Skip to main content
Top
Published in: Molecular Cancer 1/2013

Open Access 01-12-2013 | Review

USP1 deubiquitinase: cellular functions, regulatory mechanisms and emerging potential as target in cancer therapy

Authors: Iraia García-Santisteban, Godefridus J Peters, Elisa Giovannetti, Jose Antonio Rodríguez

Published in: Molecular Cancer | Issue 1/2013

Login to get access

Abstract

Reversible protein ubiquitination is emerging as a key process for maintaining cell homeostasis, and the enzymes that participate in this process, in particular E3 ubiquitin ligases and deubiquitinases (DUBs), are increasingly being regarded as candidates for drug discovery. Human DUBs are a group of approximately 100 proteins, whose cellular functions and regulatory mechanisms remain, with some exceptions, poorly characterized. One of the best-characterized human DUBs is ubiquitin-specific protease 1 (USP1), which plays an important role in the cellular response to DNA damage. USP1 levels, localization and activity are modulated through several mechanisms, including protein-protein interactions, autocleavage/degradation and phosphorylation, ensuring that USP1 function is carried out in a properly regulated spatio-temporal manner. Importantly, USP1 expression is deregulated in certain types of human cancer, suggesting that USP1 could represent a valid target in cancer therapy. This view has gained recent support with the finding that USP1 inhibition may contribute to revert cisplatin resistance in an in vitro model of non-small cell lung cancer (NSCLC). Here, we describe the current knowledge on the cellular functions and regulatory mechanisms of USP1. We also summarize USP1 alterations found in cancer, combining data from the literature and public databases with our own data. Finally, we discuss the emerging potential of USP1 as a target, integrating published data with our novel findings on the effects of the USP1 inhibitor pimozide in combination with cisplatin in NSCLC cells.
Appendix
Available only for authorised users
Literature
1.
go back to reference Hershko A: Ubiquitin: roles in protein modification and breakdown. Cell. 1983, 34 (1): 11-12. 10.1016/0092-8674(83)90131-9CrossRefPubMed Hershko A: Ubiquitin: roles in protein modification and breakdown. Cell. 1983, 34 (1): 11-12. 10.1016/0092-8674(83)90131-9CrossRefPubMed
2.
go back to reference Chen ZJ, Sun LJ: Nonproteolytic functions of ubiquitin in cell signaling. Mol Cell. 2009, 33 (3): 275-286. 10.1016/j.molcel.2009.01.014CrossRefPubMed Chen ZJ, Sun LJ: Nonproteolytic functions of ubiquitin in cell signaling. Mol Cell. 2009, 33 (3): 275-286. 10.1016/j.molcel.2009.01.014CrossRefPubMed
3.
go back to reference Pickart CM: Mechanisms underlying ubiquitination. Annu Rev Biochem. 2001, 70: 503-533. 10.1146/annurev.biochem.70.1.503CrossRefPubMed Pickart CM: Mechanisms underlying ubiquitination. Annu Rev Biochem. 2001, 70: 503-533. 10.1146/annurev.biochem.70.1.503CrossRefPubMed
4.
go back to reference Ikeda F, Dikic I: Atypical ubiquitin chains: new molecular signals, protein modifications: beyond the usual suspects’ review series. EMBO Rep. 2008, 9 (6): 536-542. 10.1038/embor.2008.93PubMedCentralCrossRefPubMed Ikeda F, Dikic I: Atypical ubiquitin chains: new molecular signals, protein modifications: beyond the usual suspects’ review series. EMBO Rep. 2008, 9 (6): 536-542. 10.1038/embor.2008.93PubMedCentralCrossRefPubMed
5.
go back to reference Kirisako T, Kamei K, Murata S, Kato M, Fukumoto H, Kanie M, Sano S, Tokunaga F, Tanaka K, Iwai K: A ubiquitin ligase complex assembles linear polyubiquitin chains. EMBO J. 2006, 25 (20): 4877-4887. 10.1038/sj.emboj.7601360PubMedCentralCrossRefPubMed Kirisako T, Kamei K, Murata S, Kato M, Fukumoto H, Kanie M, Sano S, Tokunaga F, Tanaka K, Iwai K: A ubiquitin ligase complex assembles linear polyubiquitin chains. EMBO J. 2006, 25 (20): 4877-4887. 10.1038/sj.emboj.7601360PubMedCentralCrossRefPubMed
6.
go back to reference Komander D, Rape M: The ubiquitin code. Annu Rev Biochem. 2012, 81: 203-229. 10.1146/annurev-biochem-060310-170328CrossRefPubMed Komander D, Rape M: The ubiquitin code. Annu Rev Biochem. 2012, 81: 203-229. 10.1146/annurev-biochem-060310-170328CrossRefPubMed
7.
go back to reference Trempe JF: Reading the ubiquitin postal code. Curr Opin Struct Biol. 2011, 21 (6): 792-801. 10.1016/j.sbi.2011.09.009CrossRefPubMed Trempe JF: Reading the ubiquitin postal code. Curr Opin Struct Biol. 2011, 21 (6): 792-801. 10.1016/j.sbi.2011.09.009CrossRefPubMed
8.
go back to reference Kulathu Y, Komander D: Atypical ubiquitylation-the unexplored world of polyubiquitin beyond Lys48 and Lys63 linkages. Nat Rev Mol Cell Biol. 2012, 13 (8): 508-523. 10.1038/nrm3394CrossRefPubMed Kulathu Y, Komander D: Atypical ubiquitylation-the unexplored world of polyubiquitin beyond Lys48 and Lys63 linkages. Nat Rev Mol Cell Biol. 2012, 13 (8): 508-523. 10.1038/nrm3394CrossRefPubMed
9.
go back to reference Kessler BM: Ubiquitin-omics reveals novel networks and associations with human disease. Curr Opin Chem Biol. 2013, 17 (1): 59-65. 10.1016/j.cbpa.2012.12.024CrossRefPubMed Kessler BM: Ubiquitin-omics reveals novel networks and associations with human disease. Curr Opin Chem Biol. 2013, 17 (1): 59-65. 10.1016/j.cbpa.2012.12.024CrossRefPubMed
10.
go back to reference Reyes-Turcu FE, Ventii KH, Wilkinson KD: Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu Rev Biochem. 2009, 78: 363-397. 10.1146/annurev.biochem.78.082307.091526CrossRefPubMed Reyes-Turcu FE, Ventii KH, Wilkinson KD: Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu Rev Biochem. 2009, 78: 363-397. 10.1146/annurev.biochem.78.082307.091526CrossRefPubMed
11.
go back to reference Edelmann MJ, Iphöfer A, Akutsu M, Altun M, di Gleria K, Kramer HB, Fiebiger E, Dhe-Paganon S, Kessler BM: Structural basis and specificity of human otubain 1-mediated deubiquitination. Biochem J. 2009, 418 (2): 379-390. 10.1042/BJ20081318CrossRefPubMed Edelmann MJ, Iphöfer A, Akutsu M, Altun M, di Gleria K, Kramer HB, Fiebiger E, Dhe-Paganon S, Kessler BM: Structural basis and specificity of human otubain 1-mediated deubiquitination. Biochem J. 2009, 418 (2): 379-390. 10.1042/BJ20081318CrossRefPubMed
12.
go back to reference Virdee S, Ye Y, Nguyen DP, Komander D, Chin JW: Engineered diubiquitin synthesis reveals Lys29-isopeptide specificity of an OTU deubiquitinase. Nat Chem Biol. 2010, 6 (10): 750-757. 10.1038/nchembio.426CrossRefPubMed Virdee S, Ye Y, Nguyen DP, Komander D, Chin JW: Engineered diubiquitin synthesis reveals Lys29-isopeptide specificity of an OTU deubiquitinase. Nat Chem Biol. 2010, 6 (10): 750-757. 10.1038/nchembio.426CrossRefPubMed
13.
go back to reference Faesen AC, Luna-Vargas MP, Geurink PP, Clerici M, Merkx R, Van Dijk WJ, Hameed DS, El Oualid F, Ovaa H, Sixma TK: The differential modulation of USP activity by internal regulatory domains, interactors and eight ubiquitin chain types. Chem Biol. 2011, 18 (12): 1550-1561. 10.1016/j.chembiol.2011.10.017CrossRefPubMed Faesen AC, Luna-Vargas MP, Geurink PP, Clerici M, Merkx R, Van Dijk WJ, Hameed DS, El Oualid F, Ovaa H, Sixma TK: The differential modulation of USP activity by internal regulatory domains, interactors and eight ubiquitin chain types. Chem Biol. 2011, 18 (12): 1550-1561. 10.1016/j.chembiol.2011.10.017CrossRefPubMed
14.
go back to reference Rivkin E, Almeida SM, Ceccarelli DF, Juang YC, MacLean TA, Srikumar T, Huang H, Dunham WH, Fukumura R, Xie G, Gondo Y, Raught B, Gingras AC, Sicheri F, Cordes SP: The linear ubiquitin-specific deubiquitinase gumby regulates angiogenesis. Nature. 2013, 498 (7454): 318-324. 10.1038/nature12296CrossRefPubMed Rivkin E, Almeida SM, Ceccarelli DF, Juang YC, MacLean TA, Srikumar T, Huang H, Dunham WH, Fukumura R, Xie G, Gondo Y, Raught B, Gingras AC, Sicheri F, Cordes SP: The linear ubiquitin-specific deubiquitinase gumby regulates angiogenesis. Nature. 2013, 498 (7454): 318-324. 10.1038/nature12296CrossRefPubMed
15.
go back to reference Keusekotten K, Elliott PR, Glockner L, Fiil BK, Damgaard RB, Kulathu Y, Wauer T, Hospenthal MK, Gyrd-Hansen M, Krappmann D, Hofmann K, Komander D: OTULIN antagonizes LUBAC signaling by specifically hydrolyzing Met1-linked polyubiquitin. Cell. 2013, 153 (6): 1312-1326. 10.1016/j.cell.2013.05.014PubMedCentralCrossRefPubMed Keusekotten K, Elliott PR, Glockner L, Fiil BK, Damgaard RB, Kulathu Y, Wauer T, Hospenthal MK, Gyrd-Hansen M, Krappmann D, Hofmann K, Komander D: OTULIN antagonizes LUBAC signaling by specifically hydrolyzing Met1-linked polyubiquitin. Cell. 2013, 153 (6): 1312-1326. 10.1016/j.cell.2013.05.014PubMedCentralCrossRefPubMed
16.
17.
go back to reference Nakayama KI, Nakayama K: Ubiquitin ligases: cell-cycle control and cancer. Nat Rev Canc. 2006, 6 (5): 369-381. 10.1038/nrc1881.CrossRef Nakayama KI, Nakayama K: Ubiquitin ligases: cell-cycle control and cancer. Nat Rev Canc. 2006, 6 (5): 369-381. 10.1038/nrc1881.CrossRef
18.
go back to reference Vucic D, Dixit VM, Wertz IE: Ubiquitylation in apoptosis: a post-translational modification at the edge of life and death. Nat Rev Mol Cell Biol. 2011, 12 (7): 439-452. 10.1038/nrm3143CrossRefPubMed Vucic D, Dixit VM, Wertz IE: Ubiquitylation in apoptosis: a post-translational modification at the edge of life and death. Nat Rev Mol Cell Biol. 2011, 12 (7): 439-452. 10.1038/nrm3143CrossRefPubMed
19.
go back to reference Ulrich HD, Walden H: Ubiquitin signalling in DNA replication and repair. Nat Rev Mol Cell Biol. 2010, 11 (7): 479-489. 10.1038/nrm2921CrossRefPubMed Ulrich HD, Walden H: Ubiquitin signalling in DNA replication and repair. Nat Rev Mol Cell Biol. 2010, 11 (7): 479-489. 10.1038/nrm2921CrossRefPubMed
20.
go back to reference Schaefer A, Nethe M, Hordijk PL: Ubiquitin links to cytoskeletal dynamics, cell adhesion and migration. Biochem J. 2012, 442 (1): 13-25. 10.1042/BJ20111815CrossRefPubMed Schaefer A, Nethe M, Hordijk PL: Ubiquitin links to cytoskeletal dynamics, cell adhesion and migration. Biochem J. 2012, 442 (1): 13-25. 10.1042/BJ20111815CrossRefPubMed
21.
go back to reference Chen D, Frezza M, Schmitt S, Kanwar J, Dou QP: Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives. Curr Canc Drug Targets. 2011, 11 (3): 239-253. 10.2174/156800911794519752.CrossRef Chen D, Frezza M, Schmitt S, Kanwar J, Dou QP: Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives. Curr Canc Drug Targets. 2011, 11 (3): 239-253. 10.2174/156800911794519752.CrossRef
22.
go back to reference Hoeller D, Dikic I: Targeting the ubiquitin system in cancer therapy. Nature. 2009, 458 (7237): 438-444. 10.1038/nature07960CrossRefPubMed Hoeller D, Dikic I: Targeting the ubiquitin system in cancer therapy. Nature. 2009, 458 (7237): 438-444. 10.1038/nature07960CrossRefPubMed
23.
go back to reference Mattern MR, Wu J, Nicholson B: Ubiquitin-based anticancer therapy: carpet bombing with proteasome inhibitors vs surgical strikes with E1, E2, E3, or DUB inhibitors. Biochim Biophys Acta. 2012, 1823 (11): 2014-2021. 10.1016/j.bbamcr.2012.05.005CrossRefPubMed Mattern MR, Wu J, Nicholson B: Ubiquitin-based anticancer therapy: carpet bombing with proteasome inhibitors vs surgical strikes with E1, E2, E3, or DUB inhibitors. Biochim Biophys Acta. 2012, 1823 (11): 2014-2021. 10.1016/j.bbamcr.2012.05.005CrossRefPubMed
24.
go back to reference Edelmann MJ, Nicholson B, Kessler BM: Pharmacological targets in the ubiquitin system offer new ways of treating cancer, neurodegenerative disorders and infectious diseases. Expert Rev Mol Med. 2011, 13: e35-PubMedCentralCrossRefPubMed Edelmann MJ, Nicholson B, Kessler BM: Pharmacological targets in the ubiquitin system offer new ways of treating cancer, neurodegenerative disorders and infectious diseases. Expert Rev Mol Med. 2011, 13: e35-PubMedCentralCrossRefPubMed
25.
go back to reference Fulda S, Rajalingam K, Dikic I: Ubiquitylation in immune disorders and cancer: from molecular mechanisms to therapeutic implications. EMBO Mol Med. 2012, 4 (7): 545-556. 10.1002/emmm.201100707PubMedCentralCrossRefPubMed Fulda S, Rajalingam K, Dikic I: Ubiquitylation in immune disorders and cancer: from molecular mechanisms to therapeutic implications. EMBO Mol Med. 2012, 4 (7): 545-556. 10.1002/emmm.201100707PubMedCentralCrossRefPubMed
26.
go back to reference D’Arcy P, Linder S: Proteasome deubiquitinases as novel targets for cancer therapy. Int J Biochem Cell Biol. 2012, 44 (11): 1729-1738. 10.1016/j.biocel.2012.07.011CrossRefPubMed D’Arcy P, Linder S: Proteasome deubiquitinases as novel targets for cancer therapy. Int J Biochem Cell Biol. 2012, 44 (11): 1729-1738. 10.1016/j.biocel.2012.07.011CrossRefPubMed
27.
go back to reference Nijman SM, Luna-Vargas MP, Velds A, Brummelkamp TR, Dirac AM, Sixma TK, Bernards R: A genomic and functional inventory of deubiquitinating enzymes. Cell. 2005, 123 (5): 773-786. 10.1016/j.cell.2005.11.007CrossRefPubMed Nijman SM, Luna-Vargas MP, Velds A, Brummelkamp TR, Dirac AM, Sixma TK, Bernards R: A genomic and functional inventory of deubiquitinating enzymes. Cell. 2005, 123 (5): 773-786. 10.1016/j.cell.2005.11.007CrossRefPubMed
28.
go back to reference Komander D, Clague MJ, Urbé S: Breaking the chains: structure and function of the deubiquitinases. Nat Rev Mol Cell Biol. 2009, 10 (8): 550-563. 10.1038/nrm2731CrossRefPubMed Komander D, Clague MJ, Urbé S: Breaking the chains: structure and function of the deubiquitinases. Nat Rev Mol Cell Biol. 2009, 10 (8): 550-563. 10.1038/nrm2731CrossRefPubMed
29.
go back to reference Fraile JM, Quesada V, Rodríguez D, Freije JM, López-Otín C: Deubiquitinases in cancer: new functions and therapeutic options. Oncogene. 2012, 31 (19): 2373-2388. 10.1038/onc.2011.443CrossRefPubMed Fraile JM, Quesada V, Rodríguez D, Freije JM, López-Otín C: Deubiquitinases in cancer: new functions and therapeutic options. Oncogene. 2012, 31 (19): 2373-2388. 10.1038/onc.2011.443CrossRefPubMed
30.
go back to reference Sacco JJ, Coulson JM, Clague MJ, Urbé S: Emerging roles of deubiquitinases in cancer-associated pathways. IUBMB Life. 2010, 62 (2): 140-157.PubMed Sacco JJ, Coulson JM, Clague MJ, Urbé S: Emerging roles of deubiquitinases in cancer-associated pathways. IUBMB Life. 2010, 62 (2): 140-157.PubMed
31.
go back to reference Hussain S, Zhang Y, Galardy PJ: DUBs and cancer: the role of deubiquitinating enzymes as oncogenes, non-oncogenes and tumor suppressors. Cell Cycle. 2009, 8 (11): 1688-1697. 10.4161/cc.8.11.8739CrossRefPubMed Hussain S, Zhang Y, Galardy PJ: DUBs and cancer: the role of deubiquitinating enzymes as oncogenes, non-oncogenes and tumor suppressors. Cell Cycle. 2009, 8 (11): 1688-1697. 10.4161/cc.8.11.8739CrossRefPubMed
32.
go back to reference Hock AK, Vigneron AM, Carter S, Ludwig RL, Vousden KH: Regulation of p53 stability and function by the deubiquitinating enzyme USP42. EMBO J. 2011, 30 (24): 4921-4930. 10.1038/emboj.2011.419PubMedCentralCrossRefPubMed Hock AK, Vigneron AM, Carter S, Ludwig RL, Vousden KH: Regulation of p53 stability and function by the deubiquitinating enzyme USP42. EMBO J. 2011, 30 (24): 4921-4930. 10.1038/emboj.2011.419PubMedCentralCrossRefPubMed
33.
go back to reference Aggarwal K, Massagué J: Ubiquitin removal in the TGF-β pathway. Nat Cell Biol. 2012, 14 (7): 656-657. 10.1038/ncb2534CrossRefPubMed Aggarwal K, Massagué J: Ubiquitin removal in the TGF-β pathway. Nat Cell Biol. 2012, 14 (7): 656-657. 10.1038/ncb2534CrossRefPubMed
34.
go back to reference Clague MJ, Urbé S: Endocytosis: the DUB version. Trends Cell Biol. 2006, 16 (11): 551-559. 10.1016/j.tcb.2006.09.002CrossRefPubMed Clague MJ, Urbé S: Endocytosis: the DUB version. Trends Cell Biol. 2006, 16 (11): 551-559. 10.1016/j.tcb.2006.09.002CrossRefPubMed
35.
go back to reference Liu Z, Zanata SM, Kim J, Peterson MA, Di Vizio D, Chirieac LR, Pyne S, Agostini M, Freeman MR, Loda M: The ubiquitin-specific protease USP2a prevents endocytosis-mediated EGFR degradation. Oncogene. 2013, 32 (13): 1660-1669. 10.1038/onc.2012.188CrossRefPubMed Liu Z, Zanata SM, Kim J, Peterson MA, Di Vizio D, Chirieac LR, Pyne S, Agostini M, Freeman MR, Loda M: The ubiquitin-specific protease USP2a prevents endocytosis-mediated EGFR degradation. Oncogene. 2013, 32 (13): 1660-1669. 10.1038/onc.2012.188CrossRefPubMed
36.
go back to reference Pareja F, Ferraro DA, Rubin C, Cohen-Dvashi H, Zhang F, Aulmann S, Ben-Chetrit N, Pines G, Navon R, Crosetto N, Köstler W, Carvalho S, Lavi S, Schmitt F, Dikic I, Yakhini Z, Sinn P, Mills GB, Yarden Y: Deubiquitination of EGFR by Cezanne-1 contributes to cancer progression. Oncogene. 2012, 31 (43): 4599-4608. 10.1038/onc.2011.587PubMedCentralCrossRefPubMed Pareja F, Ferraro DA, Rubin C, Cohen-Dvashi H, Zhang F, Aulmann S, Ben-Chetrit N, Pines G, Navon R, Crosetto N, Köstler W, Carvalho S, Lavi S, Schmitt F, Dikic I, Yakhini Z, Sinn P, Mills GB, Yarden Y: Deubiquitination of EGFR by Cezanne-1 contributes to cancer progression. Oncogene. 2012, 31 (43): 4599-4608. 10.1038/onc.2011.587PubMedCentralCrossRefPubMed
37.
go back to reference Song MS, Salmena L, Carracedo A, Egia A, Lo-Coco F, Teruya-Feldstein J, Pandolfi PP: The deubiquitinylation and localization of PTEN are regulated by a HAUSP-PML network. Nature. 2008, 455 (7214): 813-817. 10.1038/nature07290PubMedCentralCrossRefPubMed Song MS, Salmena L, Carracedo A, Egia A, Lo-Coco F, Teruya-Feldstein J, Pandolfi PP: The deubiquitinylation and localization of PTEN are regulated by a HAUSP-PML network. Nature. 2008, 455 (7214): 813-817. 10.1038/nature07290PubMedCentralCrossRefPubMed
38.
go back to reference Yang WL, Jin G, Li CF, Jeong YS, Moten A, Xu D, Feng Z, Chen W, Cai Z, Darnay B, Gu W, Lin HK: Cycles of ubiquitination and deubiquitination critically regulate growth factor-mediated activation of Akt signaling. Sci Signal. 2013, 6 (257): ra3- 10.1126/scisignal.2003197CrossRefPubMed Yang WL, Jin G, Li CF, Jeong YS, Moten A, Xu D, Feng Z, Chen W, Cai Z, Darnay B, Gu W, Lin HK: Cycles of ubiquitination and deubiquitination critically regulate growth factor-mediated activation of Akt signaling. Sci Signal. 2013, 6 (257): ra3- 10.1126/scisignal.2003197CrossRefPubMed
39.
go back to reference Zhang L, Zhou F, Drabsch Y, Gao R, Snaar-Jagalska BE, Mickanin C, Huang H, Sheppard KA, Porter JA, Lu CX, Ten Dijke P: USP4 is regulated by AKT phosphorylation and directly deubiquitylates TGF-β type I receptor. Nat Cell Biol. 2012, 14 (7): 717-726. 10.1038/ncb2522CrossRefPubMed Zhang L, Zhou F, Drabsch Y, Gao R, Snaar-Jagalska BE, Mickanin C, Huang H, Sheppard KA, Porter JA, Lu CX, Ten Dijke P: USP4 is regulated by AKT phosphorylation and directly deubiquitylates TGF-β type I receptor. Nat Cell Biol. 2012, 14 (7): 717-726. 10.1038/ncb2522CrossRefPubMed
40.
go back to reference Jackson SP, Durocher D: Regulation of DNA damage responses by Ubiquitin and SUMO. Mol Cell. 2013, 49 (5): 795-807. 10.1016/j.molcel.2013.01.017CrossRefPubMed Jackson SP, Durocher D: Regulation of DNA damage responses by Ubiquitin and SUMO. Mol Cell. 2013, 49 (5): 795-807. 10.1016/j.molcel.2013.01.017CrossRefPubMed
41.
go back to reference Colland F: The therapeutic potential of deubiquitinating enzyme inhibitors. Biochem Soc Trans. 2010, 38 (Pt 1): 137-143.CrossRefPubMed Colland F: The therapeutic potential of deubiquitinating enzyme inhibitors. Biochem Soc Trans. 2010, 38 (Pt 1): 137-143.CrossRefPubMed
42.
go back to reference Altun M, Kramer HB, Willems LI, McDermott JL, Leach CA, Goldenberg SJ, Kumar KG, Konietzny R, Fischer R, Kogan E, Mackeen MM, McGouran J, Khoronenkova SV, Parsons JL, Dianov GL, Nicholson B, Kessler BM: Activity-based chemical proteomics accelerates inhibitor development for deubiquitylating enzymes. Chem Biol. 2011, 18 (11): 1401-1412. 10.1016/j.chembiol.2011.08.018CrossRefPubMed Altun M, Kramer HB, Willems LI, McDermott JL, Leach CA, Goldenberg SJ, Kumar KG, Konietzny R, Fischer R, Kogan E, Mackeen MM, McGouran J, Khoronenkova SV, Parsons JL, Dianov GL, Nicholson B, Kessler BM: Activity-based chemical proteomics accelerates inhibitor development for deubiquitylating enzymes. Chem Biol. 2011, 18 (11): 1401-1412. 10.1016/j.chembiol.2011.08.018CrossRefPubMed
43.
go back to reference Kapuria V, Peterson LF, Fang D, Bornmann WG, Talpaz M, Donato NJ: Deubiquitinase inhibition by small-molecule WP1130 triggers aggresome formation and tumor cell apoptosis. Canc Res. 2010, 70 (22): 9265-9276. 10.1158/0008-5472.CAN-10-1530.CrossRef Kapuria V, Peterson LF, Fang D, Bornmann WG, Talpaz M, Donato NJ: Deubiquitinase inhibition by small-molecule WP1130 triggers aggresome formation and tumor cell apoptosis. Canc Res. 2010, 70 (22): 9265-9276. 10.1158/0008-5472.CAN-10-1530.CrossRef
44.
go back to reference D’Arcy P, Brnjic S, Olofsson MH, Fryknäs M, Lindsten K, De Cesare M, Perego P, Sadeghi B, Hassan M, Larsson R, Linder S: Inhibition of proteasome deubiquitinating activity as a new cancer therapy. Nat Med. 2011, 17 (12): 1636-1640. 10.1038/nm.2536CrossRefPubMed D’Arcy P, Brnjic S, Olofsson MH, Fryknäs M, Lindsten K, De Cesare M, Perego P, Sadeghi B, Hassan M, Larsson R, Linder S: Inhibition of proteasome deubiquitinating activity as a new cancer therapy. Nat Med. 2011, 17 (12): 1636-1640. 10.1038/nm.2536CrossRefPubMed
45.
go back to reference Ohayon S, Spasser L, Aharoni A, Brik A: Targeting deubiquitinases enabled by chemical synthesis of proteins. J Am Chem Soc. 2012, 134 (6): 3281-3289. 10.1021/ja2116712CrossRefPubMed Ohayon S, Spasser L, Aharoni A, Brik A: Targeting deubiquitinases enabled by chemical synthesis of proteins. J Am Chem Soc. 2012, 134 (6): 3281-3289. 10.1021/ja2116712CrossRefPubMed
46.
go back to reference Ernst A, Avvakumov G, Tong J, Fan Y, Zhao Y, Alberts P, Persaud A, Walker JR, Neculai AM, Neculai D, Vorobyov A, Garg P, Beatty L, Chan PK, Juang YC, Landry MC, Yeh C, Zeqiraj E, Karamboulas K, Allali-Hassani A, Vedadi M, Tyers M, Moffat J, Sicheri F, Pelletier L, Durocher D, Raught B, Rotin D, Yang J, Moran MF, Dhe-Paganon S, Sidhu SS: A strategy for modulation of enzymes in the ubiquitin system. Science. 2013, 339 (6119): 590-595. 10.1126/science.1230161CrossRefPubMed Ernst A, Avvakumov G, Tong J, Fan Y, Zhao Y, Alberts P, Persaud A, Walker JR, Neculai AM, Neculai D, Vorobyov A, Garg P, Beatty L, Chan PK, Juang YC, Landry MC, Yeh C, Zeqiraj E, Karamboulas K, Allali-Hassani A, Vedadi M, Tyers M, Moffat J, Sicheri F, Pelletier L, Durocher D, Raught B, Rotin D, Yang J, Moran MF, Dhe-Paganon S, Sidhu SS: A strategy for modulation of enzymes in the ubiquitin system. Science. 2013, 339 (6119): 590-595. 10.1126/science.1230161CrossRefPubMed
47.
go back to reference Bouwman P, Jonkers J: The effects of deregulated DNA damage signalling on cancer chemotherapy response and resistance. Nat Rev Canc. 2012, 12 (9): 587-598. 10.1038/nrc3342.CrossRef Bouwman P, Jonkers J: The effects of deregulated DNA damage signalling on cancer chemotherapy response and resistance. Nat Rev Canc. 2012, 12 (9): 587-598. 10.1038/nrc3342.CrossRef
48.
go back to reference Curtin NJ: DNA repair dysregulation from cancer driver to therapeutic target. Nat Rev Canc. 2012, 12 (12): 801-817. 10.1038/nrc3399.CrossRef Curtin NJ: DNA repair dysregulation from cancer driver to therapeutic target. Nat Rev Canc. 2012, 12 (12): 801-817. 10.1038/nrc3399.CrossRef
49.
go back to reference Hofmann K: Ubiquitin-binding domains and their role in the DNA damage response. DNA Repair (Amst). 2009, 8 (4): 544-556. 10.1016/j.dnarep.2009.01.003.CrossRef Hofmann K: Ubiquitin-binding domains and their role in the DNA damage response. DNA Repair (Amst). 2009, 8 (4): 544-556. 10.1016/j.dnarep.2009.01.003.CrossRef
50.
go back to reference Kirchmaier AL: Ub-family modifications at the replication fork: regulating PCNA-interacting components. FEBS Lett. 2011, 585 (18): 2920-2928. 10.1016/j.febslet.2011.08.008CrossRefPubMed Kirchmaier AL: Ub-family modifications at the replication fork: regulating PCNA-interacting components. FEBS Lett. 2011, 585 (18): 2920-2928. 10.1016/j.febslet.2011.08.008CrossRefPubMed
51.
go back to reference Kim H, D’Andrea AD: Regulation of DNA cross-link repair by the Fanconi anemia/BRCA pathway. Genes Dev. 2012, 26 (13): 1393-1408. 10.1101/gad.195248.112PubMedCentralCrossRefPubMed Kim H, D’Andrea AD: Regulation of DNA cross-link repair by the Fanconi anemia/BRCA pathway. Genes Dev. 2012, 26 (13): 1393-1408. 10.1101/gad.195248.112PubMedCentralCrossRefPubMed
52.
go back to reference Fujiwara T, Saito A, Suzuki M, Shinomiya H, Suzuki T, Takahashi E, Tanigami A, Ichiyama A, Chung CH, Nakamura Y, Tanaka K: Identification and chromosomal assignment of USP1, a novel gene encoding a human ubiquitin-specific protease. Genomics. 1998, 54 (1): 155-158. 10.1006/geno.1998.5554CrossRefPubMed Fujiwara T, Saito A, Suzuki M, Shinomiya H, Suzuki T, Takahashi E, Tanigami A, Ichiyama A, Chung CH, Nakamura Y, Tanaka K: Identification and chromosomal assignment of USP1, a novel gene encoding a human ubiquitin-specific protease. Genomics. 1998, 54 (1): 155-158. 10.1006/geno.1998.5554CrossRefPubMed
53.
go back to reference Villamil MA, Chen J, Liang Q, Zhuang Z: A noncanonical cysteine protease USP1 is activated through active site modulation by USP1-associated factor 1. Biochemistry. 2012, 51 (13): 2829-2839. 10.1021/bi3000512CrossRefPubMed Villamil MA, Chen J, Liang Q, Zhuang Z: A noncanonical cysteine protease USP1 is activated through active site modulation by USP1-associated factor 1. Biochemistry. 2012, 51 (13): 2829-2839. 10.1021/bi3000512CrossRefPubMed
54.
go back to reference Békés M, Huang T: Ubiquitin-specific peptidase 1. Handbook of proteolytic enzymes. Volume 1. Edited by: Rawlings ND, Salvesen G. 2013, 2079-2085. 3,CrossRef Békés M, Huang T: Ubiquitin-specific peptidase 1. Handbook of proteolytic enzymes. Volume 1. Edited by: Rawlings ND, Salvesen G. 2013, 2079-2085. 3,CrossRef
55.
go back to reference Nijman SM, Huang TT, Dirac AM, Brummelkamp TR, Kerkhoven RM, D’Andrea AD, Bernards R: The deubiquitinating enzyme USP1 regulates the Fanconi anemia pathway. Mol Cell. 2005, 17 (3): 331-339. 10.1016/j.molcel.2005.01.008CrossRefPubMed Nijman SM, Huang TT, Dirac AM, Brummelkamp TR, Kerkhoven RM, D’Andrea AD, Bernards R: The deubiquitinating enzyme USP1 regulates the Fanconi anemia pathway. Mol Cell. 2005, 17 (3): 331-339. 10.1016/j.molcel.2005.01.008CrossRefPubMed
56.
go back to reference Huang TT, Nijman SM, Mirchandani KD, Galardy PJ, Cohn MA, Haas W, Gygi SP, Ploegh HL, Bernards R, D’Andrea AD: Regulation of monoubiquitinated PCNA by DUB autocleavage. Nat Cell Biol. 2006, 8 (4): 339-347.CrossRefPubMed Huang TT, Nijman SM, Mirchandani KD, Galardy PJ, Cohn MA, Haas W, Gygi SP, Ploegh HL, Bernards R, D’Andrea AD: Regulation of monoubiquitinated PCNA by DUB autocleavage. Nat Cell Biol. 2006, 8 (4): 339-347.CrossRefPubMed
57.
go back to reference Williams SA, Maecker HL, French DM, Liu J, Gregg A, Silverstein LB, Cao TC, Carano RA, Dixit VM: USP1 deubiquitinates ID proteins to preserve a mesenchymal stem cell program in osteosarcoma. Cell. 2011, 146 (6): 918-930. 10.1016/j.cell.2011.07.040CrossRefPubMed Williams SA, Maecker HL, French DM, Liu J, Gregg A, Silverstein LB, Cao TC, Carano RA, Dixit VM: USP1 deubiquitinates ID proteins to preserve a mesenchymal stem cell program in osteosarcoma. Cell. 2011, 146 (6): 918-930. 10.1016/j.cell.2011.07.040CrossRefPubMed
58.
go back to reference Cohn MA, Kowal P, Yang K, Haas W, Huang TT, Gygi SP, D’Andrea AD: A UAF1-containing multisubunit protein complex regulates the Fanconi anemia pathway. Mol Cell. 2007, 28 (5): 786-797. 10.1016/j.molcel.2007.09.031CrossRefPubMed Cohn MA, Kowal P, Yang K, Haas W, Huang TT, Gygi SP, D’Andrea AD: A UAF1-containing multisubunit protein complex regulates the Fanconi anemia pathway. Mol Cell. 2007, 28 (5): 786-797. 10.1016/j.molcel.2007.09.031CrossRefPubMed
59.
60.
go back to reference Sims AE, Spiteri E, Sims RJ, Arita AG, Lach FP, Landers T, Wurm M, Freund M, Neveling K, Hanenberg H, Auerbach AD, Huang TT: FANCI is a second monoubiquitinated member of the Fanconi anemia pathway. Nat Struct Mol Biol. 2007, 14 (6): 564-567. 10.1038/nsmb1252CrossRefPubMed Sims AE, Spiteri E, Sims RJ, Arita AG, Lach FP, Landers T, Wurm M, Freund M, Neveling K, Hanenberg H, Auerbach AD, Huang TT: FANCI is a second monoubiquitinated member of the Fanconi anemia pathway. Nat Struct Mol Biol. 2007, 14 (6): 564-567. 10.1038/nsmb1252CrossRefPubMed
61.
go back to reference Parmar K, Kim J, Sykes SM, Shimamura A, Stuckert P, Zhu K, Hamilton A, Deloach MK, Kutok JL, Akashi K, Gilliland DG, D’andrea A: Hematopoietic stem cell defects in mice with deficiency of Fancd2 or Usp1. Stem Cells. 2010, 28 (7): 1186-1195. 10.1002/stem.437PubMedCentralCrossRefPubMed Parmar K, Kim J, Sykes SM, Shimamura A, Stuckert P, Zhu K, Hamilton A, Deloach MK, Kutok JL, Akashi K, Gilliland DG, D’andrea A: Hematopoietic stem cell defects in mice with deficiency of Fancd2 or Usp1. Stem Cells. 2010, 28 (7): 1186-1195. 10.1002/stem.437PubMedCentralCrossRefPubMed
62.
go back to reference Oestergaard VH, Langevin F, Kuiken HJ, Pace P, Niedzwiedz W, Simpson LJ, Ohzeki M, Takata M, Sale JE, Patel KJ: Deubiquitination of FANCD2 is required for DNA crosslink repair. Mol Cell. 2007, 28 (5): 798-809. 10.1016/j.molcel.2007.09.020PubMedCentralCrossRefPubMed Oestergaard VH, Langevin F, Kuiken HJ, Pace P, Niedzwiedz W, Simpson LJ, Ohzeki M, Takata M, Sale JE, Patel KJ: Deubiquitination of FANCD2 is required for DNA crosslink repair. Mol Cell. 2007, 28 (5): 798-809. 10.1016/j.molcel.2007.09.020PubMedCentralCrossRefPubMed
63.
go back to reference Kim JM, Parmar K, Huang M, Weinstock DM, Ruit CA, Kutok JL, D’Andrea AD: Inactivation of murine Usp1 results in genomic instability and a Fanconi anemia phenotype. Dev Cell. 2009, 16 (2): 314-320. 10.1016/j.devcel.2009.01.001PubMedCentralCrossRefPubMed Kim JM, Parmar K, Huang M, Weinstock DM, Ruit CA, Kutok JL, D’Andrea AD: Inactivation of murine Usp1 results in genomic instability and a Fanconi anemia phenotype. Dev Cell. 2009, 16 (2): 314-320. 10.1016/j.devcel.2009.01.001PubMedCentralCrossRefPubMed
64.
go back to reference Murai J, Yang K, Dejsuphong D, Hirota K, Takeda S, D’Andrea AD: The USP1/UAF1 complex promotes double-strand break repair through homologous recombination. Mol Cell Biol. 2011, 31 (12): 2462-2469. 10.1128/MCB.05058-11PubMedCentralCrossRefPubMed Murai J, Yang K, Dejsuphong D, Hirota K, Takeda S, D’Andrea AD: The USP1/UAF1 complex promotes double-strand break repair through homologous recombination. Mol Cell Biol. 2011, 31 (12): 2462-2469. 10.1128/MCB.05058-11PubMedCentralCrossRefPubMed
65.
go back to reference Hoege C, Pfander B, Moldovan GL, Pyrowolakis G, Jentsch S: RAD6-dependent DNA repair is linked to modification of PCNA by ubiquitin and SUMO. Nature. 2002, 419 (6903): 135-141. 10.1038/nature00991CrossRefPubMed Hoege C, Pfander B, Moldovan GL, Pyrowolakis G, Jentsch S: RAD6-dependent DNA repair is linked to modification of PCNA by ubiquitin and SUMO. Nature. 2002, 419 (6903): 135-141. 10.1038/nature00991CrossRefPubMed
66.
go back to reference Friedberg EC, Lehmann AR, Fuchs RP: Trading places: how do DNA polymerases switch during translesion DNA synthesis?. Mol Cell. 2005, 18 (5): 499-505. 10.1016/j.molcel.2005.03.032CrossRefPubMed Friedberg EC, Lehmann AR, Fuchs RP: Trading places: how do DNA polymerases switch during translesion DNA synthesis?. Mol Cell. 2005, 18 (5): 499-505. 10.1016/j.molcel.2005.03.032CrossRefPubMed
67.
go back to reference Terai K, Abbas T, Jazaeri AA, Dutta A: CRL4Cdt2 E3 ubiquitin ligase monoubiquitinates PCNA to promote translesion DNA synthesis. Mol Cell. 2010, 37 (1): 143-149. 10.1016/j.molcel.2009.12.018PubMedCentralCrossRefPubMed Terai K, Abbas T, Jazaeri AA, Dutta A: CRL4Cdt2 E3 ubiquitin ligase monoubiquitinates PCNA to promote translesion DNA synthesis. Mol Cell. 2010, 37 (1): 143-149. 10.1016/j.molcel.2009.12.018PubMedCentralCrossRefPubMed
68.
go back to reference Jones MJ, Colnaghi L, Huang TT: Dysregulation of DNA polymerase κ recruitment to replication forks results in genomic instability. EMBO J. 2012, 31 (4): 908-918.PubMedCentralCrossRefPubMed Jones MJ, Colnaghi L, Huang TT: Dysregulation of DNA polymerase κ recruitment to replication forks results in genomic instability. EMBO J. 2012, 31 (4): 908-918.PubMedCentralCrossRefPubMed
69.
go back to reference Yokota Y, Mori S: Role of Id family proteins in growth control. J Cell Physiol. 2002, 190 (1): 21-28. 10.1002/jcp.10042CrossRefPubMed Yokota Y, Mori S: Role of Id family proteins in growth control. J Cell Physiol. 2002, 190 (1): 21-28. 10.1002/jcp.10042CrossRefPubMed
70.
go back to reference Perk J, Iavarone A, Benezra R: Id family of helix-loop-helix proteins in cancer. Nat Rev Canc. 2005, 5 (8): 603-614. 10.1038/nrc1673.CrossRef Perk J, Iavarone A, Benezra R: Id family of helix-loop-helix proteins in cancer. Nat Rev Canc. 2005, 5 (8): 603-614. 10.1038/nrc1673.CrossRef
71.
go back to reference Cotto-Rios XM, Jones MJ, Busino L, Pagano M, Huang TT: APC/CCdh1-dependent proteolysis of USP1 regulates the response to UV-mediated DNA damage. J Cell Biol. 2011, 194 (2): 177-186. 10.1083/jcb.201101062PubMedCentralCrossRefPubMed Cotto-Rios XM, Jones MJ, Busino L, Pagano M, Huang TT: APC/CCdh1-dependent proteolysis of USP1 regulates the response to UV-mediated DNA damage. J Cell Biol. 2011, 194 (2): 177-186. 10.1083/jcb.201101062PubMedCentralCrossRefPubMed
72.
go back to reference Cataldo F, Peche LY, Klaric E, Brancolini C, Myers MP, Demarchi F, Schneider C: CAPNS1 Regulates USP1 Stability and the Maintenance of Genome Integrity. Mol Cell Biol. 2013, 33 (12): 2485-2496. 10.1128/MCB.01406-12PubMedCentralCrossRefPubMed Cataldo F, Peche LY, Klaric E, Brancolini C, Myers MP, Demarchi F, Schneider C: CAPNS1 Regulates USP1 Stability and the Maintenance of Genome Integrity. Mol Cell Biol. 2013, 33 (12): 2485-2496. 10.1128/MCB.01406-12PubMedCentralCrossRefPubMed
73.
go back to reference Garcia-Santisteban I, Zorroza K, Rodriguez JA: Two nuclear localization signals in USP1 mediate nuclear import of the USP1/UAF1 complex. PLoS One. 2012, 7 (6): e38570- 10.1371/journal.pone.0038570PubMedCentralCrossRefPubMed Garcia-Santisteban I, Zorroza K, Rodriguez JA: Two nuclear localization signals in USP1 mediate nuclear import of the USP1/UAF1 complex. PLoS One. 2012, 7 (6): e38570- 10.1371/journal.pone.0038570PubMedCentralCrossRefPubMed
74.
go back to reference Villamil MA, Liang Q, Chen J, Choi YS, Hou S, Lee KH, Zhuang Z: Serine phosphorylation is critical for the activation of ubiquitin-specific protease 1 and its interaction with WD40-repeat protein UAF1. Biochemistry. 2012, 51 (45): 9112-9123. 10.1021/bi300845sPubMedCentralCrossRefPubMed Villamil MA, Liang Q, Chen J, Choi YS, Hou S, Lee KH, Zhuang Z: Serine phosphorylation is critical for the activation of ubiquitin-specific protease 1 and its interaction with WD40-repeat protein UAF1. Biochemistry. 2012, 51 (45): 9112-9123. 10.1021/bi300845sPubMedCentralCrossRefPubMed
75.
go back to reference Cohn MA, Kee Y, Haas W, Gygi SP, D’Andrea AD: UAF1 is a subunit of multiple deubiquitinating enzyme complexes. J Biol Chem. 2009, 284 (8): 5343-5351.PubMedCentralCrossRefPubMed Cohn MA, Kee Y, Haas W, Gygi SP, D’Andrea AD: UAF1 is a subunit of multiple deubiquitinating enzyme complexes. J Biol Chem. 2009, 284 (8): 5343-5351.PubMedCentralCrossRefPubMed
76.
go back to reference Yang K, Moldovan GL, Vinciguerra P, Murai J, Takeda S, D’Andrea AD: Regulation of the Fanconi anemia pathway by a SUMO-like delivery network. Genes Dev. 2011, 25 (17): 1847-1858. 10.1101/gad.17020911PubMedCentralCrossRefPubMed Yang K, Moldovan GL, Vinciguerra P, Murai J, Takeda S, D’Andrea AD: Regulation of the Fanconi anemia pathway by a SUMO-like delivery network. Genes Dev. 2011, 25 (17): 1847-1858. 10.1101/gad.17020911PubMedCentralCrossRefPubMed
77.
go back to reference Lee KY, Yang K, Cohn MA, Sikdar N, D’Andrea AD, Myung K: Human ELG1 regulates the level of ubiquitinated proliferating cell nuclear antigen (PCNA) through its interactions with PCNA and USP1. J Biol Chem. 2010, 285 (14): 10362-10369. 10.1074/jbc.M109.092544PubMedCentralCrossRefPubMed Lee KY, Yang K, Cohn MA, Sikdar N, D’Andrea AD, Myung K: Human ELG1 regulates the level of ubiquitinated proliferating cell nuclear antigen (PCNA) through its interactions with PCNA and USP1. J Biol Chem. 2010, 285 (14): 10362-10369. 10.1074/jbc.M109.092544PubMedCentralCrossRefPubMed
78.
go back to reference García-Santisteban I, Bañuelos S, Rodríguez JA: A global survey of CRM1-dependent nuclear export sequences in the human deubiquitinase family. Biochem J. 2012, 441 (1): 209-217. 10.1042/BJ20111300CrossRefPubMed García-Santisteban I, Bañuelos S, Rodríguez JA: A global survey of CRM1-dependent nuclear export sequences in the human deubiquitinase family. Biochem J. 2012, 441 (1): 209-217. 10.1042/BJ20111300CrossRefPubMed
79.
go back to reference Rego MA, Harney JA, Mauro M, Shen M, Howlett NG: Regulation of the activation of the Fanconi anemia pathway by the p21 cyclin-dependent kinase inhibitor. Oncogene. 2012, 31 (3): 366-375. 10.1038/onc.2011.237PubMedCentralCrossRefPubMed Rego MA, Harney JA, Mauro M, Shen M, Howlett NG: Regulation of the activation of the Fanconi anemia pathway by the p21 cyclin-dependent kinase inhibitor. Oncogene. 2012, 31 (3): 366-375. 10.1038/onc.2011.237PubMedCentralCrossRefPubMed
80.
go back to reference Piatkov KI, Colnaghi L, Békés M, Varshavsky A, Huang TT: The auto-generated fragment of the Usp1 deubiquitylase is a physiological substrate of the N-end rule pathway. Mol Cell. 2012, 48 (6): 926-933. 10.1016/j.molcel.2012.10.012PubMedCentralCrossRefPubMed Piatkov KI, Colnaghi L, Békés M, Varshavsky A, Huang TT: The auto-generated fragment of the Usp1 deubiquitylase is a physiological substrate of the N-end rule pathway. Mol Cell. 2012, 48 (6): 926-933. 10.1016/j.molcel.2012.10.012PubMedCentralCrossRefPubMed
81.
go back to reference Cotto-Rios XM, Békés M, Chapman J, Ueberheide B, Huang TT: Deubiquitinases as a signaling target of oxidative stress. Cell Rep. 2012, 2 (6): 1475-1484. 10.1016/j.celrep.2012.11.011PubMedCentralCrossRefPubMed Cotto-Rios XM, Békés M, Chapman J, Ueberheide B, Huang TT: Deubiquitinases as a signaling target of oxidative stress. Cell Rep. 2012, 2 (6): 1475-1484. 10.1016/j.celrep.2012.11.011PubMedCentralCrossRefPubMed
82.
go back to reference Lee JG, Baek K, Soetandyo N, Ye Y: Reversible inactivation of deubiquitinases by reactive oxygen species in vitro and in cells. Nat Commun. 2013, 4: 1568-PubMedCentralCrossRefPubMed Lee JG, Baek K, Soetandyo N, Ye Y: Reversible inactivation of deubiquitinases by reactive oxygen species in vitro and in cells. Nat Commun. 2013, 4: 1568-PubMedCentralCrossRefPubMed
83.
go back to reference Kulathu Y, Garcia FJ, Mevissen TE, Busch M, Arnaudo N, Carroll KS, Barford D, Komander D: Regulation of A20 and other OTU deubiquitinases by reversible oxidation. Nat Commun. 2013, 4: 1569-PubMedCentralCrossRefPubMed Kulathu Y, Garcia FJ, Mevissen TE, Busch M, Arnaudo N, Carroll KS, Barford D, Komander D: Regulation of A20 and other OTU deubiquitinases by reversible oxidation. Nat Commun. 2013, 4: 1569-PubMedCentralCrossRefPubMed
84.
go back to reference Cotto-Rios XM, Jones MJ, Huang TT: Insights into phosphorylation-dependent mechanisms regulating USP1 protein stability during the cell cycle. Cell Cycle. 2011, 10 (23): 4009-4016. 10.4161/cc.10.23.18501PubMedCentralCrossRefPubMed Cotto-Rios XM, Jones MJ, Huang TT: Insights into phosphorylation-dependent mechanisms regulating USP1 protein stability during the cell cycle. Cell Cycle. 2011, 10 (23): 4009-4016. 10.4161/cc.10.23.18501PubMedCentralCrossRefPubMed
85.
go back to reference Bignell GR, Warren W, Seal S, Takahashi M, Rapley E, Barfoot R, Green H, Brown C, Biggs PJ, Lakhani SR, Jones C, Hansen J, Blair E, Hofmann B, Siebert R, Turner G, Evans DG, Schrander-Stumpel C, Beemer FA, van Den Ouweland A, Halley D, Delpech B, Cleveland MG, Leigh I, Leisti J, Rasmussen S: Identification of the familial cylindromatosis tumour-suppressor gene. Nat Genet. 2000, 25 (2): 160-165. 10.1038/76006CrossRefPubMed Bignell GR, Warren W, Seal S, Takahashi M, Rapley E, Barfoot R, Green H, Brown C, Biggs PJ, Lakhani SR, Jones C, Hansen J, Blair E, Hofmann B, Siebert R, Turner G, Evans DG, Schrander-Stumpel C, Beemer FA, van Den Ouweland A, Halley D, Delpech B, Cleveland MG, Leigh I, Leisti J, Rasmussen S: Identification of the familial cylindromatosis tumour-suppressor gene. Nat Genet. 2000, 25 (2): 160-165. 10.1038/76006CrossRefPubMed
88.
go back to reference Toffalorio F, Giovannetti E, De Pas T, Radice D, Pelosi G, Manzotti M, Minocci D, Spaggiari L, Spitaleri G, Noberasco C, Catania C, Boselli S, Danesi R, De Braud F: Expression of gemcitabine- and cisplatin-related genes in non-small-cell lung cancer. Pharmacogenomics J. 2010, 10 (3): 180-190. 10.1038/tpj.2009.53CrossRefPubMed Toffalorio F, Giovannetti E, De Pas T, Radice D, Pelosi G, Manzotti M, Minocci D, Spaggiari L, Spitaleri G, Noberasco C, Catania C, Boselli S, Danesi R, De Braud F: Expression of gemcitabine- and cisplatin-related genes in non-small-cell lung cancer. Pharmacogenomics J. 2010, 10 (3): 180-190. 10.1038/tpj.2009.53CrossRefPubMed
89.
go back to reference Luise C, Capra M, Donzelli M, Mazzarol G, Jodice MG, Nuciforo P, Viale G, Di Fiore PP, Confalonieri S: An atlas of altered expression of deubiquitinating enzymes in human cancer. PLoS One. 2011, 6 (1): e15891- 10.1371/journal.pone.0015891PubMedCentralCrossRefPubMed Luise C, Capra M, Donzelli M, Mazzarol G, Jodice MG, Nuciforo P, Viale G, Di Fiore PP, Confalonieri S: An atlas of altered expression of deubiquitinating enzymes in human cancer. PLoS One. 2011, 6 (1): e15891- 10.1371/journal.pone.0015891PubMedCentralCrossRefPubMed
91.
go back to reference Liu Y, Luo X, Hu H, Wang R, Sun Y, Zeng R, Chen H: Integrative proteomics and tissue microarray profiling indicate the association between overexpressed serum proteins and non-small cell lung cancer. PLoS One. 2012, 7 (12): e51748- 10.1371/journal.pone.0051748PubMedCentralCrossRefPubMed Liu Y, Luo X, Hu H, Wang R, Sun Y, Zeng R, Chen H: Integrative proteomics and tissue microarray profiling indicate the association between overexpressed serum proteins and non-small cell lung cancer. PLoS One. 2012, 7 (12): e51748- 10.1371/journal.pone.0051748PubMedCentralCrossRefPubMed
92.
go back to reference Zhiqiang Z, Qinghui Y, Yongqiang Z, Jian Z, Xin Z, Haiying M, Yuepeng G: USP1 regulates AKT phosphorylation by modulating the stability of PHLPP1 in lung cancer cells. J Canc Res Clin Oncol. 2012, 138 (7): 1231-1238. 10.1007/s00432-012-1193-3. 10.1007/s00432-012-1193-3CrossRef Zhiqiang Z, Qinghui Y, Yongqiang Z, Jian Z, Xin Z, Haiying M, Yuepeng G: USP1 regulates AKT phosphorylation by modulating the stability of PHLPP1 in lung cancer cells. J Canc Res Clin Oncol. 2012, 138 (7): 1231-1238. 10.1007/s00432-012-1193-3. 10.1007/s00432-012-1193-3CrossRef
93.
go back to reference Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M, Kroemer G: Molecular mechanisms of cisplatin resistance. Oncogene. 2012, 31 (15): 1869-1883. 10.1038/onc.2011.384CrossRefPubMed Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M, Kroemer G: Molecular mechanisms of cisplatin resistance. Oncogene. 2012, 31 (15): 1869-1883. 10.1038/onc.2011.384CrossRefPubMed
94.
go back to reference Chen J, Dexheimer TS, Ai Y, Liang Q, Villamil MA, Inglese J, Maloney DJ, Jadhav A, Simeonov A, Zhuang Z: Selective and cell-active inhibitors of the USP1/UAF1 deubiquitinase complex reverse cisplatin resistance in non-small cell lung cancer cells. Chem Biol. 2011, 18 (11): 1390-1400. 10.1016/j.chembiol.2011.08.014PubMedCentralCrossRefPubMed Chen J, Dexheimer TS, Ai Y, Liang Q, Villamil MA, Inglese J, Maloney DJ, Jadhav A, Simeonov A, Zhuang Z: Selective and cell-active inhibitors of the USP1/UAF1 deubiquitinase complex reverse cisplatin resistance in non-small cell lung cancer cells. Chem Biol. 2011, 18 (11): 1390-1400. 10.1016/j.chembiol.2011.08.014PubMedCentralCrossRefPubMed
95.
go back to reference Chou TC, Talalay P: Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 1984, 22: 27-55.CrossRefPubMed Chou TC, Talalay P: Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 1984, 22: 27-55.CrossRefPubMed
96.
go back to reference Bijnsdorp IV, Giovannetti E, Peters GJ: Analysis of drug interactions. Meth Mol Biol. 2011, 731: 421-434. 10.1007/978-1-61779-080-5_34.CrossRef Bijnsdorp IV, Giovannetti E, Peters GJ: Analysis of drug interactions. Meth Mol Biol. 2011, 731: 421-434. 10.1007/978-1-61779-080-5_34.CrossRef
97.
go back to reference Skehan P, Storeng R, Scudiero D, Monks A, McMahon J, Vistica D, Warren JT, Bokesch H, Kenney S, Boyd MR: New colorimetric cytotoxicity assay for anticancer-drug screening. J Natl Canc Inst. 1990, 82 (13): 1107-1112. 10.1093/jnci/82.13.1107.CrossRef Skehan P, Storeng R, Scudiero D, Monks A, McMahon J, Vistica D, Warren JT, Bokesch H, Kenney S, Boyd MR: New colorimetric cytotoxicity assay for anticancer-drug screening. J Natl Canc Inst. 1990, 82 (13): 1107-1112. 10.1093/jnci/82.13.1107.CrossRef
98.
go back to reference Sowa ME, Bennett EJ, Gygi SP, Harper JW: Defining the human deubiquitinating enzyme interaction landscape. Cell. 2009, 138 (2): 389-403. 10.1016/j.cell.2009.04.042PubMedCentralCrossRefPubMed Sowa ME, Bennett EJ, Gygi SP, Harper JW: Defining the human deubiquitinating enzyme interaction landscape. Cell. 2009, 138 (2): 389-403. 10.1016/j.cell.2009.04.042PubMedCentralCrossRefPubMed
Metadata
Title
USP1 deubiquitinase: cellular functions, regulatory mechanisms and emerging potential as target in cancer therapy
Authors
Iraia García-Santisteban
Godefridus J Peters
Elisa Giovannetti
Jose Antonio Rodríguez
Publication date
01-12-2013
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2013
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-12-91

Other articles of this Issue 1/2013

Molecular Cancer 1/2013 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine