and conducted the biochemical and cellular experiments to demonstrate target degradation. C.Z. conducted the modeling study. C.Z. and M.A.C. characterized the compounds. I.A.R. and A.G.C. conducted the experiments to demonstrate the effectiveness of the GFP-based degradation platform. A.M., F.K. and X.Z. conducted the proteomics studies and performed data analysis. X.Z. supervised the project and, with contributions from all authors, wrote the paper.

Corresponding author

Correspondence to Xiaoyu Zhang.

Ethics declarations

Competing interests

A.A.B. and X.Z. are named on a patent application related to TPD, held by Northwestern University (US provisional patent application number 63/538,637). The other authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Milka Kostic, Brian Liau, Xiaobao Yang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Generation of CRISPR-Cas9 transcriptional activation cells for the discovery of E3 ligases supporting targeted protein degradation.

a, The construct of FKBP12-EGFP and a schematic representation of the generation of FKBP12-EGFP expressing HEK293T cells. b, Structures of Len-SLF and SLF. c, Fluorescence quantification of FKBP12-EGFP levels in HEK293T cells treated with 2 μM of Len-SLF or 20 μM of SLF for 24 hours. Data are presented as mean values +/��� SEM (n = 3 biological independent samples). The statistical significance was evaluated through unpaired two-tailed Student’s t-tests, comparing cells treated with Len-SLF or SLF to DMSO. Statistical significance denoted as ***P < 0.001 and ns: not significant. P value is 0.00026. d, The constructs used for the CRISPR-Cas9 transcriptional activation screen. e, Quantitative PCR analysis of IL1B mRNA levels subsequent to the transduction of sgRNAs targeting the promoter regions of the IL1B gene in HEK293T CRISPR-Cas9 transcriptional activation cells. The bar graph (n = 4 technical replicates) is representative of two independent experiments.

Source data

Extended Data Fig. 2 Gating strategy and procedure of fluorescence-activated cell sorting for the CRISPR-Cas9 transcriptional activation screen.

Cells were gated for singlets using forward and side scatter. GFP+ cells were gated for the subsequent sorting. Cells from the bottom 15% of the GFP population were sorted and harvested.

Extended Data Fig. 3 Compound screening to identify candidates for the CRIPSR activation screen.

a, HEK293T cell viability after treatment of FKBP12-directed bifunctional compounds (10 µM, 24 hours). Data are presented as mean values +/− SEM (n = 3 biological independent samples). b, Structures of five FKBP12-directed bifunctional compounds that show no significant cytotoxicity (cell viability > 50%) at 10 µM. c, The constructs of FKBP12-EGFP with SFFV and hPGK promoters. d, Fluorescence quantification of FKBP12-EGFP and mCherry levels in HEK293T cells stably expressing FKBP12-EGFP with SFFV or hPGK promoter. The bar graph (n = 8 technical replicates) is representative of two independent experiments with similar results. e, Fluorescence quantification of FKBP12-EGFP/mCherry levels in HEK293T cells stably expressing FKBP12-EGFP with SFFV or hPGK promoter, treated with 2 or 5 μM of candidate bifunctional compounds for 24 hours. Data are presented as mean values +/− SEM (n = 3 biological independent samples for compound treatment, n = 8 biological independent samples for DMSO treatment). The statistical significance was evaluated through unpaired two-tailed Student’s t-tests, comparing cells treated with 22-SLF or Len-SLF to DMSO. Statistical significance denoted as *P < 0.05, ***P < 0.001 and ns: not significant. P values are 0.000011 (2 μM Len-SLF in SFFV), 0.000051 (5 μM Len-SLF in SFFV), 0.039 (2 μM 22-SLF in hPGK), 0.031 (5 μM 22-SLF in hPGK), 0.00011 (2 μM Len-SLF in hPGK) and 0.000014 (5 μM Len-SLF in hPGK). f. Flow cytometry analysis of DMSO-treated cells revealed a silenced GFP population. The gating strategy was the same as described in Extended Data Fig. 2. The result is representative of two independent experiments with similar results.

Source data

Extended Data Fig. 4 An E3 ligase focused CRISPR-Cas9 transcriptional activation screen identifies DCAF16 supporting KB02-SLF-induced degradation of FKBP12-EGFP_NLS.

a, The construct of FKBP12-EGFP_NLS and a schematic representation of the steps in the CRISPR-Cas9 transcriptional activation screen. b, Volcano plot showing the E3 ligase focused CRISPR-Cas9 transcriptional activation screen for FKBP12-EGFP_NLS degradation after treatment of 2 μM KB02-SLF in HEK293T CRISPR-Cas9 transcriptional activation cells for 24 hours (n = 3 biological independent samples). P values were calculated by two-sided t test without adjustment.

Extended Data Fig. 5 22-SLF promotes FBXO22-dependent proteasomal degradation of FKBP12.

a, Gene expression ratio values of FBXO22 and CRBN between tumor and normal samples. Data is obtained from GEPIA (http://gepia.cancer-pku.cn/). Full names of the abbreviations are shown in Supplementary Table 1. b, Genomic PCR confirms FBXO22 knockout in A549, MDA-MB-231 and PC3 cells. The result is representative of two independent experiments with similar results. c, Global proteomic analysis confirms FBXO22 knockout in A549, MDA-MB-231 and PC3 cells. The result is representative of two independent experiments with similar results. d, 22-SLF promoted reduction in FKBP12 levels in MDA-MB-231 and PC3 wildtype, but not FBXO22 knockout cells. The bar graph represents quantification of the FLAG-FKBP12/HSP90 protein content. Data are presented as mean values (n = 2 biological independent samples). e, Global proteomic analysis in A549 wildtype and FBXO22 knockout cells treated with 22-SLF (2 μM, 24 hours) (n = 3 biological independent samples). P values were calculated by two-sided t test and adjusted using Benjamini-Hochberg correction for multiple comparisons. f, Bar graph quantification showing the change in KDM4A and KDM4B upon 22-SLF treatment in A549 wildtype and FBXO22 knockout cells. Data are presented as mean values +/− SEM (n = 2 biological independent samples for DMSO treated samples, n = 3 biological independent samples for 22-SLF samples). The statistical significance was evaluated through unpaired two-tailed Student’s t-tests, comparing cells treated with 22-SLF to DMSO. Statistical significance denoted as *P < 0.05 and ns: not significant. P value is 0.044.

Source data

Extended Data Fig. 6 22-SLF rescues Len-SLF-induced FKBP12 degradation in HEK293T FBXO22 knockout cells.

a, 22-biotin-conjugated streptavidin pull-down with lysates of HA-FBXO22-expressing HEK293T cells followed by proteomic analysis revealed FBXO22 as one of the protein targets bound by 22-biotin. Data are presented as mean values (n = 2 biological independent samples). b, HEK293T FBXO22 knockout cells pretreated with 22-SLF (0.1–25 µM, 2 hours) were treated with 0.5 µM of Len-SLF for 4 hours. The graph represents quantification of the FLAG-FKBP12/HSP90 protein content. Data are presented as mean values (n = 2 biological independent samples).

Source data

Extended Data Fig. 7 FBXO22 C227 and C228 are involved in 22-SLF-mediated degradation of FKBP12.

a, Interactome studies of FBXO22 wildtype and C227A/C228A double mutant revealed that both FBXO22 wildtype and C227A/C228A double mutant were assembled into the SKP1-CUL1-RBX1 E3 complex. Data are presented as mean values (n = 2 biological independent samples). b, Global proteomic analysis in HEK293T cells expressing HA-FBXO22 wildtype versus C227A/C228A double mutant treated with 22-SLF (0.5 μM, 24 hours) (n = 2 biological independent samples for DMSO treated samples, n = 3 biological independent samples for 22-SLF samples). P values were calculated by two-sided t test and adjusted using Benjamini-Hochberg correction for multiple comparisons.

Extended Data Fig. 8 Evaluation of the impact of FBXO22 K125, V230, and N257 on FKBP12 degradation induced by 22-SLF.

Single mutation of K125, V230 or N257 to alanine in FBXO22 did not block 22-SLF-induced degradation of FKBP12. The bar graph represents quantification of the FLAG-FKBP12/HSP90 protein content. Data are presented as mean values (n = 2 biological independent samples).

Source data

Extended Data Fig. 9 Global proteomic analysis in H2228 wildtype and FBXO22 knockout cells.

Proteome in H2228 wildtype and FBXO22 knockout cells was extracted, digested by LysC and trypsin, labeled by TMT tags, and analyzed via MS. Data are presented as mean values (n = 2 biological independent samples).

Extended Data Fig. 10 The acrylamide variant of 22-SLF, 22a-SLF, induced the degradation of FKBP12.

a, Structure of 22a-SLF. b, Comparison of FKBP12 degradation by 22-SLF and 22a-SLF. HEK293T cells expressing HA-FBXO22 were treated with 0.5, 1, or 2 µM of 22-SLF or 22a-SLF for 8 hours. The graph represents quantification of the FLAG-FKBP12/HSP90 protein content. Data are presented as mean values (n = 2 biological independent samples).

Source data

Supplementary information

Supplementary Information

Supplementary Table 1 and Note 1.

Reporting Summary

Supplementary Data 1

Supplementary data (see descriptions within file).

Source data

Source Data Fig. 1

Statistical source data for Fig. 1d,e.

Source Data Fig. 2

Statistical source data for Fig. 2a–d.

Source Data Fig. 2

Unprocessed western blots for Fig. 2.

Source Data Fig. 3

Statistical source data for Fig. 3b,c,g,h.

Source Data Fig. 3

Unprocessed western blots for Fig. 3.

Source Data Fig. 4

Statistical source data for Fig. 4a.

Source Data Fig. 4

Unprocessed western blots for Fig. 4.

Source Data Fig. 5

Statistical source data for Fig. 5b,e.

Source Data Fig. 5

Unprocessed western blots for Fig. 5.

Source Data Extended Data Fig. 1

Statistical source data for Extended Data Fig. 1c,e.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3a,d,e.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5a,c,d,f.

Source Data Extended Data Fig. 5

Unprocessed western blots for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical source data for Extended Data Fig. 6b.

Source Data Extended Data Fig. 6

Unprocessed western blots for Extended Data Fig. 6.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 8

Unprocessed western blots for Extended Data Fig. 8.

Source Data Extended Data Fig. 10

Statistical source data for Extended Data Fig. 10b.

Source Data Extended Data Fig. 10

Unprocessed western blots for Extended Data Fig. 10.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Basu, A.A., Zhang, C., Riha, I.A. et al. A CRISPR activation screen identifies FBXO22 supporting targeted protein degradation. Nat Chem Biol (2024). https://doi.org/10.1038/s41589-024-01655-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41589-024-01655-9

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research