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 -- 2178 2022-05-17 20:13:23 |
2 format correct Meta information modification 2178 2022-05-18 04:34:21 |

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.
Caba, C.; , . Small Molecule Deubiquitinase Enzymes Inhibitors. Encyclopedia. Available online: https://encyclopedia.pub/entry/23024 (accessed on 29 March 2024).
Caba C,  . Small Molecule Deubiquitinase Enzymes Inhibitors. Encyclopedia. Available at: https://encyclopedia.pub/entry/23024. Accessed March 29, 2024.
Caba, Cody, . "Small Molecule Deubiquitinase Enzymes Inhibitors" Encyclopedia, https://encyclopedia.pub/entry/23024 (accessed March 29, 2024).
Caba, C., & , . (2022, May 17). Small Molecule Deubiquitinase Enzymes Inhibitors. In Encyclopedia. https://encyclopedia.pub/entry/23024
Caba, Cody and . "Small Molecule Deubiquitinase Enzymes Inhibitors." Encyclopedia. Web. 17 May, 2022.
Small Molecule Deubiquitinase Enzymes Inhibitors
Edit

Ubiquitination is reversed by the activity of deubiquitinase enzymes (DUBs). About 100 human DUBs are known, and they are divided into seven major families: the cysteine proteases of the USP (ubiquitin-specific proteases), UCH (ubiquitin C-terminal hydrolases), OTU (ovarian tumor), MJD (Machado-Joseph domain-containing proteases), MINDY (motif interacting with the Ub-containing novel DUB family), and ZUFSP (zinc finger with the UFM1-specific peptidase domain protein) families and the Zn-dependent metalloproteases of the JAMM (JAB1/MPN/MOV34 domain-associated) family. DUBs play a role in seemingly every biological process and are central to many human pathologies, thus rendering them very desirable and challenging therapeutic targets. Despite significant drug discovery efforts, only approximately 15 chemical probe-quality small molecule inhibitors have been reported, hitting just 6 of about 100 known DUBs.

deubiquitinase (DUB) ubiquitin (Ub) inhibitor

1. Introduction

Over 40 years ago, the seminal works of Avram Hershko, Aaron Ciechanover, and Irwin Rose (and, later, Alexander Varshavsky) engendered the ubiquitin (Ub) field as we know it today. A first-hand account of these early years, for which the 2004 Nobel Prize in Chemistry was awarded, has been delineated by Varshavsky [1]. It is now well-understood that ubiquitination is a tightly regulated and highly specific post-translational modification (PTM) involved in seemingly all biological processes. Covalent attachment of the small 76-amino acid Ub to a protein’s lysine ε-amine or N-terminal α-amine, via an iso- or α-peptide bond, respectively, is diverse in consequences. Proteasome-targeted degradation is the canonical role of this modification, but countless studies have shed light on its criticality for signal transduction, trafficking, gene expression, and other cellular processes [2].
The complexity of molecular outcomes as a result of ubiquitination presents a ubiquitin code [3][4]. Since Ub itself can be ubiquitinated at any of its seven lysine residues (K6, K11, K27, K29, K33, K48, and K63), as well as its N-terminus (M1), in theory, this modification is architecturally limitless. Adding another layer of complexity is the fact that Ub can also be post-translationally modified by phosphorylation, as in the case of phospho-Ser-65 during mitophagy [5][6][7][8][9] or the phospho-Thr-12 for DNA damage response [10], lysine acetylation for inhibiting Ub chain elongation [11], and by Ub-like modifiers (Ubls), for which the physiological significance is not well-known [2][12]. The encoding and decoding of the ubiquitin code have been an intense subject of research.

1.1. Deubiquitinase Enzymes: Classification and Activity Regulation

Ubiquitination is reversed by the activity of deubiquitinase enzymes (DUBs) (Figure 1). About 100 human DUBs are known, and they are divided into seven major families: the cysteine proteases of the USP (ubiquitin-specific proteases), UCH (ubiquitin C-terminal hydrolases), OTU (ovarian tumor), MJD (Machado-Joseph domain-containing proteases), MINDY (motif interacting with the Ub-containing novel DUB family), and ZUFSP (zinc finger with the UFM1-specific peptidase domain protein) families and the Zn-dependent metalloproteases of the JAMM (JAB1/MPN/MOV34 domain-associated) family [4][13][14][15][16]. The USP family is the largest, with 58 constituents. The members of each family are related through structurally homologous catalytic domains but can vary greatly by the presence of insertions, deletions, and additional domains serving diverse functions. Cysteine protease DUBs utilize a catalytic triad or dyad to facilitate a nucleophilic attack and hydrolysis of the scissile bond connecting the C-terminal Gly-76 of Ub to a substrate or other Ub of a poly-Ub chain. Contrarily, zinc-dependent metalloprotease DUBs (metalloDUBs) utilize a coordinated zinc ion and an activated, nucleophilic water molecule for catalysis [4].
DUBs are essential for maintaining Ub homeostasis by processing Ub precursors (UBC, UBB, UBA52, and UBA80) [17] and unconjugated chains [14]; recycling Ub at the proteasome [18]; and deubiquitinating various targets. Additionally, they function as interpreters and editors of a highly complex code. E3-DUB complexes are common and couple conjugation and deconjugation to facilitate chain editing and fine-tuning of the molecular output [19]. In the rare case of A20 (an OTU family member), both E3 and DUB activities are encoded into a single multifunctional enzyme [20].
The factors dictating DUB activity and substrate specificity include the structure of the catalytic domain, the presence of additional domains for substrate recruitment or Ub binding [21][22][23], alternative splicing [24], allostery [25], oligomerization [26], PTMs [27], and subcellular localization [4]. Importantly, not all DUBs are catalytically active. Approximately 10% are pseudo-enzymes currently found in the USP, OTU, JAMM, and MINDY families [28]. Pseudo-DUBs are critical components of large macromolecular complexes or as allosteric modulators of active DUBs. Most famously, PSMD7, a pseudo-DUB, dimerizes with the active DUB PSMD14. This complex is a critical component of the 19S proteasome regulatory particle [29][30][31]. Without detectable activity, pseudo-DUBs are most readily identified by sequence and structural similarity to catalytically competent homologs.
DUBs can discriminate the linkage type, chain length, modifier (Ub or Ubl), and/or the substrate to which Ub is conjugated [32]. Furthermore, they can remove poly-Ub chains in different modes: iteratively at the distal end (exo-cleavage), from within a chain (endo-cleavage), or all at once (en bloc-cleavage) [33]. Members of the USP family are generally linkage-nonspecific and often referred to as promiscuous [21][34][35]; however, using tool reagents, including small molecule compounds and activity-based probes, a group and others have revealed surprising linkage-dependent processing activities for USP9X [36] and USP7 [37], two USPs previously believed to be nonspecific. Whether or not other members of the USP family have a similar specificity remains to be explored. Contrarily, members of the OTU, MINDY, ZUFSP, and JAMM families tend to be linkage-specific [4][14][22][35][38][39][40][41][42]. For OTUD1, a C-terminal Ub-interacting motif (UIM) endows specificity for K63 linkages by forming a proximal Ub-binding site (S1’) to orient the scissile bond across the catalytic site [22]. The structural determinants of the linkage specificity can also be leveraged for chain length specificity. The five Ub-binding sites of the catalytic domain of MINDY1/2 render strict K48 linkage specificity and the ability to sense the poly-Ub chain length to modulate the exo- (<6 Ub) and endo-cleavage (>5 Ub) modes [43]. UCH DUBs preferentially cleave Ub from small or unstructured C-terminal leaving groups. This represents a sort-of substrate specificity that results from an active site crossover loop limiting the size of ubiquitinated substrates able to occupy the catalytic cleft [44].
Figure 1. Ub conjugation and deconjugation. Ubiquitination is an ATP-dependent process orchestrated by an E1–E2–E3 enzymatic cascade utilizing free, mono-Ub. The N-terminus or any of the seven lysine residues of Ub can be ubiquitinated to facilitate poly-Ub chain assembly following the sequential rounds of conjugation. The removal or deconjugation of Ub is then mediated by DUBs, for which seven families have been classified. The structure of the DUB catalytic domain of a representative family member is shown: USP, USP7 pdb 4m5w [45]; UCH, UCH-L3 pdb 1uch [46]; OTU, OTUD1 pdb 4bop [22]; MJD, Jos-2 pdb 6pgv [47]; MINDY, MINDY-1 pdb 5jqs [38]; ZUFSP, ZUP1 pdb 6ei1 [48]; JAMM, STAMBPL1 pdb 7l97 [49]. The most well-known member of each family is presented based on the number of publications curated from the NCBI Entrez database. * The reader should be aware that reported Ub chain specificities are assay-dependent, and many exceptions exist.

1.2. Challenges and Missteps When Studying DUBs at the Cellular Level

Since ubiquitination is involved in almost all biological processes, including protein homeostasis, transcription regulation, DNA repair, endocytosis and endolysosomal sorting, autophagy, immune response, and stem cell renewal, unsurprisingly, DUBs are also major players in these diverse cellular functions. Therefore, their dysregulation is associated with many diseases. This has been the subject of multiple recent reviews, either about their general functions [14][15][50] or on specific disease areas such as neuronal [51], cardiovascular [52], developmental [53], autoimmune [54] diseases, and cancers [55][56] and is beyond the scope of current content. Instead, these content highlight the importance of using quality tool reagents when exploring the biology of DUBs at the cellular level.
Due to the scarcity of quality DUB inhibitors, studies often use unvalidated, weak, or semi-selective inhibitors when probing for cellular functions. These practices can lead to spurious conclusions and add to the reproducibility crisis [57][58]. For example, compounds WP1130 and G9 have been used to examine the biological roles of USP9X, a large, 250-kDa USP family DUB. Studies using these compounds have claimed to elucidate numerous interactors and functions [59]; however, many of these interactors (for example, E3 ligases ITCH [60] and FBXW7 [61]; Halabelian and Tong unpublished data) and functions (Michael Clague personal communication) cannot be validated. One inhibitor, G9, which cross-reacts with USP5 and USP24 [62], has been used by many separate studies to assess the functions of USP9X [63][64][65], USP5 [66][67], or USP24 [68]. It is difficult to exclude the possibility of off-target effects, crosstalk, or functional compensation between these DUBs. On the other hand, FT709, a probe-quality inhibitor with a nanomolar affinity for USP9X, has provided much clearer evidence of its involvement in ribosomal stalling and confirmed its interaction with two other E3 ligases, ZNF598 and MKRN2 [69]. Another example is spautin-1 [70], which is widely used as a specific autophagy inhibitor for its inhibition of both USP10 and USP13. These two DUBs have very different structures. Whereas USP10 has a typical USP fold without insertions, USP13 has two ubiquitin-associated (UBA) domains inserted in the catalytic domain. USP13 is closer in sequence and domain architecture to USP5 than USP10. No orthogonal biophysical methods, such as the surface plasmon response (SPR) or isothermal titration calorimetry (ITC), were used to validate the binding of spautin-1 with USP10 or USP13.

2. Small Molecule DUB Inhibitors

Despite a decade of intensive investment from industry and academic labs, most reported DUB inhibitors have a low binding affinity in the micromolar range and lack selectivity. This was demonstrated by a comprehensive profiling study using the MALDI-TOF mass spectrometry of 11 inhibitors against 42 DUBs [34] and by Medivir’s DUB platform [71]. ML323, an inhibitor of the USP1/UAF1 complex, was developed by Zhuang Lab [72][73] and stood to be the only USP family DUB inhibitor that met the chemical probe criteria [74] by 2016. However, the exact structural details on how ML323 binds to the USP1/UAF1 complex remained elusive, which prevented further structure-based optimization of the inhibitor. When several groups developed a series of high-quality, structurally defined USP7 inhibitors, the efforts in DUB inhibitor discovery were reinvigorated. The progress on USP7 inhibitors was nicely summarized in Pozhidaeva and Bezsonova’s review [75]. The demonstration of the druggability of USPs and the identification of structurally defined DUB inhibitors opened many opportunities to pharmacologically interfere with their functions in vivo and to explore their biology in disease states. The advances have been the subject of several recent excellent reviews [76][77][78][79] and will not be repeated here. 
In the last several years, the chemical biology community has been advocating a stringent evaluation of chemical tool reagents for the study of the biology of protein targets [76][80][81]. These criteria include <100 nM in vitro potency, >30-fold selectivity against other members of the same protein family, off-target profiling, and cellular on-target effects at <1 ¦ÌM. A combination of orthogonal validation methods to confirm target engagement is essential, especially considering the various shortcomings of some popular inhibitors (discussed above).
Having kept track of the high-quality chemical inhibitors of DUBs using a publicly accessible repository called UbiHub [82]. So far, only four USPs (USP7, USP1, USP9X, and USP30); one UCH (UCHL1); and one JAMM family DUB (CSN5) have chemical probe-quality small molecule inhibitors (Table 1). However, it is noteworthy that some of these are covalent inhibitors, such as FT385 for USP30 [83] and IMP–1710 for UCHL1 [84][85], which both utilize a cyanopyrrolidine warhead for modifying the catalytic cysteine residue. Despite their selectivity over other DUBs, they may have off-target reactivity towards other unrelated enzymes, such as dehydrogenases, as demonstrated for the UCHL1 inhibitor MT16-205 [84]. Thus, extra care must be exercised when using these inhibitors to probe the biology of the intended DUB targets. The best practice for small molecule inhibitors in studying the biology of protein targets is well-established [86]. Readers are encouraged to follow the do’s and don’ts when using chemical probes to obtain credible results.
Table 1. Potent small molecule DUB inhibitors.

References

  1. Varshavsky, A. The Early History of the Ubiquitin Field. Protein Sci. Publ. Protein Soc. 2006, 15, 647–654.
  2. Swatek, K.N.; Komander, D. Ubiquitin Modifications. Cell Res. 2016, 26, 399–422.
  3. Komander, D.; Rape, M. The Ubiquitin Code. Annu. Rev. Biochem. 2012, 81, 203–229.
  4. Mevissen, T.E.T.; Komander, D. Mechanisms of Deubiquitinase Specificity and Regulation. Annu. Rev. Biochem. 2017, 86, 159–192.
  5. Koyano, F.; Okatsu, K.; Kosako, H.; Tamura, Y.; Go, E.; Kimura, M.; Kimura, Y.; Tsuchiya, H.; Yoshihara, H.; Hirokawa, T.; et al. Ubiquitin Is Phosphorylated by PINK1 to Activate Parkin. Nature 2014, 510, 162–166.
  6. Okatsu, K.; Sato, Y.; Yamano, K.; Matsuda, N.; Negishi, L.; Takahashi, A.; Yamagata, A.; Goto-Ito, S.; Mishima, M.; Ito, Y.; et al. Structural Insights into Ubiquitin Phosphorylation by PINK1. Sci. Rep. 2018, 8, 10382.
  7. Ordureau, A.; Heo, J.-M.; Duda, D.M.; Paulo, J.A.; Olszewski, J.L.; Yanishevski, D.; Rinehart, J.; Schulman, B.A.; Harper, J.W. Defining Roles of PARKIN and Ubiquitin Phosphorylation by PINK1 in Mitochondrial Quality Control Using a Ubiquitin Replacement Strategy. Proc. Natl. Acad. Sci. USA 2015, 112, 6637–6642.
  8. Wauer, T.; Swatek, K.N.; Wagstaff, J.L.; Gladkova, C.; Pruneda, J.N.; Michel, M.A.; Gersch, M.; Johnson, C.M.; Freund, S.M.; Komander, D. Ubiquitin Ser65 Phosphorylation Affects Ubiquitin Structure, Chain Assembly and Hydrolysis. EMBO J. 2015, 34, 307–325.
  9. Hepowit, N.L.; Kolbe, C.-C.; Zelle, S.R.; Latz, E.; MacGurn, J.A. Regulation of Ubiquitin and Ubiquitin-like Modifiers by Phosphorylation. FEBS J. 2021.
  10. Walser, F.; Mulder, M.P.C.; Bragantini, B.; Burger, S.; Gubser, T.; Gatti, M.; Botuyan, M.V.; Villa, A.; Altmeyer, M.; Neri, D.; et al. Ubiquitin Phosphorylation at Thr12 Modulates the DNA Damage Response. Mol. Cell 2020, 80, 423–436.e9.
  11. Ohtake, F.; Saeki, Y.; Sakamoto, K.; Ohtake, K.; Nishikawa, H.; Tsuchiya, H.; Ohta, T.; Tanaka, K.; Kanno, J. Ubiquitin Acetylation Inhibits Polyubiquitin Chain Elongation. EMBO Rep. 2015, 16, 192–201.
  12. Pérez Berrocal, D.A.; Witting, K.F.; Ovaa, H.; Mulder, M.P.C. Hybrid Chains: A Collaboration of Ubiquitin and Ubiquitin-Like Modifiers Introducing Cross-Functionality to the Ubiquitin Code. Front. Chem. 2020, 7, 931.
  13. Clague, M.J.; Heride, C.; Urbé, S. The Demographics of the Ubiquitin System. Trends Cell Biol. 2015, 25, 417–426.
  14. Clague, M.J.; Urbé, S.; Komander, D. Breaking the Chains: Deubiquitylating Enzyme Specificity Begets Function. Nat. Rev. Mol. Cell Biol. 2019, 20, 338–352.
  15. Clague, M.J.; Barsukov, I.; Coulson, J.M.; Liu, H.; Rigden, D.J.; Urbé, S. Deubiquitylases from Genes to Organism. Physiol. Rev. 2013, 93, 1289–1315.
  16. Vlasschaert, C.; Cook, D.; Xia, X.; Gray, D.A. The Evolution and Functional Diversification of the Deubiquitinating Enzyme Superfamily. Genome Biol. Evol. 2017, 9, 558–573.
  17. Grou, C.P.; Pinto, M.P.; Mendes, A.V.; Domingues, P.; Azevedo, J.E. The de Novo Synthesis of Ubiquitin: Identification of Deubiquitinases Acting on Ubiquitin Precursors. Sci. Rep. 2015, 5, 12836.
  18. de Poot, S.A.H.; Tian, G.; Finley, D. Meddling with Fate: The Proteasomal Deubiquitinating Enzymes. J. Mol. Biol. 2017, 429, 3525–3545.
  19. Nielsen, C.P.; MacGurn, J.A. Coupling Conjugation and Deconjugation Activities to Achieve Cellular Ubiquitin Dynamics. Trends Biochem. Sci. 2020, 45, 427–439.
  20. Wertz, I.E.; O’Rourke, K.M.; Zhou, H.; Eby, M.; Aravind, L.; Seshagiri, S.; Wu, P.; Wiesmann, C.; Baker, R.; Boone, D.L.; et al. De-Ubiquitination and Ubiquitin Ligase Domains of A20 Downregulate NF-ΚB Signalling. Nature 2004, 430, 694–699.
  21. Faesen, A.C.; Luna-Vargas, M.P.A.; 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.
  22. Mevissen, T.E.T.; Hospenthal, M.K.; Geurink, P.P.; Elliott, P.R.; Akutsu, M.; Arnaudo, N.; Ekkebus, R.; Kulathu, Y.; Wauer, T.; El Oualid, F.; et al. OTU Deubiquitinases Reveal Mechanisms of Linkage Specificity and Enable Ubiquitin Chain Restriction Analysis. Cell 2013, 154, 169–184.
  23. Sowa, M.E.; Bennett, E.J.; Gygi, S.P.; Harper, J.W. Defining the Human Deubiquitinating Enzyme Interaction Landscape. Cell 2009, 138, 389–403.
  24. Leznicki, P.; Natarajan, J.; Bader, G.; Spevak, W.; Schlattl, A.; Rehman, S.A.A.; Pathak, D.; Weidlich, S.; Zoephel, A.; Bordone, M.C.; et al. Expansion of DUB Functionality Generated by Alternative Isoforms-USP35, a Case Study. J. Cell Sci. 2018, 131, jcs212753.
  25. Reyes-Turcu, F.E.; Ventii, K.H.; Wilkinson, K.D. Regulation and Cellular Roles of Ubiquitin-Specific Deubiquitinating Enzymes. Annu. Rev. Biochem. 2009, 78, 363–397.
  26. Gersch, M.; Wagstaff, J.L.; Toms, A.V.; Graves, B.; Freund, S.M.V.; Komander, D. Distinct USP25 and USP28 Oligomerization States Regulate Deubiquitinating Activity. Mol. Cell 2019, 74, 436–451.e7.
  27. Sanchez-Bailon, M.P.; Choi, S.-Y.; Dufficy, E.R.; Sharma, K.; McNee, G.S.; Gunnell, E.; Chiang, K.; Sahay, D.; Maslen, S.; Stewart, G.S.; et al. Arginine Methylation and Ubiquitylation Crosstalk Controls DNA End-Resection and Homologous Recombination Repair. Nat. Commun. 2021, 12, 6313.
  28. Walden, M.; Masandi, S.K.; Pawłowski, K.; Zeqiraj, E. Pseudo-DUBs as Allosteric Activators and Molecular Scaffolds of Protein Complexes. Biochem. Soc. Trans. 2018, 46, 453–466.
  29. Yao, T.; Cohen, R.E. A Cryptic Protease Couples Deubiquitination and Degradation by the Proteasome. Nature 2002, 419, 403–407.
  30. Pathare, G.R.; Nagy, I.; Śledź, P.; Anderson, D.J.; Zhou, H.-J.; Pardon, E.; Steyaert, J.; Förster, F.; Bracher, A.; Baumeister, W. Crystal Structure of the Proteasomal Deubiquitylation Module Rpn8-Rpn11. Proc. Natl. Acad. Sci. USA 2014, 111, 2984–2989.
  31. Worden, E.J.; Padovani, C.; Martin, A. Structure of the Rpn11–Rpn8 Dimer Reveals Mechanisms of Substrate Deubiquitination during Proteasomal Degradation. Nat. Struct. Mol. Biol. 2014, 21, 220–227.
  32. Ronau, J.A.; Beckmann, J.F.; Hochstrasser, M. Substrate Specificity of the Ubiquitin and Ubl Proteases. Cell Res. 2016, 26, 441–456.
  33. Komander, D.; Clague, M.J.; Urbé, S. Breaking the Chains: Structure and Function of the Deubiquitinases. Nat. Rev. Mol. Cell Biol. 2009, 10, 550–563.
  34. 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.
  35. Takahashi, H.; Yamanaka, S.; Kuwada, S.; Higaki, K.; Kido, K.; Sato, Y.; Fukai, S.; Tokunaga, F.; Sawasaki, T. A Human DUB Protein Array for Clarification of Linkage Specificity of Polyubiquitin Chain and Application to Evaluation of Its Inhibitors. Biomedicines 2020, 8, 152.
  36. Paudel, P.; Zhang, Q.; Leung, C.; Greenberg, H.C.; Guo, Y.; Chern, Y.-H.; Dong, A.; Li, Y.; Vedadi, M.; Zhuang, Z.; et al. Crystal Structure and Activity-Based Labeling Reveal the Mechanisms for Linkage-Specific Substrate Recognition by Deubiquitinase USP9X. Proc. Natl. Acad. Sci. USA 2019, 116, 7288–7297.
  37. Kategaya, L.; Di Lello, P.; Rougé, 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.
  38. Abdul Rehman, S.A.; Kristariyanto, Y.A.; Choi, S.-Y.; Nkosi, P.J.; Weidlich, S.; Labib, K.; Hofmann, K.; Kulathu, Y. MINDY-1 Is a Member of an Evolutionarily Conserved and Structurally Distinct New Family of Deubiquitinating Enzymes. Mol. Cell 2016, 63, 146–155.
  39. Kwasna, D.; Abdul Rehman, S.A.; Natarajan, J.; Matthews, S.; Madden, R.; De Cesare, V.; Weidlich, S.; Virdee, S.; Ahel, I.; Gibbs-Seymour, I.; et al. Discovery and Characterization of ZUFSP/ZUP1, a Distinct Deubiquitinase Class Important for Genome Stability. Mol. Cell 2018, 70, 150–164.e6.
  40. Komander, D.; Reyes-Turcu, F.; Licchesi, J.D.F.; Odenwaelder, P.; Wilkinson, K.D.; Barford, D. Molecular Discrimination of Structurally Equivalent Lys 63-Linked and Linear Polyubiquitin Chains. EMBO Rep. 2009, 10, 466–473.
  41. McCullough, J.; Row, P.E.; Lorenzo, Ó.; Doherty, M.; Beynon, R.; Clague, M.J.; Urbé, S. Activation of the Endosome-Associated Ubiquitin Isopeptidase AMSH by STAM, a Component of the Multivesicular Body-Sorting Machinery. Curr. Biol. 2006, 16, 160–165.
  42. Hermanns, T.; Pichlo, C.; Baumann, U.; Hofmann, K. A Structural Basis for the Diverse Linkage Specificities within the ZUFSP Deubiquitinase Family. Nat. Commun. 2022, 13, 401.
  43. Abdul Rehman, S.A.; Armstrong, L.A.; Lange, S.M.; Kristariyanto, Y.A.; Gräwert, T.W.; Knebel, A.; Svergun, D.I.; Kulathu, Y. Mechanism of Activation and Regulation of Deubiquitinase Activity in MINDY1 and MINDY2. Mol. Cell 2021, 81, 4176–4190.e6.
  44. Misaghi, S.; Galardy, P.J.; Meester, W.J.N.; Ovaa, H.; Ploegh, H.L.; Gaudet, R. Structure of the Ubiquitin Hydrolase UCH-L3 Complexed with a Suicide Substrate. J. Biol. Chem. 2005, 280, 1512–1520.
  45. Molland, K.; Zhou, Q.; Mesecar, A.D. A 2.2 Å Resolution Structure of the USP7 Catalytic Domain in a New Space Group Elaborates upon Structural Rearrangements Resulting from Ubiquitin Binding. Acta Crystallogr. Sect. F Struct. Biol. Commun. 2014, 70, 283–287.
  46. Crystal Structure of a Deubiquitinating Enzyme (Human UCH-L3) at 1.8 å Resolution. EMBO J. 1997, 16, 3787–3796.
  47. Grasty, K.C.; Weeks, S.D.; Loll, P.J. Structural Insights into the Activity and Regulation of Human Josephin-2. J. Struct. Biol. X 2019, 3, 100011.
  48. Hermanns, T.; Pichlo, C.; Woiwode, I.; Klopffleisch, K.; Witting, K.F.; Ovaa, H.; Baumann, U.; Hofmann, K. A Family of Unconventional Deubiquitinases with Modular Chain Specificity Determinants. Nat. Commun. 2018, 9, 799.
  49. Guo, Y.; Liu, Q.; Mallette, E.; Caba, C.; Hou, F.; Fux, J.; LaPlante, G.; Dong, A.; Zhang, Q.; Zheng, H.; et al. Structural and Functional Characterization of Ubiquitin Variant Inhibitors for the JAMM-Family Deubiquitinases STAMBP and STAMBPL1. J. Biol. Chem. 2021, 297, 101107.
  50. Snyder, N.A.; Silva, G.M. Deubiquitinating Enzymes (DUBs): Regulation, Homeostasis, and Oxidative Stress Response. J. Biol. Chem. 2021, 297, 101077.
  51. Amer-Sarsour, F.; Kordonsky, A.; Berdichevsky, Y.; Prag, G.; Ashkenazi, A. Deubiquitylating Enzymes in Neuronal Health and Disease. Cell Death Dis. 2021, 12, 1–11.
  52. Wang, B.; Cai, W.; Ai, D.; Zhang, X.; Yao, L. The Role of Deubiquitinases in Vascular Diseases. J. Cardiovasc. Transl. Res. 2020, 13, 131–141.
  53. Basar, M.A.; Beck, D.B.; Werner, A. Deubiquitylases in Developmental Ubiquitin Signaling and Congenital Diseases. Cell Death Differ. 2021, 28, 538–556.
  54. Parihar, N.; Bhatt, L.K. Deubiquitylating Enzymes: Potential Target in Autoimmune Diseases. Inflammopharmacology 2021, 29, 1683–1699.
  55. Cruz, L.; Soares, P.; Correia, M. Ubiquitin-Specific Proteases: Players in Cancer Cellular Processes. Pharmaceuticals 2021, 14, 848.
  56. Kim, S.-H.; Baek, K.-H. Regulation of Cancer Metabolism by Deubiquitinating Enzymes: The Warburg Effect. Int. J. Mol. Sci. 2021, 22, 6173.
  57. Errington, T.M.; Mathur, M.; Soderberg, C.K.; Denis, A.; Perfito, N.; Iorns, E.; Nosek, B.A. Investigating the Replicability of Preclinical Cancer Biology. eLife 2021, 10, e71601.
  58. Errington, T.M.; Denis, A.; Perfito, N.; Iorns, E.; Nosek, B.A. Challenges for Assessing Replicability in Preclinical Cancer Biology. eLife 2021, 10, e67995.
  59. Murtaza, M.; Jolly, L.A.; Gecz, J.; Wood, S.A. La FAM Fatale: USP9X in Development and Disease. Cell. Mol. Life Sci. CMLS 2015, 72, 2075–2089.
  60. Mouchantaf, R.; Azakir, B.A.; McPherson, P.S.; Millard, S.M.; Wood, S.A.; Angers, A. The Ubiquitin Ligase Itch Is Auto-Ubiquitylated in Vivo and in Vitro but Is Protected from Degradation by Interacting with the Deubiquitylating Enzyme FAM/USP9X. J. Biol. Chem. 2006, 281, 38738–38747.
  61. Khan, O.M.; Carvalho, J.; Spencer-Dene, B.; Mitter, R.; Frith, D.; Snijders, A.P.; Wood, S.A.; Behrens, A. The Deubiquitinase USP9X Regulates FBW7 Stability and Suppresses Colorectal Cancer. J. Clin. Investig. 2018, 128, 1326–1337.
  62. Donato, N.J.; Talpaz, M.; Peterson, L.; Young, M.; Showalter, H.D.; Wobus, C.; O’Riordan, M.X.D.; Ermann, M. Deubiquitinase Inhibitors and Methods for Use of the Same. WO2015054555A1, 16 April 2015.
  63. Akiyama, H.; Umezawa, Y.; Watanabe, D.; Okada, K.; Ishida, S.; Nogami, A.; Miura, O. Inhibition of USP9X Downregulates JAK2-V617F and Induces Apoptosis Synergistically with BH3 Mimetics Preferentially in Ruxolitinib-Persistent JAK2-V617F-Positive Leukemic Cells. Cancers 2020, 12, 406.
  64. Potu, H.; Kandarpa, M.; Peterson, L.F.; Durham, A.; Donato, N.J.; Talpaz, M. Downregulation of SOX2 by Inhibition of Usp9X Induces Apoptosis in Melanoma. Oncotarget 2021, 12, 160–172.
  65. Jaiswal, A.; Murakami, K.; Elia, A.; Shibahara, Y.; Done, S.J.; Wood, S.A.; Donato, N.J.; Ohashi, P.S.; Reedijk, M. Therapeutic Inhibition of USP9x-Mediated Notch Signaling in Triple-Negative Breast Cancer. Proc. Natl. Acad. Sci. USA 2021, 118, e2101592118.
  66. Zhang, Z.; Cui, Z.; Xie, Z.; Li, C.; Xu, C.; Guo, X.; Yu, J.; Chen, T.; Facchinetti, F.; Bohnenberger, H.; et al. Deubiquitinase USP5 Promotes Non-Small Cell Lung Cancer Cell Proliferation by Stabilizing Cyclin D1. Transl. Lung Cancer Res. 2021, 10, 3995–4011.
  67. Potu, H.; Kandarpa, M.; Peterson, L.F.; Donato, N.J.; Talpaz, M. Tumor Necrosis Factor Related Apoptosis Inducing Ligand (TRAIL) Regulates Deubiquitinase USP5 in Tumor Cells. Oncotarget 2019, 10, 5745–5754.
  68. Peterson, L.F.; Sun, H.; Liu, Y.; Potu, H.; Kandarpa, M.; Ermann, M.; Courtney, S.M.; Young, M.; Showalter, H.D.; Sun, D.; et al. Targeting Deubiquitinase Activity with a Novel Small-Molecule Inhibitor as Therapy for B-Cell Malignancies. Blood 2015, 125, 3588–3597.
  69. Clancy, A.; Heride, C.; Pinto-Fernández, A.; Elcocks, H.; Kallinos, A.; Kayser-Bricker, K.J.; Wang, W.; Smith, V.; Davis, S.; Fessler, S.; et al. The Deubiquitylase USP9X Controls Ribosomal Stalling. J. Cell Biol. 2021, 220, e202004211.
  70. Liu, J.; Xia, H.; Kim, M.; Xu, L.; Li, Y.; Zhang, L.; Cai, Y.; Norberg, H.V.; Zhang, T.; Furuya, T.; et al. Beclin1 Controls the Levels of P53 by Regulating the Deubiquitination Activity of USP10 and USP13. Cell 2011, 147, 223–234.
  71. Lundgren, S.; Albertella, M.; Belda, O.; Derbyshire, D.; Henderson, I.; Lindberg, J.; Odrzywol, E.; Stoor, C.; Strömberg, K.; Unnerståle, S. Comprehensive Profiling of DUB Inhibitors Using the Medivir DUB Platform; Discovery on Target: Boston, MA, USA, 2016.
  72. Dexheimer, T.S.; Rosenthal, A.S.; Liang, Q.; Chen, J.; Villamil, M.A.; Kerns, E.H.; Simeonov, A.; Jadhav, A.; Zhuang, Z.; Maloney, D.J. Discovery of ML323 as a Novel Inhibitor of the USP1/UAF1 Deubiquitinase Complex. In Probe Reports from the NIH Molecular Libraries Program; National Center for Biotechnology Information: Bethesda, MD, USA, 2010.
  73. Liang, Q.; Dexheimer, T.S.; Zhang, P.; Rosenthal, A.S.; Villamil, M.A.; You, C.; Zhang, Q.; Chen, J.; Ott, C.A.; Sun, H.; et al. A Selective USP1-UAF1 Inhibitor Links Deubiquitination to DNA Damage Responses. Nat. Chem. Biol. 2014, 10, 298–304.
  74. Arrowsmith, C.H.; Audia, J.E.; Austin, C.; Baell, J.; Bennett, J.; Blagg, J.; Bountra, C.; Brennan, P.E.; Brown, P.J.; Bunnage, M.E.; et al. The Promise and Peril of Chemical Probes. Nat. Chem. Biol. 2015, 11, 541.
  75. Pozhidaeva, A.; Bezsonova, I. USP7: Structure, Substrate Specificity, and Inhibition. DNA Repair 2019, 76, 30–39.
  76. Lange, S.M.; Armstrong, L.A.; Kulathu, Y. Deubiquitinases: From Mechanisms to Their Inhibition by Small Molecules. Mol. Cell 2021, 82, 15–29.
  77. Wertz, I.E.; Murray, J.M. Structurally-Defined Deubiquitinase Inhibitors Provide Opportunities to Investigate Disease Mechanisms. Drug Discov. Today Technol. 2019, 31, 109–123.
  78. Varca, A.C.; Casalena, D.; Chan, W.C.; Hu, B.; Magin, R.S.; Roberts, R.M.; Liu, X.; Zhu, H.; Seo, H.-S.; Dhe-Paganon, S.; et al. Identification and Validation of Selective Deubiquitinase Inhibitors. Cell Chem. Biol. 2021, 28, 1758–1771.e13.
  79. 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.
  80. Mulder, M.P.C.; Witting, K.F.; Ovaa, H. Cracking the Ubiquitin Code: The Ubiquitin Toolbox. Curr. Issues Mol. Biol. 2020, 37, 1–20.
  81. Pruneda, J.N.; Komander, D. Evaluating Enzyme Activities and Structures of DUBs. Methods Enzymol. 2019, 618, 321–341.
  82. Liu, L.; Damerell, D.R.; Koukouflis, L.; Tong, Y.; Marsden, B.D.; Schapira, M. UbiHub: A Data Hub for the Explorers of Ubiquitination Pathways. Bioinformatics 2019, 35, 2882–2884.
  83. Rusilowicz-Jones, E.V.; Jardine, J.; Kallinos, A.; Pinto-Fernandez, A.; Guenther, F.; Giurrandino, M.; Barone, F.G.; McCarron, K.; Burke, C.J.; Murad, A.; et al. USP30 Sets a Trigger Threshold for PINK1–PARKIN Amplification of Mitochondrial Ubiquitylation. Life Sci. Alliance 2020, 3.
  84. Panyain, N.; Godinat, A.; Thawani, A.R.; Lachiondo-Ortega, S.; Mason, K.; Elkhalifa, S.; Smith, L.M.; Harrigan, J.A.; Tate, E.W. Activity-Based Protein Profiling Reveals Deubiquitinase and Aldehyde Dehydrogenase Targets of a Cyanopyrrolidine Probe. RSC Med. Chem. 2021, 12, 1935–1943.
  85. Panyain, N.; Godinat, A.; Lanyon-Hogg, T.; Lachiondo-Ortega, S.; Will, E.J.; Soudy, C.; Mondal, M.; Mason, K.; Elkhalifa, S.; Smith, L.M.; et al. Discovery of a Potent and Selective Covalent Inhibitor and Activity-Based Probe for the Deubiquitylating Enzyme UCHL1, with Antifibrotic Activity. J. Am. Chem. Soc. 2020, 142, 12020–12026.
  86. Blagg, J.; Workman, P. Choose and Use Your Chemical Probe Wisely to Explore Cancer Biology. Cancer Cell 2017, 32, 9–25.
  87. Schlierf, A.; Altmann, E.; Quancard, J.; Jefferson, A.B.; Assenberg, R.; Renatus, M.; Jones, M.; Hassiepen, U.; Schaefer, M.; Kiffe, M.; et al. Targeted Inhibition of the COP9 Signalosome for Treatment of Cancer. Nat. Commun. 2016, 7, 13166.
  88. Altmann, E.; Erbel, P.; Renatus, M.; Schaefer, M.; Schlierf, A.; Druet, A.; Kieffer, L.; Sorge, M.; Pfister, K.; Hassiepen, U.; et al. Azaindoles as Zinc-Binding Small-Molecule Inhibitors of the JAMM Protease CSN5. Angew. Chem. Int. Ed. Engl. 2017, 56, 1294–1297.
  89. 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.
  90. 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.
  91. O’Dowd, C.R.; Helm, M.D.; Rountree, J.S.S.; Flasz, J.T.; Arkoudis, E.; Miel, H.; Hewitt, P.R.; Jordan, L.; Barker, O.; Hughes, C.; et al. Identification and Structure-Guided Development of Pyrimidinone Based USP7 Inhibitors. ACS Med. Chem. Lett. 2018, 9, 238–243.
  92. Schauer, N.J.; Liu, X.; Magin, R.S.; Doherty, L.M.; Chan, W.C.; Ficarro, S.B.; Hu, W.; Roberts, R.M.; Iacob, R.E.; Stolte, B.; et al. Selective USP7 Inhibition Elicits Cancer Cell Killing through a P53-Dependent Mechanism. Sci. Rep. 2020, 10, 5324.
  93. Lamberto, I.; Liu, X.; Seo, H.-S.; Schauer, N.J.; Iacob, R.E.; Hu, W.; 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.
  94. 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. Bioorg. Med. Chem. Lett. 2018, 28, 2655–2659.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 732
Revisions: 2 times (View History)
Update Date: 18 May 2022
1000/1000