Next Article in Journal
Cold Stress Induced Liver Injury of Mice through Activated NLRP3/Caspase-1/GSDMD Pyroptosis Signaling Pathway
Previous Article in Journal
Role of Pyroptosis in Gynecological Oncology and Its Therapeutic Regulation
Previous Article in Special Issue
On the Study of Deubiquitinases: Using the Right Tools for the Job
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

The Multifaceted Role of the Ubiquitin Proteasome System in Pathogenesis and Diseases

1
Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, 50 Stone Rd. E, Guelph, ON N1G 2W1, Canada
2
Department of Biology, York University, Room 327B Life Science Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
3
CIFAR Azrieli Global Scholars Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
*
Authors to whom correspondence should be addressed.
Biomolecules 2022, 12(7), 925; https://doi.org/10.3390/biom12070925
Submission received: 28 June 2022 / Accepted: 29 June 2022 / Published: 1 July 2022
(This article belongs to the Special Issue The Ubiquitin Proteasome System (UPS) in Pathogenesis and Diseases)

Abstract

:
Ubiquitin is a small protein that is conjugated to target proteins to signal a great number of critical biological processes. Impaired ubiquitin signaling and defects in the ubiquitin proteasome system (UPS) surveillance are implicated in many human diseases, including cancer. Characterization of the physiological roles of UPS components and their regulatory mechanisms is therefore vital for the identification of therapeutic targets and the development of tools and paradigms to better understand and treat human diseases. In this Special Issue, we assembled seven original research and review articles to provide insights on the multifaceted role of the UPS in pathogenesis and disease, covering the areas of molecular and cellular mechanisms of UPS enzymes, biochemical and biophysical characterization strategies, drug development, and targeted protein degradation.

Ubiquitination is a well-conserved protein post-translational modification (PTM), characterized by the covalent attachment of ubiquitin (Ub) or poly-Ub chains to substrate proteins, leading to proteasomal degradation or changes in cellular signaling pathways [1]. This process involves a series of ordered steps, mediated by the enzymatic functions of E1 activating enzymes, E2 conjugating enzymes and E3 ligases (E1→ E2→ E3) [2,3]. Ubiquitination is a dynamic and reversible process in which deubiquitinases (DUBs) can cleave ubiquitin from target proteins [4]. Collectively, all of these functional components form the ubiquitin-proteasome system (UPS), a regulated protein degradation system present in all eukaryotes [5]. The UPS is responsible for the degradation of nuclear, cytosolic, and myofibrillar proteins, therefore critical for the proper maintenance of protein homeostasis in human cells [6]. This Special Issue is dedicated to the new paradigm of ubiquitin biology in diseases and recent developments in therapeutic approaches targeting components of UPS.
Malfunction of the UPS is associated with numerous human diseases including cancer, cardiovascular disease and neurodegenerative disease [5,6]. Particularly, E3 ligases and DUBs are frequently implicated in the progression of these diseases, as they catalyze specific ubiquitination and deubiquitination events that are critical to maintaining normal cellular function. In this issue, Diaz et al. highlighted recent findings regarding the physiological and pathological roles of Cullin-RING E3 ubiquitin ligases (CRLs), which play essential roles in regulating the cardiovascular system through UPS [7]. Further research in this field could lead to the development of therapeutic intervention strategies targeting cardiovascular diseases.
The UPS also plays a critical role in virus-induced cell signaling and immune responses [8,9,10,11]. An emerging area of research has been on SARS-CoV-2 (Severe acute respiratory syndrome coronavirus 2), the virus causing COVID-19. PTMs such as ubiquitination and ISGylation are involved in mediating host-pathogen interactions and antiviral cellular signaling through modulation of key events in innate immunity activation [12]. SARS-CoV-2 encodes a papain-like protease (PLpro) that removes Ub and/or ISG15 (an Ub-like modifier) from human substrate proteins. This allows viral replication and avoidance of host immune system surveillance [12]. An in-depth review in this issue by Vere et al. focused on how human and viral DUBs orchestrate the ubiquitination and ISGylation pathways involved in host-pathogen interactions. These proteins can serve as potential targets for therapeutic development to treat COVID-19 patients.
In the past few years, a combinatorial structure-based protein engineering strategy has been successfully used to develop Ub variants (UbVs) to manipulate UPS components in human cells [3,13,14]. Caba et al. provided a comprehensive overview of DUB pharmacological modulators and tools such as UbVs and activity-based probes (ABPs). These tools can be used to better understand DUBs at the cellular level. Specifically, UbV technology is a promising approach to expanding the library of known DUB inhibitors and can be used as a combinatorial platform for structure-guided drug design [15]. For instance, Hewitt et al. utilized a computational approach to create a UbV-based ABP for a DUB UCHL1 (Ubiquitin C-Terminal Hydrolase L1) as UCHL1 overexpression has been associated with various cancer types and neurodegenerative disorders [16]. UCHL3 is another UCH-family DUB that regulates DNA repair and is overexpressed in many cancers [17,18]. Building on their previous UCHL1 work, Hewitt and colleagues generated a selective triple-mutant UbV-ABP for UCHL3 and validated its function in multiple human cell lines [19].
E3 ligases are involved in many biological activities, as they can control protein stability, function and localization [20]. Targeted inhibition of E3 ligases by small molecules provides opportunities for therapeutic development. MDM2 is an E3 ligase that negatively regulates the tumor suppressor p53 and other proteins involved in DNA repair, cell cycle control and apoptosis pathways. Ilic et al. performed virtual screening against a natural product library, identified, and validated that Hinokiflavone (a biflavonoid) is a potent intracellular inhibitor of MDM2 activity [21]. This study sheds light on developing other E3 ligase inhibitors using natural compounds.
Approximately 20 years ago, researchers devised an attractive strategy to induce targeted protein degradation by artificially recruiting an E3 ligase to a non-native substrate. This study was the first to establish the efficacy of developing proteolysis targeting chimeras (PROTACs) for drug development [22]. Since then, many small-molecule ligands that redirect protein homeostasis machinery were developed for the targeted degradation of disease-related proteins for therapeutics [23]. While there are over 600 E3 ligases encoded by the human genome, the capability of many has yet to be established for this type of application [20,24]. In this issue, Aminu et al. utilized structure-based protein engineering to create what they termed Ubiquitin Variant Induced Proximity (UbVIP). The lab first generated non-inhibitory UbV binders for a selected panel of E3 ligases and screened a library of UbVIPs targeting a DNA damage response protein 53BP1, before identifying two E3 ligases (RFWD3 and NEDD4L) that can be leveraged for targeted protein degradation. This work provides new insights into the identification of novel E3 ligases for PROTACs design [25].
Last but not least, PTMs such as methylation, acetylation and phosphorylation are crucial in regulating various cellular processes [26]. Mass spectrometry-based proteomics studies are the key driving force that led to the detection of the majority of the PTM sites known to date [27,28,29]. In this issue, Lacoursiere and colleagues revealed the Ub and UPS PTM landscape, which was discovered through biochemical, biophysical, and proteomics assays. The potential impacts of these PTMs have on ubiquitin signaling are also discussed, along with implications in human diseases [26].
In conclusion, this Special Issue highlights emerging trends in the UPS field, including new biological functions of UPS components, novel strategies to modulate E3 ligases and DUBs in human diseases, targeted protein degradation, and the functional impacts of UPS PTM events.

Author Contributions

Writing—original draft preparation, Z.S., Y.S. and W.Z.; writing—review and editing, Z.S., Y.S. and W.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NSERC (Natural Sciences and Engineering Research Council of Canada) Discovery Grants awarded to Y.S. (RGPIN-2017-05709) and W.Z. (RGPIN-2019-05721).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We thank Daniel Martinho for proofreading of the manuscript. We also thank suggestions from members of the Sheng laboratory and the Zhang laboratory. We greatly appreciate the researchers who contributed to this Special Issue.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Martín-Villanueva, S.; Gutiérrez, G.; Kressler, D.; de la Cruz, J. Ubiquitin and Ubiquitin-Like Proteins and Domains in Ribosome Production and Function: Chance or Necessity? Int. J. Mol. Sci. 2021, 22, 4359. [Google Scholar] [CrossRef] [PubMed]
  2. Sluimer, J.; Distel, B. Regulating the Human HECT E3 Ligases. Cell. Mol. Life Sci. 2018, 75, 3121–3141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Zhang, W.; Wu, K.-P.; Sartori, M.A.; Kamadurai, H.B.; Ordureau, A.; Jiang, C.; Mercredi, P.Y.; Murchie, R.; Hu, J.; Persaud, A.; et al. System-Wide Modulation of HECT E3 Ligases with Selective Ubiquitin Variant Probes. Mol. Cell 2016, 62, 121–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Wang, Y.; Wang, F. Post-Translational Modifications of Deubiquitinating Enzymes: Expanding the Ubiquitin Code. Front. Pharmacol. 2021, 12, 685011. [Google Scholar] [CrossRef]
  5. Gong, B.; Radulovic, M.; Figueiredo-Pereira, M.E.; Cardozo, C. The Ubiquitin-Proteasome System: Potential Therapeutic Targets for Alzheimer’s Disease and Spinal Cord Injury. Front. Mol. Neurosci. 2016, 9, 4. [Google Scholar] [CrossRef] [Green Version]
  6. Mearini, G.; Schlossarek, S.; Willis, M.S.; Carrier, L. The Ubiquitin–Proteasome System in Cardiac Dysfunction. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2008, 1782, 749–763. [Google Scholar] [CrossRef] [Green Version]
  7. Diaz, S.; Wang, K.; Sjögren, B.; Liu, X. Roles of Cullin-RING Ubiquitin Ligases in Cardiovascular Diseases. Biomolecules 2022, 12, 416. [Google Scholar] [CrossRef]
  8. Flotho, A.; Melchior, F. Sumoylation: A Regulatory Protein Modification in Health and Disease. Annu. Rev. Biochem. 2013, 82, 357–385. [Google Scholar] [CrossRef]
  9. Geiss-Friedlander, R.; Melchior, F. Concepts in Sumoylation: A Decade On. Nat. Rev. Mol. Cell Biol. 2007, 8, 947–956. [Google Scholar] [CrossRef]
  10. Skaug, B.; Chen, Z.J. Emerging Role of ISG15 in Antiviral Immunity. Cell 2010, 143, 187–190. [Google Scholar] [CrossRef] [Green Version]
  11. Zhang, D.; Zhang, D.-E. Interferon-Stimulated Gene 15 and the Protein ISGylation System. J. Interferon Cytokine Res. 2011, 31, 119–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Vere, G.; Alam, M.R.; Farrar, S.; Kealy, R.; Kessler, B.M.; O’Brien, D.P.; Pinto-Fernández, A. Targeting the Ubiquitylation and ISGylation Machinery for the Treatment of COVID-19. Biomolecules 2022, 12, 300. [Google Scholar] [CrossRef] [PubMed]
  13. Ernst, A.; Avvakumov, G.; Tong, J.; Fan, Y.; Zhao, Y.; Alberts, P.; Persaud, A.; Walker, J.R.; Neculai, A.-M.; Neculai, D.; et al. A Strategy for Modulation of Enzymes in the Ubiquitin System. Science 2013, 339, 590–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gorelik, M.; Orlicky, S.; Sartori, M.A.; Tang, X.; Marcon, E.; Kurinov, I.; Greenblatt, J.F.; Tyers, M.; Moffat, J.; Sicheri, F.; et al. Inhibition of SCF Ubiquitin Ligases by Engineered Ubiquitin Variants That Target the Cul1 Binding Site on the Skp1–F-Box Interface. Proc. Natl. Acad. Sci. USA 2016, 113, 3527–3532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Caba, C.; Mohammadzadeh, A.; Tong, Y. On the Study of Deubiquitinases: Using the Right Tools for the Job. Biomolecules 2022, 12, 703. [Google Scholar] [CrossRef]
  16. Hewitt, C.S.; Krabill, A.D.; Das, C.; Flaherty, D.P. Development of Ubiquitin Variants with Selectivity for Ubiquitin C-Terminal Hydrolase Deubiquitinases. Biochemistry 2020, 59, 3447–3462. [Google Scholar] [CrossRef]
  17. Liao, C.; Beveridge, R.; Hudson, J.J.R.; Parker, J.D.; Chiang, S.-C.; Ray, S.; Ashour, M.E.; Sudbery, I.; Dickman, M.J.; El-Khamisy, S.F. UCHL3 Regulates Topoisomerase-Induced Chromosomal Break Repair by Controlling TDP1 Proteostasis. Cell Rep. 2018, 23, 3352–3365. [Google Scholar] [CrossRef]
  18. Song, Z.; Li, J.; Zhang, L.; Deng, J.; Fang, Z.; Xiang, X.; Xiong, J. UCHL3 Promotes Pancreatic Cancer Progression and Chemo-Resistance through FOXM1 Stabilization. Am. J. Cancer Res. 2019, 9, 1970–1981. [Google Scholar]
  19. Hewitt, C.S.; Das, C.; Flaherty, D.P. Rational Development and Characterization of a Ubiquitin Variant with Selectivity for Ubiquitin C-Terminal Hydrolase L3. Biomolecules 2022, 12, 62. [Google Scholar] [CrossRef]
  20. Koganti, P.; Levy-Cohen, G.; Blank, M. Smurfs in Protein Homeostasis, Signaling, and Cancer. Front. Oncol. 2018, 8, 295. [Google Scholar] [CrossRef] [Green Version]
  21. Ilic, V.K.; Egorova, O.; Tsang, E.; Gatto, M.; Wen, Y.; Zhao, Y.; Sheng, Y. Hinokiflavone Inhibits MDM2 Activity by Targeting the MDM2-MDMX RING Domain. Biomolecules 2022, 12, 643. [Google Scholar] [CrossRef] [PubMed]
  22. Sakamoto, K.M.; Kim, K.B.; Kumagai, A.; Mercurio, F.; Crews, C.M.; Deshaies, R.J. Protacs: Chimeric Molecules That Target Proteins to the Skp1–Cullin–F Box Complex for Ubiquitination and Degradation. Proc. Natl. Acad. Sci. USA 2001, 98, 8554–8559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Chamberlain, P.P.; Hamann, L.G. Development of Targeted Protein Degradation Therapeutics. Nat. Chem. Biol. 2019, 15, 937–944. [Google Scholar] [CrossRef] [PubMed]
  24. Iconomou, M.; Saunders, D.N. Systematic Approaches to Identify E3 Ligase Substrates. Biochem. J. 2016, 473, 4083–4101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Aminu, B.; Fux, J.; Mallette, E.; Petersen, N.; Zhang, W. Targeted Degradation of 53BP1 Using Ubiquitin Variant Induced Proximity. Biomolecules 2022, 12, 479. [Google Scholar] [CrossRef]
  26. Lacoursiere, R.E.; Hadi, D.; Shaw, G.S. Acetylation, Phosphorylation, Ubiquitination (Oh My!): Following Post-Translational Modifications on the Ubiquitin Road. Biomolecules 2022, 12, 467. [Google Scholar] [CrossRef] [PubMed]
  27. Tsai, C.-F.; Wang, Y.-T.; Yen, H.-Y.; Tsou, C.-C.; Ku, W.-C.; Lin, P.-Y.; Chen, H.-Y.; Nesvizhskii, A.I.; Ishihama, Y.; Chen, Y.-J. Large-Scale Determination of Absolute Phosphorylation Stoichiometries in Human Cells by Motif-Targeting Quantitative Proteomics. Nat. Commun. 2015, 6, 6622. [Google Scholar] [CrossRef] [Green Version]
  28. Mertins, P.; Qiao, J.W.; Patel, J.; Udeshi, N.D.; Clauser, K.R.; Mani, D.R.; Burgess, M.W.; Gillette, M.A.; Jaffe, J.D.; Carr, S.A. Integrated Proteomic Analysis of Post-Translational Modifications by Serial Enrichment. Nat. Methods 2013, 10, 634–637. [Google Scholar] [CrossRef]
  29. Choudhary, C.; Kumar, C.; Gnad, F.; Nielsen, M.L.; Rehman, M.; Walther, T.C.; Olsen, J.V.; Mann, M. Lysine Acetylation Targets Protein Complexes and Co-Regulates Major Cellular Functions. Science 2009, 325, 834–840. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Stekel, Z.; Sheng, Y.; Zhang, W. The Multifaceted Role of the Ubiquitin Proteasome System in Pathogenesis and Diseases. Biomolecules 2022, 12, 925. https://doi.org/10.3390/biom12070925

AMA Style

Stekel Z, Sheng Y, Zhang W. The Multifaceted Role of the Ubiquitin Proteasome System in Pathogenesis and Diseases. Biomolecules. 2022; 12(7):925. https://doi.org/10.3390/biom12070925

Chicago/Turabian Style

Stekel, Zane, Yi Sheng, and Wei Zhang. 2022. "The Multifaceted Role of the Ubiquitin Proteasome System in Pathogenesis and Diseases" Biomolecules 12, no. 7: 925. https://doi.org/10.3390/biom12070925

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop