Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1780 word(s) 1780 2021-05-18 06:19:33 |
2 format change Meta information modification 1780 2021-06-11 04:35:35 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Chen, S. Ubiquitin-Specific Peptidase (USP) Inhibitors. Encyclopedia. Available online: https://encyclopedia.pub/entry/10735 (accessed on 19 April 2024).
Chen S. Ubiquitin-Specific Peptidase (USP) Inhibitors. Encyclopedia. Available at: https://encyclopedia.pub/entry/10735. Accessed April 19, 2024.
Chen, Shiyao. "Ubiquitin-Specific Peptidase (USP) Inhibitors" Encyclopedia, https://encyclopedia.pub/entry/10735 (accessed April 19, 2024).
Chen, S. (2021, June 10). Ubiquitin-Specific Peptidase (USP) Inhibitors. In Encyclopedia. https://encyclopedia.pub/entry/10735
Chen, Shiyao. "Ubiquitin-Specific Peptidase (USP) Inhibitors." Encyclopedia. Web. 10 June, 2021.
Ubiquitin-Specific Peptidase (USP) Inhibitors
Edit

Ubiquitylation and deubiquitylation are reversible protein post-translational modification (PTM) processes involving the regulation of protein degradation under physiological conditions. Loss of balance in this regulatory system can lead to a wide range of diseases, such as cancer and inflammation. As the main members of the deubiquitinases (DUBs) family, ubiquitin-specific peptidases (USPs) are closely related to biological processes through a variety of molecular signaling pathways, including DNA damage repair, p53 and transforming growth factor-β (TGF-β) pathways.

Ubiquitin-Specific Peptidases Signaling Pathways Drug Screening USP Inhibitors

1. Introduction

As a complex regulatory mechanism of biological functions, post-translational modification (PTM) is essential for cell growth and stress response. Generally, intracellular proteins will experience multiple types of modifications after translation, such as phosphorylation, acetylation, methylation, and ubiquitylation, each corresponding to one or more specific functions [1]. Among them, ubiquitylation is responsible for regulating protein–protein interactions, cellular localization, and enzymatic activities of its protein substrates, and it is also related to proteasome-mediated protein degradation. A large number of studies have identified the ubiquitin-driven degradation pathways as one of the most important ways to help maintain protein balance within eukaryotic cells [1][2]. Therefore, the ubiquitylation of proteins plays indispensable regulatory roles in various biological phenomena [2].
Eukaryotic cells are equipped to recognize and degrade proteins by the ubiquitin–proteasome system (UPS). Upon conjugated to chains of ubiquitin, proteins are then directed to the 26S proteasome, a macromolecular protease, and degraded [3]. Ubiquitin is a small peptide (8.5 kDa) consisting of 76 amino acids that is ubiquitous in eukaryotic cells. The peptide sequence is highly conserved and contains seven lysine sites (Lys6, Lys11, Lys27, Lys29, Lys33, Lys48, and Lys63), a glycine site at the C-terminus, and a methionine at the N-terminus (Met1). In general, the poly-ubiquitin chain linked by Lys48 is a degradation marker for proteasome, while the Lys63-linked poly-ubiquitin chain usually works with non-proteasome pathways (such as DNA repair, DNA replication, and cell signal transduction) [4][5][6]. It has been reported that the ubiquitin chains connected to the target protein through Lys6, Lys11, Lys27, Lys29 or Lys33 are also related to proteasome-mediated degradation [7]. In addition, under certain circumstances Lys63-linked ubiquitin chains can also bind and target proteins that need to be degraded by the proteasome [8].
Similar to other PTMs, the ubiquitin modification of protein is a dynamic and reversible process. Ubiquitin modification can be removed by a series of ubiquitin-specific proteases, which is called deubiquitylation. These proteases are named deubiquitinases (DUBs). Deubiquitinases specifically recognize and excise the tumbling molecules on the target protein, and also participate in the editing of poly-ubiquitin, thus playing an important role in the cleavage of ubiquitin precursors and ubiquitin monomers [9][10]. DUBs also regulate gene expression, apoptosis, cell cycle, DNA repair, and cytokines [11][12][13][14][15].
There are nearly a hundred known DUBs including cysteine proteases (USPs, UCHs, MJDs and OTUs) and metalloproteinases (containing metal catalytic domains) according to different catalytic mechanisms. They are divided into the following superfamilies: ubiquitin-specific protease (USP), ubiquitin C-terminal hydrolase (UCH), ovarian tumor protease (OTU), Machado-Josephin domain superfamily (MJD), and zinc-containing metalloproteases [16][17].
In recent years, the vast majority of DUBs have been shown to be associated with a variety of diseases, including cancer, diabetes, neurodegenerative diseases, and infectious diseases [18][19][20][21]. As the largest superfamily with over 50 members, USPs have aroused increasing attention as potential therapeutic targets in recent years [22]. It is interesting to compare USPs with kinases as drug targets since they are both involved in protein posttranslational modifications. Discovery in protein phosphorylation was awarded the Nobel Prize in 1992, and in the past thirty years numerous efforts have been invested in kinase inhibitors which resulted in a good number of clinically approved drugs including Gleevec. However, the research in the ubiquitin system caught the attention of medicinal chemists much later, and only in the past ten years USPs inhibitors have started to gradually emerge. We envision that USPs represent a new reservoir of therapeutic targets, which will reach its prime time in the twenty years to come. To date, no USP inhibitor has yet been approved for clinical use. In this review, we focus on advances in the development of USP inhibitors within the past decade.

2. Roles of USPs in Cancers

Aberrant regulation of protein ubiquitylation is closely related to the occurrence and development of tumorigenesis and other pathologies such as neurodegenerative diseases, autoimmunity, inflammatory disorders, infection, muscle dystrophies, etc. [23]. Given that the target proteins for USPs contain a large number of cell homeostasis regulators, as well as products of known oncogenes or tumor suppressor genes, USPs might be attractive and promising targets for the development of novel cancer therapies.
Studies have shown the involvement of USPs in the regulation of multiple known cancer-related pathways, including p53, transforming growth factor-β (TGF-β), protein kinase B (Akt), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), Janus kinase/signal transducers and activators of transcription (JAKs/STATs), and G protein-coupled receptor (GPCR). For example, the overexpression of USP2a stabilizes p53-murine double minute 2 (MDM2) through direct deubiquitylation, without reducing MDM2-mediated p53 ubiquitylation, and thus enhances p53 degradation [24]. Since p53 functions as a tumor suppressor and is vital for normal cellular process controlling, such downregulation of p53 can ultimately cause tumor progression [24]. USP7, however, deubiquitylates both MDM2 and p53, while its affinity to MDM2 is confirmed to be higher [25][26]. Another notable example is USP26. It has been reported to be a novel negative regulator of the TGF-β pathway and the loss of USP26 expression may be an important factor in glioblastoma pathogenesis and breast cancer [27]. Low levels of USP26 degrade drosophila mothers against decapentaplegic protein 7 (SMAD7) and stabilize TGFβ, while high levels of USP26 stabilize SMAD7 by deubiquitylation and form a complex with SMAD ubiquitylation regulatory factor 2 (SMURF2), which degrades the TGF-β receptor by ubiquitylation [27].
Here, in Table 1 we summarized the roles of USPs implicated in tumorigenesis according to the different signaling pathways on which they act.
Table 1. Cancer-related pathways regulated by USPs.
Pathway USPs Involved Refs
DNA damage repair USP1, USP28 [28][29]
TGF-β USP2a, USP4, USP9X, USP15, USP26 [27][30][31][32][33]
Wnt/β-catenin USP4, USP5, USP9X, USP14 [34][35][36][37][38]
p53 USP2, USP4, USP5, USP7, USP10, USP15, USP24, USP42 [24][25][26][39][40][41][42][43][44][45][46][47][48]
c-Myc USP2, USP10, USP22, USP28, USP36, USP37 [49][50][51][52][53][54]
Akt USP4, USP12, USP14, USP22, USP46 [55][56][57][58][59][60]
JAKs-STATs USP7 [61]
NF-κB USP4, USP11, USP14, USP15, USP18, USP19, USP20, USP35, USP24, USP48 [62][63][64][65][66][67][68][69][70][71][72][73][74][75]
GPCR USP4, USP8, USP14, USP20, USP30 [76][77][78][79]

3. Methods in Screening and Identification of Inhibitors for USPs

In order to enable the continuous discovery and development of inhibitors for USPs, a number of biological testing methods have been developed to screen and identify small molecule inhibitors (Figure 1).
Figure 1. USPs inhibitor screening methods. (A) Activity-based probe (ABP) structures; (B) Ub-7-amino-4-methylcoumarin (AMC) structure; (C) Ub-phospholipase A2 (PLA2) assay; (D) Time-resolved fluorescence resonance energy transfer (TR-FRET) assay; (E) UBA52 structure and the SDS-PAGE-Coomassie assay; (F) Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) method.

3.1. Activity-Based Probes

Activity-based probes (ABPs), in which an electrophilic warhead is introduced onto the C-terminal glycine of ubiquitin, provide a way to test compounds in a cellular environment. Currently reported probe molecules include ubiquitin-vinylmethyl sulfone (Ub-VS) [80], ubiquitin-vinylmethyl ester (Ub-VME) [81], and ubiquitin-propargylic acid (Ub-PA) [82]. They can covalently label the nucleophilic cysteine of DUBs, resulting in a band shift on SDS-PAGE.
The advantage of this method is that it is closely related to the physiological environment of cells. The disadvantage is that it is time-consuming and laborious, so it is not currently recommended for screening USP inhibitors.
Interestingly, in order to overcome the lack of target selectivity, a novel Ub-based activity probe (Rh-M20-PA) bearing specific mutations to achieve selectivity for USP16 was developed by combining structural modelling and computation. A number of USP16-specific inhibitors were successfully discovered using these USP16-selective ABPs [83].

3.2. Ub-AMC

Ub-AMC, which has the C-terminus of a ubiquitin molecule linked to 7-amino-4-methylcoumarin (AMC), is a rather simple method, and has been widely applied in the determination of deubiquitinating enzyme activities [84]. The advantages of the Ub-AMC method are low cost per test and commercial availability. However, Ub-AMC is an unnatural substrate, and light-emitting substances (auto-fluorescence or fluorescence quenchers) can interfere with the reading. Later, researchers improved the method by replacing AMC with rhodamine-110 (Rho110) or tetramethylrhodamine, so the wavelength was red-shifted and the interference was reduced [85][86].

3.3. Ub-PLA2

In the Ub-phospholipase A2 (PLA2) method, the PLA2 does not directly emit fluorescence after being cleaved from the ubiquitin chain, but it acts on a fluorescent substrate and causes it to emit fluorescence [87]. It is also called the Ub-CHOP method and makes the screening at lower enzyme concentrations possible by amplifying the activity of deubiquitinating enzymes [88]. Its signal intensity and duration are better than the Ub-AMC. Besides, the excitation wavelength is not in the ultraviolet region, and currently there are commercial kits. However, the Ub-PLA2 method is not sensitive to some deubiquitinating enzymes of the UCH family, and its price is higher than the Ub-AMC method. USP inhibitors identified by this method include shionone and P22077 [88].

3.4. TR-FRET

The time-resolved fluorescence resonance energy transfer (TR-FRET) method is based on a full-length ubiquitin substrate that is site-specifically labeled with a yellow fluorescent protein (YFP) at the N-terminus and a terbium donor at the C-terminus. This substrate has strong fluorescence resonance energy transfer (FRET) between the two groups, while the cleavage by USPs will decrease the extent of FRET [89][90][91].
The advantage of the TR-FRET method is that it is equally sensitive to the four deubiquitinating enzymes of the UCH family. However, there is no commercial kit available.
The expansion of this application is to use diubiquitin molecules (diUb) as substrates, and this diubiquitin molecule can be connected through different lysine sites to simulate different ubiquitin chain forms [92].

3.5. SDS-PAGE-Coomassie

The development of a highly reliable assay based on a readily available SDS-PAGE-Coomassie system using UBA52 as the substrate protein has been reported recently [93]. A number of effective USP2 inhibitors were identified using this assay. Natural substrate UBA52 was used and quantitative measurement was based on the infrared emission of Coomassie dye on SDS-PAGE.
This method uses readily available and inexpensive materials and has excellent reproducibility without the interference problem that is intrinsic to any fluorescence-based approaches. It also has the advantage of using a natural protein substrate, avoiding any artifacts that may be introduced by unnatural substrates. However, this assay was not amenable to high-throughput screening. It is useful for the accurate determination of IC50 values during fine-tuning of the structures during the structure-activity studies.

3.6. MADAL-TOF

A sensitive and fast assay to quantify in vitro DUBs enzyme activities using matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry has been developed [94]. This method realized the high specificity of many members of the OTU and JAB/MPN/Mov34 metalloenzyme DUB families. It used unmodified substrates, such as di-ubiquitin topoisomers, and can be used to assess the potency and specificity of deubiquitylation inhibitors.

References

  1. Millar, A.H.; Heazlewood, J.L.; Giglione, C.; Holdsworth, M.J.; Bachmair, A.; Schulze, W.X. The scope, functions, and dynamics of posttranslational protein modifications. Annu. Rev. Plant Biol. 2019, 70, 119–151.
  2. Swatek, K.N.; Komander, D. Ubiquitin modifications. Cell Res. 2016, 26, 399–422.
  3. Kleiger, G.; Mayor, T. Perilous journey: A tour of the ubiquitin-proteasome system. Trends Cell Biol. 2014, 24, 352–359.
  4. Chau, V.; Tobias, J.W.; Bachmair, A.; Marriott, D.; Ecker, D.J.; Gonda, D.K.; Varshavsky, A. A multiubiquitin chain is confined to specific lysine in a targeted short-lived protein. Science 1989, 243, 1576–1583.
  5. Hershko, A.; Ciechanover, A. The ubiquitin system. Annu. Rev. Biochem. 1998, 67, 425–479.
  6. Haglund, K.; Dikic, I. Ubiquitylation and cell signaling. EMBO J. 2005, 24, 3353–3359.
  7. Xu, P.; Duong, D.M.; Seyfried, N.T.; Cheng, D.; Xie, Y.; Robert, J.; Rush, J.; Hochstrasser, M.; Finley, D.; Peng, J. Quantitative proteomics reveals the function of unconventional ubiquitin chains in proteasomal degradation. Cell 2009, 137, 133–145.
  8. Saeki, Y.; Kudo, T.; Sone, T.; Kikuchi, Y.; Yokosawa, H.; Tohe, A.; Tanaka, K. Lysine 63-linked polyubiquitin chain may serve as a targeting signal for the 26S proteasome. EMBO J. 2009, 28, 359–371.
  9. Pickart, C.M.; Rose, I.A. Ubiquitin carboxyl-terminal hydrolase acts on ubiquitin carboxyl-terminal amides. J. Biol. Chem. 1985, 260, 7903–7910.
  10. Mevissen, T.E.T.; Komander, D. Mechanisms of deubiquitinase specificity and regulation. Annu. Rev. Biochem. 2017, 86, 159–192.
  11. Frappier, L.; Verrijzer, C.P. Gene expression control by protein deubiquitinases. Curr. Opin. Genet. Dev. 2011, 21, 207–213.
  12. He, M.; Zhou, Z.; Wu, G.; Chen, Q.; Wan, Y. Emerging role of DUBs in tumor metastasis and apoptosis: Therapeutic implication. Pharmacol. Ther. 2017, 177, 96–107.
  13. Darling, S.; Fielding, A.B.; Sabat-Pospiech, D.; Prior, I.A.; Coulson, J.M. Regulation of the cell cycle and centrosome biology by deubiquitylases. Biochem. Soc. Trans. 2017, 45, 1125–1136.
  14. Kee, Y.; Huang, T.T. Role of deubiquitinating enzymes in DNA repair. Mol. Cell. Biol. 2016, 36, 524–544.
  15. Woo, B.; Baek, K.H. Regulatory interplay between deubiquitinating enzymes and cytokines. Cytokine Growth Factor Rev. 2019, 48, 40–51.
  16. Komander, D.; Clague, M.J.; Urbe, S. Breaking the chains: Structure and function of the deubiquitinases. Nat. Rev. Mol. Cell Biol. 2009, 10, 550–563.
  17. Clague, M.J.; Barsukov, I.; Coulson, J.M.; Liu, H.; Rigden, D.J.; Urbe, S. Deubiquitylases from genes to organism. Physiol. Rev. 2013, 93, 1289–1315.
  18. D’Arcy, P.; Wang, X.; Linder, S. Deubiquitinase inhibition as a cancer therapeutic strategy. Pharmacol. Ther. 2015, 147, 32–54.
  19. Balaji, V.; Pokrzywa, W.; Hoppe, T. Ubiquitylation pathways in insulin signaling and organismal homeostasis. Bioessays 2018, 40, e1700223.
  20. Ruan, J.; Schluter, D.; Wang, X. Deubiquitinating enzymes (DUBs): DoUBle-edged swords in CNS autoimmunity. J. Neuroinflamm. 2020, 17, 102.
  21. Nanduri, B.; Suvarnapunya, A.E.; Venkatesan, M.; Edelmann, M.J. Deubiquitinating enzymes as promising drug targets for infectious diseases. Curr. Pharm. Des. 2013, 19, 3234–3247.
  22. Young, M.J.; Hsu, K.C.; Lin, T.E.; Chang, W.C.; Hung, J.J. The role of ubiquitin-specific peptidases in cancer progression. J. Biomed. Sci. 2019, 26, 42.
  23. Popovic, D.; Vucic, D.; Dikic, I. Ubiquitination in disease pathogenesis and treatment. Nat. Med. 2014, 20, 1242–1253.
  24. Stevenson, L.F.; Sparks, A.; Allende-Vega, N.; Xirodimas, D.P.; Lane, D.P.; Saville, M.K. The deubiquitinating enzyme USP2a regulates the p53 pathway by targeting Mdm2. EMBO J. 2007, 26, 976–986.
  25. Hu, M.; Gu, L.; Li, M.; Jeffrey, P.D.; Gu, W.; Shi, Y. Structural basis of competitive recognition of p53 and MDM2 by HAUSP/USP7: Implications for the regulation of the p53-MDM2 pathway. PLoS Biol. 2006, 4, e27.
  26. Meulmeester, E.; Pereg, Y.; Shiloh, Y.; Jochemsen, A.G. ATM-mediated phosphorylations inhibit Mdmx/Mdm2 stabilization by HAUSP in favor of p53 activation. Cell Cycle 2005, 4, 1166–1170.
  27. Kit Leng Lui, S.; Iyengar, P.V.; Jaynes, P.; Isa, Z.; Pang, B.; Tan, T.Z.; Eichhorn, P.J.A. USP26 regulates TGF-β signaling by deubiquitinating and stabilizing SMAD7. EMBO Rep. 2017, 18, 797–808.
  28. Dianov, G.L.; Meisenberg, C.; Parsons, J.L. Regulation of DNA repair by ubiquitylation. Biochemistry 2011, 76, 69–79.
  29. Zhang, D.; Zaugg, K.; Mak, T.W.; Elledge, S.J. A role for the deubiquitinating enzyme USP28 in control of the DNA-damage response. Cell 2006, 126, 529–542.
  30. Zhao, Y.; Wang, X.; Wang, Q.; Deng, Y.; Li, K.; Zhang, M.; Zhang, Q.; Zhou, J.; Wang, H.Y.; Bai, P.; et al. USP2a supports metastasis by tuning TGF-β signaling. Cell Rep. 2018, 22, 2442–2454.
  31. Zhang, L.; Zhou, F.; Drabsch, Y.; Gao, R.; Snaar-Jagalska, B.E.; Mickanin, C.; Huang, H.; Sheppard, K.A.; Porter, J.A.; Lu, C.X.; et al. USP4 is regulated by AKT phosphorylation and directly deubiquitylates TGF-β type I receptor. Nat. Cell Biol. 2012, 14, 717–726.
  32. Dupont, S.; Mamidi, A.; Cordenonsi, M.; Montagner, M.; Zacchigna, L.; Adorno, M.; Martello, G.; Stinchfield, M.J.; Soligo, S.; Morsut, L.; et al. FAM/USP9x, a deubiquitinating enzyme essential for TGFβ signaling, controls Smad4 monoubiquitination. Cell 2009, 136, 123–135.
  33. Iyengar, P.V.; Jaynes, P.; Rodon, L.; Lama, D.; Law, K.P.; Lim, Y.P.; Verma, C.; Seoane, J.; Eichhorn, P.J. USP15 regulates SMURF2 kinetics through C-lobe mediated deubiquitination. Sci. Rep. 2015, 5, 14733.
  34. Yun, S.I.; Kim, H.H.; Yoon, J.H.; Park, W.S.; Hahn, M.J.; Kim, H.C.; Chung, C.H.; Kim, K.K. Ubiquitin specific protease 4 positively regulates the WNT/β-catenin signaling in colorectal cancer. Mol. Oncol. 2015, 9, 1834–1851.
  35. Chen, Y.; Li, Y.; Xue, J.; Gong, A.; Yu, G.; Zhou, A.; Lin, K.; Zhang, S.; Zhang, N.; Gottardi, C.J.; et al. Wnt-induced deubiquitination FoxM1 ensures nucleus β-catenin transactivation. EMBO J. 2016, 35, 668–684.
  36. Premarathne, S.; Murtaza, M.; Matigian, N.; Jolly, L.A.; Wood, S.A. Loss of Usp9x disrupts cell adhesion, and components of the Wnt and Notch signaling pathways in neural progenitors. Sci. Rep. 2017, 7, 8109.
  37. Yang, B.; Zhang, S.; Wang, Z.; Yang, C.; Ouyang, W.; Zhou, F.; Zhou, Y.; Xie, C. Deubiquitinase USP9X deubiquitinates β-catenin and promotes high grade glioma cell growth. Oncotarget 2016, 7, 79515–79525.
  38. Huang, G.; Li, L.; Zhou, W. USP14 activation promotes tumor progression in hepatocellular carcinoma. Oncol. Rep. 2015, 34, 2917–2924.
  39. Wang, C.L.; Wang, J.Y.; Liu, Z.Y.; Ma, X.M.; Wang, X.W.; Jin, H.; Zhang, X.P.; Fu, D.; Hou, L.J.; Lu, Y.C. Ubiquitin-specific protease 2a stabilizes MDM4 and facilitates the p53-mediated intrinsic apoptotic pathway in glioblastoma. Carcinogenesis 2014, 35, 1500–1509.
  40. Li, Z.; Hao, Q.; Luo, J.; Xiong, J.; Zhang, S.; Wang, T.; Bai, L.; Wang, W.; Chen, M.; Wang, W.; et al. USP4 inhibits p53 and NF-κB through deubiquitinating and stabilizing HDAC2. Oncogene 2016, 35, 2902–2912.
  41. Zhang, X.N.; Berger, F.G.; Yang, J.H.; Lu, X.B. USP4 inhibits p53 through deubiquitinating and stabilizing ARF-BP1. EMBO J. 2011, 30, 2177–2189.
  42. Dayal, S.; Sparks, A.; Jacob, J.; Allende-Vega, N.; Lane, D.P.; Saville, M.K. Suppression of the deubiquitinating enzyme USP5 causes the accumulation of unanchored polyubiquitin and the activation of p53. J. Biol. Chem. 2009, 284, 5030–5041.
  43. Potu, H.; Peterson, L.F.; Pal, A.; Verhaegen, M.; Cao, J.; Talpaz, M.; Donato, N.J. Usp5 links suppression of p53 and FAS levels in melanoma to the BRAF pathway. Oncotarget 2014, 5, 5559–5569.
  44. Tavana, O.; Sun, H.; Gu, W. Targeting HAUSP in both p53 wildtype and p53-mutant tumors. Cell Cycle 2018, 17, 823–828.
  45. Takayama, K.I.; Suzuki, T.; Fujimura, T.; Takahashi, S.; Inoue, S. Association of USP10 with G3BP2 inhibits p53 signaling and contributes to poor outcome in prostate cancer. Mol. Cancer Res. 2018, 16, 846–856.
  46. Zou, Q.; Jin, J.; Hu, H.; Li, H.S.; Romano, S.; Xiao, Y.; Nakaya, M.; Zhou, X.; Cheng, X.; Yang, P.; et al. USP15 stabilizes MDM2 to mediate cancer-cell survival and inhibit antitumor T cell responses. Nat. Immunol. 2014, 15, 562–570.
  47. Zhang, L.; Nemzow, L.; Chen, H.; Lubin, A.; Rong, X.; Sun, Z.Y.; Harris, T.K.; Gong, F. The deubiquitinating enzyme USP24 is a regulator of the UV damage response. Cell Rep. 2015, 10, 140–147.
  48. Hock, A.K.; Vigneron, A.M.; Carter, S.; Ludwig, R.L.; Vousden, K.H. Regulation of p53 stability and function by the deubiquitinating enzyme USP42. EMBO J. 2011, 30, 4921–4930.
  49. Benassi, B.; Flavin, R.; Marchionni, L.; Zanata, S.; Pan, Y.; Chowdhury, D.; Marani, M.; Strano, S.; Muti, P.; Blandino, G.; et al. MYC is activated by USP2a-mediated modulation of microRNAs in prostate cancer. Cancer Discov. 2012, 2, 236–247.
  50. Lin, Z.; Yang, H.; Tan, C.; Li, J.; Liu, Z.; Quan, Q.; Kong, S.; Ye, J.; Gao, B.; Fang, D. USP10 antagonizes c-Myc transcriptional activation through SIRT6 stabilization to suppress tumor formation. Cell Rep. 2013, 5, 1639–1649.
  51. Kim, D.; Hong, A.; Park, H.I.; Shin, W.H.; Yoo, L.; Jeon, S.J.; Chung, K.C. Deubiquitinating enzyme USP22 positively regulates c-Myc stability and tumorigenic activity in mammalian and breast cancer cells. J. Cell. Physiol. 2017, 232, 3664–3676.
  52. Weili, Z.; Zhikun, L.; Jianmin, W.; Qingbao, T. Knockdown of USP28 enhances the radiosensitivity of esophageal cancer cells via the c-Myc/hypoxia-inducible factor-1 α pathway. J. Cell. Biochem. 2019, 120, 201–212.
  53. Sun, X.X.; Sears, R.C.; Dai, M.S. Deubiquitinating c-Myc: USP36 steps up in the nucleolus. Cell Cycle 2015, 14, 3786–3793.
  54. Pan, J.; Deng, Q.; Jiang, C.; Wang, X.; Niu, T.; Li, H.; Chen, T.; Jin, J.; Pan, W.; Cai, X.; et al. USP37 directly deubiquitinates and stabilizes c-Myc in lung cancer. Oncogene 2015, 34, 3957–3967.
  55. Xing, C.; Lu, X.X.; Guo, P.D.; Shen, T.; Zhang, S.; He, X.S.; Gan, W.J.; Li, X.M.; Wang, J.R.; Zhao, Y.Y.; et al. Ubiquitin-specific protease 4-mediated deubiquitination and stabilization of PRL-3 is required for potentiating colorectal oncogenesis. Cancer Res. 2016, 76, 83–95.
  56. McClurg, U.L.; Summerscales, E.E.; Harle, V.J.; Gaughan, L.; Robson, C.N. Deubiquitinating enzyme Usp12 regulates the interaction between the androgen receptor and the Akt pathway. Oncotarget 2014, 5, 7081–7092.
  57. Xu, D.; Shan, B.; Lee, B.H.; Zhu, K.; Zhang, T.; Sun, H.; Liu, M.; Shi, L.; Liang, W.; Qian, L.; et al. Phosphorylation and activation of ubiquitin-specific protease-14 by Akt regulates the ubiquitin-proteasome system. eLife 2015, 4, e10510.
  58. Zhuang, Y.J.; Liao, Z.W.; Yu, H.W.; Song, X.L.; Liu, Y.; Shi, X.Y.; Lin, X.D.; Zhou, T.C. ShRNA-mediated silencing of the ubiquitin-specific protease 22 gene restrained cell progression and affected the Akt pathway in nasopharyngeal carcinoma. Cancer Biol. Ther. 2015, 16, 88–96.
  59. Gui, D.; Peng, W.; Jiang, W.; Huang, G.; Liu, G.; Ye, Z.; Wang, Y.; Xu, Z.; Fu, J.; Luo, S.; et al. Ubiquitin-specific peptidase 46 (USP46) suppresses renal cell carcinoma tumorigenesis through AKT pathway inactivation. Biochem. Biophys. Res. Commun. 2019, 519, 689–696.
  60. Li, X.; Stevens, P.D.; Yang, H.; Gulhati, P.; Wang, W.; Evers, B.M.; Gao, T. The deubiquitination enzyme USP46 functions as a tumor suppressor by controlling PHLPP-dependent attenuation of Akt signaling in colon cancer. Oncogene 2013, 32, 471–478.
  61. Yang, Z.; Huo, S.; Shan, Y.; Liu, H.; Xu, Y.; Yao, K.; Li, X.; Zhang, X. STAT3 repressed USP7 expression is crucial for colon cancer development. FEBS Lett. 2012, 586, 3013–3017.
  62. Mahul-Mellier, A.L.; Pazarentzos, E.; Datler, C.; Iwasawa, R.; AbuAli, G.; Lin, B.; Grimm, S. De-ubiquitinating protease USP2a targets RIP1 and TRAF2 to mediate cell death by TNF. Cell Death Differ. 2012, 19, 891–899.
  63. Xiao, N.; Li, H.; Luo, J.; Wang, R.; Chen, H.; Chen, J.; Wang, P. Ubiquitin-specific protease 4 (USP4) targets TRAF2 and TRAF6 for deubiquitination and inhibits TNFα-induced cancer cell migration. Biochem. J. 2012, 441, 979–986.
  64. Xu, C.; Peng, Y.; Zhang, Q.; Xu, X.P.; Kong, X.M.; Shi, W.F. USP4 positively regulates RLR-induced NF-κB activation by targeting TRAF6 for K48-linked deubiquitination and inhibits enterovirus 71 replication. Sci. Rep. 2018, 8, 13418.
  65. Sun, W.; Tan, X.; Shi, Y.; Xu, G.; Mao, R.; Gu, X.; Fan, Y.; Yu, Y.; Burlingame, S.; Zhang, H.; et al. USP11 negatively regulates TNFα-induced NF-κB activation by targeting on IκBα. Cell. Signal. 2010, 22, 386–394.
  66. Mialki, R.K.; Zhao, J.; Wei, J.; Mallampalli, D.F.; Zhao, Y. Overexpression of USP14 protease reduces I-κB protein levels and increases cytokine release in lung epithelial cells. J. Biol. Chem. 2013, 288, 15437–15441.
  67. Meng, Q.; Cai, C.; Sun, T.; Wang, Q.; Xie, W.; Wang, R.; Cui, J. Reversible ubiquitination shapes NLRC5 function and modulates NF-κB activation switch. J. Cell Biol. 2015, 211, 1025–1040.
  68. Villeneuve, N.F.; Tian, W.; Wu, T.; Sun, Z.; Lau, A.; Chapman, E.; Fang, D.; Zhang, D.D. USP15 negatively regulates Nrf2 through deubiquitination of Keap1. Mol. Cell 2013, 51, 68–79.
  69. Liu, X.; Li, H.; Zhong, B.; Blonska, M.; Gorjestani, S.; Yan, M.; Tian, Q.; Zhang, D.E.; Lin, X.; Dong, C. USP18 inhibits NF-κB and NFAT activation during Th17 differentiation by deubiquitinating the TAK1-TAB1 complex. J. Exp. Med. 2013, 210, 1575–1590.
  70. Yang, Z.; Xian, H.; Hu, J.; Tian, S.; Qin, Y.; Wang, R.F.; Cui, J. USP18 negatively regulates NF-κB signaling by targeting TAK1 and NEMO for deubiquitination through distinct mechanisms. Sci. Rep. 2015, 5, 12738.
  71. Lei, C.Q.; Wu, X.; Zhong, X.; Jiang, L.; Zhong, B.; Shu, H.B. USP19 inhibits TNF-α- and IL-1β-triggered NF-κB activation by deubiquitinating TAK1. J. Immunol. 2019, 203, 259–268.
  72. Yasunaga, J.; Lin, F.C.; Lu, X.; Jeang, K.T. Ubiquitin-specific peptidase 20 targets TRAF6 and human T cell leukemia virus type 1 tax to negatively regulate NF-κB signaling. J. Virol. 2011, 85, 6212–6219.
  73. Liu, C.; Wang, L.; Chen, W.; Zhao, S.; Yin, C.; Lin, Y.; Jiang, A.; Zhang, P. USP35 activated by miR let-7a inhibits cell proliferation and NF-κB activation through stabilization of ABIN-2. Oncotarget 2015, 6, 27891–27906.
  74. Wang, Y.C.; Wu, Y.S.; Hung, C.Y.; Wang, S.A.; Young, M.J.; Hsu, T.I.; Hung, J.J. USP24 induces IL-6 in tumor-associated microenvironment by stabilizing p300 and β-TrCP and promotes cancer malignancy. Nat. Commun. 2018, 9, 3996.
  75. Schweitzer, K.; Naumann, M. CSN-associated USP48 confers stability to nuclear NF-κB/RelA by trimming K48-linked Ub-chains. Biochim. Biophys. Acta 2015, 1853, 453–469.
  76. Milojevic, T.; Reiterer, V.; Stefan, E.; Korkhov, V.M.; Dorostkar, M.M.; Ducza, E.; Ogris, E.; Boehm, S.; Freissmuth, M.; Nanoff, C. The ubiquitin-specific protease Usp4 regulates the cell surface level of the A2A receptor. Mol. Pharmacol. 2006, 69, 1083–1094.
  77. Berlin, I.; Higginbotham, K.M.; Dise, R.S.; Sierra, M.I.; Nash, P.D. The deubiquitinating enzyme USP8 promotes trafficking and degradation of the chemokine receptor 4 at the sorting endosome. J. Biol. Chem. 2010, 285, 37895–37908.
  78. Mines, M.A.; Goodwin, J.S.; Limbird, L.E.; Cui, F.F.; Fan, G.H. Deubiquitination of CXCR4 by USP14 is critical for both CXCL12-induced CXCR4 degradation and chemotaxis but not ERK ativation. J. Biol. Chem. 2009, 284, 5742–5752.
  79. Berthouze, M.; Venkataramanan, V.; Li, Y.; Shenoy, S.K. The deubiquitinases USP33 and USP20 coordinate β2 adrenergic receptor recycling and resensitization. EMBO J. 2009, 28, 1684–1696.
  80. Borodovsky, A.; Kessler, B.M.; Casagrande, R.; Overkleeft, H.S.; Wilkinson, K.D.; Ploegh, H.L. A novel active site-directed probe specific for deubiquitylating enzymes reveals proteasome association of USP14. EMBO J. 2001, 20, 5187–5196.
  81. Borodovsky, A.; Ovaa, H.; Kolli, N.; Gan-Erdene, T.; Wilkinson, K.D.; Ploegh, H.L.; Kessler, B.M. Chemistry-based functional proteomics reveals novel members of the deubiquitinating enzyme family. Chem. Biol. 2002, 9, 1149–1159.
  82. Sommer, S.; Weikart, N.D.; Linne, U.; Mootz, H.D. Covalent inhibition of SUMO and ubiquitin-specific cysteine proteases by an in situ thiol-alkyne addition. Bioorg. Med. Chem. 2013, 21, 2511–2517.
  83. Gjonaj, L.; Sapmaz, A.; Flierman, D.; Janssen, G.M.C.; van Veelen, P.A.; Ovaa, H. Development of a DUB-selective fluorogenic substrate. Chem. Sci. 2019, 10, 10290–10296.
  84. Dang, L.C.; Melandri, F.D.; Stein, R.L. Kinetic and mechanistic studies on the hydrolysis of ubiquitin C-terminal 7-amido-4-methylcoumarin by deubiquitinating enzymes. Biochemistry 1998, 37, 1868–1879.
  85. Hassiepen, U.; Eidhoff, U.; Meder, G.; Bulber, J.F.; Hein, A.; Bodendorf, U.; Lorthiois, E.; Martoglio, B. A sensitive fluorescence intensity assay for deubiquitinating proteases using ubiquitin-rhodamine110-glycine as substrate. Anal. Biochem. 2007, 371, 201–207.
  86. Tirat, A.; Schilb, A.; Riou, V.; Leder, L.; Gerhartz, B.; Zimmermann, J.; Worpenberg, S.; Eldhoff, U.; Freuler, F.; Stettler, T.; et al. Synthesis and characterization of fluorescent ubiquitin derivatives as highly sensitive substrates for the deubiquitinating enzymes UCH-L3 and USP-2. Anal. Biochem. 2005, 343, 244–255.
  87. Nicholson, B.; Leach, C.A.; Goldenberg, S.J.; Francis, D.M.; Kodrasov, M.P.; Tian, X.; Shanks, J.; Sterner, D.E.; Bernal, A.; Mattern, M.R.; et al. Characterization of ubiquitin and ubiquitin-like-protein isopeptidase activities. Protein Sci. 2008, 17, 1035–1043.
  88. Goldenberg, S.J.; McDermott, J.L.; Butt, T.R.; Mattern, M.R.; Nicholson, B. Strategies for the identification of novel inhibitors of deubiquitinating enzymes. Biochem. Soc. Trans. 2008, 36, 828–832.
  89. Geurink, P.P.; El Oualid, F.; Jonker, A.; Hameed, D.S.; Ovaa, H. A general chemical ligation approach towards isopeptide-linked ubiquitin and ubiquitin-like assay reagents. ChemBioChem 2012, 13, 293–297.
  90. Ohayon, S.; Spasser, L.; Aharoni, A.; Brik, A. Targeting deubiquitinases enabled by chemical synthesis of proteins. J. Am. Chem. Soc. 2012, 134, 3281–3289.
  91. Horton, R.A.; Strachan, E.A.; Vogel, K.W.; Riddle, S.M. A substrate for deubiquitinating enzymes based on time-resolved fluorescence resonance energy transfer between terbium and yellow fluorescent protein. Anal. Biochem. 2007, 360, 138–143.
  92. Faesen, A.C.; Luna-Vargas, M.P.; Geurink, P.P.; Clerici, M.; Merkx, R.; van Dijk, W.J.; Hameed, D.S.; El Oualid, F.; Ovaa, H.; Sixma, T.K. The differential modulation of USP activity by internal regulatory domains, interactors and eight ubiquitin chain types. Chem. Biol. 2011, 18, 1550–1561.
  93. Wang, Z.; Xie, W.; Zhu, M.; Zhou, H. Development of a highly reliable assay for ubiquitin-specific protease 2 inhibitors. Bioorg. Med. Chem. Lett. 2017, 27, 4015–4018.
  94. Ritorto, M.S.; Ewan, R.; Perez-Oliva, A.B.; Knebel, A.; Buhrlage, S.J.; Wightman, M.; Kelly, S.M.; Wood, N.T.; Virdee, S.; Gray, N.S.; et al. Screening of DUB activity and specificity by MALDI-TOF mass spectrometry. Nat. Commun. 2014, 5, 4763.
More
Information
Subjects: Others
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 1.5K
Revisions: 2 times (View History)
Update Date: 11 Jun 2021
1000/1000