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:

Harnessing the anti-cancer natural product nimbolide for targeted protein degradation

Abstract

Nimbolide, a terpenoid natural product derived from the Neem tree, impairs cancer pathogenicity; however, the direct targets and mechanisms by which nimbolide exerts its effects are poorly understood. Here, we used activity-based protein profiling (ABPP) chemoproteomic platforms to discover that nimbolide reacts with a novel functional cysteine crucial for substrate recognition in the E3 ubiquitin ligase RNF114. Nimbolide impairs breast cancer cell proliferation in-part by disrupting RNF114-substrate recognition, leading to inhibition of ubiquitination and degradation of tumor suppressors such as p21, resulting in their rapid stabilization. We further demonstrate that nimbolide can be harnessed to recruit RNF114 as an E3 ligase in targeted protein degradation applications and show that synthetically simpler scaffolds are also capable of accessing this unique reactive site. Our study highlights the use of ABPP platforms in uncovering unique druggable modalities accessed by natural products for cancer therapy and targeted protein degradation applications.

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: Nimbolide impairs breast cancer cell proliferation and survival.
Fig. 2: isoTOP-ABPP analysis of nimbolide in 231MFP breast cancer cell proteomes reveal RNF114 as a target.
Fig. 3: Nimbolide reacts covalently with C8 of RNF114.
Fig. 4: Nimbolide inhibits RNF114 activity through disrupting substrate recognition.
Fig. 5: Nimbolide can be used to recruit RNF114 for targeted protein degradation of BRD4.

Similar content being viewed by others

Data availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Nomura, D. K. & Maimone, T. J. Target identification of bioactive covalently acting natural products. Curr. Top. Microbiol. Immunol. 420, 351–374 (2018).

    Google Scholar 

  2. Drahl, C., Cravatt, B. F. & Sorensen, E. J. Protein-reactive natural products. Angew. Chem. Int. Ed Engl. 44, 5788–5809 (2005).

    Article  CAS  Google Scholar 

  3. Liu, J. et al. Calcineurin is a common target of cyclophilin-cyclosporin A and FKBP-FK506 complexes. Cell 66, 807–815 (1991).

    Article  CAS  Google Scholar 

  4. Cohen, E., Quistad, G. B. & Casida, J. E. Cytotoxicity of nimbolide, epoxyazadiradione and other limonoids from neem insecticide. Life Sci. 58, 1075–1081 (1996).

    Article  CAS  Google Scholar 

  5. Bodduluru, L. N., Kasala, E. R., Thota, N., Barua, C. C. & Sistla, R. Chemopreventive and therapeutic effects of nimbolide in cancer: the underlying mechanisms. Toxicol. In Vitro 28, 1026–1035 (2014).

    Article  CAS  Google Scholar 

  6. Subramani, R. et al. Nimbolide inhibits pancreatic cancer growth and metastasis through ROS-mediated apoptosis and inhibition of epithelial-to-mesenchymal transition. Sci. Rep. 6, 19819 (2016).

    Article  CAS  Google Scholar 

  7. Hao, F., Kumar, S., Yadav, N. & Chandra, D. Neem components as potential agents for cancer prevention and treatment. Biochim. Biophys. Acta 1846, 247–257 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gupta, S. C., Prasad, S., Tyagi, A. K., Kunnumakkara, A. B. & Aggarwal, B. B. Neem (Azadirachta indica): an Indian traditional panacea with modern molecular basis. Phytomedicine 34, 14–20 (2017).

    Article  CAS  Google Scholar 

  9. Burslem, G. M. & Crews, C. M. Small-molecule modulation of protein homeostasis. Chem. Rev. 117, 11269–11301 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. Bianchini, G., Balko, J. M., Mayer, I. A., Sanders, M. E. & Gianni, L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat. Rev. Clin. Oncol. 13, 674–690 (2016).

    Article  CAS  Google Scholar 

  12. Weerapana, E. et al. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–795 (2010).

    Article  CAS  Google Scholar 

  13. Roberts, A. M., Ward, C. C. & Nomura, D. K. Activity-based protein profiling for mapping and pharmacologically interrogating proteome-wide ligandable hotspots. Curr. Opin. Biotechnol. 43, 25–33 (2017).

    Article  CAS  Google Scholar 

  14. Grossman, E. A. et al. Covalent ligand discovery against druggable hotspots targeted by anti-cancer natural products. Cell Chem. Biol. 24, 1368–1376.e4 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  16. Wang, C., Weerapana, E., Blewett, M. M. & Cravatt, B. F. A chemoproteomic platform to quantitatively map targets of lipid-derived electrophiles. Nat. Methods 11, 79–85 (2014).

    Article  Google Scholar 

  17. Han, J. et al. ZNF313 is a novel cell cycle activator with an E3 ligase activity inhibiting cellular senescence by destabilizingp21(WAF1.). Cell Death Differ. 20, 1055–1067 (2013).

    Article  CAS  Google Scholar 

  18. Lee, M.-G. et al. XAF1 directs apoptotic switch of p53 signaling through activation of HIPK2 and ZNF313. Proc. Natl Acad. Sci. USA 111, 15532–15537 (2014).

    Article  CAS  Google Scholar 

  19. Huang, S. et al. The UbL-UBA Ubiquilin4 protein functions as a tumor suppressor in gastric cancer by p53-dependent and p53-independent regulation of p21. Cell Death Differ. 26, 516–530 (2019).

    Article  CAS  Google Scholar 

  20. Abbas, T. & Dutta, A. p21 in cancer: intricate networks and multiple activities. Nat. Rev. Cancer 9, 400–414 (2009).

    Article  CAS  Google Scholar 

  21. Guo, H., Tian, T., Nan, K. & Wang, W. p57: a multifunctional protein in cancer (Review). Int. J. Oncol. 36, 1321–1329 (2010).

    Article  CAS  Google Scholar 

  22. Zengerle, M., Chan, K.-H. & Ciulli, A. Selective small molecule induced degradation of the BET bromodomain protein BRD4. ACS Chem. Biol. 10, 1770–1777 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  24. Havens, C. G. & Walter, J. C. Mechanism of CRL4(Cdt2), a PCNA-dependent E3 ubiquitin ligase. Genes Dev. 25, 1568–1582 (2011).

    Article  CAS  Google Scholar 

  25. Kitagawa, K., Kotake, Y. & Kitagawa, M. Ubiquitin-mediated control of oncogene and tumor suppressor gene products. Cancer Sci. 100, 1374–1381 (2009).

    Article  CAS  Google Scholar 

  26. Biswas, K. et al. The E3 ligase CHIP mediates p21 degradation to maintain radioresistance. Mol. Cancer Res. 15, 651–659 (2017).

    Article  CAS  Google Scholar 

  27. Rodriguez, M. S. et al. The RING ubiquitin E3 RNF114 interacts with A20 and modulates NF-κB activity and T-cell activation. Cell Death Dis. 5, e1399 (2014).

    Article  CAS  Google Scholar 

  28. Yang, Y. et al. The E3 ubiquitin ligase RNF114 and TAB1 degradation are required for maternal-to-zygotic transition. EMBO Rep. 18, 205–216 (2017).

    Article  CAS  Google Scholar 

  29. Rape, M. Ubiquitylation at the crossroads of development and disease. Nat. Rev. Mol. Cell Biol. 19, 59–70 (2018).

    Article  CAS  Google Scholar 

  30. Hughes, S. J. & Ciulli, A. Molecular recognition of ternary complexes: a new dimension in the structure-guided design of chemical degraders. Essays Biochem. 61, 505–516 (2017).

    Article  Google Scholar 

  31. Gadd, M. S. et al. Structural basis of PROTAC cooperative recognition for selective protein degradation. Nat. Chem. Biol. 13, 514–521 (2017).

    Article  CAS  Google Scholar 

  32. Nowak, R. P. et al. Plasticity in binding confers selectivity in ligand-induced protein degradation. Nat. Chem. Biol. 14, 706–714 (2018).

    Article  CAS  Google Scholar 

  33. Jessani, N. et al. Carcinoma and stromal enzyme activity profiles associated with breast tumor growth in vivo. Proc. Natl Acad. Sci. USA 101, 13756–13761 (2004).

    Article  CAS  Google Scholar 

  34. Anderson, K. E., To, M., Olzmann, J. A. & Nomura, D. K. Chemoproteomics-enabled covalent ligand screening reveals a thioredoxin-caspase 3 interaction disruptor that impairs breast cancer pathogenicity. ACS Chem. Biol. 12, 2522–2528 (2017).

    Article  CAS  Google Scholar 

  35. Smith, P. K. et al. Measurement of protein using bicinchoninic acid. Anal. Biochem. 150, 76–85 (1985).

    Article  CAS  Google Scholar 

  36. Xu, T. et al. ProLuCID: an improved SEQUEST-like algorithm with enhanced sensitivity and specificity. J. Proteomics 129, 16–24 (2015).

    Article  CAS  Google Scholar 

  37. Bateman, L. A. et al. Chemoproteomics-enabled covalent ligand screen reveals a cysteine hotspot in reticulon 4 that impairs ER morphology and cancer pathogenicity. Chem. Commun. (Camb.) 53, 7234–7237 (2017).

    Article  CAS  Google Scholar 

  38. Counihan, J. L.., Wiggenhorn, A. L.., Anderson, K. E.. & Nomura, D. K.. Chemoproteomics-enabled covalent ligand screening reveals ALDH3A1 as a lung cancer therapy target. ACS Chem. Biol. 13, 1970–1977 (2018).

    Article  CAS  Google Scholar 

  39. Roberts, A. M. et al. Chemoproteomic screening of covalent ligands reveals UBA5 As a novel pancreatic cancer target. ACS Chem. Biol. 12, 899–904 (2017).

    Article  CAS  Google Scholar 

  40. Kokosza, K., Balzarini, J. & Piotrowska, D. G. Novel 5-arylcarbamoyl-2-methylisoxazolidin-3-yl-3-phosphonates as nucleotide analogues. Nucleosides Nucleotides Nucleic Acids 33, 552–582 (2014).

    Article  CAS  Google Scholar 

  41. Talaty, E. R., Young, S. M., Dain, R. P. & Stipdonk, M. J. V. A study of fragmentation of protonated amides of some acylated amino acids by tandem mass spectrometry: observation of an unusual nitrilium ion. Rapid Commun. Mass Spectrom. 25, 1119–1129 (2011).

    Article  CAS  Google Scholar 

  42. Timokhin, V. I., Gastaldi, S., Bertrand, M. P. & Chatgilialoglu, C. Rate constants for the β-elimination of tosyl radical from a variety of substituted carbon-centered radicals. J. Org. Chem. 68, 3532–3537 (2003).

    Article  CAS  Google Scholar 

  43. Cee, V. J. et al. Systematic study of the glutathione (GSH) reactivity of N-arylacrylamides: 1. Effects of aryl substitution. J. Med. Chem. 58, 9171–9178 (2015).

    Article  CAS  Google Scholar 

  44. Le Sann, C., Huddleston, J. & Mann, J. Synthesis and preliminary evaluation of novel analogues of quindolines as potential stabilisers of telomeric G-quadruplex DNA. Tetrahedron 63, 12903–12911 (2007).

    Article  Google Scholar 

  45. Ikoma, M., Oikawa, M. & Sasaki, M. Synthesis and domino metathesis of functionalized 7-oxanorbornene analogs toward cis-fused heterocycles. Tetrahedron 64, 2740–2749 (2008).

    Article  CAS  Google Scholar 

  46. Cho, S.-D. et al. A one-pot synthesis of pyrido[2,3-b][1,4]oxazin-2-ones. J. Org. Chem. 68, 7918–7920 (2003).

    Article  CAS  Google Scholar 

  47. Magolan, J., Carson, C. A. & Kerr, M. A. Total synthesis of (±)-mersicarpine. Org. Lett. 10, 1437–1440 (2008).

    Article  CAS  Google Scholar 

  48. Longo, P. A., Kavran, J. M., Kim, M.-S. & Leahy, D. J. Transient mammalian cell transfection with polyethylenimine (PEI). Methods Enzymol 529, 227–240 (2013).

    Article  CAS  Google Scholar 

  49. Li, C. et al. FastCloning: a highly simplified, purification-free, sequence- and ligation-independent PCR cloning method. BMC Biotechnol. 11, 92 (2011).

    Article  CAS  Google Scholar 

  50. Thomas, J. R. et al. A photoaffinity labeling-based chemoproteomics strategy for unbiased target deconvolution of small molecule drug candidates. Methods Mol. Biol. 1647, 1–18 (2017).

    Article  Google Scholar 

  51. Käll, L., Canterbury, J. D., Weston, J., Noble, W. S. & MacCoss, M. J. Semi-supervised learning for peptide identification from shotgun proteomics datasets. Nat. Methods 4, 923–925 (2007).

    Article  Google Scholar 

Download references

Acknowledgements

We thank the members of the Nomura Research Group, the Maimone laboratory and Novartis Institutes for BioMedical Research for critical reading of the manuscript. We acknowledge M. Moeller and A. Olding for assistance in nimbolide isolation studies. This work was supported by Novartis Institutes for BioMedical Research and the Novartis-Berkeley Center for Proteomics and Chemistry Technologies (NB-CPACT) for all listed authors. This work was also supported by grants from the National Institutes of Health (no. R01CA172667 for D.K.N., J.N.S., C.C.W. and L.O.; no. F31CA225173 for C.C.W.; no. F31CA239327 for J.N.S. and no. R01GM112948 for J.A.O.). This work was also supported by the Mark Foundation for Cancer Research ASPIRE award. J.A.O. is a Chan Zuckerberg Biohub investigator.

Author information

Authors and Affiliations

Authors

Contributions

J.N.S., T.J.M. and D.K.N. conceived the project and wrote the paper. J.N.S., X.H., C.C.W., M.D.J., D.E.B., J.R.T., J.A.T., J.M.K., M.S., T.J.M. and D.K.N. provided intellectual contributions and insights into project direction. J.N.S., X.H., S.M.B., M.T., J.A.O., M.S., T.J.M. and D.K.N. designed the experiments and analyzed data. C.C.W. and M.D.J. developed bioinformatic methods and analyzed data for proteomics experiments. J.N.S., X.H., S.M.B., L.O., M.T., A.P., E.O., M.W. and D.K.N. performed experiments and analyzed data. A.P., E.O., M.W. and D.E.B. provided pure RNF114 protein. J.N.S., X.H. and T.J.M. designed and synthesized compounds. J.N.S., S.M.B., M.D.J., J.A.O., D.E.B., J.R.T., J.A.T., J.M.K., M.S., T.J.M. and D.K.N. edited the paper.

Corresponding authors

Correspondence to Thomas J. Maimone or Daniel K. Nomura.

Ethics declarations

Competing interests

S.M.B., M.D.J., A.P., E.O., M.W., D.E.B., J.A.T., J.M.K. and M.S. are employees of Novartis Institutes for BioMedical Research. J.R.T. was an employee of Novartis Institutes for BioMedical Research when this study was submitted, but is now an employee of Vertex Pharmaceuticals. This study was funded by the Novartis Institutes for BioMedical Research and the Novartis-Berkeley Center for Proteomics and Chemistry Technologies. D.K.N. is a co-founder, share-holder and adviser for Artris Therapeutics and Frontier Medicines.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figures 1–13

Reporting Summary

Supplementary Note

Synthetic Procedures

Supplementary Dataset 1

IsoTOP-ABPP analysis of nimbolide treatment in situ in 231MFP breast cancer cells.

Supplementary Dataset 2

TMT-based quantitative proteomic analysis of proteins enriched by nimbolide-alkyne probe in situ treatment in 231MFP breast cancer cells.

Supplementary Dataset 3

TMT-based quantitative proteomic profiling of XH2 treatment in 231MFP breast cancer cells.

Supplementary Dataset 4

Structures of covalent ligands screened against RNF114.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Spradlin, J.N., Hu, X., Ward, C.C. et al. Harnessing the anti-cancer natural product nimbolide for targeted protein degradation. Nat Chem Biol 15, 747–755 (2019). https://doi.org/10.1038/s41589-019-0304-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-019-0304-8

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