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 + 2555 word(s) 2555 2021-06-16 05:52:26 |
2 format correction Meta information modification 2555 2021-06-23 03:47:35 |

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.
Lee, B. Small-Molecule Inhibitors Targeting Proteasome-Associated Deubiquitinases. Encyclopedia. Available online: https://encyclopedia.pub/entry/11144 (accessed on 28 March 2024).
Lee B. Small-Molecule Inhibitors Targeting Proteasome-Associated Deubiquitinases. Encyclopedia. Available at: https://encyclopedia.pub/entry/11144. Accessed March 28, 2024.
Lee, Byung-Hoon. "Small-Molecule Inhibitors Targeting Proteasome-Associated Deubiquitinases" Encyclopedia, https://encyclopedia.pub/entry/11144 (accessed March 28, 2024).
Lee, B. (2021, June 22). Small-Molecule Inhibitors Targeting Proteasome-Associated Deubiquitinases. In Encyclopedia. https://encyclopedia.pub/entry/11144
Lee, Byung-Hoon. "Small-Molecule Inhibitors Targeting Proteasome-Associated Deubiquitinases." Encyclopedia. Web. 22 June, 2021.
Small-Molecule Inhibitors Targeting Proteasome-Associated Deubiquitinases
Edit

The 26S proteasome is the principal protease for regulated intracellular proteolysis. This multi-subunit complex is also pivotal for clearance of harmful proteins that are produced throughout the lifetime of eukaryotes. Recent structural and kinetic studies have revealed a multitude of conformational states of the proteasome in substrate-free and substrate-engaged forms. These conformational transitions demonstrate that proteasome is a highly dynamic machinery during substrate processing that can be also controlled by a number of proteasome-associated factors. Essentially, three distinct family of deubiquitinases–USP14, RPN11, and UCH37–are associated with the 19S regulatory particle of human proteasome. USP14 and UCH37 are capable of editing ubiquitin conjugates during the process of their dynamic engagement into the proteasome prior to the catalytic commitment. In contrast, RPN11-mediated deubiquitination is directly coupled to substrate degradation by sensing the proteasome’s conformational switch into the commitment steps. 

proteasome proteolysis deubiquitinase USP14 UCH37 RPN11 small-molecule inhibitors IU1 capzimin

1. Introduction

The ubiquitin-proteasome system (UPS) represents a crucial cellular mechanism for highly regulated proteolysis and protein quality control process in eukaryotes [1][2]. The 26S proteasome is a large multi-subunit protease of ~2.5 MDa for selective degradation of intracellular proteins that are tagged by ubiquitins [3][4]. Recent findings indicate that proteasome is actively adapted to a large network of protein interactions for discrete degradation events, and such adaptability may also be controlled through a multitude of proteasome’s conformational transitions [5][6][7][8][9]. Notably, deubiquitinases (DUBs), which exclusively reverse the ubiquitination process in the UPS, are also critically associated with the proteasome [10][11][12]. In mammals, the regulatory particle (RP) of the 26S proteasome contains three major classes of DUBs–USP14 (Ubp6 in budding yeast), RPN11 (also known as PSMD14), and UCH37 (also known as UCH-L5) (Figure 1) [3][11][12][13]. USP14/Ubp6 is a reversible interactor with the proteasome, and its activity can be highly enhanced by association with the proteasome [12][14][15][16]. USP14 is capable of sparing the substrates from degradation prior to the proteasome’s commitment step and shows remarkable preference for multi-chain bearing ubiquitin conjugates [16][17][18]. By contrast, RPN11 is an integral subunit of the proteasome, and this metalloprotease is essentially coupled to substrate degradation in an ATP-dependent manner [11][19][20]. Although USP14 and RPN11 may mediate opposite proteolytic consequences, both of the enzymes apparently share a similar en bloc or proximal cleavage mechanism [11][17]. The function of UCH37 on the proteasome remains to be further established because this DUB may distally trim the ubiquitin chains for rescuing the substrates from degradation but also can selectively debranch the K48-linkage among a complex mixture of bifurcate ubiquitin conjugates for enhanced substrate degradation [12][21][22]. DUBs are emerging as attractive therapeutic targets because they may control the turnover rate of a number of intracellular proteins, including ones that might be highly deregulated in the disease states [23][24]. The isopeptidase activities of DUBs can be selectively inhibited by catalytic site-directed drug-like compounds. Moreover, recent advances in developing robust screening technologies with more refined chemical libraries have successfully yielded promising small-molecule DUB antagonists of active site-directed inhibitors as well as allosteric inhibitors [24][25][26][27]. Specific DUB inhibition on the proteasome is particularly appealing because each proteasome-associated DUB can exert distinct influence over the proteolytic outputs (Figure 1A). Therefore, it is not surprising that considerable efforts from academia and industry have also been put towards developing drug-like molecules for targeting proteasome-associated DUB activities [23][24][28]. Such specific DUB inhibitors at the proteasome not only offer exciting degradation-based therapeutic strategies but also serve as valuable chemical tools to reveal novel deubiquitination biology for dynamic proteasome function.Ijms 22 06213 g001 550
Figure 1. Proposed working mechanisms of proteasomal deubiquitinase inhibitors and their comparison to proteasome inhibitor and PROTAC. (A) (Top) USP14’s multi-chain specific cleavage activity can be selectively targeted by USP14 inhibitors (e.g., IU1 is shown as an example), resulting in induced degradation of substrates. (Middle) Degradation-coupled RPN11 activity can be selectively inhibited such as by capzimin as shown. RPN11 inhibition can strongly suppress the proteasome-mediated substrate degradation. (Bottom) UCH37 specific inhibitors–which have not been developed yet–may exert differential effects on proteolysis depending on the type of ubiquitin conjugates. Unbranched or poorly ubiquitinated substrates might be highly subject to UCH37’s trimming activity, and its specific inhibition may lead to induced protein degradation. By contrast, degradation of branched ubiquitin conjugates is likely to be attenuated by UCH37 inhibition. (B,C) Proteasome inhibitor (e.g., bortezomib as shown) and PROTAC are depicted as examples of proteolysis suppressor and inducer, respectively. Color-coded circles in proteasome at B indicate each pair of proteasome’s active sites. PROTAC is a chimeric compound closely linking E3 and target substrate, thus facilitating the ubiquitination process. Inh, inhibitor. See the text for more details.

2. Proteasomal Deubiquitinases as Therapeutic Targets

For the past decades, the UPS has been clearly recognized among the most important drug targets because of its critical contribution to protein homeostasis, signaling pathways, and cellular physiology; its deregulation or genetic alteration is intimately associated with human pathogenesis [29][30][31][32][33]. The success story of proteasome inhibitors for cancer therapy highlights the clinical importance of the UPS as valid targets that can be even further expanded into various aspects of the proteolytic system and other types of pathophysiology [34][35]. In fact, a recently emerging novel paradigm of “induced proteolysis”, such as by PROteolysis TArgeting Chimera (PROTAC), can chemically harness the endogenous ubiquitination machinery for targeted protein degradation (Figure 1B) [36][37].
As opposed to proteasome inhibition, this new concept defines the UPS as yet another class of extraordinary drug target for effectively disposing of the conventionally intractable or “undruggable” disease-associated proteins (Figure 1) [24][38]. In the similar context, deubiquitylation reactions may offer exciting opportunities for developing promising drug candidates due to their key roles in the proteolytic pathways as well as other biological processes [23][24]. Although the development of specific DUB inhibitors is challenging per se and still in its early stage, recently performed a series of elegant works have produced nice examples of highly selective small-molecule inhibitors for targeting USP7 and USP30 [24][39][40][41][42][43][44].
Targeting DUBs on the proteasome may also represent unique therapeutic strategies for actively regulating the proteasome-mediated proteolysis in a dynamic manner. Individual or ensemble of deubiquitination activities can exert distinct and multiple impacts on the proteasome before or throughout substrate processing (Figure 1A); such DUB-imposed regulation may render the proteasomal activities to be highly versatile, and in this sense, the proteasome acts as a critical hub as well as a rate limiting step for the ubiquitin-dependent degradation pathways [11][12]. Recent high resolution cryo-electron microscopy (cryo-EM) studies also have identified a number of conformational states of substrate-free and substrate-bound proteasomes, in which the proteasome-associated DUBs are likely to actively and differentially modulate the degradation events by sensing those conformational dynamics [4][5][12].
Among three major proteasomal DUBs, USP14 or its yeast ortholog Ubp6 is a thiol protease that is only transiently associated with the proteasome; thus, this enzyme may favor the specific conformational states of the proteasome [14][15][16][45][46][47]. Earlier genetic studies have revealed that USP14/Ubp6 is a sensitive responder to ubiquitin and proteasome stress, and also to proteotoxic stress, although in general its deficiency is tolerable for cell survival [48][49][50][51][52][53][54]. During the mouse development, however, this isopeptidase is critically involved in the motor neuron function partially through the noncatalytic mechanism [55][56][57][58]. As a therapeutic target, USP14 has been best studied in neurological disorders and cancers [23][59]. USP14 and its inhibitors (as discussed in Section 3) have been reported to regulate several pathological targets, such as Tau, ATXN3, TDP-43, GFAP, and PrP that are highly implicated in neurodegenerative diseases [16][60][61][62]. Intriguingly, apart from its inhibitory role on the proteasome, USP14 was also found to negatively regulate autophagy in mammalian cells and basal mitophagy in fly models [63][64]. USP14 expression is upregulated in several cancers including lung adenocarcinoma, ovarian cancers, esophageal squamous cell carcinoma, and pancreatic ductal adenocarcinoma, in which this enzyme is often positively correlated with tumor recurrence, metastasis, and poor patient survival [65][66][67][68][69]. Albeit seemingly a promising clinical target, the underlying mechanisms of how USP14 participates in those disease processes still remain to be elucidated.
In contrast to USP14, RPN11/PSMD14 (also known as POH1) is an essential subunit of the proteasome that belongs to JAMM/MPN metalloprotease class of DUB [19][20][70]. Recent cryo-EM studies provide the structural basis of how RPN11′s DUB activity can be coupled to substrate translocation and degradation in an ATP-dependent fashion [6][11][13][47][71][72][73]. Along with other RP components, this metalloprotease undergoes noticeable structural changes during the transitions from the substrate-free state to the substrate-processing states of the proteasome. This conformational switch drives RPN11 to be catalytically productive for the committed substrates in both repositioning on the proteasome and reshaping the local structure of its featured Ins-1 loop. Due to its strict requirement for proteasomal degradation, the genetic depletion or catalytic mutation of RPN11 causes the lethality [19][49][51][74][75][76]. Therefore, the successful targeting strategy for cancer therapy by proteasome inhibitors might be also similarly applied to RPN11-mediated inhibition of proteasomal degradation [77]. The key difference here, however, is that in contrast to the core particle (CP)-directed catalytic inhibition, RPN11 inhibition will occur on the RP, and thus is likely to show more specific effects. Besides, RPN11 has been implicated in oncogenesis as a potential drug target; its expression level is positively correlated with tumor formation and metastasis, while the genetic depletion or pharmacological inhibition showing the opposite effects–such as in hepatocellular carcinoma, multiple myeloma, breast cancer, esophageal cancer, colorectal cancer, and prostate cancer [78][79][80][81][82][83].
Like USP14, UCH37/UCH-L5 is a thiol protease class of DUB that is reversibly associated with the 19S RP of the proteasome; its binding is mediated by RPN13/ADRM1, a ubiquitin receptor which can also markedly enhance the UCH37′s activity [84][85][86]. An intriguing feature of UCH37 is that this enzyme belongs to both the proteasome and the INO80 chromatin-remodeling complex in a mutually exclusive manner; its DUB activity can be selectively activated only when bound to the proteasome [87][88][89]. UCH37 was reported to trim the distal ubiquitin from erroneously ubiquitinated proteins for their rescue [21], or it does so to release proteasome-occupying unanchored chains for the productive round of substrate loading [90]. Interestingly, a recent study demonstrated that UCH37 on the proteasome can selectively cleave the K48-linked branched chains to promote the degradation of substrates [12][22]. In any case, the exact physiological functions of UCH37 remain largely elusive. Like USP14 and RPN11, several lines of studies have reported that UCH37 expression is elevated in a number of cancers including esophageal squamous cell carcinoma, hepatocellular carcinoma, epithelial ovarian cancer, endometrial cancer, and lung adenocarcinoma, in which this protease is associated with tumor progression and poor patient survival [91][92][93][94][95][96].

3. Proteasomal Deubiquitinase Inhibitors

3.1. USP14 Inhibitors

In 2010, Finley and colleagues have identified IU1, the first highly selective inhibitor of proteasome-bound USP14 by ubiquitin-7-amido-4-methylcoumarin (Ub-AMC) hydrolysis assay-based high-throughput screening (Table 1) [16]. Because Ub-AMC is preferentially cleaved by UCH37 over USP14, the assay was performed by reconstituting ubiquitin-vinyl sulfone (Ub-VS)-pretreated human proteasome with recombinant USP14–to quench the basal UCH37 activity by Ub-VS and isolate the authentic USP14 activity on the proteasome [16][18][97]. IU1 specifically inhibits the proteasome-bound form of USP14 with an IC50 of 4–5 μM and shows good selectivity against a panel of eight other DUBs. A subsequent medicinal chemistry led to identification of more potent IU1 derivatives, such as IU1-2, IU1-33, and IU1-47 (IC50s = 1.7 μM, 1.1 μM, and 0.6 μM, respectively against USP14), all of which exhibited better selectivity over IsoT (Table 1) [62]. From this structure-activity relationship (SAR) study, some key functional moieties in the parental compound were revealed to further improve the inhibitory activity.
Table 1. Representative examples of the reported proteasomal deubiquitinase inhibitors.

Target

Compound Name

Structure

Notes

Reference

USP14

IU1

Ijms 22 06213 i001

IC50: 4.7 μM

(Ub-AMC)

Lee et al., 2010 [16]

IU1-2

Ijms 22 06213 i002

IC50: 1.7 μM

(Ub-AMC)

Boselli et al., 2017 [62]

IU1-33

Ijms 22 06213 i003

IC50: 1.1 μM

(Ub-AMC)

IU1-47

Ijms 22 06213 i004

IC50: 0.6 μM

(Ub-AMC)

IU1-206

Ijms 22 06213 i005

N/A

Wang et al., 2018 [98]

IU1-248

Ijms 22 06213 i006

IC50: 0.83 μM

(Ub-AMC)

1B10

Ijms 22 06213 i007

N/A

Palmer et al., 2018 [99]

1D18

Ijms 22 06213 i008

N/A

Compound 162

Ijms 22 06213 i009

IC50: <0.5 μM

(Ub-AMC)

WO/2015/073528 [100]

Compound 335

(SB1-B-57)

Ijms 22 06213 i010

IC50: <0.5 μM

(Ub-AMC)

Compound 83

Ijms 22 06213 i011

IC50: <0.5 μM

(Ub-AMC)

WO/2020/006269 [101]

Compound 2B

Ijms 22 06213 i012

IC50: <0.05 μM

(Ub-AMC)

WO/2020/006296 [102]

IU2-6

Ijms 22 06213 i013

74% inhibition at 8 μM

(Ub-AMC)

WO/2012/012712 [103];

Kemp, 2016 [104]

Compound 3

Ijms 22 06213 i014

IC50: 0.5 μM

(Ub-AMC)

WO/2013/112651 [105];

Kemp, 2016 [104]

RPN11

8-TQ *

Ijms 22 06213 i015

IC50: 2.4 μM

(Ub4-pepOG)

Li et al., 2017 [106];

Perez et al., 2017 [107]

Capzimin

Ijms 22 06213 i016

IC50: 0.34 μM

(Ub4-pepOG)

Thiolutin *

Ijms 22 06213 i017

IC50: 0.53 μM

(Ub4-pepOG)

Lauinger et al., 2017 [108]

SOP6 *

Ijms 22 06213 i018

IC50: 3.8 μM

(Fluorescent UbnGST-Wbp2)

Li et al., 2018 [109]

SOP11 *

Ijms 22 06213 i019

IC50: 1.3 μM

(Fluorescent UbnGST-Wbp2)

Promiscuous proteasomal DUB inhibitors

b-AP15

(USP14/

UCH37)

Ijms 22 06213 i020

19S RP IC50:

6.5 μM

(Ub-Rho)

D‘Arcy et al., 2011 [110];

Wang et al., 2015 [111]

VLX1570

(USP14/

UCH37)

Ijms 22 06213 i021

19S RP IC50:

6.4 μM

(Ub-Rho)

Wang et al., 2015 [111]

WP1130

(USP9x/USP5/

USP14/UCH37)

Ijms 22 06213 i022

IC50s: <5~10 μM

(Ub-AMC

& Ub-VS)

Kapuria et al., 2010 [112]

AC17

(19S RP)

Ijms 22 06213 i023

19S RP IC50:

4.23 μM

(Ub-AMC)

Zhou et al., 2013 [113]

Auranofin **

(TrxR/19S RP; 19S RP at higher dosage than TrxR)

Ijms 22 06213 i024

TrxR system inhibition at ~1 μM

(HCT-116 cell)

Reduced 19S RP labeling at 5 μM

(Ub-VS)

Liu et al., 2014 [114];

Stafford et al., 2018 [115];

Zhang et al., 2019 [116]

* These inhibitors also inhibit other JAMM metalloproteases. ** This inhibitor may have its non-DUB target in pharmacological dosage.

3.2. RPN11 Inhibitors

The first selective RPN11 inhibitor was reported by the Deshaies and Cohen groups in 2017 [106][107]. To measure robust RPN11 activity, the researchers established an elegant fluorescence polarization-based assay with the tandem tetraubiquitin tagged-peptide Oregon Green (Ub4-pepOG) as the DUB substrate. By employing two types of chemical libraries–1) metal binding pharmacophores-focused fragment library of 351 compounds and 2) high-throughput screening library of 330,000 compounds, they identified the hits of 8-thioquinoline (8TQ) and H18, which is actually a thioester derivative of 8TQ (Table 1). They also demonstrated that 8TQ and H18 inhibit RPN11 by chelating the metal coordination of the active site Zn2+ ion [106][107]. Subsequent SAR study was conducted to optimize the functional moiety of 8TQ, and the lead compound capzimin was successfully developed (Table 1). Capzimin (8-mercapto-N-(2-(thiazol-2-yl)ethyl)quinoline-3-carboxamide) was seven-fold more potent than 8TQ for RPN11 (IC50 = 0.34 μM), and showed good selectivity over other JAMM metalloproteases with a range of 6 to 80-fold in IC50s. Intriguingly, the inhibitory mechanism of capzimin is reversible and uncompetitive for RPN11, while displaying the competitive inhibition against AMSH and BRCC36. When treated in cells, capzimin strongly elicited the formation of aggresomes and the accumulation of ubiquitinated conjugates, UbG76V-GFP model substrate, and endogenous proteasomal substrates, as observed in proteasome inhibitor treatment.

3.3. Other Proteasomal Deubiquitinase Inhibitors

UCH37 specific inhibitors have not been developed yet, and so in this Section, we will primarily discuss some nonspecific DUB inhibitors which also target UCH37 activity. b-AP15 (originally known as NSC687852) was first identified from cell-based chemical screening by the Linder group as a small-molecule that can induce lysosomal and p53-independent apoptosis (Table 1) [117][118]. In 2011, the same group reported that b-AP15 inhibits 19S RP DUB activity, specifically USP14 and UCH37, and such dual inhibition leads to the accumulation of polyubiquitinated conjugates through proteasome inhibition [110]. The reported IC50s of b-AP15 against 19S RP have been somewhat inconsistent in the literatures: 2.1 μM or 16.8 μM for Ub-AMC [110][111][119], or 6.5 μM for ubiquitin-rhodamine (Ub-Rho) [111]. The researchers also developed VLX1570, an azepane-cored b-AP derivative by performing SAR studies (Table 1) [111]. IC50s of VLX1570 against 19S RP DUB activity were obtained as 13 μM for Ub-AMC and 6.4 μM for Ub-Rho [111]. The authors argued that VLX1570 may inhibit those two thiol protease of DUBs by making Michael’s addition-based covalent interactions with the catalytic cysteines despite its reversible binding mode [119][120]. Curiously, they further found that VLX1570 preferentially inhibits USP14 over UCH37 in active site-directed ubiquitin probe competition assays, which is distinct from its comparable inhibition of both of the DUBs from the fluorescent ubiquitin adduct cleavage assays [120].

References

  1. Hershko, A.; Ciechanover, A. The ubiquitin system for protein degradation. Annu. Rev. Biochem. 1992, 61, 761–807.
  2. Goldberg, A.L. Protein degradation and protection against misfolded or damaged proteins. Nature 2003, 426, 895–899.
  3. Finley, D. Recognition and processing of ubiquitin-protein conjugates by the proteasome. Annu. Rev. Biochem. 2009, 78, 477–513.
  4. Mao, Y. Structure, Dynamics and Function of the 26S Proteasome. Subcell. Biochem. 2021, 96, 1–151.
  5. Finley, D.; Prado, M.A. The Proteasome and Its Network: Engineering for Adaptability. Cold Spring Harb. Perspect. Biol. 2020, 12, a033985.
  6. Greene, E.R.; Dong, K.C.; Martin, A. Understanding the 26S proteasome molecular machine from a structural and conformational dynamics perspective. Curr. Opin. Struct. Biol. 2020, 61, 33–41.
  7. Chen, X.; Htet, Z.M.; Lopez-Alfonzo, E.; Martin, A.; Walters, K.J. Proteasome interaction with ubiquitinated substrates: From mechanisms to therapies. FEBS J. 2020.
  8. Davis, C.; Spaller, B.L.; Matouschek, A. Mechanisms of substrate recognition by the 26S proteasome. Curr. Opin. Struct. Biol. 2020, 67, 161–169.
  9. Sahu, I.; Glickman, M.H. Proteasome in action: Substrate degradation by the 26S proteasome. Biochem. Soc. Trans. 2021, 49, 629–644.
  10. Mevissen, T.E.T.; Komander, D. Mechanisms of Deubiquitinase Specificity and Regulation. Annu. Rev. Biochem. 2017, 86, 159–192.
  11. De Poot, S.A.H.; Tian, G.; Finley, D. Meddling with Fate: The Proteasomal Deubiquitinating Enzymes. J. Mol. Biol. 2017, 429, 3525–3545.
  12. Shin, J.Y.; Muniyappan, S.; Tran, N.N.; Park, H.; Lee, S.B.; Lee, B.H. Deubiquitination Reactions on the Proteasome for Proteasome Versatility. Int. J. Mol. Sci. 2020, 21, 5312.
  13. Bard, J.A.M.; Goodall, E.A.; Greene, E.R.; Jonsson, E.; Dong, K.C.; Martin, A. Structure and Function of the 26S Proteasome. Annu. Rev. Biochem. 2018, 87, 697–724.
  14. Leggett, D.S.; Hanna, J.; Borodovsky, A.; Crosas, B.; Schmidt, M.; Baker, R.T.; Walz, T.; Ploegh, H.; Finley, D. Multiple associated proteins regulate proteasome structure and function. Mol. Cell 2002, 10, 495–507.
  15. Hanna, J.; Hathaway, N.A.; Tone, Y.; Crosas, B.; Elsasser, S.; Kirkpatrick, D.S.; Leggett, D.S.; Gygi, S.P.; King, R.W.; Finley, D. Deubiquitinating enzyme Ubp6 functions noncatalytically to delay proteasomal degradation. Cell 2006, 127, 99–111.
  16. Lee, B.H.; Lee, M.J.; Park, S.; Oh, D.C.; Elsasser, S.; Chen, P.C.; Gartner, C.; Dimova, N.; Hanna, J.; Gygi, S.P.; et al. Enhancement of proteasome activity by a small-molecule inhibitor of USP14. Nature 2010, 467, 179–184.
  17. Lee, B.H.; Lu, Y.; Prado, M.A.; Shi, Y.; Tian, G.; Sun, S.; Elsasser, S.; Gygi, S.P.; King, R.W.; Finley, D. USP14 deubiquitinates proteasome-bound substrates that are ubiquitinated at multiple sites. Nature 2016, 532, 398–401.
  18. Muniyappan, S.; Lee, B.H. In vitro analysis of proteasome-associated USP14 activity for substrate degradation and deubiquitylation. Methods Enzymol. 2019, 619, 249–268.
  19. Verma, R.; Aravind, L.; Oania, R.; McDonald, W.H.; Yates, J.R., III; Koonin, E.V.; Deshaies, R.J. Role of Rpn11 metalloprotease in deubiquitination and degradation by the 26S proteasome. Science 2002, 298, 611–615.
  20. Yao, T.; Cohen, R.E. A cryptic protease couples deubiquitination and degradation by the proteasome. Nature 2002, 419, 403–407.
  21. Lam, Y.A.; Xu, W.; DeMartino, G.N.; Cohen, R.E. Editing of ubiquitin conjugates by an isopeptidase in the 26S proteasome. Nature 1997, 385, 737–740.
  22. Deol, K.K.; Crowe, S.O.; Du, J.; Bisbee, H.A.; Guenette, R.G.; Strieter, E.R. Proteasome-Bound UCH37/UCHL5 Debranches Ubiquitin Chains to Promote Degradation. Mol. Cell 2020, 80, 796–809.e9.
  23. Harrigan, J.A.; Jacq, X.; Martin, N.M.; Jackson, S.P. Deubiquitylating enzymes and drug discovery: Emerging opportunities. Nat. Rev. Drug Discov. 2018, 17, 57–78.
  24. Moon, S.; Lee, B.H. Chemically Induced Cellular Proteolysis: An Emerging Therapeutic Strategy for Undruggable Targets. Mol. Cells 2018, 41, 933–942.
  25. Ndubaku, C.; Tsui, V. Inhibiting the deubiquitinating enzymes (DUBs). J. Med. Chem. 2015, 58, 1581–1595.
  26. Gopinath, P.; Ohayon, S.; Nawatha, M.; Brik, A. Chemical and semisynthetic approaches to study and target deubiquitinases. Chem. Soc. Rev. 2016, 45, 4171–4198.
  27. Schauer, N.J.; Magin, R.S.; Liu, X.; Doherty, L.M.; Buhrlage, S.J. Advances in Discovering Deubiquitinating Enzyme (DUB) Inhibitors. J. Med. Chem. 2020, 63, 2731–2750.
  28. Wertz, I.E.; Wang, X. From Discovery to Bedside: Targeting the Ubiquitin System. Cell Chem. Biol. 2019, 26, 156–177.
  29. Cohen, P.; Tcherpakov, M. Will the Ubiquitin System Furnish as Many Drug Targets as Protein Kinases? Cell 2010, 143, 686–693.
  30. Popovic, D.; Vucic, D.; Dikic, I. Ubiquitination in disease pathogenesis and treatment. Nat. Med. 2014, 20, 1242–1253.
  31. Schmidt, M.; Finley, D. Regulation of proteasome activity in health and disease. BBA Mol. Cell Res. 2014, 1843, 13–25.
  32. Weathington, N.M.; Mallampalli, R.K. Emerging therapies targeting the ubiquitin proteasome system in cancer. J. Clin. Investig. 2014, 124, 6–12.
  33. Rape, M. Ubiquitylation at the crossroads of development and disease. Nat. Rev. Mol. Cell Biol. 2018, 19, 59–70.
  34. Richardson, P.G.; Hideshima, T.; Anderson, K.C. Bortezomib (PS-341): A novel, first-in-class proteasome inhibitor for the treatment of multiple myeloma and other cancers. Cancer Control 2003, 10, 361–369.
  35. Chen, D.; Frezza, M.; Schmitt, S.; Kanwar, J.; Dou, Q.P. Bortezomib as the First Proteasome Inhibitor Anticancer Drug: Current Status and Future Perspectives. Curr. Cancer Drug Targets 2011, 11, 239–253.
  36. Burslem, G.M.; Crews, C.M. Proteolysis-Targeting Chimeras as Therapeutics and Tools for Biological Discovery. Cell 2020, 181, 102–114.
  37. Verma, R.; Mohl, D.; Deshaies, R.J. Harnessing the Power of Proteolysis for Targeted Protein Inactivation. Mol. Cell 2020, 77, 446–460.
  38. Huang, X.D.; Dixit, V.M. Drugging the undruggables: Exploring the ubiquitin system for drug development. Cell Res. 2016, 26, 484–498.
  39. Kategaya, L.; Di Lello, P.; Rouge, L.; Pastor, R.; Clark, K.R.; Drummond, J.; Kleinheinz, T.; Lin, E.; Upton, J.P.; Prakash, S.; et al. USP7 small-molecule inhibitors interfere with ubiquitin binding. Nature 2017, 550, 534–538.
  40. Lamberto, I.; Liu, X.X.; Seo, H.S.; Schauer, N.J.; Iacob, R.E.; Hu, W.Y.; Das, D.; Mikhailova, T.; Weisberg, E.L.; Engen, J.R.; et al. Structure-Guided Development of a Potent and Selective Non-covalent Active-Site Inhibitor of USP7. Cell Chem. Biol. 2017, 24, 1490–1500.e11.
  41. Turnbull, A.P.; Ioannidis, S.; Krajewski, W.W.; Pinto-Fernandez, A.; Heride, C.; Martin, A.C.L.; Tonkin, L.M.; Townsend, E.C.; Buker, S.M.; Lancia, D.R.; et al. Molecular basis of USP7 inhibition by selective small-molecule inhibitors. Nature 2017, 550, 481–486.
  42. Gavory, G.; O’Dowd, C.R.; Helm, M.D.; Flasz, J.; Arkoudis, E.; Dossang, A.; Hughes, C.; Cassidy, E.; McClelland, K.; Odrzywol, E.; et al. Discovery and characterization of highly potent and selective allosteric USP7 inhibitors. Nat. Chem. Biol. 2018, 14, 118–125.
  43. Kluge, A.F.; Lagu, B.R.; Maiti, P.; Jaleel, M.; Webb, M.; Malhotra, J.; Mallat, A.; Srinivas, P.A.; Thompson, J.E. Novel highly selective inhibitors of ubiquitin specific protease 30 (USP30) accelerate mitophagy. Bioorganic Med. Chem. Lett. 2018, 28, 2655–2659.
  44. Li, P.; Liu, H.-M. Recent advances in the development of ubiquitin-specific-processing protease 7 (USP7) inhibitors. Eur. J. Med. Chem. 2020, 191, 112107.
  45. Borodovsky, A.; Kessler, B.; 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.
  46. Aufderheide, A.; Beck, F.; Stengel, F.; Hartwig, M.; Schweitzer, A.; Pfeifer, G.; Goldberg, A.L.; Sakata, E.; Baumeister, W.; Förster, F. Structural characterization of the interaction of Ubp6 with the 26S proteasome. Proc. Natl. Acad. Sci. USA 2015, 112, 8626–8631.
  47. Huang, X.; Luan, B.; Wu, J.; Shi, Y. An atomic structure of the human 26S proteasome. Nat. Struct. Mol. Biol. 2016, 23, 778–785.
  48. Hanna, J.; Leggett, D.S.; Finley, D. Ubiquitin Depletion as a Key Mediator of Toxicity by Translational Inhibitors. Mol. Cell. Biol. 2003, 23, 9251–9261.
  49. Guterman, A.; Glickman, M. Complementary Roles for Rpn11 and Ubp6 in Deubiquitination and Proteolysis by the Proteasome. J. Biol. Chem. 2004, 279, 1729–1738.
  50. Hanna, J.; Meides, A.; Zhang, D.P.; Finley, D. A Ubiquitin Stress Response Induces Altered Proteasome Composition. Cell 2007, 129, 747–759.
  51. Koulich, E.; Li, X.; DeMartino, G.N. Relative Structural and Functional Roles of Multiple Deubiquitylating Proteins Associated with Mammalian 26S Proteasome. Mol. Biol. Cell 2008, 19, 1072–1082.
  52. Torres, E.M.; Dephoure, N.; Panneerselvam, A.; Tucker, C.M.; Whittaker, C.A.; Gygi, S.P.; Dunham, M.J.; Amon, A. Identification of Aneuploidy-Tolerating Mutations. Cell 2010, 143, 71–83.
  53. Oromendia, A.B.; Dodgson, S.E.; Amon, A. Aneuploidy causes proteotoxic stress in yeast. Genes Dev. 2012, 26, 2696–2708.
  54. Dephoure, N.; Hwang, S.; O’Sullivan, C.; Dodgson, S.E.; Gygi, S.P.; Amon, A.; Torres, E.M. Quantitative proteomic analysis reveals posttranslational responses to aneuploidy in yeast. eLife 2014, 3, e03023.
  55. Wilson, S.; Bhattacharyya, B.; Rachel, R.A.; Coppola, V.; Tessarollo, L.; Householder, D.B.; Fletcher, C.F.; Miller, R.J.; Copeland, N.G.; Jenkins, N.A. Synaptic defects in ataxia mice result from a mutation in Usp14, encoding a ubiquitin-specific protease. Nat. Genet. 2002, 32, 420–425.
  56. Chen, P.-C.; Qin, L.-N.; Li, X.-M.; Walters, B.J.; Wilson, J.A.; Mei, L.; Wilson, S. The Proteasome-Associated Deubiquitinating Enzyme Usp14 Is Essential for the Maintenance of Synaptic Ubiquitin Levels and the Development of Neuromuscular Junctions. J. Neurosci. 2009, 29, 10909–10919.
  57. Walters, B.J.; Hallengren, J.J.; Theile, C.S.; Ploegh, H.L.; Wilson, S.M.; Dobrunz, L.E. A catalytic independent function of the deubiquitinating enzyme USP14 regulates hippocampal synaptic short-term plasticity and vesicle number. J. Physiol. 2014, 592, 571–586.
  58. Vaden, J.H.; Watson, J.A.; Howard, A.D.; Echen, P.-C.; Wilson, J.A.; Wilson, S.M. Distinct effects of ubiquitin overexpression on NMJ structure and motor performance in mice expressing catalytically inactive USP14. Front. Mol. Neurosci. 2015, 8, 11.
  59. Wertz, I.E.; Murray, J.M. Structurally-defined deubiquitinase inhibitors provide opportunities to investigate disease mechanisms. Drug Discov. Today Technol. 2019, 31, 109–123.
  60. Homma, T.; Ishibashi, D.; Nakagaki, T.; Fuse, T.; Mori, T.; Satoh, K.; Atarashi, R.; Nishida, N. Ubiquitin-specific protease 14 modulates degradation of cellular prion protein. Sci. Rep. 2015, 5, 11028.
  61. McKinnon, C.; Goold, R.G.; Andre, R.; Devoy, A.; Ortega, Z.; Moonga, J.; Linehan, J.M.; Brandner, S.; Lucas, J.J.; Collinge, J.; et al. Prion-mediated neurodegeneration is associated with early impairment of the ubiquitin–proteasome system. Acta Neuropathol. 2016, 131, 411–425.
  62. Boselli, M.; Lee, B.-H.; Robert, J.; Prado, M.A.; Min, S.-W.; Cheng, C.; Silva, M.C.; Seong, C.; Elsasser, S.; Hatle, K.M.; et al. An inhibitor of the proteasomal deubiquitinating enzyme USP14 induces tau elimination in cultured neurons. J. Biol. Chem. 2017, 292, 19209–19225.
  63. 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.
  64. Chakraborty, J.; Von Stockum, S.; Marchesan, E.; Caicci, F.; Ferrari, V.; Rakovic, A.; Klein, C.; Antonini, A.; Bubacco, L.; Ziviani, E. USP14 inhibition corrects an in vivo model of impaired mitophagy. EMBO Mol. Med. 2018, 10, 11.
  65. Wu, N.; Liu, C.; Bai, C.; Han, Y.P.; Cho, W.C.S.; Li, Q. Over-Expression of Deubiquitinating Enzyme USP14 in Lung Adenocarcinoma Promotes Proliferation through the Accumulation of beta-Catenin. Int. J. Mol. Sci. 2013, 14, 10749–10760.
  66. Wang, Y.; Wang, J.; Zhong, J.; Deng, Y.; Xi, Q.; He, S.; Yang, S.; Jiang, L.; Huang, M.; Tang, C.; et al. Ubiquitin-specific protease 14 (USP14) regulates cellular proliferation and apoptosis in epithelial ovarian cancer. Med. Oncol. 2015, 32, 1–10.
  67. Zhu, Y.; Zhang, C.; Gu, C.; Li, Q.; Wu, N. Function of Deubiquitinating Enzyme USP14 as Oncogene in Different Types of Cancer. Cell. Physiol. Biochem. 2016, 38, 993–1002.
  68. Zhang, B.; Li, M.; Huang, P.; Guan, X.-Y.; Zhu, Y.-H. Overexpression of ubiquitin specific peptidase 14 predicts unfavorable prognosis in esophageal squamous cell carcinoma. Thorac. Cancer 2017, 8, 344–349.
  69. Hang, C.; Gong, C.; Fang, Y.; Chen, L.; Zhu, J. Ubiquitin-specific protease 14 (USP14) promotes proliferation and metastasis in pancreatic ductal adenocarcinoma. J. Mol. Histol. 2021, 52, 1–10.
  70. Lander, G.C.; Estrin, E.; Matyskiela, M.E.; Bashore, C.; Nogales, E.; Martin, A. Complete subunit architecture of the proteasome regulatory particle. Nat. Cell Biol. 2012, 482, 186–191.
  71. Chen, S.; Wu, J.; Lu, Y.; Ma, Y.-B.; Lee, B.-H.; Yu, Z.; Ouyang, Q.; Finley, D.J.; Kirschner, M.W.; Mao, Y. Structural basis for dynamic regulation of the human 26S proteasome. Proc. Natl. Acad. Sci. USA 2016, 113, 12991–12996.
  72. de la Peña, A.H.; Goodall, E.A.; Gates, S.N.; Lander, G.C.; Martin, A. Substrate-engaged 26S proteasome structures reveal mechanisms for ATP-hydrolysis–driven translocation. Science 2018, 362, eaav0725.
  73. Dong, Y.; Zhang, S.; Wu, Z.; Li, X.; Wang, W.L.; Zhu, Y.; Stoilova-McPhie, S.; Lu, Y.; Finley, D.; Mao, Y. Cryo-EM structures and dynamics of substrate-engaged human 26S proteasome. Nat. Cell Biol. 2018, 565, 49–55.
  74. Maytal-Kivity, V.; Reis, N.; Hofmann, K.; Glickman, M.H. MPN+, a putative catalytic motif found in a subset of MPN domain proteins from eukaryotes and prokaryotes, is critical for Rpn11 function. BMC Biochem. 2002, 3, 28.
  75. Lundgren, J.; Masson, P.; Realini, C.A.; Young, P. Use of RNA Interference and Complementation to Study the Function of the Drosophila and Human 26S Proteasome Subunit S13. Mol. Cell. Biol. 2003, 23, 5320–5330.
  76. Gallery, M.; Blank, J.L.; Lin, Y.; Gutierrez, J.A.; Pulido, J.C.; Rappoli, D.; Badola, S.; Rolfe, M.; Macbeth, K.J. The JAMM motif of human deubiquitinase Poh1 is essential for cell viability. Mol. Cancer Ther. 2007, 6, 262–268.
  77. Deshaies, R.J. Proteotoxic crisis, the ubiquitin-proteasome system, and cancer therapy. BMC Biol. 2014, 12, 94.
  78. Wang, B.; Ma, A.; Zhang, L.; Jin, W.-L.; Qiang, X.; Xu, G.; Qiu, B.; Yang, Z.; Liu, Y.; Xia, Q.; et al. POH1 deubiquitylates and stabilizes E2F1 to promote tumour formation. Nat. Commun. 2015, 6, 8704.
  79. Song, Y.; Ray, A.; Das, D.S.; Samur, M.K.; Carrasco, R.D.; Munshi, N.C.; Chauhan, D.; Anderson, K.C. Deubiquitylating Enzyme Rpn11/POH1/PSMD14 As Therapeutic Target in Multiple Myeloma. Blood 2016, 128, 4469.
  80. Luo, G.; Hu, N.; Xia, X.; Zhou, J.; Ye, C. RPN11 deubiquitinase promotes proliferation and migration of breast cancer cells. Mol. Med. Rep. 2017, 16, 331–338.
  81. Wang, C.-H.; Lu, S.-X.; Liu, L.-L.; Li, Y.; Yang, X.; He, Y.-F.; Chen, S.-L.; Cai, S.-H.; Wang, H.; Yun, J.-P. POH1 Knockdown Induces Cancer Cell Apoptosis via p53 and Bim. Neoplasia 2018, 20, 411–424.
  82. Wang, B.S.; Xu, X.L.; Yang, Z.J.; Zhang, L.; Liu, Y.; Ma, A.H.; Xu, G.Q.; Tang, M.; Jing, T.T.; Wu, L.; et al. POH1 contributes to hyperactivation of TGF-beta signaling and facilitates hepatocellular carcinoma metastasis through deubiquitinating TGF-beta receptors and caveolin-1. Ebiomedicine 2019, 41, 320–332.
  83. Yu, W.; Li, J.; Wang, Q.; Wang, B.; Zhang, L.; Liu, Y.; Tang, M.; Xu, G.; Yang, Z.; Wang, X.; et al. Targeting POH1 inhibits prostate cancer cell growth and enhances the suppressive efficacy of androgen deprivation and docetaxel. Prostate 2019, 79, 1304–1315.
  84. Hamazaki, J.; Iemura, S.-I.; Natsume, T.; Yashiroda, H.; Tanaka, K.; Murata, S. A novel proteasome interacting protein recruits the deubiquitinating enzyme UCH37 to 26S proteasomes. EMBO J. 2006, 25, 4524–4536.
  85. Qiu, X.B.; Ouyang, S.Y.; Li, C.J.; Miao, S.Y.; Wang, L.F.; Goldberg, A.L. hRpn13/ADRM1/GP110 is a novel proteasome subunit that binds the deubiquitinating enzyme, UCH37. EMBO J 2006, 25, 5742–5753.
  86. Yao, T.T.; Song, L.; Xu, W.; DeMartino, G.N.; Florens, L.; Swanson, S.K.; Washburn, M.P.; Conaway, R.C.; Conaway, J.W.; Cohen, R.E. Proteasome recruitment and activation of the Uch37 deubiquitinating enzyme by Adrm1. Nat. Cell Biol. 2006, 8, 994–1002.
  87. Yao, T.; Song, L.; Jin, J.; Cai, Y.; Takahashi, H.; Swanson, S.K.; Washburn, M.P.; Florens, L.; Conaway, R.C.; Cohen, R.E.; et al. Distinct Modes of Regulation of the Uch37 Deubiquitinating Enzyme in the Proteasome and in the Ino80 Chromatin-Remodeling Complex. Mol. Cell 2008, 31, 909–917.
  88. Sahtoe, D.D.; Van Dijk, W.J.; El Oualid, F.; Ekkebus, R.; Ovaa, H.; Sixma, T.K. Mechanism of UCH-L5 Activation and Inhibition by DEUBAD Domains in RPN13 and INO80G. Mol. Cell 2015, 57, 887–900.
  89. VanderLinden, R.T.; Hemmis, C.W.; Schmitt, B.; Ndoja, A.; Whitby, F.G.; Robinson, H.; Cohen, R.E.; Yao, T.; Hill, C.P. Structural Basis for the Activation and Inhibition of the UCH37 Deubiquitylase. Mol. Cell 2015, 57, 901–911.
  90. Zhang, N.Y.; Jacobson, A.D.; Macfadden, A.; Liu, C.W. Ubiquitin chain trimming recycles the substrate binding sites of the 26 S proteasome and promotes degradation of lysine 48-linked polyubiquitin conjugates. J. Biol. Chem. 2011, 286, 25540–25546.
  91. Chen, Y.; Fu, D.; Xi, J.; Ji, Z.; Liu, T.; Ma, Y.; Zhao, Y.; Dong, L.; Wang, Q.; Shen, X. Expression and Clinical Significance of UCH37 in Human Esophageal Squamous Cell Carcinoma. Dig. Dis. Sci. 2012, 57, 2310–2317.
  92. Fang, Y.; Fu, D.; Tang, W.; Cai, Y.; Ma, D.; Wang, H.; Xue, R.; Liu, T.; Huang, X.; Dong, L.; et al. Ubiquitin C-terminal Hydrolase 37, a novel predictor for hepatocellular carcinoma recurrence, promotes cell migration and invasion via interacting and deubiquitinating PRP19. Biochim. Biophys. Acta (BBA) Bioenerg. 2013, 1833, 559–572.
  93. Wang, L.; Chen, Y.-J.; Xu, K.; Wang, Y.-Y.; Shen, X.-Z.; Tu, R.-Q. High expression of UCH37 is significantly associated with poor prognosis in human epithelial ovarian cancer. Tumor Biol. 2014, 35, 11427–11433.
  94. Fang, Y.; Shen, X. Ubiquitin carboxyl-terminal hydrolases: Involvement in cancer progression and clinical implications. Cancer Metastasis Rev. 2017, 36, 669–682.
  95. Liu, D.; Song, Z.X.; Wang, X.Y.; Ouyang, L. Ubiquitin C-Terminal Hydrolase L5 (UCHL5) Accelerates the Growth of Endometrial Cancer via Activating the Wnt/beta-Catenin Signaling Pathway. Front. Oncol. 2020, 10, 865.
  96. Zhang, J.; Xu, H.; Yang, X.; Zhao, Y.; Xu, X.; Zhang, L.; Xuan, X.; Ma, C.; Qian, W.; Li, D. Deubiquitinase UCHL5 is elevated and associated with a poor clinical outcome in lung adenocarcinoma (LUAD). J. Cancer 2020, 11, 6675–6685.
  97. Lee, B.; Finley, D.; King, R. A High-Throughput Screening Method for Identification of Inhibitors of the Deubiquitinating Enzyme USP14. Curr. Protoc. Chem. Biol. 2012, 4, 311–330.
  98. Wang, Y.; Jiang, Y.; Ding, S.; Li, J.; Song, N.; Ren, Y.; Hong, D.; Wu, C.; Li, B.; Wang, F.; et al. Small molecule inhibitors reveal allosteric regulation of USP14 via steric blockade. Cell Res. 2018, 28, 1186–1194.
  99. Palmer, A.L.; De Jong, A.; Leestemaker, Y.; Geurink, P.P.; Wijdeven, R.H.; Ovaa, H.; Dolan, B.P. Inhibition of the Deubiquitinase Usp14 Diminishes Direct MHC Class I Antigen Presentation. J. Immunol. 2018, 200, 928–936.
  100. Cullen, M.; Hauck, S.; Geng, B.; Bastos, C.M.; Munoz, B.; Haeberlein, M. Proteasome Activity Enhancing Compounds. WO/2015/073528, 21 May 2015.
  101. Cullen, M.; Hauck, S.; Foley, M.; Tait, B.; Haeberlein, M. Proteasome Activity Enhancing Compounds. WO/2020/006269, 2 January 2020.
  102. Cullen, M.; Bastos, C.M.; Parks, D.; Munoz, B. Proteasome Activity Enhancing Compounds. WO/2020/006296, 2 January 2020.
  103. Finley, D.; King, R.W.; Lee, B.-H.; Lee, M.J.; Gahman, T.C. Tricyclic Proteasome Activity Enhancing Compounds. WO/2012/012712, 26 January 2012.
  104. Kemp, M. Recent Advances in the Discovery of Deubiquitinating Enzyme Inhibitors. Prog. Med. Chem. 2016, 55, 149–192.
  105. Chambers, R.J.; Foley, M.; Tait, B. Proteasome Activity Modulating Compounds. WO/2013/112651, 1 August 2013.
  106. Li, J.; Yakushi, T.; Parlati, F.; MacKinnon, A.L.; Perez, C.; Ma, Y.; Carter, K.P.; Colayco, S.; Magnuson, G.; Brown, B.; et al. Capzimin is a potent and specific inhibitor of proteasome isopeptidase Rpn11. Nat. Chem. Biol. 2017, 13, 486–493.
  107. Perez, C.; Li, J.; Parlati, F.; Rouffet, M.; Ma, Y.; MacKinnon, A.L.; Chou, T.-F.; Deshaies, R.J.; Cohen, S.M.; Parlati, F.; et al. Discovery of an Inhibitor of the Proteasome Subunit Rpn11. J. Med. Chem. 2017, 60, 1343–1361.
  108. Lauinger, L.; Li, J.; Shostak, A.; Cemel, I.A.; Ha, N.; Zhang, Y.; E Merkl, P.; Obermeyer, S.; Stankovic-Valentin, N.; Schafmeier, T.; et al. Thiolutin is a zinc chelator that inhibits the Rpn11 and other JAMM metalloproteases. Nat. Chem. Biol. 2017, 13, 709–714.
  109. Li, J.; Zhang, Y.; Santos, B.D.S.S.D.; Wang, F.; Ma, Y.; Perez, C.; Yang, Y.; Peng, J.; Cohen, S.M.; Chou, T.-F.; et al. Epidithiodiketopiperazines Inhibit Protein Degradation by Targeting Proteasome Deubiquitinase Rpn11. Cell Chem. Biol. 2018, 25, 1350–1358.e9.
  110. D’Arcy, P.; Brnjic, S.; Olofsson, M.H.; Fryknäs, M.; Lindsten, K.; De Cesare, M.; Perego, P.; Sadeghi, B.; Hassan, M.; Larsson, R.; et al. Inhibition of proteasome deubiquitinating activity as a new cancer therapy. Nat. Med. 2011, 17, 1636–1640.
  111. Wang, X.; D’Arcy, P.; Caulfield, T.R.; Paulus, A.; Chitta, K.; Mohanty, C.; Gullbo, J.; Chanan-Khan, A.; Linder, S. Synthesis and Evaluation of Derivatives of the Proteasome Deubiquitinase Inhibitor b-AP15. Chem. Biol. Drug Des. 2015, 86, 1036–1048.
  112. Kapuria, V.; Peterson, L.F.; Fang, D.; Bornmann, W.G.; Talpaz, M.; Donato, N.J. Deubiquitinase Inhibition by Small-Molecule WP1130 Triggers Aggresome Formation and Tumor Cell Apoptosis. Cancer Res. 2010, 70, 9265–9276.
  113. Zhou, B.; Zuo, Y.; Li, B.; Wang, H.; Liu, H.; Wang, X.; Qiu, X.; Hu, Y.; Wen, S.; Du, J.; et al. Deubiquitinase inhibition of 19S regulatory particles by 4-arylidene curcumin analog AC17 causes NF-kappaB inhibition and p53 reactivation in human lung cancer cells. Mol. Cancer Ther. 2013, 12, 1381–1392.
  114. Liu, N.; Li, X.; Huang, H.; Zhao, C.; Liao, S.; Yang, C.; Liu, S.; Song, W.; Lu, X.; Lan, X.; et al. Clinically used antirheumatic agent auranofin is a proteasomal deubiquitinase inhibitor and inhibits tumor growth. Oncotarget 2014, 5, 5453–5471.
  115. Stafford, W.C.; Peng, X.; Olofsson, M.H.; Zhang, X.; Luci, D.K.; Lu, L.; Cheng, Q.; Trésaugues, L.; Dexheimer, T.S.; Coussens, N.P.; et al. Irreversible inhibition of cytosolic thioredoxin reductase 1 as a mechanistic basis for anticancer therapy. Sci. Transl. Med. 2018, 10, eaaf7444.
  116. Zhang, X.; Selvaraju, K.; Saei, A.A.; D’Arcy, P.; Zubarev, R.; Arnér, E.S.; Linder, S. Repurposing of auranofin: Thioredoxin reductase remains a primary target of the drug. Biochimie 2019, 162, 46–54.
  117. Erdal, H.; Berndtsson, M.; Castro, J.; Brunk, U.; Shoshan, M.C.; Linder, S. Induction of lysosomal membrane permeabilization by compounds that activate p53-independent apoptosis. Proc. Natl. Acad. Sci. USA 2005, 102, 192–197.
  118. Berndtsson, M.; Beaujouin, M.; Rickardson, L.; Havelka, A.M.; Larsson, R.; Westman, J.; Liaudet-Coopman, E.; Linder, S. Induction of the lysosomal apoptosis pathway by inhibitors of the ubiquitin-proteasome system. Int. J. Cancer 2009, 124, 1463–1469.
  119. Wang, X.; Stafford, W.; Mazurkiewicz, M.; Fryknäs, M.; Brjnic, S.; Zhang, X.; Gullbo, J.; Larsson, R.; Arnér, E.S.J.; D’Arcy, P.; et al. The 19S Deubiquitinase Inhibitor b-AP15 Is Enriched in Cells and Elicits Rapid Commitment to Cell Death. Mol. Pharmacol. 2014, 85, 932–945.
  120. Wang, X.; Mazurkiewicz, M.; Hillert, E.K.; Olofsson, M.H.; Pierrou, S.; Hillertz, P.; Gullbo, J.; Selvaraju, K.; Paulus, A.; Akhtar, S.; et al. The proteasome deubiquitinase inhibitor VLX1570 shows selectivity for ubiquitin-specific protease-14 and induces apoptosis of multiple myeloma cells. Sci. Rep. 2016, 6, 1–15.
More
Information
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: 391
Revisions: 2 times (View History)
Update Date: 23 Jun 2021
1000/1000