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.

  • Review Article
  • Published:

Targeted protein degradation as a powerful research tool in basic biology and drug target discovery

Abstract

Controlled perturbation of protein activity is essential to study protein function in cells and living organisms. Small molecules that hijack the cellular protein ubiquitination machinery to selectively degrade proteins of interest, so-called degraders, have recently emerged as alternatives to selective chemical inhibitors, both as therapeutic modalities and as powerful research tools. These systems offer unprecedented temporal and spatial control over protein function. Here, we review recent developments in this field, with a particular focus on the use of degraders as research tools to interrogate complex biological problems.

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: Schematic representations of key concepts in protein degradation.
Fig. 2: An overview of degradation tag fusion strategies for targeted protein degradation.
Fig. 3: Schematic representations of targeted protein degradation strategies mediated by macromolecular conjugates.
Fig. 4: Validation methods for targeted protein degradation at different stages of the ubiquitin–proteasome pathway.

Similar content being viewed by others

References

  1. Ciechanover, A. Intracellular protein degradation: from a vague idea, through the lysosome and the ubiquitin-proteasome system, and onto human diseases and drug targeting (Nobel lecture). Angew. Chem. Int. Ed. Engl. 44, 5944–5967 (2005).

    CAS  PubMed  Google Scholar 

  2. Dikic, I. Proteasomal and autophagic degradation systems. Annu. Rev. Biochem. 86, 193–224 (2017).

    CAS  PubMed  Google Scholar 

  3. Zheng, N. & Shabek, N. Ubiquitin ligases: structure, function, and regulation. Annu. Rev. Biochem. 86, 129–157 (2017).

    CAS  PubMed  Google Scholar 

  4. King, R. W., Glotzer, M. & Kirschner, M. W. Mutagenic analysis of the destruction signal of mitotic cyclins and structural characterization of ubiquitinated intermediates. Mol. Biol. Cell 7, 1343–1357 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Takeuchi, J., Chen, H., Hoyt, M. A. & Coffino, P. Structural elements of the ubiquitin-independent proteasome degron of ornithine decarboxylase. Biochem. J. 410, 401–407 (2008).

    CAS  PubMed  Google Scholar 

  6. Park, E. C., Finley, D. & Szostak, J. W. A strategy for the generation of conditional mutations by protein destabilization. Proc. Natl Acad. Sci. USA 89, 1249–1252 (1992).

    CAS  PubMed  Google Scholar 

  7. Pfaff, P., Samarasinghe, K. T. G., Crews, C. M. & Carreira, E. M. Reversible spatiotemporal control of induced protein degradation by bistable photoPROTACs. ACS Cent. Sci. 5, 1682–1690 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Martin, R. et al. PHOTACs enable optical control of protein degradation. Sci. Adv. 6, eaay5064 (2020).

    Google Scholar 

  9. Renicke, C., Schuster, D., Usherenko, S., Essen, L.-O. & Taxis, C. A LOV2 domain-based optogenetic tool to control protein degradation and cellular function. Chem. Biol. 20, 619–626 (2013).

    CAS  PubMed  Google Scholar 

  10. Caussinus, E., Kanca, O. & Affolter, M. Fluorescent fusion protein knockout mediated by anti-GFP nanobody. Nat. Struct. Mol. Biol. 19, 117–121 (2012). This paper describes the engineering of a nanobody-based CRL substrate receptor for the degradation of GFP-fusion proteins.

    CAS  Google Scholar 

  11. Chung, H. K. et al. Tunable and reversible drug control of protein production via a self-excising degron. Nat. Chem. Biol. 11, 713–720 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Banaszynski, L. A., Chen, L. C., Maynard-Smith, L. A., Ooi, A. G. & Wandless, T. J. A rapid, reversible, and tunable method to regulate protein function in living cells using synthetic small molecules. Cell 126, 995–1004 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Nishimura, K., Fukagawa, T., Takisawa, H., Kakimoto, T. & Kanemaki, M. An auxin-based degron system for the rapid depletion of proteins in nonplant cells. Nat. Methods 6, 917–922 (2009).

    CAS  PubMed  Google Scholar 

  14. Koduri, V. et al. Peptidic degron for IMiD-induced degradation of heterologous proteins. Proc. Natl Acad. Sci. USA 116, 2539–2544 (2019).

    CAS  PubMed  Google Scholar 

  15. Nabet, B. et al. The dTAG system for immediate and target-specific protein degradation. Nat. Chem. Biol. 14, 431–441 (2018). This work describes the dTAG system and demonstrates its utility in target validation by degrading KRAS G12V.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Buckley, D. L. et al. HaloPROTACS: use of small molecule PROTACs to induce degradation of HaloTag fusion proteins. ACS Chem. Biol. 10, 1831–1837 (2015). This paper describes the development of HaloPROTAC for the degradation of Halo-tagged proteins.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Sakamoto, K. M. et al. Protacs: chimeric molecules that target proteins to the Skp1–Cullin–F box complex for ubiquitination and degradation. Proc. Natl Acad. Sci. USA 98, 8554–8559 (2001). This paper shows the first example of using chimeric molecules to redirect the specificity of a ubiquitin ligase toward a target protein of interest.

    CAS  PubMed  Google Scholar 

  18. Gray, W. M., Kepinski, S., Rouse, D., Leyser, O. & Estelle, M. Auxin regulates SCF(TIR1)-dependent degradation of AUX/IAA proteins. Nature 414, 271–276 (2001).

    CAS  PubMed  Google Scholar 

  19. Pettersson, M. & Crews, C. M. PROteolysis TArgeting Chimeras (PROTACs)—past, present and future. Drug Discov. Today Technol. 31, 15–27 (2019).

    PubMed  PubMed Central  Google Scholar 

  20. Churcher, I. Protac-induced protein degradation in drug discovery: breaking the rules or just making new ones? J. Med. Chem. 61, 444–452 (2018).

    CAS  PubMed  Google Scholar 

  21. Watt, G. F., Scott-Stevens, P. & Gaohua, L. Targeted protein degradation in vivo with Proteolysis Targeting Chimeras: current status and future considerations. Drug Discov. Today Technol. 31, 69–80 (2019).

    PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  23. Salami, J. & Crews, C. M. Waste disposal—an attractive strategy for cancer therapy. Science 355, 1163–1167 (2017).

    CAS  PubMed  Google Scholar 

  24. Bondeson, D. P. et al. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat. Chem. Biol. 11, 611–617 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Lu, J. et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem. Biol. 22, 755–763 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Krönke, J. et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, 301–305 (2014). This study and the concurrent study by Lu et al. (ref. 28) demonstrate how thalidomide promotes degradation of IKZF1/3 transcription factors.

    PubMed  Google Scholar 

  28. Lu, G. et al. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science 343, 305–309 (2014). This study and the concurrent study by Krönke et al. (ref. 27) demonstrate how thalidomide promotes degradation of IKZF1/3 transcription factors.

    CAS  PubMed  Google Scholar 

  29. Krönke, J., Hurst, S. N. & Ebert, B. L. Lenalidomide induces degradation of IKZF1 and IKZF3. Oncoimmunology 3, e941742 (2014).

    PubMed  PubMed Central  Google Scholar 

  30. Petzold, G., Fischer, E. S. & Thomä, N. H. Structural basis of lenalidomide-induced CK1α degradation by the CRL4CRBN ubiquitin ligase. Nature 532, 127–130 (2016).

    CAS  PubMed  Google Scholar 

  31. Matyskiela, M. E. et al. A novel cereblon modulator recruits GSPT1 to the CRL4CRBN ubiquitin ligase. Nature 535, 252–257 (2016).

    CAS  PubMed  Google Scholar 

  32. Donovan, K. A. et al. Thalidomide promotes degradation of SALL4, a transcription factor implicated in Duane Radial Ray syndrome. Elife 7, e38430 (2018).

    PubMed  PubMed Central  Google Scholar 

  33. Ito, T. et al. Identification of a primary target of thalidomide teratogenicity. Science 327, 1345–1350 (2010). This study identified cereblon as a target of thalidomide.

    CAS  PubMed  Google Scholar 

  34. Tan, X. et al. Mechanism of auxin perception by the TIR1 ubiquitin ligase. Nature 446, 640–645 (2007). This paper provides the structural basis for auxin perception, the first proof-of-principle study for a ‘molecular glue’ regulatory mechanism.

    CAS  PubMed  Google Scholar 

  35. Faust, T. B. et al. Structural complementarity facilitates E7820-mediated degradation of RBM39 by DCAF15. Nat. Chem. Biol. 16, 7–14 (2020).

    CAS  PubMed  Google Scholar 

  36. Bussiere, D. E. et al. Structural basis of indisulam-mediated RBM39 recruitment to DCAF15 E3 ligase complex. Nat. Chem. Biol. 16, 15–23 (2020).

    CAS  PubMed  Google Scholar 

  37. Du, X. et al. Structural basis and kinetic pathway of RBM39 recruitment to DCAF15 by a sulfonamide molecular glue E7820. Structure 27, 1625–1633.e3 (2019).

    CAS  PubMed  Google Scholar 

  38. Ting, T. C. et al. Aryl sulfonamides degrade RBM39 and RBM23 by recruitment to CRL4-DCAF15. Cell Rep. 29, 1499–1510.e6 (2019).

    CAS  PubMed  Google Scholar 

  39. Sheard, L. B. et al. Jasmonate perception by inositol-phosphate-potentiated COI1–JAZ co-receptor. Nature 468, 400–405 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Sievers, Q. L. et al. Defining the human C2H2 zinc finger degrome targeted by thalidomide analogs through CRBN. Science 362, eaat0572 (2018).

    PubMed  PubMed Central  Google Scholar 

  41. Huang, H.-T. et al. A chemoproteomic approach to query the degradable kinome using a multi-kinase degrader. Cell Chem. Biol. 25, 88–99.e6 (2018).

    CAS  PubMed  Google Scholar 

  42. Bondeson, D. P. et al. Lessons in PROTAC design from selective degradation with a promiscuous warhead. Cell Chem. Biol. 25, 78–87.e5 (2018).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Farnaby, W. et al. BAF complex vulnerabilities in cancer demonstrated via structure-based PROTAC design. Nat. Chem. Biol. 15, 672–680 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Pozo, J. C., Timpte, C., Tan, S., Callis, J. & Estelle, M. The ubiquitin-related protein RUB1 and auxin response in. Arabidopsis. Science 280, 1760–1763 (1998).

    CAS  PubMed  Google Scholar 

  47. Schwechheimer, C. et al. Interactions of the COP9 signalosome with the E3 ubiquitin ligase SCFTIR1 in mediating auxin response. Science 292, 1379–1382 (2001).

    CAS  PubMed  Google Scholar 

  48. D’Amato, R. J., Loughnan, M. S., Flynn, E. & Folkman, J. Thalidomide is an inhibitor of angiogenesis. Proc. Natl Acad. Sci. USA 91, 4082–4085 (1994).

    PubMed  Google Scholar 

  49. Pan, B. & Lentzsch, S. The application and biology of immunomodulatory drugs (IMiDs) in cancer. Pharmacol. Ther. 136, 56–68 (2012).

    CAS  PubMed  Google Scholar 

  50. Teo, S. et al. Thalidomide in the treatment of leprosy. Microbes Infect. 4, 1193–1202 (2002).

    CAS  PubMed  Google Scholar 

  51. Thomas, D. A. & Kantarjian, H. M. Current role of thalidomide in cancer treatment. Curr. Opin. Oncol. 12, 564–573 (2000).

    CAS  PubMed  Google Scholar 

  52. An, J. et al. pSILAC mass spectrometry reveals ZFP91 as IMiD-dependent substrate of the CRL4CRBN ubiquitin ligase. Nat. Commun. 8, 15398 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Gandhi, A. K. et al. Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4CRBN. Br. J. Haematol. 164, 811–821 (2014).

    CAS  PubMed  Google Scholar 

  54. Li, Y. et al. In vivo assessment of the effect of CYP1A2 inhibition and induction on pomalidomide pharmacokinetics in healthy subjects. J. Clin. Pharmacol. 58, 1295–1304 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Chen, N., Zhou, S. & Palmisano, M. Clinical pharmacokinetics and pharmacodynamics of lenalidomide. Clin. Pharmacokinet. 56, 139–152 (2017).

    CAS  PubMed  Google Scholar 

  56. Fink, E. C. et al. Crbn I391V is sufficient to confer in vivo sensitivity to thalidomide and its derivatives in mice. Blood 132, 1535–1544 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Uehara, T. et al. Selective degradation of splicing factor CAPERα by anticancer sulfonamides. Nat. Chem. Biol. 13, 675–680 (2017). This paper and the concurrent study by Han et al. (ref. 58) demonstrate how aryl-sulfonamides promote degradation of RBM39.

    CAS  PubMed  Google Scholar 

  58. Han, T. et al. Anticancer sulfonamides target splicing by inducing RBM39 degradation via recruitment to DCAF15. Science 356, eaal3755 (2017). This paper and the concurrent study by Uehara et al. (ref. 57) demonstrate how aryl-sulfonamides promote degradation of RBM39.

    PubMed  Google Scholar 

  59. Jia, X. et al. pSILAC method coupled with two complementary digestion approaches reveals PRPF39 as a new E7070-dependent DCAF15 substrate. J. Proteomics 210, 103545 (2020).

    PubMed  Google Scholar 

  60. Fischer, E. S. et al. Structure of the DDB1–CRBN E3 ubiquitin ligase in complex with thalidomide. Nature 512, 49–53 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Itoh, Y., Ishikawa, M., Naito, M. & Hashimoto, Y. Protein knockdown using methyl bestatin−ligand hybrid molecules: design and synthesis of inducers of ubiquitination-mediated degradation of cellular retinoic acid-binding proteins. J. Am. Chem. Soc. 132, 5820–5826 (2010).

    CAS  PubMed  Google Scholar 

  63. 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).

    CAS  PubMed  Google Scholar 

  64. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Ward, C. C. et al. Covalent ligand screening uncovers a RNF4 E3 ligase recruiter for targeted protein degradation applications. ACS Chem. Biol. 14, 2430–2440 (2019).

    CAS  PubMed  Google Scholar 

  67. Burslem, G. M., Song, J., Chen, X., Hines, J. & Crews, C. M. Enhancing antiproliferative activity and selectivity of a FLT-3 inhibitor by proteolysis targeting chimera conversion. J. Am. Chem. Soc. 140, 16428–16432 (2018).

    CAS  PubMed  Google Scholar 

  68. Burslem, G. M. et al. The advantages of targeted protein degradation over inhibition: an RTK case study. Cell Chem. Biol. 25, 67–77.e3 (2018).

    CAS  PubMed  Google Scholar 

  69. Li, W. et al. Phthalimide conjugations for the degradation of oncogenic PI3K. Eur. J. Med. Chem. 151, 237–247 (2018).

    CAS  Google Scholar 

  70. Tinworth, C. P. et al. PROTAC-mediated degradation of Bruton’s tyrosine kinase is inhibited by covalent binding. ACS Chem. Biol. 14, 342–347 (2019).

    CAS  PubMed  Google Scholar 

  71. Salami, J. et al. Androgen receptor degradation by the proteolysis-targeting chimera ARCC-4 outperforms enzalutamide in cellular models of prostate cancer drug resistance. Commun. Biol. 1, 100 (2018).

    PubMed  PubMed Central  Google Scholar 

  72. Yang, K. et al. Development of the first small molecule histone deacetylase 6 (HDAC6) degraders. Bioorg. Med. Chem. Lett. 28, 2493–2497 (2018).

    CAS  PubMed  Google Scholar 

  73. Bassi, Z. I. et al. Modulating PCAF/GCN5 immune cell function through a PROTAC approach. ACS Chem. Biol. 13, 2862–2867 (2018).

    CAS  PubMed  Google Scholar 

  74. Brand, M. et al. Homolog-selective degradation as a strategy to probe the function of CDK6 in AML. Cell Chem. Biol. 26, 300–306.e9 (2019).

    CAS  PubMed  Google Scholar 

  75. Jiang, B. et al. Development of dual and selective degraders of cyclin-dependent kinases 4 and 6. Angew. Chem. Int. Ed. Engl. 58, 6321–6326 (2019).

    CAS  PubMed  Google Scholar 

  76. Chessum, N. E. A. et al. Demonstrating in-cell target engagement using a pirin protein degradation probe (CCT367766). J. Med. Chem. 61, 918–933 (2018).

    CAS  PubMed  Google Scholar 

  77. Liu, J. et al. Light-induced control of protein destruction by opto-PROTAC. Sci. Adv. 6, eaay5154 (2020).

    PubMed  PubMed Central  Google Scholar 

  78. Xue, G., Wang, K., Zhou, D., Zhong, H. & Pan, Z. Light-induced protein degradation with photocaged PROTACs. J. Am. Chem. Soc. 141, 18370–18374 (2019).

    CAS  PubMed  Google Scholar 

  79. Naro, Y., Darrah, K. & Deiters, A. Optical control of small molecule-induced protein degradation. J. Am. Chem. Soc. 142, 2193–2197 (2020).

    CAS  PubMed  Google Scholar 

  80. Jin, Y. et al. Azo-PROTAC: novel light-controlled small-molecule tool for protein knockdown. J. Med. Chem. 63, 4644–4654 (2020).

    CAS  PubMed  Google Scholar 

  81. Zeng, M. et al. Exploring targeted degradation strategy for oncogenic KRASG12C. Cell Chem. Biol. 27, 19–31.e6 (2020).

    CAS  PubMed  Google Scholar 

  82. Li, S., Prasanna, X., Salo, V. T., Vattulainen, I. & Ikonen, E. An efficient auxin-inducible degron system with low basal degradation in human cells. Nat. Methods 16, 866–869 (2019).

    CAS  PubMed  Google Scholar 

  83. Madeira da Silva, L., Owens, K. L., Murta, S. M. F. & Beverley, S. M. Regulated expression of the Leishmania major surface virulence factor lipophosphoglycan using conditionally destabilized fusion proteins. Proc. Natl Acad. Sci. USA 106, 7583–7588 (2009).

    CAS  PubMed  Google Scholar 

  84. Armstrong, C. M. & Goldberg, D. E. An FKBP destabilization domain modulates protein levels in Plasmodium falciparum. Nat. Methods 4, 1007–1009 (2007).

    CAS  PubMed  Google Scholar 

  85. An, W. et al. Engineering FKBP-based destabilizing domains to build sophisticated protein regulation systems. PLoS ONE 10, e0145783 (2015).

    PubMed  PubMed Central  Google Scholar 

  86. Huang, H. T. et al. MELK is not necessary for the proliferation of basal-like breast cancer cells. Elife 6, e26693 (2017).

    PubMed  PubMed Central  Google Scholar 

  87. Erb, M. A. et al. Transcription control by the ENL YEATS domain in acute leukaemia. Nature 543, 270–274 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Neklesa, T. K. et al. Small-molecule hydrophobic tagging-induced degradation of HaloTag fusion proteins. Nat. Chem. Biol. 7, 538–543 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Raina, K. et al. Targeted protein destabilization reveals an estrogen-mediated ER stress response. Nat. Chem. Biol. 10, 957–962 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Los, G. V. et al. HaloTag: a novel protein labeling technology for cell imaging and protein analysis. ACS Chem. Biol. 3, 373–382 (2008).

    CAS  PubMed  Google Scholar 

  91. Tomoshige, S., Naito, M., Hashimoto, Y. & Ishikawa, M. Degradation of HaloTag-fused nuclear proteins using bestatin-HaloTag ligand hybrid molecules. Org. Biomol. Chem. 13, 9746–9750 (2015).

    CAS  PubMed  Google Scholar 

  92. Tovell, H. et al. Rapid and reversible knockdown of endogenously tagged endosomal proteins via an optimized HaloPROTAC degrader. ACS Chem. Biol. 14, 882–892 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. BasuRay, S., Wang, Y., Smagris, E., Cohen, J. C. & Hobbs, H. H. Accumulation of PNPLA3 on lipid droplets is the basis of associated hepatic steatosis. Proc. Natl Acad. Sci. USA 116, 9521–9526 (2019).

    CAS  PubMed  Google Scholar 

  94. Shin, Y. J. et al. Nanobody-targeted E3-ubiquitin ligase complex degrades nuclear proteins. Sci. Rep. 5, 14269 (2015).

    CAS  PubMed  Google Scholar 

  95. Clift, D. et al. A method for the acute and rapid degradation of endogenous proteins. Cell 171, 1692–1706.e18 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Chen, X. et al. Degradation of endogenous proteins and generation of a null-like phenotype in zebrafish using Trim-Away technology. Genome Biol. 20, 19 (2019).

    PubMed  PubMed Central  Google Scholar 

  97. Banik, S., Pedram, K., Wisnovsky, S., Riley, N. & Bertozzi, C. Lysosome targeting chimeras (LYTACs) for the degradation of secreted and membrane proteins. Preprint available at chemRxiv https://doi.org/10.26434/chemrxiv.7927061.v2 (2019). This paper shows the method of using chimeric macromolecular conjugates to target proteins for degradation by the lysosomal pathway.

  98. Fan, X., Jin, W. Y., Lu, J., Wang, J. & Wang, Y. T. Rapid and reversible knockdown of endogenous proteins by peptide-directed lysosomal degradation. Nat. Neurosci. 17, 471–480 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Li, W. et al. Chaperone-mediated autophagy: advances from bench to bedside. Neurobiol. Dis. 122, 41–48 (2019).

    CAS  PubMed  Google Scholar 

  100. Riching, K. M. et al. Quantitative live-cell kinetic degradation and mechanistic profiling of PROTAC mode of action. ACS Chem. Biol. 13, 2758–2770 (2018).

    CAS  PubMed  Google Scholar 

  101. Takahashi, D. et al. AUTACs: cargo-specific degraders using selective autophagy. Mol. Cell 76, 797–810.e10 (2019). This paper shows the first examples of small molecule degraders that target proteins to the lysosomal pathway for degradation.

    CAS  PubMed  Google Scholar 

  102. Matyskiela, M. E. et al. SALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrate. Nat. Chem. Biol. 14, 981–987 (2018).

    CAS  PubMed  Google Scholar 

  103. Hagner, P. R. et al. CC-122, a pleiotropic pathway modifier, mimics an interferon response and has antitumor activity in DLBCL. Blood 126, 779–789 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Matyskiela, M. E. et al. A cereblon modulator (CC-220) with improved degradation of Ikaros and Aiolos. J. Med. Chem. 61, 535–542 (2018).

    CAS  PubMed  Google Scholar 

  105. Gemechu, Y. et al. Humanized cereblon mice revealed two distinct therapeutic pathways of immunomodulatory drugs. Proc. Natl Acad. Sci. USA 115, 11802–11807 (2018).

    CAS  PubMed  Google Scholar 

  106. Nakazawa, N., Arakawa, O. & Yanagida, M. Condensin locates at transcriptional termination sites in mitosis, possibly releasing mitotic transcripts. Open Biol. 9, 190125 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Yoshiba, S. et al. HsSAS-6-dependent cartwheel assembly ensures stabilization of centriole intermediates. J. Cell Sci. 132, jcs217521 (2019).

    CAS  PubMed  Google Scholar 

  108. Goto, H. et al. Chk1-mediated Cdc25A degradation as a critical mechanism for normal cell cycle progression. J. Cell Sci. 132, jcs223123 (2019).

    CAS  PubMed  Google Scholar 

  109. Boija, A. et al. Transcription factors activate genes through the phase-separation capacity of their activation domains. Cell 175, 1842–1855.e16 (2018).

    CAS  PubMed  Google Scholar 

  110. Brunetti, L. et al. Mutant NPM1 maintains the leukemic state through HOX expression. Cancer Cell 34, 499–512.e9 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Fay, E. J. et al. Engineered small-molecule control of influenza A virus replication. J. Virol. 93, e01677–18 (2019).

    CAS  PubMed  Google Scholar 

  112. Rago, F. et al. Degron mediated BRM/SMARCA2 depletion uncovers novel combination partners for treatment of BRG1/SMARCA4-mutant cancers. Biochem. Biophys. Res. Commun. 508, 109–116 (2019).

    CAS  PubMed  Google Scholar 

  113. Zhu, W. et al. Precisely controlling endogenous protein dosage in hPSCs and derivatives to model FOXG1 syndrome. Nat. Commun. 10, 928 (2019).

    PubMed  PubMed Central  Google Scholar 

  114. Wu, Y., Yang, L., Chang, T., Kandeel, F. & Yee, J.-K. A small molecule-controlled Cas9 repressible system. Mol. Ther. Nucleic Acids 19, 922–932 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Roy, M. J. et al. SPR-measured dissociation kinetics of PROTAC ternary complexes influence target degradation rate. ACS Chem. Biol. 14, 361–368 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Robers, M. B. et al. Quantitative, real-time measurements of intracellular target engagement using energy transfer. Methods Mol. Biol. 1888, 45–71 (2019).

    CAS  PubMed  Google Scholar 

  117. Hjerpe, R. et al. Efficient protection and isolation of ubiquitylated proteins using tandem ubiquitin-binding entities. EMBO Rep. 10, 1250–1258 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Emanuele, M. J. et al. Global identification of modular cullin-RING ligase substrates. Cell 147, 459–474 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Feng, S. et al. Improved split fluorescent proteins for endogenous protein labeling. Nat. Commun. 8, 370 (2017).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  121. Schlierf, A. et al. Targeted inhibition of the COP9 signalosome for treatment of cancer. Nat. Commun. 7, 13166 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Anderson, D. J. et al. Targeting the AAA ATPase p97 as an approach to treat cancer through disruption of protein homeostasis. Cancer Cell 28, 653–665 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Hyer, M. L. et al. A small-molecule inhibitor of the ubiquitin activating enzyme for cancer treatment. Nat. Med. 24, 186–193 (2018).

    CAS  PubMed  Google Scholar 

  124. Goodsell, D. S., Autin, L. & Olson, A. J. Illustrate: software for biomolecular illustration. Structure 27, 1716–1720.e1 (2019).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all members of the Fischer lab for discussions and insights. This work was supported by NIH grants NCI R01CA214608 and R01CA218278 (to E.S.F.) and a Mark Foundation Emerging Leader Award (to E.S.F.). E.S.F. is a Damon Runyon-Rachleff Innovator supported in part by the Damon Runyon Cancer Research Foundation (DRR-50–18). H.Y. is supported by a Chleck Foundation fellowship.

Author information

Authors and Affiliations

Authors

Contributions

T.W., H.Y., Y.X., S.D-C., R.P.N. and E.S.F. contributed to writing the manuscript and preparing display items and have approved the final version of the manuscript.

Corresponding author

Correspondence to Eric S. Fischer.

Ethics declarations

Competing interests

E.S.F. is a founder, scientific advisory board (SAB) member and equity holder of Civetta Therapeutics. E.S.F. is a SAB member and equity holder of C4 Therapeutics. E.S.F. is or has consulted for Novartis, AbbVie, Astellas, Deerfield, EcoR1 and Pfizer. The Fischer lab receives or has received research funding from Novartis, Deerfield and Astellas.

Additional information

Editor recognition statement Katarzyna Marcinkiewicz and Anke Sparmann were the primary editors on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, T., Yoon, H., Xiong, Y. et al. Targeted protein degradation as a powerful research tool in basic biology and drug target discovery. Nat Struct Mol Biol 27, 605–614 (2020). https://doi.org/10.1038/s41594-020-0438-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-020-0438-0

This article is cited by

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