2,176
Views
83
CrossRef citations to date
0
Altmetric
Review

Deubiquitinases (DUBs) and DUB inhibitors: a patent review

, , , , , & show all

Bibliography

  • Komander D. Mechanism, Specificity and structure of the deubiquitinases. in: subcellular biochemistry. Deubiquitiantion 2010;54:69-87
  • Komander D. The emerging complexity of protein ubiquitination. Biochem Soc Trans 2009;37(Pt 5):937-53
  • Matias AC, Ramos PC, Dohmen RJ. Chaperone-assisted assembly of the proteasome core particle. Biochem Soc Trans 2010;38(Pt 1):29-33
  • Peth A, Besche HC, Goldberg AL. Ubiquitinated proteins activate the proteasome by binding to Usp14/Ubp6, which causes 20S gate opening. Mol Cell 2009;36(5):794-804
  • Schmitt S, Deshmukh RR, Dou PQ. Resistance to Proteasome Inhibitors in Cancer. Springer; 2014
  • Dou QP, Zonder JA. Overview of proteasome inhibitor-based anti-cancer therapies: perspective on bortezomib and second generation proteasome inhibitors versus future generation inhibitors of ubiquitin-proteasome system. Cur Cancer Drug Targets 2014;14(6):517-36
  • Shen M, Schmitt S, Buac D, Dou QP. Targeting the ubiquitin-proteasome system for cancer therapy. Expert Opin Ther Targets 2013;17(9):1091-108
  • Lilienbaum A. Relationship between the proteasomal system and autophagy. Int J Biochem Mol Biol 2013;4(1):1-26
  • Wang XJ, Yu J, Wong SH, et al. A novel crosstalk between two major protein degradation systems: regulation of proteasomal activity by autophagy. Autophagy 2013;9(10):1500-8
  • Atkin G, Paulson H. Ubiquitin pathways in neurodegenerative disease. Front Mol Neurosci 2014;7:63
  • Nedelsky NB, Todd PK, Taylor JP. Autophagy and the ubiquitin-proteasome system: collaborators in neuroprotection. Biochim Biophys Acta 2008;1782(12):691-9
  • Suryadinata R, Roesley SN, Yang G, Sarcevic B. Mechanisms of generating polyubiquitin chains of different topology. Cells 2014;3(3):674-89
  • Komander D, Clague MJ, Urbe S. Breaking the chains: structure and function of the deubiquitinases. Nat Rev Mol Cell Biol 2009;10(8):550-63
  • Amerik A, Swaminathan S, Krantz BA, et al. In vivo disassembly of free polyubiquitin chains by yeast Ubp14 modulates rates of protein degradation by the proteasome. EMBO J 1997;16(16):4826-38
  • Wilkinson KD. Regulation of ubiquitin-dependent processes by deubiquitinating enzymes. FASEB J 1997;11(14):1245-56
  • Baek KH. Conjugation and deconjugation of ubiquitin regulating the destiny of proteins. Exp Mol Med 2003;35(1):1-7
  • Wilkinson KD. DUBs at a glance. J Cell Sci 2009;122(Pt 14):2325-9
  • Meray RK, Lansbury PTJr. Reversible monoubiquitination regulates the Parkinson disease-associated ubiquitin hydrolase UCH-L1. J Biol Chem 2007;282(14):10567-75
  • Nguyen LK, Kolch W, Kholodenko BN. When ubiquitination meets phosphorylation: a systems biology perspective of EGFR/MAPK signalling. Cell Commun Signal 2013;11:52
  • Millard SM, Wood SA. Riding the DUBway: regulation of protein trafficking by deubiquitylating enzymes. J Cell Biol 2006;173(4):463-8
  • Phillips D, Aponte AM, Covian R, Balaban RS. Intrinsic protein kinase activity in mitochondrial oxidative phosphorylation complexes. Biochemistry 2011;50(13):2515-29
  • Reyes-Turcu FE, Ventii KH, Wilkinson KD. Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu Rev Biochem 2009;78:363-97
  • Bonifacino JS, Weissman AM. Ubiquitin and the control of protein fate in the secretory and endocytic pathways. Annu Rev Cell Dev Biol 1998;14:19-57
  • Iwai K, Fujita H, Sasaki Y. Linear ubiquitin chains: NF-kappaB signalling, cell death and beyond. Nat Rev Mol Cell Biol 2014;15(8):503-8
  • Hurley JH, Lee S, Prag G. Ubiquitin-binding domains. Biochem J 2006;399(3):361-72
  • Nijman SM, Luna-Vargas MP, Velds A, et al. A genomic and functional inventory of deubiquitinating enzymes. Cell 2005;123(5):773-86
  • Fraile JM, Quesada V, Rodriguez D, et al. Deubiquitinases in cancer: new functions and therapeutic options. Oncogene 2012;31(19):2373-88
  • D’Arcy P, Wang X, Linder S. Deubiquitinase inhibition as a cancer therapeutic strategy. Pharmacol Ther 2014;147:32-54
  • Fortelny N, Cox JH, Kappelhoff R, et al. Network analyses reveal pervasive functional regulation between proteases in the human protease web. PLoS Biol 2014;12(5):e1001869
  • Sgorbissa A, Potu H, Brancolini C. Isopeptidases in anticancer therapy: looking for inhibitors. Am J Transl Res 2010;2(3):235-47
  • Katz EJ, Isasa M, Crosas B. A new map to understand deubiquitination. Biochem Soc Trans 2010;38(Pt 1):21-8
  • Bernassola F, Ciechanover A, Melino G. The ubiquitin proteasome system and its involvement in cell death pathways. Cell Death Differ 2010;17(1):1-3
  • Chauhan D, Hideshima T, Mitsiades C, et al. Proteasome inhibitor therapy in multiple myeloma. Mol Cancer Ther 2005;4(4):686-92
  • Crosas B. Deubiquitinating enzyme inhibitors and their potential in cancer therapy. Curr Cancer Drug Targets 2014;14(6):506-16
  • Lim KH, Baek KH. Deubiquitinating enzymes as therapeutic targets in cancer. Curr Pharm Des 2013;19(22):4039-52
  • Chauhan D, Catley L, Li G, et al. A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from Bortezomib. Cancer Cell 2005;8(5):407-19
  • Ahmad N, Abdel Malek M, Driscoll JJ. Resistance to proteasome inhibitors in cancer. Springer; 2014
  • Buac D, Shen M, Schmitt S, et al. From bortezomib to other inhibitors of the proteasome and beyond. Curr Pharm Des 2013;19(22):4025-38
  • Nicholson B, Marblestone JG, Butt TR, Mattern MR. Deubiquitinating enzymes as novel anticancer targets. Future Oncol 2007;3(2):191-9
  • Wong BR, Parlati F, Qu K, et al. Drug discovery in the ubiquitin regulatory pathway. Drug Discov Today 2003;8(16):746-54
  • Wada T, Yamashita Y, Saga Y, et al. Screening for genetic abnormalities involved in ovarian carcinogenesis using retroviral expression libraries. Int J Oncol 2009;35(5):973-6
  • Maiti TK, Permaul M, Boudreaux DA, et al. Crystal structure of the catalytic domain of UCHL5, a proteasome-associated human deubiquitinating enzyme, reveals an unproductive form of the enzyme. FEBS J 2011;278(24):4917-26
  • Shinji S, Naito Z, Ishiwata S, et al. Ubiquitin-specific protease 14 expression in colorectal cancer is associated with liver and lymph node metastases. Oncol Rep 2006;15(3):539-43
  • Mines MA, Goodwin JS, Limbird LE, et al. Deubiquitination of CXCR4 by USP14 is critical for both CXCL12-induced CXCR4 degradation and chemotaxis but not ERK ativation. J Biol Chem 2009;284(9):5742-52
  • Wicks SJ, Haros K, Maillard M, et al. The deubiquitinating enzyme UCH37 interacts with Smads and regulates TGF-beta signalling. Oncogene 2005;24(54):8080-4
  • Chen Y, Fu D, Xi J, et al. Expression and clinical significance of UCH37 in human esophageal squamous cell carcinoma. Dig Dis Sci 2012;57(9):2310-17
  • Byun S, Lee SY, Lee J, et al. USP8 is a novel target for overcoming gefitinib resistance in lung cancer. Clin Cancer Res 2013;19(14):3894-904
  • Tian Z, D’Arcy P, Wang X, et al. A novel small molecule inhibitor of deubiquitylating enzyme USP14 and UCHL5 induces apoptosis in multiple myeloma and overcomes bortezomib resistance. Blood 2014;123(5):706-16
  • D’Arcy P, Linder S. Proteasome deubiquitinases as novel targets for cancer therapy. Int J Biochem Cell Biol 2012;44(11):1729-38
  • Costa Mdo C, Paulson HL. Toward understanding Machado-Joseph disease. Prog Neurobiol 2012;97(2):239-57
  • Ragland M, Hutter C, Zabetian C, Edwards K. Association between the ubiquitin carboxyl-terminal esterase L1 gene (UCHL1) S18Y variant and Parkinson’s Disease: a HuGE review and meta-analysis. Am J Epidemiol 2009;170(11):1344-57
  • Miyake Y, Tanaka K, Fukushima W, et al. UCHL1 S18Y variant is a risk factor for Parkinson’s disease in Japan. BMC Neurol 2012;12:62
  • Adorno M, Sikandar S, Mitra SS, et al. Usp16 contributes to somatic stem-cell defects in Down’s syndrome. Nature 2013;501(7467):380-4
  • Edelmann MJ, Kessler BM. Ubiquitin and ubiquitin-like specific proteases targeted by infectious pathogens: Emerging patterns and molecular principles. Biochim Biophys Acta 2008;1782(12):809-16
  • Majumdar I, Paul J. The deubiquitinase A20 in immunopathology of autoimmune diseases. Autoimmunity 2014;47(5):307-19
  • Atanassov BS, Koutelou E, Dent SY. The role of deubiquitinating enzymes in chromatin regulation. FEBS Lett 2011;585(13):2016-23
  • Love KR, Catic A, Schlieker C, Ploegh HL. Mechanisms, biology and inhibitors of deubiquitinating enzymes. Nat Chem Biol 2007;3(11):697-705
  • Colland F. The therapeutic potential of deubiquitinating enzyme inhibitors. Biochem Soc Trans 2010;38(Pt 1):137-43
  • Mermerian AH, Case A, Stein RL, Cuny GD. Structure-activity relationship, kinetic mechanism, and selectivity for a new class of ubiquitin C-terminal hydrolase-L1 (UCH-L1) inhibitors. Bioorg Med Chem Lett 2007;17(13):3729-32
  • Liu Y, Lashuel HA, Choi S, et al. Discovery of inhibitors that elucidate the role of UCH-L1 activity in the H1299 lung cancer cell line. Chem Biol 2003;10(9):837-46
  • Brnjic S, Mazurkiewicz M, Fryknas M, et al. Induction of tumor cell apoptosis by a proteasome deubiquitinase inhibitor is associated with oxidative stress. Antioxid Redox Signal 2014;21(17):2271-85
  • Slavica BRNJIC PDA. Rolf Larsson, Stig Linder. Proteasome deubiquinating inhibitor screening. WO2013039438; 2012
  • Grande E, Earl J, Fuentes R, Carrato A. New targeted approaches against the ubiquitin-proteasome system in gastrointestinal malignancies. Expert Rev Anticancer Ther 2012;12(4):457-67
  • Aftab O, Engskog MK, Haglof J, et al. NMR spectroscopy-based metabolic profiling of drug-induced changes in vitro can discriminate between pharmacological classes. J Chem Inform Model 2014;54(11):3251-8
  • Lynn Kirkpatrick GP. Sarah Welsh. Regulation of HIF protein levels via deubiquitination pathway. WO007828; 2005
  • Powis G, Wipf P, Lynch SM, et al. Molecular pharmacology and antitumor activity of palmarumycin-based inhibitors of thioredoxin reductase. Mol Cancer Ther 2006;5(3):630-6
  • Semenza GL. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci 2012;33(4):207-14
  • Xiong M, Elson G, Legarda D, Leibovich SJ. Production of vascular endothelial growth factor by murine macrophages: regulation by hypoxia, lactate, and the inducible nitric oxide synthase pathway. Am J Pathol 1998;153(2):587-98
  • Gutschner T, Diederichs S. The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol 2012;9(6):703-19
  • Hoffman MA, Ohh M, Yang H, et al. von Hippel-Lindau protein mutants linked to type 2C VHL disease preserve the ability to downregulate HIF. Human Mol Gen 2001;10(10):1019-27
  • Zundel W, Schindler C, Haas-Kogan D, et al. Loss of PTEN facilitates HIF-1-mediated gene expression. Genes Develop 2000;14(4):391-6
  • Semenza GL. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J Clin Invest 2013;123(9):3664-71
  • Finley D. Recognition and processing of ubiquitin-protein conjugates by the proteasome. Annu Rev Biochem 2009;78:477-513
  • Daniel Finley RW, Byung-Hoon L, Min JL, Gahman TC. Compositions and methods for enhancing proteasome activity. WO094545; 2011
  • Stig Linder RL. Method for inhibition of deubiquitinating activity In. WO058691; 2013
  • Lesinski GB, Raig ET, Guenterberg K, et al. IFN-alpha and bortezomib overcome Bcl-2 and Mcl-1 overexpression in melanoma cells by stimulating the extrinsic pathway of apoptosis. Cancer Res 2008;68(20):8351-60
  • Perry JW, Ahmed M, Chang KO, et al. Antiviral activity of a small molecule deubiquitinase inhibitor occurs via induction of the unfolded protein response. PLoS Pathog 2012;8(7):e1002783
  • Donato NJ, Wobus CU, Showalter HD, et al. Deubiquitinase inhibitors and methods for use of the same. WO040527A3; 2012
  • Maloney DJ, Rosenthal AS, Jadhav AU, et al. Inhibitors of the usp1/uaf1 deubiquitinase complex and uses thereof. WO105952; 2014
  • Andrea Alan DD. Deubiquitinating enzymes that regulate cell growth. WO006247; 1997
  • Anna Borodovsky PG, Gan-Erdene T, Hemelaar J, et al. Semisynthetic protein-based site-directed probes for identification and inhibition of active sites, and methods therefor. WO091411; 2003
  • Borodovsky A, Kessler BM, Casagrande R, et al. A novel active site-directed probe specific for deubiquitylating enzymes reveals proteasome association of USP14. EMBO J 2001;20(18):5187-96
  • Cajee UF, Hull R, Ntwasa M. Modification by ubiquitin-like proteins: significance in apoptosis and autophagy pathways. Int J Mol Sci 2012;13(9):11804-31
  • David Komander YY. Fluorescent -labelled diubiquitin substrate for a deubiquitinase assay. WO2011157982 A1
  • Shanmugham A, Ovaa H. DUBs and disease: activity assays for inhibitor development. Cur Opin Drug Disc Devel 2008;11(5):688-96
  • Altun M, Walter TS, Kramer HB, et al. The human otubain2-ubiquitin structure provides insights into the cleavage specificity of poly-ubiquitin-linkages. PLoS ONE 2015;10(1):e0115344
  • Rene Bernards TR, Annette MD, Sebastian MN. Modulation of deubiquitinase family members. WO064711; 2004
  • Gamper AM, Qiao X, Kim J, et al. Regulation of KLF4 turnover reveals an unexpected tissue-specific role of pVHL in tumorigenesis. Mol Cell 2012;45(2):233-43
  • Koyama D, Kikuchi J, Hiraoka N, et al. Proteasome inhibitors exert cytotoxicity and increase chemosensitivity via transcriptional repression of Notch1 in T-cell acute lymphoblastic leukemia. Leukemia 2014;28(6):1216-26
  • Harhaj EW, Dixit VM. Deubiquitinases in the regulation of NF-kappaB signaling. Cell Res 2011;21(1):22-39
  • Dattananda Chelur ME, Craig L, Steven JO, et al. Kathryn Longenecker Di- and poly-ubiquitin deubiquitinase substrates and uses thereof. US20120107857; 2013
  • Wei Gu ML. Ubiquitin hydrolase/deubiquitinase (HAUSP-Mdm2) complexes; p53 binding proteins; for diagnosis/treatment of neoplasia/carcinoma; drug screening; kits. WO2005098449; 2004
  • John W, Tobias AJ. Ubiquitin-specific protease. WO017245 A1; 1991
  • Jung J. Peptide inhibitors of hausp deubiquitinase. US0073585 A1; 2014
  • Cvek B, Milacic V, Taraba J, Dou QP. Ni(II), Cu(II), and Zn(II) diethyldithiocarbamate complexes show various activities against the proteasome in breast cancer cells. J Med Chem 2008;51(20):6256-8
  • Daniel KG, Gupta P, Harbach RH, et al. Organic copper complexes as a new class of proteasome inhibitors and apoptosis inducers in human cancer cells. Biochem Pharmacol 2004;67(6):1139-51
  • Verani CN. Metal complexes as inhibitors of the 26S proteasome in tumor cells. J Inorg Biochem 2012;106(1):59-67
  • Skrott Z, Cvek B. Diethyldithiocarbamate complex with copper: the mechanism of action in cancer cells. Mini Rev Med Chem 2012;12(12):1184-92
  • Chen D, Cui QC, Yang H, Dou QP. Disulfiram, a clinically used anti-alcoholism drug and copper-binding agent, induces apoptotic cell death in breast cancer cultures and xenografts via inhibition of the proteasome activity. Cancer Res 2006;66(21):10425-33
  • Chen D, Cui QC, Yang H, et al. Clioquinol, a therapeutic agent for Alzheimer’s disease, has proteasome-inhibitory, androgen receptor-suppressing, apoptosis-inducing, and antitumor activities in human prostate cancer cells and xenografts. Cancer Res 2007;67(4):1636-44
  • Milacic V, Chen D, Giovagnini L, et al. Pyrrolidine dithiocarbamate-zinc(II) and -copper(II) complexes induce apoptosis in tumor cells by inhibiting the proteasomal activity. Toxicol Appl Pharmacol 2008;231(1):24-33
  • Liu N, Liu C, Li X, et al. A novel proteasome inhibitor suppresses tumor growth via targeting both 19S proteasome deubiquitinases and 20S proteolytic peptidases. Sci Rep 2014;4:5240
  • Chandler CJ, Segel IH. Mechanism of the antimicrobial action of pyrithione: effects on membrane transport, ATP levels, and protein synthesis. Antimicrob Agents Chemother 1978;14(1):60-8
  • Davies TW. The dissolution rate of zinc pyrithione. Int J Cosmet Sci 1985;7(4):153-6
  • Liu N, Li X, Huang H, et al. Clinically used antirheumatic agent auranofin is a proteasomal deubiquitinase inhibitor and inhibits tumor growth. Oncotarget 2014;5(14):5453-71
  • Fiskus W, Saba N, Shen M, et al. Auranofin induces lethal oxidative and endoplasmic reticulum stress and exerts potent preclinical activity against chronic lymphocytic leukemia. Cancer Res 2014;74(9):2520-32
  • Mirabelli CK, Johnson RK, Hill DT, et al. Correlation of the in vitro cytotoxic and in vivo antitumor activities of gold(I) coordination complexes. J Med Chem 1986;29(2):218-23
  • Mirabelli CK, Johnson RK, Sung CM, et al. Evaluation of the in vivo antitumor activity and in vitro cytotoxic properties of auranofin, a coordinated gold compound, in murine tumor models. Cancer Res 1985;45(1):32-9
  • Potts BC, Albitar MX, Anderson KC, et al. Marizomib, a proteasome inhibitor for all seasons: preclinical profile and a framework for clinical trials. Curr Cancer Drug Targets 2011;11(3):254-84
  • Heideker J, Wertz IE. DUBs, the regulation of cell identity and disease. Biochem J 2015;465(1):1-26
  • Pfoh R, Lacdao IK, Saridakis V. Deubiquitinases and the new therapeutic opportunities offered to cancer. Endocr Relat Cancer 2015;22(1):T35-54

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.