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:

Drug discovery in the ubiquitin–proteasome system

Key Points

  • The ubiquitin–proteasome pathway promotes the degradation of proteins by first catalysing the formation of a polyubiquitin chain on the target protein. This chain is then recognized by the proteasome, allowing the target protein to be degraded. Cancer cells seem to be much more sensitive to inhibition of the proteasome than normal cells. Recent studies have revealed that peptide inhibitors of the proteasome have efficacy as antitumour agents in humans for particular types of cancer (for example, multiple myeloma). This brings the ubiquitin–proteasome system to the forefront as a target for drug discovery.

  • The ubiquitin–proteasome pathway contains a large number of components which function in specific biochemical pathways. Some of these components, such as E1-activating enzymes, are conventional enzymes and are therefore potentially amenable to drug development, whereas other components of the system are non-conventional targets for which extensive work will be required to assess their suitability as drug targets.

  • Much of our understanding of how the ubiquitin–proteasome pathway is deregulated in human disease comes from the cancer field, where it is found that certain ubiquitin ligases are overexpressed in tumours and promote the degradation of negative regulators of cell proliferation. In principle, inhibition of some of these central regulators (SKP2 and MDM2) could serve to reduce cell proliferation, and these types of molecules represent important clinical targets.

  • The other class of alteration represents mutations in ubiquitin ligases which negatively affect their activities. In the cancer setting, mutations have been found in ubiquitin ligases in cancer, including the BRCA1 and FBW7 proteins. These types of defects will be much harder to develop drugs for because it would require reactivation of a non-functional allele. Such mutations have also been seen in other classes of diseases, including Parkinson's disease in which the parkin ubiquitin ligase has been found to be mutated in its E2-binding domain.

  • Recent work using chemical library approaches to identify compounds that inhibit protein degradation have led to the realization that the polyubiquitin chain itself can be a drug target. Ubistatin is a small molecule that interacts specificially with polyubiquitin chains and blocks their interactions with receptors on the proteasome. This opens up a new area in which a non-traditional target may be employed to inhibit protein turnover, although how this type of inhibitor could achieve specificity is not clear at present.

Abstract

Regulated protein turnover via the ubiquitin–proteasome system (UPS) underlies a wide variety of signalling pathways, from cell-cycle control and transcription to development. Recent evidence that pharmacological inhibition of the proteasome can be efficacious in the treatment of human cancers has set the stage for attempts to selectively inhibit the activities of disease-specific components of the UPS. Here, we review recent advances linking UPS components with specific human diseases, most prominently cancer and neurodegenerative disorders, and emphasize potential sites of therapeutic intervention along the regulated protein-degradation pathway.

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: Overview of the ubiquitin–proteasome system (UPS).
Figure 2: Biochemical steps in the ubiquitin-activation reaction.
Figure 3: Ubiquitin-activating enzymes as potential pharmacological targets.
Figure 4: Ubiquitin ligases as ubiquitylation specificity modules.
Figure 5: RING-finger domain.
Figure 6: p53–MDM2 interaction as a therapeutic target in human cancer.
Figure 7: Recognition of phosphorylated substrates by SCF ubiquitin ligases.
Figure 8: Structure of HECT-domain ubiquitin ligases.

Similar content being viewed by others

References

  1. Pickart, C. M. Back to the future with ubiquitin. Cell 116, 181–190 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Wilkinson, K. D. Ubiquitin: a Nobel protein. Cell 119, 741–745 (2004).

    CAS  PubMed  Google Scholar 

  3. Hershko, A., Ciechanover, A. & Varshavsky, A. Basic Medical Research Award. The ubiquitin system. Nature Med. 6, 1073–1081 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Ang, X. L. & Harper, J. W. Interwoven ubiquitination oscillators and control of cell cycle transitions. Sci. STKE pe31 (2004).

  5. Moberg, K. H., Bell, D. W., Wahrer, D. C., Haber, D. A. & Hariharan, I. K. Archipelago regulates Cyclin E levels in Drosophila and is mutated in human cancer cell lines. Nature 413, 311–316 (2001).

    Article  CAS  PubMed  Google Scholar 

  6. Koh, M. S., Ittmann, M., Kadmon, D., Thompson, T. C. & Leach, F. S. CDC4 gene expression as potential biomarker for targeted therapy in prostate cancer. Cancer Biol. Ther. 5, 78–83 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Ruffner, H., Joazeiro, C. A., Hemmati, D., Hunter, T. & Verma, I. M. Cancer-predisposing mutations within the RING domain of BRCA1: loss of ubiquitin protein ligase activity and protection from radiation hypersensitivity. Proc. Natl Acad. Sci. USA 98, 5134–5139 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ekholm-Reed, S. et al. Mutation of hCDC4 leads to cell cycle deregulation of cyclin E in cancer. Cancer Res. 64, 795–800 (2004).

    Article  PubMed  Google Scholar 

  9. Gandhi, S. & Wood, N. W. Molecular pathogenesis of Parkinson's disease. Hum. Mol. Genet. 14 (Spec. No. 2), 2749–2755 (2005).

    Article  PubMed  CAS  Google Scholar 

  10. Gasser, T. Genetics of Parkinson's disease. Curr. Opin. Neurol. 18, 363–369 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Dauer, W. & Przedborski, S. Parkinson's disease: mechanisms and models. Neuron 39, 889–909 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Vila, M. & Przedborski, S. Genetic clues to the pathogenesis of Parkinson's disease. Nature Med. 10 (Suppl.), S58–S62 (2004).

    Article  PubMed  CAS  Google Scholar 

  13. Tanaka, K., Suzuki, T., Hattori, N. & Mizuno, Y. Ubiquitin, proteasome and parkin. Biochim. Biophys. Acta 1695, 235–247 (2004).

    Article  CAS  PubMed  Google Scholar 

  14. Al-Kuraya, K. et al. Prognostic relevance of gene amplifications and coamplifications in breast cancer. Cancer Res. 64, 8534–8540 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. Honda, R., Tanaka, H. & Yasuda, H. Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett. 420, 25–27 (1997).

    Article  CAS  PubMed  Google Scholar 

  16. Iwakuma, T. & Lozano, G. MDM2, an introduction. Mol. Cancer Res. 1, 993–1000 (2003).

    CAS  PubMed  Google Scholar 

  17. Nakayama, T. et al. MDM2 gene amplification in bone and soft-tissue tumors: association with tumor progression in differentiated adipose-tissue tumors. Int. J. Cancer 64, 342–346 (1995).

    Article  CAS  PubMed  Google Scholar 

  18. Shieh, S. Y., Ikeda, M., Taya, Y. & Prives, C. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell 91, 325–334 (1997).

    Article  CAS  PubMed  Google Scholar 

  19. Kussie, P. H. et al. Structure of the MDM2 oncoprotein bound to the p53 tumor suppressor transactivation domain. Science 274, 948–953 (1996).

    Article  CAS  PubMed  Google Scholar 

  20. Montes de Oca Luna, R., Wagner, D. S. & Lozano, G. Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature 378, 203–206 (1995).

    Article  CAS  PubMed  Google Scholar 

  21. Li, M. et al. Mono- versus polyubiquitination: differential control of p53 fate by Mdm2. Science 302, 1972–1975 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Carrano, A. C., Eytan, E., Hershko, A. & Pagano, M. SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27. Nature Cell Biol. 1, 193–199 (1999).

    Article  CAS  PubMed  Google Scholar 

  23. Hershko, D. et al. Inverse relation between levels of p27(Kip1) and of its ubiquitin ligase subunit Skp2 in colorectal carcinomas. Cancer 91, 1745–1751 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Kossatz, U. et al. Skp2-dependent degradation of p27kip1 is essential for cell cycle progression. Genes Dev. 18, 2602–2607 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Nakayama, K. et al. Targeted disruption of Skp2 results in accumulation of cyclin E and p27(Kip1), polyploidy and centrosome overduplication. EMBO J. 19, 2069–2081 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nakayama, K. et al. Skp2-mediated degradation of p27 regulates progression into mitosis. Dev. Cell 6, 661–672 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Nijman, S. M. et al. A genomic and functional inventory of deubiquitinating enzymes. Cell 123, 773–786 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Rubinfeld, B. et al. Stabilization of β-catenin by genetic defects in melanoma cell lines. Science 275, 1790–1792 (1997).

    Article  CAS  PubMed  Google Scholar 

  29. Welcker, M. et al. The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation. Proc. Natl Acad. Sci. USA 101, 9085–9090 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wei, W., Jin, J., Schlisio, S., Harper, J. W. & Kaelin, W. G. Jr. The v-Jun point mutation allows c-Jun to escape GSK3-dependent recognition and destruction by the Fbw7 ubiquitin ligase. Cancer Cell 8, 25–33 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Adams, J. The development of proteasome inhibitors as anticancer drugs. Cancer Cell 5, 417–421 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Burger, A. M. & Seth, A. K. The ubiquitin-mediated protein degradation pathway in cancer: therapeutic implications. Eur. J. Cancer 40, 2217–2229 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Orlowski, R. Z. et al. Phase I trial of the proteasome inhibitor PS-341 in patients with refractory hematologic malignancies. J. Clin. Oncol. 20, 4420–4427 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Richardson, P. G. et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. N. Engl. J. Med. 348, 2609–2617 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Richardson, P. G. et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N. Engl. J. Med. 352, 2487–2498 (2005). This paper, and Reference 34, provide analysis of data from clinical trials on the proteasome inhibitor bortezomib indicating that the proteasome inhibitor is superior to high-dose dexamethasone in the treatment of relapsed multiple myeloma.

    Article  CAS  PubMed  Google Scholar 

  36. Nalepa, G. & Wade Harper, J. Therapeutic anti-cancer targets upstream of the proteasome. Cancer Treat. Rev. 29 (Suppl. 1), 49–57 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Kyle, R. A. & Rajkumar, S. V. Multiple myeloma. N. Engl. J. Med. 351, 1860–1873 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Pickart, C. M. Mechanisms underlying ubiquitination. Annu. Rev. Biochem. 70, 503–533 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Hershko, A., Heller, H., Elias, S. & Ciechanover, A. Components of ubiquitin-protein ligase system. Resolution, affinity purification & role in protein breakdown. J. Biol. Chem. 258, 8206–8214 (1983). A landmark study that used affinity chromatography with ubiquitin–sepharose to separate and reconstitute the E1–E2–E3 ubiquitin conjugation system, demonstrating that all three components are required for conjugation of ubiquitin to lysine residues in the substrate.

    Article  CAS  PubMed  Google Scholar 

  40. Ciechanover, A., Elias, S., Heller, H. & Hershko, A. 'Covalent affinity' purification of ubiquitin-activating enzyme. J. Biol. Chem. 257, 2537–2542 (1982).

    Article  CAS  PubMed  Google Scholar 

  41. Ciechanover, A., Elias, S., Heller, H., Ferber, S. & Hershko, A. Characterization of the heat-stable polypeptide of the ATP-dependent proteolytic system from reticulocytes. J. Biol. Chem. 255, 7525–7528 (1980).

    Article  CAS  PubMed  Google Scholar 

  42. Ciechanover, A., Heller, H., Elias, S., Haas, A. L. & Hershko, A. ATP-dependent conjugation of reticulocyte proteins with the polypeptide required for protein degradation. Proc. Natl Acad. Sci. USA 77, 1365–1368 (1980).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Vijay-Kumar, S., Bugg, C. E. & Cook, W. J. Structure of ubiquitin refined at 1. 8 A resolution. J. Mol. Biol. 194, 531–544 (1987).

    Article  CAS  PubMed  Google Scholar 

  44. Chau, V. et al. A multiubiquitin chain is confined to specific lysine in a targeted short-lived protein. Science 243, 1576–1583 (1989).

    Article  CAS  PubMed  Google Scholar 

  45. Gregori, L., Poosch, M. S., Cousins, G. & Chau, V. A uniform isopeptide-linked multiubiquitin chain is sufficient to target substrate for degradation in ubiquitin-mediated proteolysis. J. Biol. Chem. 265, 8354–8357 (1990).

    Article  CAS  PubMed  Google Scholar 

  46. Pickart, C. M. Ubiquitin in chains. Trends Biochem. Sci. 25, 544–548 (2000).

    Article  CAS  PubMed  Google Scholar 

  47. Huang, D. T. et al. A unique E1-E2 interaction required for optimal conjugation of the ubiquitin-like protein NEDD8. Nature Struct. Mol. Biol. 11, 927–935 (2004).

    Article  CAS  Google Scholar 

  48. Huang, D. T. et al. Structural basis for recruitment of Ubc12 by an E2 binding domain in NEDD8's E1. Mol. Cell 17, 341–350 (2005).

    Article  CAS  PubMed  Google Scholar 

  49. Lois, L. M. & Lima, C. D. Structures of the SUMO E1 provide mechanistic insights into SUMO activation and E2 recruitment to E1. EMBO J. 24, 439–451 (2005). Together with references 47 and 48, this study provided the first structural insight into E1-activating enzymes and how they activate ubiquitin-like proteins. These studies also suggested how different E2s are recognized by divergent ubiquitin-like domains located within the E1 enzyme.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. VanDemark, A. P. & Hill, C. P. E1 on the move. Mol. Cell 17, 474–475 (2005).

    Article  CAS  PubMed  Google Scholar 

  51. Ren, R. Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia. Nature Rev. Cancer 5, 172–183 (2005).

    Article  CAS  Google Scholar 

  52. Arkin, M. R. & Wells, J. A. Small-molecule inhibitors of protein-protein interactions: progressing towards the dream. Nature Rev. Drug Discov. 3, 301–317 (2004).

    Article  CAS  Google Scholar 

  53. Jones, D., Crowe, E., Stevens, T. A. & Candido, E. P. Functional and phylogenetic analysis of the ubiquitylation system in Caenorhabditis elegans: ubiquitin-conjugating enzymes, ubiquitin-activating enzymes & ubiquitin-like proteins. Genome Biol. 3, 2.1–2.15 (2002).

  54. Stickle, N. H. et al. pVHL modification by NEDD8 is required for fibronectin matrix assembly and suppression of tumor development. Mol. Cell Biol. 24, 3251–3261 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Harper, J. W. Neddylating the guardian; Mdm2 catalyzed conjugation of Nedd8 to p53. Cell 118, 2–4 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Xirodimas, D. P., Saville, M. K., Bourdon, J. C., Hay, R. T. & Lane, D. P. Mdm2-mediated NEDD8 conjugation of p53 inhibits its transcriptional activity. Cell 118, 83–97 (2004).

    Article  CAS  PubMed  Google Scholar 

  57. Hatakeyama, S., Yada, M., Matsumoto, M., Ishida, N. & Nakayama, K. I. U box proteins as a new family of ubiquitin-protein ligases. J. Biol. Chem. 276, 33111–33120 (2001).

    Article  CAS  PubMed  Google Scholar 

  58. Zheng, N. et al. Structure of the Cul1–Rbx1–Skp1-F boxSkp2 SCF ubiquitin ligase complex. Nature 416, 703–709 (2002). The first structural insight into the cullin-based RING-finger family of ubiquitin ligases. Subsequent studies revealed the structure of related complexes and demonstrate how F-box proteins bind substrates (see also References 102,127,140).

    Article  CAS  PubMed  Google Scholar 

  59. Huang, L. et al. Structure of an E6AP-UbcH7 complex: insights into ubiquitination by the E2-E3 enzyme cascade. Science 286, 1321–1326 (1999).

    Article  CAS  PubMed  Google Scholar 

  60. Petroski, M. D. & Deshaies, R. J. Function and regulation of cullin-RING ubiquitin ligases. Nat Rev Mol. Cell Biol. 6, 9–20 (2005).

    Article  CAS  PubMed  Google Scholar 

  61. Fang, S., Lorick, K. L., Jensen, J. P. & Weissman, A. M. RING finger ubiquitin protein ligases: implications for tumorigenesis, metastasis and for molecular targets in cancer. Semin. Cancer Biol. 13, 5–14 (2003).

    Article  CAS  PubMed  Google Scholar 

  62. Lane, D. P. & Lain, S. Therapeutic exploitation of the p53 pathway. Trends Mol. Med. 8, S38–S42 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. Balint, E. E. & Vousden, K. H. Activation and activities of the p53 tumour suppressor protein. Br. J. Cancer 85, 1813–1823 (2001).

    Article  CAS  PubMed Central  Google Scholar 

  64. Pavletich, N. P., Chambers, K. A. & Pabo, C. O. The DNA-binding domain of p53 contains the four conserved regions and the major mutation hot spots. Genes Dev. 7, 2556–2564 (1993).

    Article  CAS  PubMed  Google Scholar 

  65. Vousden, K. H. & Prives, C. P53 and prognosis: new insights and further complexity. Cell 120, 7–10 (2005).

    CAS  PubMed  Google Scholar 

  66. Vousden, K. H. & Lu, X. Live or let die: the cell's response to p53. Nature Rev. Cancer 2, 594–604 (2002).

    Article  CAS  Google Scholar 

  67. Zhang, Z. et al. MDM2 is a negative regulator of p21WAF1/CIP1, independent of p53. J. Biol. Chem. 279, 16000–16006 (2004).

    Article  CAS  PubMed  Google Scholar 

  68. Wang, H., Nan, L., Yu, D., Agrawal, S. & Zhang, R. Antisense anti-MDM2 oligonucleotides as a novel therapeutic approach to human breast cancer: in vitro and in vivo activities and mechanisms. Clin. Cancer Res. 7, 3613–3624 (2001).

    CAS  PubMed  Google Scholar 

  69. Prasad, G., Wang, H., Agrawal, S. & Zhang, R. Antisense anti-MDM2 oligonucleotides as a novel approach to the treatment of glioblastoma multiforme. Anticancer Res. 22, 107–116 (2002).

    CAS  PubMed  Google Scholar 

  70. Zhang, R., Wang, H. & Agrawal, S. Novel antisense anti-MDM2 mixed-backbone oligonucleotides: proof of principle, in vitro and in vivo activities & mechanisms. Curr. Cancer Drug Targets 5, 43–49 (2005).

    Article  PubMed  Google Scholar 

  71. Karlsson, G. B. et al. Activation of p53 by scaffold-stabilised expression of Mdm2-binding peptides: visualisation of reporter gene induction at the single-cell level. Br. J. Cancer 91, 1488–1494 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Bottger, A. et al. Design of a synthetic Mdm2-binding mini protein that activates the p53 response in vivo. Curr. Biol. 7, 860–869 (1997).

    Article  CAS  PubMed  Google Scholar 

  73. Vassilev, L. T. et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303, 844–848 (2004). This paper identified the first small-molecule inhibitor of the RING-finger protein MDM2, an E3 for p53. The paper demonstrated that the inhibitor blocks binding of p53 to its interaction site on MDM2, thereby blocking its ability to be ubiquitylated.

    Article  CAS  PubMed  Google Scholar 

  74. Rubinstein, L. V. et al. Comparison of in vitro anticancer-drug-screening data generated with a tetrazolium assay versus a protein assay against a diverse panel of human tumor cell lines. J. Natl Cancer Inst. 82, 1113–1118 (1990).

    Article  CAS  PubMed  Google Scholar 

  75. Issaeva, N. et al. Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors. Nature Med. 10, 1321–1328 (2004).

    Article  CAS  PubMed  Google Scholar 

  76. Gu, W. & Roeder, R. G. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 90, 595–606 (1997).

    Article  CAS  PubMed  Google Scholar 

  77. Sakaguchi, K. et al. DNA damage activates p53 through a phosphorylation-acetylation cascade. Genes Dev. 12, 2831–2841 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Grossman, S. R. et al. Polyubiquitination of p53 by a ubiquitin ligase activity of p300. Science 300, 342–344 (2003).

    Article  CAS  PubMed  Google Scholar 

  79. Zhu, Q., Yao, J., Wani, G., Wani, M. A. & Wani, A. A. Mdm2 mutant defective in binding p300 promotes ubiquitination but not degradation of p53: evidence for the role of p300 in integrating ubiquitination and proteolysis. J. Biol. Chem. 276, 29695–29701 (2001).

    Article  CAS  PubMed  Google Scholar 

  80. Krajewski, M., Ozdowy, P., D'Silva, L., Rothweiler, U. & Holak, T. A. NMR indicates that the small molecule RITA does not block p53-MDM2 binding in vitro. Nature Med. 11, 1135–1136 (2005).

    Article  CAS  PubMed  Google Scholar 

  81. Grinkevich, V., Issaeva, N., Hossain, S., Pramanik, A. & Selivanova, G. Reply to 'NMR indicates that the small molecule RITA does not block p53-MDM2 binding in vitro'. Nature Med. 11, 1136–1137 (2005).

    Article  CAS  Google Scholar 

  82. Lasne, C., Lowy, R. & Venegas, W. In vitro induction of sister-chromatid exchanges after G0 exposure of human lymphocytes to five naphthofurans. Mutagenesis 4, 27–30 (1989).

    Article  CAS  PubMed  Google Scholar 

  83. Touati, E., Krin, E., Quillardet, P. & Hofnung, M. 7-Methoxy-2-nitronaphtho[2,1-b]furan (R7000)-induced mutation spectrum in the lacI gene of Escherichia coli: influence of SOS mutagenesis. Carcinogenesis 17, 2543–2550 (1996).

    Article  CAS  PubMed  Google Scholar 

  84. Quillardet, P., Boscus, D., Touati, E. & Hofnung, M. DNA damage induced in vivo by 7-methoxy-2-nitronaphtho[2,1-b]-furan (R7000) in the lacI gene of Escherichia coli. Mutat. Res. 422, 237–245 (1998).

    Article  CAS  PubMed  Google Scholar 

  85. Quillardet, P., Michel, V., Arrault, X., Hofnung, M. & Touati, E. Mutagenic properties of a nitrofuran, 7-methoxy-2-nitronaphtho[2,1-b]furan (R7000), in lacI transgenic mice. Mutat. Res. 470, 177–188 (2000).

    Article  CAS  PubMed  Google Scholar 

  86. Bossy-Wetzel, E., Schwarzenbacher, R. & Lipton, S. A. Molecular pathways to neurodegeneration. Nature Med. 10 (Suppl.), S2–S9 (2004).

    Article  PubMed  CAS  Google Scholar 

  87. Dawson, T. M. & Dawson, V. L. Molecular pathways of neurodegeneration in Parkinson's disease. Science 302, 819–822 (2003).

    Article  CAS  PubMed  Google Scholar 

  88. Farrer, M. J. Genetics of Parkinson disease: paradigm shifts and future prospects. Nature Rev. Genet. 7, 306–318 (2006).

    Article  CAS  PubMed  Google Scholar 

  89. Giasson, B. I. & Lee, V. M. Are ubiquitination pathways central to Parkinson's disease? Cell 114, 1–8 (2003).

    Article  CAS  PubMed  Google Scholar 

  90. Kahle, P. J. & Haass, C. How does parkin ligate ubiquitin to Parkinson's disease? EMBO Rep. 5, 681–685 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Nutt, J. G. & Wooten, G. F. Clinical practice. Diagnosis and initial management of Parkinson's disease. N. Engl. J. Med. 353, 1021–1027 (2005).

    Article  CAS  PubMed  Google Scholar 

  92. Tolosa, E., Wenning, G. & Poewe, W. The diagnosis of Parkinson's disease. Lancet Neurol. 5, 75–86 (2006).

    Article  PubMed  Google Scholar 

  93. Kitada, T. et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 392, 605–608 (1998). The authors identified an autosomal recessive juvenile parkinsonism gene located on chromosome 6 as the parkin RING-finger ubiquitin ligase and multiple classes of deletion mutants in the gene. Subsequent studies identified further point mutations which also occur in the RING-finger domain, linking its E3 activity to disease.

    Article  CAS  PubMed  Google Scholar 

  94. Pesah, Y. et al. Drosophila parkin mutants have decreased mass and cell size and increased sensitivity to oxygen radical stress. Development 131, 2183–2194 (2004).

    Article  CAS  PubMed  Google Scholar 

  95. Greene, J. C., Whitworth, A. J., Andrews, L. A., Parker, T. J. & Pallanck, L. J. Genetic and genomic studies of Drosophila parkin mutants implicate oxidative stress and innate immune responses in pathogenesis. Hum. Mol. Genet. 14, 799–811 (2005).

    Article  CAS  PubMed  Google Scholar 

  96. Haywood, A. F. & Staveley, B. E. Parkin counteracts symptoms in a Drosophila model of Parkinson's disease. BMC Neurosci. 5, 14 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Yamada, M., Mizuno, Y. & Mochizuki, H. Parkin gene therapy for α-synucleinopathy: a rat model of Parkinson's disease. Hum. Gene Ther. 16, 262–270 (2005).

    Article  CAS  PubMed  Google Scholar 

  98. Shimura, H. et al. Ubiquitination of a new form of α-synuclein by parkin from human brain: implications for Parkinson's disease. Science 293, 263–269 (2001).

    Article  CAS  PubMed  Google Scholar 

  99. Ang, X. L. & Wade Harper, J. (2001). SCF-mediated protein degradation and cell cycle control. Oncogene 24, 2860–2870 (2001).

    Article  CAS  Google Scholar 

  100. Jin, J. et al. (2001). Systematic analysis and nomenclature of mammalian F-box proteins. Genes Dev. 18, 2573–2580 (2001).

    Article  CAS  Google Scholar 

  101. Latres, E., Chiaur, D. S. & Pagano, M. (2001). The human F box protein β-Trcp associates with the Cul1/Skp1 complex and regulates the stability of β-catenin. Oncogene 18, 849–854 (2001).

    Article  CAS  Google Scholar 

  102. Wu, G. et al. Structure of a β-TrCP1–Skp1-β-catenin complex: destruction motif binding and lysine specificity of the SCF(β-TrCP1) ubiquitin ligase. Mol. Cell 11, 1445–1456 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. Read, M. A. et al. Nedd8 modification of cul-1 activates SCF(β(TrCP))-dependent ubiquitination of IκBα. Mol. Cell Biol. 20, 2326–2333 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Fero, M. L. et al. A syndrome of multiorgan hyperplasia with features of gigantism, tumorigenesis & female sterility in p27(Kip1)-deficient mice. Cell 85, 733–744 (1996).

    Article  CAS  PubMed  Google Scholar 

  105. Kiyokawa, H. et al. Enhanced growth of mice lacking the cyclin-dependent kinase inhibitor function of p27(Kip1). Cell 85, 721–732 (1996).

    Article  CAS  PubMed  Google Scholar 

  106. Nakayama, K. et al. Mice lacking p27(Kip1) display increased body size, multiple organ hyperplasia, retinal dysplasia & pituitary tumors. Cell 85, 707–720 (1996).

    CAS  PubMed  Google Scholar 

  107. Bhattacharya, S. et al. SKP2 associates with p130 and accelerates p130 ubiquitylation and degradation in human cells. Oncogene 22, 2443–2451 (2003).

    Article  CAS  PubMed  Google Scholar 

  108. Tedesco, D., Lukas, J. & Reed, S. I. The pRb-related protein p130 is regulated by phosphorylation-dependent proteolysis via the protein-ubiquitin ligase SCF(Skp2). Genes Dev. 16, 2946–2957 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Bornstein, G. et al. Role of the SCFSkp2 ubiquitin ligase in the degradation of p21Cip1 in S phase. J. Biol. Chem. 278, 25752–25757 (2003).

    Article  CAS  PubMed  Google Scholar 

  110. Kamura, T. et al. Degradation of p57Kip2 mediated by SCFSkp2-dependent ubiquitylation. Proc. Natl Acad. Sci. USA 100, 10231–10236 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Dehan, E. & Pagano, M. Skp2, the FoxO1 hunter. Cancer Cell 7, 209–210 (2005).

    Article  CAS  PubMed  Google Scholar 

  112. Huang, H. et al. Skp2 inhibits FOXO1 in tumor suppression through ubiquitin-mediated degradation. Proc. Natl Acad. Sci. USA 102, 1649–1654 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Gstaiger, M. et al. Skp2 is oncogenic and overexpressed in human cancers. Proc. Natl Acad. Sci. USA 98, 5043–5048 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Shim, E. H. et al. Expression of the F-box protein SKP2 induces hyperplasia, dysplasia & low-grade carcinoma in the mouse prostate. Cancer Res. 63, 1583–1588 (2003).

    CAS  PubMed  Google Scholar 

  115. Piva, R. et al. In vivo interference with Skp1 function leads to genetic instability and neoplastic transformation. Mol. Cell Biol. 22, 8375–8387 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Sumimoto, H. et al. Gene therapy for human small-cell lung carcinoma by inactivation of Skp-2 with virally mediated RNA interference. Gene Ther. 12, 95–100 (2005).

    Article  CAS  PubMed  Google Scholar 

  117. Kudo, Y. et al. Small interfering RNA targeting of S phase kinase-interacting protein 2 inhibits cell growth of oral cancer cells by inhibiting p27 degradation. Mol. Cancer Ther. 4, 471–476 (2005).

    Article  CAS  PubMed  Google Scholar 

  118. Jiang, F., Caraway, N. P., Li, R. & Katz, R. L. RNA silencing of S-phase kinase-interacting protein 2 inhibits proliferation and centrosome amplification in lung cancer cells. Oncogene 24, 3409–3418 (2005).

    Article  CAS  PubMed  Google Scholar 

  119. Katagiri, Y., Hozumi, Y. & Kondo, S. Knockdown of Skp2 by siRNA inhibits melanoma cell growth in vitro and in vivo. J. Dermatol Sci. 42, 215–224 (2006).

    Article  CAS  PubMed  Google Scholar 

  120. Hao, B. et al. Structural basis of the Cks1-dependent recognition of p27(Kip1) by the SCF(Skp2) ubiquitin ligase. Mol. Cell 20, 9–19 (2005).

    Article  CAS  PubMed  Google Scholar 

  121. Winston, J. T. et al. The SCFβ-TRCP-ubiquitin ligase complex associates specifically with phosphorylated destruction motifs in IκBα and β-catenin and stimulates IκBα ubiquitination in vitro. Genes Dev. 13, 270–283 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Rajagopalan, H. et al. Inactivation of hCDC4 can cause chromosomal instability. Nature 428, 77–81 (2004).

    Article  CAS  PubMed  Google Scholar 

  123. Hemann, M. T. et al. Evasion of the p53 tumour surveillance network by tumour-derived MYC mutants. Nature 436, 807–811 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Kim, W. & Kaelin, W. G., Jr. The von Hippel-Lindau tumor suppressor protein: new insights into oxygen sensing and cancer. Curr. Opin. Genet. Dev. 13, 55–60 (2003).

    Article  CAS  PubMed  Google Scholar 

  125. Maxwell, P. H. et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399, 271–275 (1999).

    Article  CAS  PubMed  Google Scholar 

  126. Ohh, M. et al. Ubiquitination of hypoxia-inducible factor requires direct binding to the β-domain of the von Hippel-Lindau protein. Nature Cell Biol. 2, 423–427 (2000).

    Article  CAS  PubMed  Google Scholar 

  127. Stebbins, C. E., Kaelin, W. G., Jr. & Pavletich, N. P. Structure of the VHL-ElonginC-ElonginB complex: implications for VHL tumor suppressor function. Science 284, 455–461 (1999).

    Article  CAS  PubMed  Google Scholar 

  128. Hon, W. C. et al. Structural basis for the recognition of hydroxyproline in HIF-1 α by pVHL. Nature 417, 975–978 (2002).

    Article  CAS  PubMed  Google Scholar 

  129. Isaacs, J. S. et al. Hsp90 regulates a von Hippel Lindau-independent hypoxia-inducible factor-1 α-degradative pathway. J. Biol. Chem. 277, 29936–29944 (2002).

    Article  CAS  PubMed  Google Scholar 

  130. Isaacs, J. S., Xu, W. & Neckers, L. Heat shock protein 90 as a molecular target for cancer therapeutics. Cancer Cell 3, 213–217 (2003).

    Article  CAS  PubMed  Google Scholar 

  131. Aravind, L. & Koonin, E. V. The U box is a modified RING finger- a common domain in ubiquitination. Curr. Biol. 10, R132–R134 (2000).

    Article  CAS  PubMed  Google Scholar 

  132. Ohi, M. D., Vander Kooi, C. W., Rosenberg, J. A., Chazin, W. J. & Gould, K. L. (2003). Structural insights into the U-box, a domain associated with multi-ubiquitination. Nature Struct. Biol. 10, 250–255.

    Article  CAS  PubMed  Google Scholar 

  133. Hatakeyama, S. & Nakayama, K. I. U-box proteins as a new family of ubiquitin ligases. Biochem. Biophys. Res. Commun. 302, 635–645 (2003).

    Article  CAS  PubMed  Google Scholar 

  134. Xu, W. et al. Chaperone-dependent E3 ubiquitin ligase CHIP mediates a degradative pathway for c-ErbB2/Neu. Proc. Natl Acad. Sci. USA 99, 12847–12852 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Zhou, P. et al. ErbB2 degradation mediated by the co-chaperone protein CHIP. J. Biol. Chem. 278, 13829–13837 (2003).

    Article  CAS  PubMed  Google Scholar 

  136. Meacham, G. C., Patterson, C., Zhang, W., Younger, J. M. & Cyr, D. M. The Hsc70 co-chaperone CHIP targets immature CFTR for proteasomal degradation. Nature Cell Biol. 3, 100–105 (2001).

    Article  CAS  PubMed  Google Scholar 

  137. Imai, Y. et al. CHIP is associated with Parkin, a gene responsible for familial Parkinson's disease & enhances its ubiquitin ligase activity. Mol. Cell 10, 55–67 (2002).

    Article  CAS  PubMed  Google Scholar 

  138. Huibregtse, J. M., Scheffner, M., Beaudenon, S. & Howley, P. M. A family of proteins structurally and functionally related to the E6-AP ubiquitin-protein ligase. Proc. Natl Acad. Sci. USA 92, 2563–2567 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Anglesio, M. S. et al. Differential expression of a novel ankyrin containing E3 ubiquitin-protein ligase, Hace1, in sporadic Wilms' tumor versus normal kidney. Hum. Mol. Genet. 13, 2061–2074 (2004).

    Article  CAS  PubMed  Google Scholar 

  140. Talis, A. L., Huibregtse, J. M. & Howley, P. M. The role of E6AP in the regulation of p53 protein levels in human papillomavirus (HPV)-positive and HPV-negative cells. J. Biol. Chem. 273, 6439–6445 (1998).

    Article  CAS  PubMed  Google Scholar 

  141. Cooper, E. M., Hudson, A. W., Amos, J., Wagstaff, J. & Howley, P. M. Biochemical analysis of Angelman syndrome-associated mutations in the E3 ubiquitin ligase E6-associated protein. J. Biol. Chem. 279, 41208–41217 (2004).

    Article  CAS  PubMed  Google Scholar 

  142. Kishino, T., Lalande, M. & Wagstaff, J. UBE3A/E6-AP mutations cause Angelman syndrome. Nature Genet. 15, 70–73 (1997).

    Article  CAS  PubMed  Google Scholar 

  143. Verdecia, M. A. et al. Conformational flexibility underlies ubiquitin ligation mediated by the WWP1 HECT domain E3 ligase. Mol. Cell 11, 249–259 (2003).

    Article  CAS  PubMed  Google Scholar 

  144. Scheffner, M., Huibregtse, J. M., Vierstra, R. D. & Howley, P. M. The HPV-16 E6 and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53. Cell 75, 495–505 (1993). The first demonstration of a HECT-domain ubiquitin ligase and its direct involvement in ubiquitin transfer through a thiol-ester intermediate. Subsequent work (see Reference 138 ) demonstrated that there is a large family of HECT-domain containing proteins that all probably act to promote ubiquitin transfer through a similar mechanism.

    Article  CAS  PubMed  Google Scholar 

  145. Huibregtse, J. M., Scheffner, M. & Howley, P. M. A cellular protein mediates association of p53 with the E6 oncoprotein of human papillomavirus types 16 or 18. EMBO J. 10, 4129–4135 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Rolfe, M. et al. Reconstitution of p53-ubiquitinylation reactions from purified components: the role of human ubiquitin-conjugating enzyme UBC4 and E6-associated protein (E6AP). Proc. Natl Acad. Sci. USA 92, 3264–3268 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Gross-Mesilaty, S. et al. Basal and human papillomavirus E6 oncoprotein-induced degradation of Myc proteins by the ubiquitin pathway. Proc. Natl Acad. Sci. USA 95, 8058–8063 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Gewin, L., Myers, H., Kiyono, T. & Galloway, D. A. Identification of a novel telomerase repressor that interacts with the human papillomavirus type-16 E6/E6-AP complex. Genes Dev. 18, 2269–2282 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Matsuura, T. et al. De novo truncating mutations in E6-AP ubiquitin-protein ligase gene (UBE3A) in Angelman syndrome. Nature Genet. 15, 74–77 (1997).

    Article  CAS  PubMed  Google Scholar 

  150. Buntinx, I. M. et al. Clinical profile of Angelman syndrome at different ages. Am. J. Med. Genet. 56, 176–183 (1995).

    Article  CAS  PubMed  Google Scholar 

  151. Duensing, S. & Munger, K. Mechanisms of genomic instability in human cancer: insights from studies with human papillomavirus oncoproteins. Int. J. Cancer 109, 157–162 (2004).

    Article  CAS  PubMed  Google Scholar 

  152. Kim, Y., Cairns, M. J., Marouga, R. & Sun, L. Q. E6AP gene suppression and characterization with in vitro selected hammerhead ribozymes. Cancer Gene Ther. 10, 707–716 (2003).

    Article  CAS  PubMed  Google Scholar 

  153. Foster, S. A. & Phelps, W. C. Zn(2+) fingers and cervical cancer. J. Natl Cancer Inst. 91, 1180–1181 (1999).

    Article  CAS  PubMed  Google Scholar 

  154. Harper, D. M. et al. Sustained efficacy up to 4.5 years of a bivalent L1 virus-like particle vaccine against human papillomavirus types 16 and 18: follow-up from a randomised control trial. Lancet 367, 1247–1255 (2006).

    Article  CAS  PubMed  Google Scholar 

  155. Mayeaux, E. J., Jr. Harnessing the power of prevention: human papillomavirus vaccines. Curr. Opin. Obstet. Gynecol. 18 (Suppl. 1), S15–S21 (2006).

    Article  PubMed  Google Scholar 

  156. Lowy, D. R. & Schiller, J. T. Prophylactic human papillomavirus vaccines. J. Clin. Invest. 116, 1167–1173 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Sakamoto, K. M. et al. Development of Protacs to target cancer-promoting proteins for ubiquitination and degradation. Mol. Cell Proteomics 2, 1350–1358 (2003).

    Article  CAS  PubMed  Google Scholar 

  159. Zhou, P., Bogacki, R., McReynolds, L. & Howley, P. M. Harnessing the ubiquitination machinery to target the degradation of specific cellular proteins. Mol. Cell 6, 751–756 (2000).

    Article  CAS  PubMed  Google Scholar 

  160. Verma, R. et al. Role of Rpn11 metalloprotease in deubiquitination and degradation by the 26S proteasome. Science 298, 611–615 (2002).

    Article  CAS  PubMed  Google Scholar 

  161. Goldenberg, S. J. et al. Structure of the Cand1-Cul1-Roc1 complex reveals regulatory mechanisms for the assembly of the multisubunit cullin-dependent ubiquitin ligases. Cell 119, 517–528 (2004).

    Article  CAS  PubMed  Google Scholar 

  162. Zheng, J. et al. CAND1 binds to unneddylated CUL1 and regulates the formation of SCF ubiquitin E3 ligase complex. Mol. Cell 10, 1519–1526 (2002).

    Article  CAS  PubMed  Google Scholar 

  163. Cope, G. A. & Deshaies, R. J. COP9 signalosome: a multifunctional regulator of SCF and other cullin-based ubiquitin ligases. Cell 114, 663–671 (2003).

    Article  CAS  PubMed  Google Scholar 

  164. Wei, N. & Deng, X. W. The COP9 signalosome. Annu. Rev. Cell Dev. Biol. 19, 261–286 (2003).

    Article  CAS  PubMed  Google Scholar 

  165. Ambroggio, X. I., Rees, D. C. & Deshaies, R. J. JAMM: a metalloprotease-like zinc site in the proteasome and signalosome. PLoS Biol. 2, E2 (2004).

    Article  CAS  PubMed  Google Scholar 

  166. Cope, G. A. et al. Role of predicted metalloprotease motif of Jab1/Csn5 in cleavage of Nedd8 from Cul1. Science 298, 608–611 (2002). This study identified the machinery required to remove NEDD8 from cullins and demonstrated an important role for NEDD8-removal for the biological activity of SCF complexes.

    Article  CAS  PubMed  Google Scholar 

  167. Verma, R., Oania, R., Graumann, J. & Deshaies, R. J. Multiubiquitin chain receptors define a layer of substrate selectivity in the ubiquitin–proteasome system. Cell 118, 99–110 (2004).

    Article  CAS  PubMed  Google Scholar 

  168. Verma, R. et al. Ubistatins inhibit proteasome-dependent degradation by binding the ubiquitin chain. Science 306, 117–120 (2004).

    Article  CAS  PubMed  Google Scholar 

  169. Schwartz, D. C. & Hochstrasser, M. (2003). A superfamily of protein tags: ubiquitin, SUMO and related modifiers. Trends Biochem. Sci. 28, 321–328.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Research on the ubiquitin pathway in the Harper laboratory is supported by the National Institutes of Health Grants.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to J. Wade Harper.

Ethics declarations

Competing interests

M.R. is an employee of and owns stocks in Millennium Pharmaceuticals, Inc. J.W.H. is a consultant for Millennium Pharmaceuticals, Inc.

Related links

Related links

DATABASES

OMIM

Angelman's syndrome

chronic myelogenous leukaemia

multiple myeloma

non-Hodgkin's lymphoma

Parkinson's disease

von Hippel-Lindau syndrome

Glossary

Neddylation

Covalent attachment of the ubiquitin-like protein NEDD8 (RUB1) to another protein.

Endoreduplication

Duplication of the genome without mitosis, which results in an increase in the nuclear DNA content, permitting amplification of the genome of specialized cells.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nalepa, G., Rolfe, M. & Harper, J. Drug discovery in the ubiquitin–proteasome system. Nat Rev Drug Discov 5, 596–613 (2006). https://doi.org/10.1038/nrd2056

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd2056

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing