The Proteasome System: Comparison
Please note this is a comparison between Version 2 by Vicky Zhou and Version 3 by Vicky Zhou.

The proteasome system is a large and complex molecular machinery responsible for the degradation of misfolded, damaged, and redundant cellular proteins. When proteasome function is impaired, unwanted proteins accumulate, which can lead to several diseases including age-related and neurodegenerative diseases. Enhancing proteasome-mediated substrate degradation with small molecules may therefore be a valuable strategy for the treatment of various neurodegenerative diseases such as Parkinson’s, Alzheimer’s, and Huntington’s diseases. 

  • proteasome
  • neurodegeneration
  • ubiquitin
  • misfolded
  • disordered
  • degradation
  • protein

1. Introduction

The degradation of proteins is a continual process that is highly regulated by the two major proteolysis systems, the lysosomal degradation pathway and the proteasome-mediated pathway. Protein degradation helps maintain biological homeostasis in cells which are needed for all cell functions and for maintaining optimal conditions for enzyme function [1]. The proteasome pathway is the major pathway for the degradation of misfolded, oxidatively damaged, and redundant proteins. Dysregulation of proteasome function has been identified in the pathogenesis of several neurodegenerative diseases including Parkinson’s disease (PD) [2], Alzheimer’s disease (AD), and other neurodegenerative diseases [3]. The proteasome pathway is also involved in the regulation of several other cellular processes such as cell cycle, stress signaling, gene expression regulation, inflammatory response, cell differentiation, and apoptosis, which makes it an appealing target in the treatment of other types of diseases, including cancer [4]. Due to the critical role of the proteasome-mediated degradation pathway in cell regulation, the modulation of proteasome proteolytic activity has become a valuable strategy in the pursuit of new therapeutics to treat several neurodegenerative diseases [5][6][7][8].

1.1. The Human Proteasome

The human proteasome is a large complex protein responsible for the intracellular degradation of unwanted and damaged proteins via a ubiquitin-dependent and ubiquitin-independent degradation pathway. The most common proteolytic clearance of proteins proceeds by tagging the protein with polyubiquitin, after which it is degraded into small peptides of seven to eight amino acids by the 26S proteasome [9]. Highly disordered proteins can also be degraded in a ubiquitin-independent manner by the 20S proteasome [10].

1.2. Ubiquitin-Proteasome System

1.2.1. Ubiquitin

Ubiquitin (Ub) is a small protein (approximately 8600 Da) with 76 amino acid residues responsible for tagging a wide range of cellular proteins for proteolytic degradation. In the ubiquitin-proteasome system (UPS) (Figure 1), proteins are tagged for proteolysis by covalent ligation to ubiquitin [11]. Ubiquitination of proteins requires three enzymes in chronological order (see Figure 1a). The E1 ubiquitin-activating enzyme, just like its name, activates the C-terminal glycine residue of the ubiquitin in an ATP-dependent manner. The binding of the ubiquitin to a cysteine residue of E1 forms a Ub-E1 complex via a thioester linkage. The E2 ubiquitin-conjugating enzymes transfer the ubiquitin from the Ub-E1 complex to itself via a trans-thioesterification to form the Ub-E2 complex and release the E1 enzyme from the system. Lastly, the ubiquitin ligases E3s are responsible for selecting proteins for ubiquitin-mediated proteolysis. Humans have two E1 enzymes, about 40 E2 enzymes, and are estimated to have about 500–1000 E3s [12].
Biomolecules 11 01789 g001

References

  1. Hetz, C.; Glimcher, L.H. Protein homeostasis networks in physiology and disease. Curr. Opin. Cell. Biol. 2011, 23, 123–125.
  2. McNaught, K.S.P.; Olanow, C.W.; Halliwell, B.; Isacson, O.; Jenner, P. Failure of the ubiquitin–proteasome system in Parkinson’s disease. Nat. Rev. Neurosci. 2001, 2, 589–594.
  3. Saez, I.; Vilchez, D. The Mechanistic Links Between Proteasome Activity, Aging and Age-related Diseases. Curr. Genomics 2014, 15, 38–51.
  4. LaPlante, G.; Zhang, W. Targeting the Ubiquitin-Proteasome System for Cancer Therapeutics by Small-Molecule Inhibitors. Cancers 2021, 13, 3079.
  5. Momtaz, S.; Memariani, Z.; El-Senduny, F.F.; Sanadgol, N.; Golab, F.; Katebi, M.; Abdolghaffari, A.H.; Farzaei, M.H.; Abdollahi, M. Targeting Ubiquitin-Proteasome Pathway by Natural Products: Novel Therapeutic Strategy for Treatment of Neurodegenerative Diseases. Front. Physiol. 2020, 11, 361.
  6. Rao, G.; Croft, B.; Teng, C.; Awasthi, V. Ubiquitin-Proteasome System in Neurodegenerative Disorders. J. Drug Metab. Toxicol. 2015, 6, 187.
  7. Huang, Q.; Figueiredo-Pereira, M.E. Ubiquitin/proteasome pathway impairment in neurodegeneration: Therapeutic implications. Apoptosis 2010, 15, 1292–1311.
  8. Njomen, E.; Tepe, J.J. Proteasome activation as a new therapeutic approach to target proteotoxic disorders. J. Med. Chem. 2019, 62, 6469–6481.
  9. Kisselev, A.F.; Akopian, T.N.; Woo, K.M.; Goldberg, A.L. The sizes of peptides generated from protein by mammalian 26 and 20 S proteasomes. Implications for understanding the degradative mechanism and antigen presentation. J. Biol. Chem. 1999, 274, 3363–3371.
  10. Kumar Deshmukh, F.; Yaffe, D.; Olshina, M.A.; Ben-Nissan, G.; Sharon, M. The Contribution of the 20S Proteasome to Proteostasis. Biomolecules 2019, 9, 190.
  11. Hershko, A.; Ciechanover, A. The ubiquitin system. Annu. Rev. Biochem. 1998, 67, 425–479.
  12. Stewart, M.D.; Ritterhoff, T.; Klevit, R.E.; Brzovic, P.S. E2 enzymes: More than just middle men. Cell Res. 2016, 26, 423–440.
  13. Adapted from “Ubiquitin Proteasome System”, by BioRender.com. Available online: https://app.biorender.com/biorender-templates (accessed on 19 October 2021).
  14. Komander, D. The emerging complexity of protein ubiquitination. Biochem. Soc. Trans. 2009, 37, 937–953.
  15. Peng, J.; Schwartz, D.; Elias, J.E.; Thoreen, C.C.; Cheng, D.; Marsischky, G.; Roelofs, J.; Finley, D.; Gygi, S.P. A proteomics approach to understanding protein ubiquitination. Nat. Biotechnol. 2003, 21, 921–926.
  16. Chau, V.; Tobias, J.W.; Bachmair, A.; Marriott, D.; Ecker, D.J.; Gonda, D.K.; Varshavsky, A. A multiubiquitin chain is confined to specific lysine in a targeted short-lived protein. Science 1989, 243, 1576–1583.
  17. Tracz, M.; Bialek, W. Beyond K48 and K63: Non-canonical protein ubiquitination. Cell. Mol. Biol. Lett. 2021, 26, 1.
  18. Komander, D.; Rape, M. The Ubiquitin Code. Annu. Rev. Biochem 2012, 81, 203–229.
  19. hrower, J.S.; Hoffman, L.; Rechsteiner, M.; Pickart, C.M. Recognition of the polyubiquitin proteolytic signal. EMBO J. 2000, 19, 94–102.
  20. Dimova, N.V.; Hathaway, N.A.; Lee, B.-H.; Kirkpatrick, D.S.; Berkowitz, M.L.; Gygi, S.P.; Finley, D.; King, R.W. APC/C-mediated multiple monoubiquitylation provides an alternative degradation signal for cyclin B1. Nat. Cell Biol. 2012, 14, 168–176.
  21. Shabek, N.; Herman-Bachinsky, Y.; Buchsbaum, S.; Lewinson, O.; Haj-Yahya, M.; Hejjaoui, M.; Lashuel, H.A.; Sommer, T.; Brik, A.; Ciechanover, A. The Size of the Proteasomal Substrate Determines Whether Its Degradation Will Be Mediated by Mono- or Polyubiquitylation. Mol. Cell 2012, 48, 87–97.
  22. Kravtsova-Ivantsiv, Y.; Ciechanover, A. Non-canonical ubiquitin-based signals for proteasomal degradation. J. Cell Sci. 2012, 125, 539–548.
  23. Martinez-Fonts, K.; Davis, C.; Tomita, T.; Elsasser, S.; Nager, A.R.; Shi, Y.; Finley, D.; Matouschek, A. The proteasome 19S cap and its ubiquitin receptors provide a versatile recognition platform for substrates. Nat. Commun. 2020, 11, 477.
  24. 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.
  25. Peters, J.M.; Cejka, Z.; Harris, J.R.; Kleinschmidt, J.A.; Baumeister, W. Structural features of the 26S proteasome complex. J. Mol. Biol. 1993, 234, 932–937.
  26. da Fonseca, P.C.; Morris, E.P. Structure of the human 26S proteasome: Subunit radial displacements open the gate into the proteolytic core. J. Biol. Chem. 2008, 283, 23305–23314.
  27. Orlowski, M.; Wilk, S. Catalytic Activities of the 20S Proteasome, a Multicatalytic Proteinase Complex. Arch. Biochem. Biophys. 2000, 383, 1–16.
  28. 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. Nature 2019, 565, 49–55.
  29. Finley, D.; Chen, X.; Walters, K.J. Gates, Channels, and Switches: Elements of the Proteasome Machine. Trends Biochem. Sci 2016, 41, 77–93.
  30. Andres, 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.
  31. Eisele, M.R.; Reed, R.G.; Rudack, T.; Schweitzer, A.; Beck, F.; Nagy, I.; Pfeifer, G.; Plitzko, J.M.; Baumeister, W.; Tomko, R.J.; et al. Expanded Coverage of the 26S Proteasome Conformational Landscape Reveals Mechanisms of Peptidase Gating. Cell Rep. 2018, 24, 1301–1315.e1305.
  32. Ding, Z.; Fu, Z.; Xu, C.; Wang, Y.; Wang, Y.; Li, J.; Kong, L.; Chen, J.; Li, N.; Zhang, R.; et al. High-resolution cryo-EM structure of the proteasome in complex with ADP-AlFx. Cell Res. 2017, 27, 373–385.
  33. Huang, X.; Luan, B.; Wu, J.; Shi, Y. An atomic structure of the human 26S proteasome. Nat. Struct. Mol. Biol. 2016, 23, 778–785.
  34. 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.
  35. Ding, Z.; Xu, C.; Sahu, I.; Wang, Y.; Fu, Z.; Huang, M.; Wong, C.C.L.; Glickman, M.H.; Cong, Y. Structural Snapshots of 26S Proteasome Reveal Tetraubiquitin-Induced Conformations. Mol. Cell 2019, 73, 1150–1161.e1156.
  36. Vilchez, D.; Saez, I.; Dillin, A. The role of protein clearance mechanisms in organismal ageing and age-related diseases. Nat. Commun. 2014, 5, 5659.
  37. Bochtler, M.; Ditzel, L.; Groll, M.; Hartmann, C.; Huber, R. The proteasome. Annu Rev Biophys Biomol Struct 1999, 28, 295–317.
  38. Huber, E.M.; Heinemeyer, W.; Li, X.; Arendt, C.S.; Hochstrasser, M.; Groll, M. A unified mechanism for proteolysis and autocatalytic activation in the 20S proteasome. Nat. Commun. 2016, 7, 10900.
  39. Huang, L.; Chen, C.H. Proteasome regulators: Activators and inhibitors. Curr. Med. Chem. 2009, 16, 931–939.
  40. Kane, R.C.; Bross, P.F.; Farrell, A.T.; Pazdur, R. Velcade: U.S. FDA approval for the treatment of multiple myeloma progressing on prior therapy. Oncologist 2003, 8, 508–513.
  41. Bruna, J.; Udina, E.; Alé, A.; Vilches, J.J.; Vynckier, A.; Monbaliu, J.; Silverman, L.; Navarro, X. Neurophysiological, histological and immunohistochemical characterization of bortezomib-induced neuropathy in mice. Exp. Neurol. 2010, 223, 599–608.
  42. Gilmore, T.D.; Herscovitch, M. Inhibitors of NF-κB signaling: 785 and counting. Oncogene 2006, 25, 6887–6899.
  43. Hideshima, T.; Richardson, P.; Chauhan, D.; Palombella, V.J.; Elliott, P.J.; Adams, J.; Anderson, K.C. The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res. 2001, 61, 3071–3076.
  44. Russo, S.M.; Tepper, J.E.; Baldwin, A.S., Jr.; Liu, R.; Adams, J.; Elliott, P.; Cusack, J.C., Jr. Enhancement of radiosensitivity by proteasome inhibition: Implications for a role of NF-kappaB. Int. J. Radiat. Oncol. Biol. Phys. 2001, 50, 183–193.
  45. Sunwoo, J.B.; Chen, Z.; Dong, G.; Yeh, N.; Crowl Bancroft, C.; Sausville, E.; Adams, J.; Elliott, P.; Van Waes, C. Novel proteasome inhibitor PS-341 inhibits activation of nuclear factor-kappa B, cell survival, tumor growth, and angiogenesis in squamous cell carcinoma. Clin. Cancer Res. 2001, 7, 1419–1428.
  46. Hideshima, T.; Chauhan, D.; Richardson, P.; Mitsiades, C.; Mitsiades, N.; Hayashi, T.; Munshi, N.; Dang, L.; Castro, A.; Palombella, V.; et al. NF-kappa B as a therapeutic target in multiple myeloma. J. Biol. Chem. 2002, 277, 16639–16647.
  47. Tan, C.; Waldmann, T.A. Proteasome inhibitor PS-341, a potential therapeutic agent for adult T-cell leukemia. Cancer Res. 2002, 62, 1083–1086.
  48. Ma, M.H.; Yang, H.H.; Parker, K.; Manyak, S.; Friedman, J.M.; Altamirano, C.; Wu, Z.Q.; Borad, M.J.; Frantzen, M.; Roussos, E.; et al. The proteasome inhibitor PS-341 markedly enhances sensitivity of multiple myeloma tumor cells to chemotherapeutic agents. Clin. Cancer Res. 2003, 9, 1136–1144.
  49. Shah, S.A.; Potter, M.W.; McDade, T.P.; Ricciardi, R.; Perugini, R.A.; Elliott, P.J.; Adams, J.; Callery, M.P. 26S proteasome inhibition induces apoptosis and limits growth of human pancreatic cancer. J. Cell. Biochem. 2001, 82, 110–122.
  50. Yang, Y.; Ikezoe, T.; Saito, T.; Kobayashi, M.; Koeffler, H.P.; Taguchi, H. Proteasome inhibitor PS-341 induces growth arrest and apoptosis of non-small cell lung cancer cells via the JNK/c-Jun/AP-1 signaling. Cancer Sci. 2004, 95, 176–180.
  51. Williams, S.A.; McConkey, D.J. The proteasome inhibitor bortezomib stabilizes a novel active form of p53 in human LNCaP-Pro5 prostate cancer cells. Cancer Res. 2003, 63, 7338–7344.
  52. Li, B.; Dou, Q.P. Bax degradation by the ubiquitin/proteasome-dependent pathway: Involvement in tumor survival and progression. Proc. Natl. Acad. Sci. USA 2000, 97, 3850–3855.
  53. Breitschopf, K.; Zeiher, A.M.; Dimmeler, S. Ubiquitin-mediated degradation of the proapoptotic active form of bid. A functional consequence on apoptosis induction. J. Biol. Chem. 2000, 275, 21648–21652.
  54. Bianchi, G.; Oliva, L.; Cascio, P.; Pengo, N.; Fontana, F.; Cerruti, F.; Orsi, A.; Pasqualetto, E.; Mezghrani, A.; Calbi, V.; et al. The proteasome load versus capacity balance determines apoptotic sensitivity of multiple myeloma cells to proteasome inhibition. Blood 2009, 113, 3040–3049.
  55. Sha, Z.; Goldberg, A.L. Multiple myeloma cells are exceptionally sensitive to heat shock, which overwhelms their proteostasis network and induces apoptosis. Proc. Natl. Acad. Sci. USA 2020, 117, 21588.
  56. Suraweera, A.; Münch, C.; Hanssum, A.; Bertolotti, A. Failure of amino acid homeostasis causes cell death following proteasome inhibition. Mol. Cell 2012, 48, 242–253.
  57. Aliabadi, F.; Sohrabi, B.; Mostafavi, E.; Pazoki-Toroudi, H.; Webster, T.J. Ubiquitin–proteasome system and the role of its inhibitors in cancer therapy. Open Biol. 2021, 11, 200390.
  58. Fricker, L.D. Proteasome Inhibitor Drugs. Annu. Rev. Pharmacol. Toxicol. 2020, 60, 457–476.
  59. Gandolfi, S.; Laubach, J.P.; Hideshima, T.; Chauhan, D.; Anderson, K.C.; Richardson, P.G. The proteasome and proteasome inhibitors in multiple myeloma. Cancer Metastasis Rev. 2017, 36, 561–584.
  60. Ito, S. Proteasome Inhibitors for the Treatment of Multiple Myeloma. Cancers 2020, 12, 265.
  61. Kaplan, G.S.; Torcun, C.C.; Grune, T.; Ozer, N.K.; Karademir, B. Proteasome inhibitors in cancer therapy: Treatment regimen and peripheral neuropathy as a side effect. Free Radic. Biol. Med. 2017, 103, 1–13.
  62. Manasanch, E.E.; Orlowski, R.Z. Proteasome inhibitors in cancer therapy. Nat. Rev. Clin. Oncol. 2017, 14, 417–433.
  63. Narayanan, S.; Cai, C.-Y.; Assaraf, Y.G.; Guo, H.-Q.; Cui, Q.; Wei, L.; Huang, J.-J.; Ashby, C.R.; Chen, Z.-S. Targeting the ubiquitin-proteasome pathway to overcome anti-cancer drug resistance. Drug Resist. Updates 2020, 48, 100663.
  64. Roeten, M.S.F.; Cloos, J.; Jansen, G. Positioning of proteasome inhibitors in therapy of solid malignancies. Cancer Chemother. Pharmacol. 2018, 81, 227–243.
  65. Sherman, D.J.; Li, J. Proteasome Inhibitors: Harnessing Proteostasis to Combat Disease. Molecules 2020, 25, 671.
  66. Zhang, X.; Linder, S.; Bazzaro, M. Drug Development Targeting the Ubiquitin–Proteasome System (UPS) for the Treatment of Human Cancers. Cancers 2020, 12, 902.
  67. Hubbell, G.E.; Tepe, J.J. Natural product scaffolds as inspiration for the design and synthesis of 20S human proteasome inhibitors. RSC Chem. Biol. 2020, 1, 305–332.
  68. Jones, C.L.; Tepe, J.J. Proteasome Activation to Combat Proteotoxicity. Molecules 2019, 24, 2841.
  69. Jones, C.L.; Njomen, E.; Sjogren, B.; Dexheimer, T.S.; Tepe, J.J. Small Molecule Enhancement of 20S Proteasome Activity Targets Intrinsically Disordered Proteins. ACS Chem. Biol. 2017, 12, 2240–2247.
  70. Njomen, E.; Osmulski, P.A.; Jones, C.L.; Gaczynska, M.; Tepe, J.J. Small Molecule Modulation of Proteasome Assembly. Biochemistry 2018, 57, 4214–4224.
  71. Trippier, P.C.; Zhao, K.T.; Fox, S.G.; Schiefer, I.T.; Benmohamed, R.; Moran, J.; Kirsch, D.R.; Morimoto, R.I.; Silverman, R.B. Proteasome Activation is a Mechanism for Pyrazolone Small Molecules Displaying Therapeutic Potential in Amyotrophic Lateral Sclerosis. ACS Chem. Neurosci. 2014, 5, 823–829.
  72. Trader, D.J.; Simanski, S.; Dickson, P.; Kodadek, T. Establishment of a suite of assays that support the discovery of proteasome stimulators. Biochim. Biophys. Acta 2017, 1861, 892–899.
  73. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217.
  74. Lee, C.-K.; Klopp, R.G.; Weindruch, R.; Prolla, T.A. Gene Expression Profile of Aging and Its Retardation by Caloric Restriction. Science 1999, 285, 1390–1393.
  75. Bulteau, A.-L.; Lundberg, K.C.; Humphries, K.M.; Sadek, H.A.; Szweda, P.A.; Friguet, B.; Szweda, L.I. Oxidative Modification and Inactivation of the Proteasome during Coronary Occlusion/Reperfusion*. J. Biol. Chem. 2001, 276, 30057–30063.
  76. Wang, X.; Yen, J.; Kaiser, P.; Huang, L. Regulation of the 26S Proteasome Complex During Oxidative Stress. Sci. Signal. 2010, 3, ra88.
  77. Tonoki, A.; Kuranaga, E.; Tomioka, T.; Hamazaki, J.; Murata, S.; Tanaka, K.; Miura, M. Genetic Evidence Linking Age-Dependent Attenuation of the 26S Proteasome with the Aging Process. Mol. Cell. Biol. 2009, 29, 1095–1106.
  78. Bajorek, M.; Finley, D.; Glickman, M.H. Proteasome Disassembly and Downregulation Is Correlated with Viability during Stationary Phase. Curr. Biol. 2003, 13, 1140–1144.
  79. Kayed, R.; Dettmer, U.; Lesné, S.E. Soluble endogenous oligomeric α-synuclein species in neurodegenerative diseases: Expression, spreading, and cross-talk. J. Parkinsons Dis. 2020, 10, 791–818.
  80. Ono, K. Alzheimer’s disease as oligomeropathy. Neurochem. Int. 2018, 119, 57–70.
  81. Mroczko, B.; Groblewska, M.; Litman-Zawadzka, A.; Kornhuber, J.; Lewczuk, P. Amyloid β oligomers (AβOs) in Alzheimer’s disease. J. Neural Transm. 2018, 125, 177–191.
  82. Gulisano, W.; Maugeri, D.; Baltrons, M.A.; Fà, M.; Amato, A.; Palmeri, A.; D’Adamio, L.; Grassi, C.; Devanand, D.; Honig, L.S. Role of amyloid-β and tau proteins in Alzheimer’s disease: Confuting the amyloid cascade. J. Alzheimer’s Dis. 2018, 64, S611–S631.
  83. Ghag, G.; Bhatt, N.; Cantu, D.V.; Guerrero-Munoz, M.J.; Ellsworth, A.; Sengupta, U.; Kayed, R. Soluble tau aggregates, not large fibrils, are the toxic species that display seeding and cross-seeding behavior. Protein Sci. 2018, 27, 1901–1909.
  84. Forloni, G.; Balducci, C. Alzheimer’s disease, oligomers, and inflammation. J. Alzheimer’s Dis. 2018, 62, 1261–1276.
  85. Cline, E.N.; Bicca, M.A.; Viola, K.L.; Klein, W.L. The amyloid-β oligomer hypothesis: Beginning of the third decade. J. Alzheimer’s Dis. 2018, 64, S567–S610.
  86. Choi, M.L.; Gandhi, S. Crucial role of protein oligomerization in the pathogenesis of Alzheimer’s and Parkinson’s diseases. FEBS J. 2018, 285, 3631–3644.
  87. Castillo-Carranza, D.L.; Guerrero-Muñoz, M.J.; Sengupta, U.; Gerson, J.E.; Kayed, R. α-Synuclein oligomers induce a unique toxic tau strain. Biol. Psychiatry 2018, 84, 499–508.
  88. Shafiei, S.S.; Guerrero-Muñoz, M.J.; Castillo-Carranza, D.L. Tau oligomers: Cytotoxicity, propagation, and mitochondrial damage. Front. Aging Neurosci. 2017, 9, 83.
  89. Sengupta, U.; Nilson, A.N.; Kayed, R. The role of amyloid-β oligomers in toxicity, propagation, and immunotherapy. EBioMedicine 2016, 6, 42–49.
  90. Ingelsson, M. Alpha-synuclein oligomers—neurotoxic molecules in Parkinson’s disease and other Lewy body disorders. Front. Neurosci. 2016, 10, 408.
  91. Caárdenas-Aguayo, M.a.d.C.; Goόmez-Virgilio, L.; DeRosa, S.; Meraz-Ríos, M.A. The role of tau oligomers in the onset of Alzheimer’s disease neuropathology. ACS Chem. Neurosci. 2014, 5, 1178–1191.
  92. Katzmarski, N.; Ziegler-Waldkirch, S.; Scheffler, N.; Witt, C.; Abou-Ajram, C.; Nuscher, B.; Prinz, M.; Haass, C.; Meyer-Luehmann, M. Aβ oligomers trigger and accelerate Aβ seeding. Brain Pathol. 2020, 30, 36–45.
  93. Haass, C.; Selkoe, D.J. Soluble protein oligomers in neurodegeneration: Lessons from the Alzheimer’s amyloid β-peptide. Nat. Rev. Mol. Cell Biol. 2007, 8, 101–112.
  94. Smith, D.M. Could a common mechanism of protein degradation impairment underlie many neurodegenerative diseases? J. Exp. Neurosci. 2018, 12, 1179069518794675.
  95. Gerson, J.E.; Farmer, K.M.; Henson, N.; Castillo-Carranza, D.L.; Murillo, M.C.; Sengupta, U.; Barrett, A.; Kayed, R. Tau oligomers mediate α-synuclein toxicity and can be targeted by immunotherapy. Mol. Neurodegener. 2018, 13, 1–14.
  96. Gerson, J.E.; Sengupta, U.; Kayed, R. Tau oligomers as pathogenic seeds: Preparation and propagation in vitro and in vivo. In Tau Protein; Humana Press: New York, NY, USA, 2017; pp. 141–157.
  97. Bengoa-Vergniory, N.; Roberts, R.F.; Wade-Martins, R.; Alegre-Abarrategui, J. Alpha-synuclein oligomers: A new hope. Acta Neuropathol. 2017, 134, 819–838.
  98. Gerson, J.E.; Mudher, A.; Kayed, R. Potential mechanisms and implications for the formation of tau oligomeric strains. Crit. Rev. Biochem. Mol. Biol. 2016, 51, 482–496.
  99. Brettschneider, J.; Del Tredici, K.; Lee, V.M.; Trojanowski, J.Q. Spreading of pathology in neurodegenerative diseases: A focus on human studies. Nat. Rev. Neurosci. 2015, 16, 109–120.
  100. Rubinsztein, D.C. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 2006, 443, 780–786.
  101. Selkoe, D.J. Folding proteins in fatal ways. Nature 2003, 426, 900–904.
  102. Selkoe, D.J.; Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 2016, 8, 595–608.
  103. Cecarini, V.; Bonfili, L.; Amici, M.; Angeletti, M.; Keller, J.N.; Eleuteri, A.M. Amyloid peptides in different assembly states and related effects on isolated and cellular proteasomes. Brain Res. 2008, 1209, 8–18.
  104. Díaz-Hernández, M.; Valera, A.G.; Morán, M.A.; Gómez-Ramos, P.; Alvarez-Castelao, B.; Castaño, J.G.; Hernández, F.; Lucas, J.J. Inhibition of 26S proteasome activity by huntingtin filaments but not inclusion bodies isolated from mouse and human brain. J. Neurochem. 2006, 98, 1585–1596.
  105. Gregori, L.; Fuchs, C.; Figueiredo-Pereira, M.E.; Van Nostrand, W.E.; Goldgaber, D. Amyloid β-Protein Inhibits Ubiquitin-dependent Protein Degradation in Vitro (∗). J. Biol. Chem. 1995, 270, 19702–19708.
  106. Lindersson, E.; Beedholm, R.; Højrup, P.; Moos, T.; Gai, W.; Hendil, K.B.; Jensen, P.H. Proteasomal inhibition by α-synuclein filaments and oligomers. J. Biol. Chem. 2004, 279, 12924–12934.
  107. Bence, N.F.; Sampat, R.M.; Kopito, R.R. Impairment of the ubiquitin-proteasome system by protein aggregation. Science 2001, 292, 1552–1555.
  108. Oh, S.; Hong, H.S.; Hwang, E.; Sim, H.J.; Lee, W.; Shin, S.J.; Mook-Jung, I. Amyloid peptide attenuates the proteasome activity in neuronal cells. Mech. Ageing Dev. 2005, 126, 1292–1299.
  109. Tanaka, K.; Matsuda, N. Proteostasis and neurodegeneration: The roles of proteasomal degradation and autophagy. Biochim. Biophys. Acta 2014, 1843, 197–204.
  110. Tanaka, Y.; Engelender, S.; Igarashi, S.; Rao, R.K.; Wanner, T.; Tanzi, R.E.; Sawa, A.; Dawson, V.L.; Dawson, T.M.; Ross, C.A. Inducible expression of mutant α-synuclein decreases proteasome activity and increases sensitivity to mitochondria-dependent apoptosis. Hum. Mol. Genet. 2001, 10, 919–926.
  111. Tseng, B.P.; Green, K.N.; Chan, J.L.; Blurton-Jones, M.; LaFerla, F.M. Aβ inhibits the proteasome and enhances amyloid and tau accumulation. Neurobiol. Aging 2008, 29, 1607–1618.
  112. Emmanouilidou, E.; Stefanis, L.; Vekrellis, K. Cell-produced α-synuclein oligomers are targeted to, and impair, the 26S proteasome. Neurobiol. Aging 2010, 31, 953–968.
  113. Deriziotis, P.; André, R.; Smith, D.M.; Goold, R.; Kinghorn, K.J.; Kristiansen, M.; Nathan, J.A.; Rosenzweig, R.; Krutauz, D.; Glickman, M.H. Misfolded PrP impairs the UPS by interaction with the 20S proteasome and inhibition of substrate entry. EMBO J. 2011, 30, 3065–3077.
  114. Deriziotis, P.; Tabrizi, S.J. Prions and the proteasome. Biochim. Biophys. Acta 2008, 1782, 713–722.
  115. Kristiansen, M.; Deriziotis, P.; Dimcheff, D.E.; Jackson, G.S.; Ovaa, H.; Naumann, H.; Clarke, A.R.; van Leeuwen, F.W.; Menéndez-Benito, V.; Dantuma, N.P. Disease-associated prion protein oligomers inhibit the 26S proteasome. Mol. Cell 2007, 26, 175–188.
  116. Thibaudeau, T.A.; Anderson, R.T.; Smith, D.M. A common mechanism of proteasome impairment by neurodegenerative disease-associated oligomers. Nat. Commun. 2018, 9, 1097.
  117. Zondler, L.; Kostka, M.; Garidel, P.; Heinzelmann, U.; Hengerer, B.; Mayer, B.; Weishaupt, J.H.; Gillardon, F.; Danzer, K.M. Proteasome impairment by α-synuclein. PLoS ONE 2017, 12, e0184040.
  118. Ruegsegger, C.; Saxena, S. Proteostasis impairment in ALS. Brain Res. 2016, 1648, 571–579.
  119. Myeku, N.; Clelland, C.L.; Emrani, S.; Kukushkin, N.V.; Yu, W.H.; Goldberg, A.L.; Duff, K.E. Tau-driven 26S proteasome impairment and cognitive dysfunction can be prevented early in disease by activating cAMP-PKA signaling. Nat. Med. 2016, 22, 46–53.
  120. Papanikolopoulou, K.; Skoulakis, E. Altered proteostasis in neurodegenerative tauopathies. In Proteostasis and Disease; Barrio, R., Sutherland, J.D., Rodriguez, M.S., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 177–194.
  121. Choi, W.H.; De Poot, S.A.; Lee, J.H.; Kim, J.H.; Han, D.H.; Kim, Y.K.; Finley, D.; Lee, M.J. Open-gate mutants of the mammalian proteasome show enhanced ubiquitin-conjugate degradation. Nat. Commun. 2016, 7, 1–12.
  122. Leestemaker, Y.; de Jong, A.; Witting, K.F.; Penning, R.; Schuurman, K.; Rodenko, B.; Zaal, E.A.; van de Kooij, B.; Laufer, S.; Heck, A.J. Proteasome activation by small molecules. Cell Chem. Biol. 2017, 24, 725–736.
  123. Leestemaker, Y.; Ovaa, H. Tools to investigate the ubiquitin proteasome system. Drug Discov. Today Technol. 2017, 26, 25–31.
  124. 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. Enhancement of proteasome activity by a small-molecule inhibitor of USP14. Nature 2010, 467, 179–184.
  125. Fiolek, T.J.; Magyar, C.L.; Wall, T.J.; Davies, S.B.; Campbell, M.V.; Savich, C.J.; Tepe, J.J.; Mosey, R.A. Dihydroquinazolines enhance 20S proteasome activity and induce degradation of alpha-synuclein, an intrinsically disordered protein associated with neurodegeneration. Bioorg. Med. Chem. Lett. 2021, 36, 127821.
  126. Coleman, R.A.; Muli, C.S.; Zhao, Y.; Bhardwaj, A.; Newhouse, T.R.; Trader, D.J. Analysis of chain length, substitution patterns, and unsaturation of AM-404 derivatives as 20S proteasome stimulators. Bioorg. Med. Chem. Lett. 2019, 29, 420–423.
  127. Coleman, R.A.; Trader, D.J. Development and Application of a Sensitive Peptide Reporter to Discover 20S Proteasome Stimulators. ACS Comb. Sci. 2018, 20, 269–276.
  128. Giżynńska, M.; Witkowska, J.; Karpowicz, P.; Rostankowski, R.; Chocron, E.S.; Pickering, A.M.; Osmulski, P.; Gaczynska, M.; Jankowska, E.b. Proline-and arginine-rich peptides as flexible allosteric modulators of human proteasome activity. J. Med. Chem. 2019, 62, 359–370.
  129. Njomen, E.; Lansdell, T.; Vanecek, A.; Benham, V.; Bernard, M.; Yang, Y.-T.; Schall, P.; Isaac, D.; Alkharabsheh, O.; Al-Janadi, A. Enhancing c-MYC degradation via 20S proteasome activation induces in vivo anti-tumor efficacy. bioRxiv 2020.
  130. Chondrogianni, N.; Georgila, K.; Kourtis, N.; Tavernarakis, N.; Gonos, E.S. 20S proteasome activation promotes life span extension and resistance to proteotoxicity in Caenorhabditis elegans. FASEB J. 2015, 29, 611–622.
  131. Gonos, E. Proteasome activation as a novel anti-aging strategy. Free Radic. Biol. Med. 2014, 75, S7.
  132. Fiolek, T.J.; Keel, K.L.; Tepe, J.J. Fluspirilene Analogs Activate the 20S Proteasome and Overcome Proteasome Impairment by Intrinsically Disordered Protein Oligomers. ACS Chem. Neurosci. 2021, 12, 1438–1448.
  133. Ben-Nissan, G.; Sharon, M. Regulating the 20S proteasome ubiquitin-independent degradation pathway. Biomolecules 2014, 4, 862–884.
  134. Njomen, E.; Tepe, J.J. Regulation of Autophagic Flux by the 20S Proteasome. Cell Chem. Biol. 2019, 26, 1283–1294.
  135. Asher, G.; Reuven, N.; Shaul, Y. 20S proteasomes and protein degradation "by default". Bioessays 2006, 28, 844–849.
  136. Korovila, I.; Hugo, M.; Castro, J.P.; Weber, D.; Höhn, A.; Grune, T.; Jung, T. Proteostasis, oxidative stress and aging. Redox Biol. 2017, 13, 550–567.
  137. Höhn, T.J.; Grune, T. The proteasome and the degradation of oxidized proteins: Part III-Redox regulation of the proteasomal system. Redox Biol. 2014, 2, 388–394.
  138. Chondrogianni, N.; Petropoulos, I.; Grimm, S.; Georgila, K.; Catalgol, B.; Friguet, B.; Grune, T.; Gonos, E.S. Protein damage, repair and proteolysis. Mol. Aspects Med. 2014, 35, 1–71.
  139. Myers, N.; Olender, T.; Savidor, A.; Levin, Y.; Reuven, N.; Shaul, Y. The Disordered Landscape of the 20S Proteasome Substrates Reveals Tight Association with Phase Separated Granules. Proteomics 2018, 18, e1800076.
  140. Tsvetkov, P.; Reuven, N.; Shaul, Y. The nanny model for IDPs. Nat. Chem. Biol. 2009, 5, 778–781.
More
Video Production Service