Amyloid Fragmentation and Disaggregation in Yeast and Animals: Comparison
Please note this is a comparison between Version 2 by Bruce Ren and Version 1 by Vitaly Kushnirov.

Amyloids are filamentous protein aggregates that are associated with a number of incurable diseases, termed amyloidoses. Amyloids can also manifest as infectious or heritable particles, known as prions. While just one prion is known in humans and animals, more than ten prion amyloids have been discovered in fungi. The propagation of fungal prion amyloids requires the chaperone Hsp104, though in excess it can eliminate some prions. Even though Hsp104 acts to disassemble prion fibrils, at normal levels it fragments them into multiple smaller pieces, which ensures prion propagation and accelerates prion conversion. Animals lack Hsp104, but disaggregation is performed by the same complement of chaperones that assist Hsp104 in yeast—Hsp40, Hsp70, and Hsp110. Exogenous Hsp104 can efficiently cooperate with these chaperones in animals and promotes disaggregation, especially of large amyloid aggregates, which indicates its potential as a treatment for amyloid diseases. However, despite the significant effects, Hsp104 and its potentiated variants may be insufficient to fully dissolve amyloid.

  • amyloid
  • prion
  • chaperone
  • amyloid fragmentation
  • Sup35
  • α-synuclein
  • Hsp40
  • Hsp70
  • Hsp104
  • HSP110
Please wait, diff process is still running!

References

  1. Chiti, F.; Dobson, C.M. Protein Misfolding, Amyloid Formation, and Human Disease: A Summary of Progress over the Last Decade. Annu. Rev. Biochem. 2017, 86, 27–68.
  2. Prusiner, S.B. Biology and genetics of prions causing neurodegeneration. Annu. Rev. Genet. 2013, 47, 601–623.
  3. Brundin, P.; Melki, R.; Kopito, R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat. Rev. Mol. Cell Biol. 2010, 11, 301–307.
  4. Goedert, M.; Masuda-Suzukake, M.; Falcon, B. Like prions: The propagation of aggregated tau and α-synuclein in neurodegeneration. Brain 2017, 140, 266–278.
  5. Lam, S.; Petit, F.; Hérard, A.-S.; Boluda, S.; Eddarkaoui, S.; Guillermier, M.; Letournel, F.; Martin-Négrier, M.-L.; Faisant, M.; Godfraind, C.; et al. Transmission of amyloid-beta and tau pathologies is associated with cognitive impairments in a primate. Acta Neuropathol. Commun. 2021, 9, 165.
  6. Keleman, K.; Krüttner, S.; Alenius, M.; Dickson, B.J. Function of the Drosophila CPEB protein Orb2 in long-term courtship memory. Nat. Neurosci. 2007, 10, 1587–1593.
  7. Mastushita-Sakai, T.; White-Grindley, E.; Samuelson, J.; Seidel, C.; Si, K. Drosophila Orb2 targets genes involved in neuronal growth, synapse formation, and protein turnover. Proc. Natl. Acad. Sci. USA 2010, 107, 11987–11992.
  8. Wickner, R.B. Yeast and Fungal Prions. Cold Spring Harb. Perspect. Biol. 2016, 8, a023531.
  9. Cox, B.S. Ψ, A cytoplasmic suppressor of super-suppressor in yeast. Heredity 1965, 20, 505–521.
  10. Cox, B.; Tuite, M. The life of . Curr. Genet. 2018, 64, 1–8.
  11. Lacroute, F. Non-Mendelian mutation allowing ureidosuccinic acid uptake in yeast. J. Bacteriol. 1971, 106, 519–522.
  12. Wickner, R.B. as an altered URE2 protein: Evidence for a prion analog in Saccharomyces cerevisiae. Science 1994, 264, 566–569.
  13. Derkatch, I.L.; Bradley, M.E.; Hong, J.Y.; Liebman, S.W. Prions affect the appearance of other prions: The story of . Cell 2001, 106, 171–182.
  14. Du, Z.; Park, K.-W.; Yu, H.; Fan, Q.; Li, L. Newly identified prion linked to the chromatin-remodeling factor Swi1 in Saccharomyces cerevisiae. Nat. Genet. 2008, 40, 460–465.
  15. Patel, B.K.; Gavin-Smyth, J.; Liebman, S.W. The yeast global transcriptional co-repressor protein Cyc8 can propagate as a prion. Nat. Cell Biol. 2009, 11, 344–349.
  16. Suzuki, G.; Shimazu, N.; Tanaka, M. A yeast prion, Mod5, promotes acquired drug resistance and cell survival under environmental stress. Science 2012, 336, 355–359.
  17. Alberti, S.; Halfmann, R.; King, O.; Kapila, A.; Lindquist, S. A systematic survey identifies prions and illuminates sequence features of prionogenic proteins. Cell 2009, 137, 146–158.
  18. Halfmann, R.; Wright, J.R.; Alberti, S.; Lindquist, S.; Rexach, M. Prion formation by a yeast GLFG nucleoporin. Prion 2012, 6, 391–399.
  19. Chernova, T.A.; Kiktev, D.A.; Romanyuk, A.V.; Shanks, J.R.; Laur, O.; Ali, M.; Ghosh, A.; Kim, D.; Yang, Z.; Mang, M.; et al. Yeast Short-Lived Actin-Associated Protein Forms a Metastable Prion in Response to Thermal Stress. Cell Rep. 2017, 18, 751–761.
  20. Cereghetti, G.; Wilson-Zbinden, C.; Kissling, V.M.; Diether, M.; Arm, A.; Yoo, H.; Piazza, I.; Saad, S.; Picotti, P.; Drummond, D.A.; et al. Reversible amyloids of pyruvate kinase couple cell metabolism and stress granule disassembly. Nat. Cell Biol. 2021, 23, 1085–1094.
  21. Reichert, P.; Caudron, F. Mnemons and the memorization of past signaling events. Curr. Opin. Cell Biol. 2021, 69, 127–135.
  22. Brown, J.C.S.; Lindquist, S. A heritable switch in carbon source utilization driven by an unusual yeast prion. Genes Dev. 2009, 23, 2320–2332.
  23. Chakrabortee, S.; Byers, J.S.; Jones, S.; Garcia, D.M.; Bhullar, B.; Chang, A.; She, R.; Lee, L.; Fremin, B.; Lindquist, S.; et al. Intrinsically Disordered Proteins Drive Emergence and Inheritance of Biological Traits. Cell 2016, 167, 369–381.e12.
  24. True, H.L.; Lindquist, S.L. A yeast prion provides a mechanism for genetic variation and phenotypic diversity. Nature 2000, 407, 477–483.
  25. Namy, O.; Galopier, A.; Martini, C.; Matsufuji, S.; Fabret, C.; Rousset, J.-P. Epigenetic control of polyamines by the prion . Nat. Cell Biol. 2008, 10, 1069–1075.
  26. Tuite, M.F. Yeast models of neurodegenerative diseases. Prog. Mol. Biol. Transl. Sci. 2019, 168, 351–379.
  27. Chernoff, Y.O.; Grizel, A.V.; Rubel, A.A.; Zelinsky, A.A.; Chandramowlishwaran, P.; Chernova, T.A. Application of yeast to studying amyloid and prion diseases. Adv. Genet. 2020, 105, 293–380.
  28. Serpionov, G.V.; Alexandrov, A.I.; Antonenko, Y.N.; Ter-Avanesyan, M.D. A protein polymerization cascade mediates toxicity of non-pathological human huntingtin in yeast. Sci. Rep. 2015, 5, 18407.
  29. Kushnirov, V.V.; Ter-Avanesyan, M.D. Structure and replication of yeast prions. Cell 1998, 94, 13–16.
  30. Stansfield, I.; Jones, K.M.; Kushnirov, V.V.; Dagkesamanskaya, A.R.; Poznyakovski, A.I.; Paushkin, S.V.; Nierras, C.R.; Cox, B.S.; Ter-Avanesyan, M.D.; Tuite, M.F. The products of the SUP45 (eRF1) and SUP35 genes interact to mediate translation termination in Saccharomyces cerevisiae. EMBO J. 1995, 14, 4365–4373.
  31. Zhouravleva, G.; Frolova, L.; Le Goff, X.; Le Guellec, R.; Inge-Vechtomov, S.; Kisselev, L.; Philippe, M. Termination of translation in eukaryotes is governed by two interacting polypeptide chain release factors, eRF1 and eRF3. EMBO J. 1995, 14, 4065–4072.
  32. Ter-Avanesyan, M.D.; Kushnirov, V.V.; Dagkesamanskaya, A.R.; Didichenko, S.A.; Chernoff, Y.O.; Inge-Vechtomov, S.G.; Smirnov, V.N. Deletion analysis of the SUP35 gene of the yeast Saccharomyces cerevisiae reveals two non-overlapping functional regions in the encoded protein. Mol. Microbiol. 1993, 7, 683–692.
  33. Helsen, C.W.; Glover, J.R. Insight into molecular basis of curing of prion by overexpression of 104-kDa heat shock protein Hsp104. J. Biol. Chem. 2012, 287, 542–556.
  34. Franzmann, T.M.; Jahnel, M.; Pozniakovsky, A.; Mahamid, J.; Holehouse, A.S.; Nüske, E.; Richter, D.; Baumeister, W.; Grill, S.W.; Pappu, R.V.; et al. Phase separation of a yeast prion protein promotes cellular fitness. Science 2018, 359, 6371.
  35. Mehra, S.; Gadhe, L.; Bera, R.; Sawner, A.S.; Maji, S.K. Structural and Functional Insights into α-Synuclein Fibril Polymorphism. Biomolecules 2021, 11, 1419.
  36. Carta, M.; Aguzzi, A. Molecular foundations of prion strain diversity. Curr. Opin. Neurobiol. 2021, 72, 22–31.
  37. Derkatch, I.L.; Chernoff, Y.O.; Kushnirov, V.V.; Inge-Vechtomov, S.G.; Liebman, S.W. Genesis and variability of prion factors in Saccharomyces cerevisiae. Genetics 1996, 144, 1375–1386.
  38. Wickner, R.B.; Son, M.; Edskes, H.K. Prion Variants of Yeast are Numerous, Mutable, and Segregate on Growth, Affecting Prion Pathogenesis, Transmission Barriers, and Sensitivity to Anti-Prion Systems. Viruses 2019, 11, 238.
  39. Nizhnikov, A.A.; Ryzhova, T.A.; Volkov, K.V.; Zadorsky, S.P.; Sopova, J.V.; Inge-Vechtomov, S.G.; Galkin, A.P. Interaction of Prions Causes Heritable Traits in Saccharomyces cerevisiae. PLoS Genet. 2016, 12, e1006504.
  40. Chang, H.-Y.; Lin, J.-Y.; Lee, H.-C.; Wang, H.-L.; King, C.-Y. Strain-specific sequences required for yeast prion propagation. Proc. Natl. Acad. Sci. USA 2008, 105, 13345–13350.
  41. Huang, Y.-W.; King, C.-Y. A complete catalog of wild-type Sup35 prion variants and their protein-only propagation. Curr. Genet. 2020, 66, 97–122.
  42. Huang, Y.-W.; Kushnirov, V.V.; King, C.-Y. Mutable yeast prion variants are stabilized by a defective Hsp104 chaperone. Mol. Microbiol. 2021, 115, 774–788.
  43. Ohhashi, Y.; Yamaguchi, Y.; Kurahashi, H.; Kamatari, Y.O.; Sugiyama, S.; Uluca, B.; Piechatzek, T.; Komi, Y.; Shida, T.; Müller, H.; et al. Molecular basis for diversification of yeast prion strain conformation. Proc. Natl. Acad. Sci. USA 2018, 115, 2389–2394.
  44. Kryndushkin, D.S.; Alexandrov, I.M.; Ter-Avanesyan, M.D.; Kushnirov, V. V Yeast prion aggregates are formed by small Sup35 polymers fragmented by Hsp104. J. Biol. Chem. 2003, 278, 49636–49643.
  45. Dergalev, A.; Alexandrov, A.; Ivannikov, R.; Ter-Avanesyan, M.; Kushnirov, V. Yeast Sup35 Prion Structure: Two Types, Four Parts, Many Variants. Int. J. Mol. Sci. 2019, 20, 2633.
  46. Shewmaker, F.; Wickner, R.B.; Tycko, R. Amyloid of the prion domain of Sup35p has an in-register parallel beta-sheet structure. Proc. Natl. Acad. Sci. USA 2006, 103, 19754–19759.
  47. Shewmaker, F.; Kryndushkin, D.; Chen, B.; Tycko, R.; Wickner, R.B. Two prion variants of Sup35p have in-register parallel β-sheet structures, independent of hydration. Biochemistry 2009, 48, 5074–5082.
  48. Tuttle, M.D.; Comellas, G.; Nieuwkoop, A.J.; Covell, D.J.; Berthold, D.A.; Kloepper, K.D.; Courtney, J.M.; Kim, J.K.; Barclay, A.M.; Kendall, A.; et al. Solid-state NMR structure of a pathogenic fibril of full-length human α-synuclein. Nat. Struct. Mol. Biol. 2016, 23, 409–415.
  49. Krishnan, R.; Lindquist, S.L. Structural insights into a yeast prion illuminate nucleation and strain diversity. Nature 2005, 435, 765–772.
  50. Toyama, B.H.; Kelly, M.J.S.; Gross, J.D.; Weissman, J.S. The structural basis of yeast prion strain variants. Nature 2007, 449, 233–237.
  51. Depace, A.H.; Santoso, A.; Hillner, P.; Weissman, J.S. A Critical Role for Amino-Terminal Glutamine/Asparagine Repeats in the Formation and Propagation of a Yeast Prion. Cell 1998, 93, 1241–1252.
  52. Huang, Y.W.; Chang, Y.C.; Diaz-Avalos, R.; King, C.Y. W8, a new Sup35 prion strain, transmits distinctive information with a conserved assembly scheme. Prion 2015, 9, 207–227.
  53. Ghosh, R.; Dong, J.; Wall, J.; Frederick, K.K. Amyloid fibrils embodying distinctive yeast prion phenotypes exhibit diverse morphologies. FEMS Yeast Res. 2018, 18, foy059.
  54. Parham, S.N.; Resende, C.G.; Tuite, M.F. Oligopeptide repeats in the yeast protein Sup35p stabilize intermolecular prion interactions. EMBO J. 2001, 20, 2111–2119.
  55. Shkundina, I.S.; Kushnirov, V.V.; Tuite, M.F.; Ter-Avanesyan, M.D. The role of the N-terminal oligopeptide repeats of the yeast Sup35 prion protein in propagation and transmission of prion variants. Genetics 2006, 172, 827–835.
  56. Tanaka, M.; Collins, S.R.; Toyama, B.H.; Weissman, J.S. The physical basis of how prion conformations determine strain phenotypes. Nature 2006, 442, 585–589.
  57. Alexandrov, A.I.; Polyanskaya, A.B.; Serpionov, G.V.; Ter-Avanesyan, M.D.; Kushnirov, V.V. The Effects of Amino Acid Composition of Glutamine-Rich Domains on Amyloid Formation and Fragmentation. PLoS ONE 2012, 7, e46458.
  58. Chernoff, Y.O.; Lindquist, S.L.; Ono, B.; Inge-Vechtomov, S.G.; Liebman, S.W. Role of the chaperone protein Hsp104 in propagation of the yeast prion-like factor . Science 1995, 268, 880–884.
  59. Paushkin, S.V.; Kushnirov, V.V.; Smirnov, V.N.; Ter-Avanesyan, M.D. Propagation of the yeast prion-like determinant is mediated by oligomerization of the SUP35-encoded polypeptide chain release factor. EMBO J. 1996, 15, 3127–3134.
  60. Parsell, D.A.; Kowal, A.S.; Singer, M.A.; Lindquist, S. Protein disaggregation mediated by heat-shock protein Hsp104. Nature 1994, 372, 475–478.
  61. King, C.-Y.; Tittmann, P.; Gross, H.; Gebert, R.; Aebi, M.; Wuthrich, K. Prion-inducing domain 2-114 of yeast Sup35 protein transforms in vitro into amyloid-like filaments. Proc. Natl. Acad. Sci. USA 1997, 94, 6618–6622.
  62. Glover, J.R.; Kowal, A.S.; Schirmer, E.C.; Patino, M.M.; Liu, J.J.; Lindquist, S. Self-seeded fibers formed by Sup35, the protein determinant of , a heritable prion-like factor of S. cerevisiae. Cell 1997, 89, 811–819.
  63. Patino, M.M.; Liu, J.J.; Glover, J.R.; Lindquist, S. Support for the prion hypothesis for inheritance of a phenotypic trait in yeast. Science 1996, 273, 622–626.
  64. Paushkin, S.V.; Kushnirov, V.V.; Smirnov, V.N.; Ter-Avanesyan, M.D. In vitro propagation of the prion-like state of yeast Sup35 protein. Science 1997, 277, 381–383.
  65. Byrne, L.J.; Cole, D.J.; Cox, B.S.; Ridout, M.S.; Morgan, B.J.T.; Tuite, M.F. The number and transmission of prion seeds (Propagons) in the yeast Saccharomyces cerevisiae. PLoS ONE 2009, 4, e4670.
  66. Winkler, J.; Tyedmers, J.; Bukau, B.; Mogk, A. Hsp70 targets Hsp100 chaperones to substrates for protein disaggregation and prion fragmentation. J. Cell Biol. 2012, 198, 387–404.
  67. Tuite, M.F.; Mundy, C.R.; Cox, B.S. Agents that cause a high frequency of genetic change from to in Saccharomyces cerevisiae. Genetics 1981, 98, 691–711.
  68. Baxa, U.; Keller, P.W.; Cheng, N.; Wall, J.S.; Steven, A.C. In Sup35p filaments (the prion), the globular C-terminal domains are widely offset from the amyloid fibril backbone. Mol. Microbiol. 2011, 79, 523–532.
  69. Prusiner, S.B. Prions. Proc. Natl. Acad. Sci. USA 1998, 95, 13363–13383.
  70. Cohen, F.E.; Pan, K.M.; Huang, Z.; Baldwin, M.; Fletterick, R.J.; Prusiner, S.B. Structural clues to prion replication. Science 1994, 264, 530–531.
  71. Walker, L.C. Proteopathic Strains and the Heterogeneity of Neurodegenerative Diseases. Annu. Rev. Genet. 2016, 50, 329–346.
  72. Glover, J.R.; Lindquist, S. Hsp104, Hsp70, and Hsp40: A novel chaperone system that rescues previously aggregated proteins. Cell 1998, 94, 73–82.
  73. Bagriantsev, S.N.; Gracheva, E.O.; Richmond, J.E.; Liebman, S.W. Variant-specific infection is transmitted by Sup35 polymers within aggregates with heterogeneous protein composition. Mol. Biol. Cell 2008, 19, 2433–2443.
  74. Park, S.; Wang, X.; Xi, W.; Richardson, R.; Laue, T.M.; Denis, C.L. The non-prion SUP35 preexists in large chaperone-containing molecular complexes. Proteins 2021.
  75. Sondheimer, N.; Lopez, N.; Craig, E.A.; Lindquist, S. The role of Sis1 in the maintenance of the prion. EMBO J. 2001, 20, 2435–2442.
  76. Higurashi, T.; Hines, J.K.; Sahi, C.; Aron, R.; Craig, E.A. Specificity of the J-protein Sis1 in the propagation of 3 yeast prions. Proc. Natl. Acad. Sci. USA 2008, 105, 16596–16601.
  77. Tipton, K.A.; Verges, K.J.; Weissman, J.S. In vivo monitoring of the prion replication cycle reveals a critical role for Sis1 in delivering substrates to Hsp104. Mol. Cell 2008, 32, 584–591.
  78. Hines, J.K.; Li, X.; Du, Z.; Higurashi, T.; Li, L.; Craig, E.A. , the prion formed by the chromatin remodeling factor Swi1, is highly sensitive to alterations in Hsp70 chaperone system activity. PLoS Genet. 2011, 7, e1001309.
  79. Schilke, B.A.; Ciesielski, S.J.; Ziegelhoffer, T.; Kamiya, E.; Tonelli, M.; Lee, W.; Cornilescu, G.; Hines, J.K.; Markley, J.L.; Craig, E.A. Broadening the functionality of a J-protein/Hsp70 molecular chaperone system. PLoS Genet. 2017, 13, e1007084.
  80. Bradley, M.E.; Edskes, H.K.; Hong, J.Y.; Wickner, R.B.; Liebman, S.W. Interactions among prions and prion “strains” in yeast. Proc. Natl. Acad. Sci. USA 2002, 99 (Suppl. 4), 16392–16399.
  81. Kryndushkin, D.S.; Smirnov, V.N.; Ter-Avanesyan, M.D.; Kushnirov, V.V. Increased expression of Hsp40 chaperones, transcriptional factors, and ribosomal protein Rpp0 can cure yeast prions. J. Biol. Chem. 2002, 277, 23702–23708.
  82. Troisi, E.M.; Rockman, M.E.; Nguyen, P.P.; Oliver, E.E.; Hines, J.K. Swa2, the yeast homolog of mammalian auxilin, is specifically required for the propagation of the prion variant . Mol. Microbiol. 2015, 97, 926–941.
  83. Parsell, D.A.; Kowal, A.S.; Lindquist, S. Saccharomyces cerevisiae Hsp104 protein. Purification and characterization of ATP-induced structural changes. J. Biol. Chem. 1994, 269, 4480–4487.
  84. Shorter, J.; Southworth, D.R. Spiraling in Control: Structures and Mechanisms of the Hsp104 Disaggregase. Cold Spring Harb. Perspect. Biol. 2019, 11, a034033.
  85. Gates, S.N.; Yokom, A.L.; Lin, J.; Jackrel, M.E.; Rizo, A.N.; Kendsersky, N.M.; Buell, C.E.; Sweeny, E.A.; Mack, K.L.; Chuang, E.; et al. Ratchet-like polypeptide translocation mechanism of the AAA+ disaggregase Hsp104. Science 2017, 357, 273–279.
  86. Avellaneda, M.J.; Franke, K.B.; Sunderlikova, V.; Bukau, B.; Mogk, A.; Tans, S.J. Processive extrusion of polypeptide loops by a Hsp100 disaggregase. Nature 2020, 578, 317–320.
  87. Zeymer, C.; Werbeck, N.D.; Schlichting, I.; Reinstein, J. The molecular mechanism of Hsp100 chaperone inhibition by the prion curing agent guanidinium chloride. J. Biol. Chem. 2013, 288, 7065–7076.
  88. Kummer, E.; Oguchi, Y.; Seyffer, F.; Bukau, B.; Mogk, A. Mechanism of Hsp104/ClpB inhibition by prion curing Guanidinium hydrochloride. FEBS Lett. 2013, 587, 810–817.
  89. Escusa-Toret, S.; Vonk, W.I.M.; Frydman, J. Spatial sequestration of misfolded proteins by a dynamic chaperone pathway enhances cellular fitness during stress. Nat. Cell Biol. 2013, 15, 1231–1243.
  90. Desantis, M.E.; Leung, E.H.; Sweeny, E.A.; Jackrel, M.E.; Cushman-Nick, M.; Neuhaus-Follini, A.; Vashist, S.; Sochor, M.A.; Knight, M.N.; Shorter, J. Operational plasticity enables Hsp104 to disaggregate diverse amyloid and nonamyloid clients. Cell 2012, 151, 778–793.
  91. Erives, A.J.; Fassler, J.S. Metabolic and chaperone gene loss marks the origin of animals: Evidence for Hsp104 and Hsp78 chaperones sharing mitochondrial enzymes as clients. PLoS ONE 2015, 10, e0117192.
  92. Rampelt, H.; Kirstein-Miles, J.; Nillegoda, N.B.; Chi, K.; Scholz, S.R.; Morimoto, R.I.; Bukau, B. Metazoan Hsp70 machines use Hsp110 to power protein disaggregation. EMBO J. 2012, 31, 4221–4235.
  93. Gao, X.; Carroni, M.; Nussbaum-Krammer, C.; Mogk, A.; Nillegoda, N.B.; Szlachcic, A.; Guilbride, D.L.; Saibil, H.R.; Mayer, M.P.; Bukau, B. Human Hsp70 Disaggregase Reverses Parkinson’s-Linked α-Synuclein Amyloid Fibrils. Mol. Cell 2015, 59, 781–793.
  94. Shorter, J. The mammalian disaggregase machinery: Hsp110 synergizes with Hsp70 and Hsp40 to catalyze protein disaggregation and reactivation in a cell-free system. PLoS ONE 2011, 6, e26319.
  95. Wentink, A.S.; Nillegoda, N.B.; Feufel, J.; Ubartaitė, G.; Schneider, C.P.; de los Rios, P.; Hennig, J.; Barducci, A.; Bukau, B. Molecular dissection of amyloid disaggregation by human HSP70. Nature 2020, 587, 483–488.
  96. Roberts, B.T.; Wickner, R.B. Heritable activity: A prion that propagates by covalent autoactivation. Genes Dev. 2003, 17, 2083–2087.
  97. Zakharov, I.A.; Yarovoy, B.P. Cytoduction as a new tool in studying the cytoplasmic heredity in yeast. Mol. Cell. Biochem. 1977, 14, 15–18.
  98. Urakov, V.N.; Vishnevskaya, A.B.; Alexandrov, I.M.; Kushnirov, V.V.; Smirnov, V.N.; Ter-Avanesyan, M.D. Interdependence of amyloid formation in yeast: Implications for polyglutamine disorders and biological functions. Prion 2010, 4, 45–52.
  99. Kato, M.; Han, T.W.; Xie, S.; Shi, K.; Du, X.; Wu, L.C.; Mirzaei, H.; Goldsmith, E.J.; Longgood, J.; Pei, J.; et al. Cell-free Formation of RNA Granules: Low Complexity Sequence Domains Form Dynamic Fibers within Hydrogels. Cell 2012, 149, 753–767.
  100. Murakami, T.; Qamar, S.; Lin, J.Q.; Schierle, G.S.K.; Rees, E.; Miyashita, A.; Costa, A.R.; Dodd, R.B.; Chan, F.T.S.; Michel, C.H.; et al. ALS/FTD Mutation-Induced Phase Transition of FUS Liquid Droplets and Reversible Hydrogels into Irreversible Hydrogels Impairs RNP Granule Function. Neuron 2015, 88, 678–690.
  101. Yang, Y.-S.; Kato, M.; Wu, X.; Litsios, A.; Sutter, B.M.; Wang, Y.; Hsu, C.-H.; Wood, N.E.; Lemoff, A.; Mirzaei, H.; et al. Yeast Ataxin-2 Forms an Intracellular Condensate Required for the Inhibition of TORC1 Signaling during Respiratory Growth. Cell 2019, 177, 697–710.e17.
  102. Lagaudrière-Gesbert, C.; Newmyer, S.L.; Gregers, T.F.; Bakke, O.; Ploegh, H.L. Uncoating ATPase Hsc70 is recruited by invariant chain and controls the size of endocytic compartments. Proc. Natl. Acad. Sci. USA 2002, 99, 1515–1520.
  103. Chakravarty, A.K.; Smejkal, T.; Itakura, A.K.; Garcia, D.M.; Jarosz, D.F. A Non-amyloid Prion Particle that Activates a Heritable Gene Expression Program. Mol. Cell 2020, 77, 251–265.e9.
  104. Cox, B.; Ness, F.; Tuite, M. Analysis of the generation and segregation of propagons: Entities that propagate the prion in yeast. Genetics 2003, 165, 23–33.
  105. Ghaemmaghami, S.; Huh, W.-K.; Bower, K.; Howson, R.W.; Belle, A.; Dephoure, N.; O’Shea, E.K.; Weissman, J.S. Global analysis of protein expression in yeast. Nature 2003, 425, 737–741.
  106. Ness, F.; Cox, B.S.; Wongwigkarn, J.; Naeimi, W.R.; Tuite, M.F. Over-expression of the molecular chaperone Hsp104 in Saccharomyces cerevisiae results in the malpartition of propagons. Mol. Microbiol. 2017, 104, 125–143.
  107. Kushnirov, V.V.; Kochneva-Pervukhova, N.V.; Chechenova, M.B.; Frolova, N.S.; Ter-Avanesyan, M.D. Prion properties of the Sup35 protein of yeast Pichia methanolica. EMBO J. 2000, 19, 324–331.
  108. Gorkovskiy, A.; Reidy, M.; Masison, D.C.; Wickner, R.B. Hsp104 disaggregase at normal levels cures many prion variants in a process promoted by Sti1p, Hsp90, and Sis1p. Proc. Natl. Acad. Sci. USA 2017, 114, E4193–E4202.
  109. Moriyama, H.; Edskes, H.K.; Wickner, R.B. prion propagation in Saccharomyces cerevisiae: Requirement for chaperone Hsp104 and curing by overexpressed chaperone Ydj1p. Mol. Cell. Biol. 2000, 20, 8916–8922.
  110. Derkatch, I.L.; Bradley, M.E.; Masse, S.V.; Zadorsky, S.P.; Polozkov, G.V.; Inge-Vechtomov, S.G.; Liebman, S.W. Dependence and independence of and : A two-prion system in yeast? EMBO J. 2000, 19, 1942–1952.
  111. Matveenko, A.G.; Barbitoff, Y.A.; Jay-Garcia, L.M.; Chernoff, Y.O.; Zhouravleva, G.A. Differential effects of chaperones on yeast prions: CURrent view. Curr. Genet. 2018, 64, 317–325.
  112. Kushnirov, V.V.; Kryndushkin, D.S.; Boguta, M.; Smirnov, V.N.; Ter-Avanesyan, M.D. Chaperones that cure yeast artificial and their prion-specific effects. Curr. Biol. 2000, 10, 1443–1446.
  113. Greene, L.E.; Saba, F.; Silberman, R.E.; Zhao, X. Mechanisms for curing yeast prions. Int. J. Mol. Sci. 2020, 21, 6536.
  114. Villali, J.; Dark, J.; Brechtel, T.M.; Pei, F.; Sindi, S.S.; Serio, T.R. Nucleation seed size determines amyloid clearance and establishes a barrier to prion appearance in yeast. Nat. Struct. Mol. Biol. 2020, 27, 540–549.
  115. Frederick, K.K.; Debelouchina, G.T.; Kayatekin, C.; Dorminy, T.; Jacavone, A.C.; Griffin, R.G.; Lindquist, S. Distinct prion strains are defined by amyloid core structure and chaperone binding site dynamics. Chem. Biol. 2014, 21, 295–305.
  116. Newnam, G.P.; Birchmore, J.L.; Chernoff, Y.O. Destabilization and recovery of a yeast prion after mild heat shock. J. Mol. Biol. 2011, 408, 432–448.
  117. Howie, R.L.; Jay-Garcia, L.M.; Kiktev, D.A.; Faber, Q.L.; Murphy, M.; Rees, K.A.; Sachwani, N.; Chernoff, Y.O. Role of the Cell Asymmetry Apparatus and Ribosome-Associated Chaperones in the Destabilization of a Saccharomyces cerevisiae Prion by Heat Shock. Genetics 2019, 212, 757–771.
  118. Alexandrov, I.M.; Vishnevskaya, A.B.; Ter-Avanesyan, M.D.; Kushnirov, V.V. Appearance and propagation of polyglutamine-based amyloids in yeast: Tyrosine residues enable polymer fragmentation. J. Biol. Chem. 2008, 283, 15185–15192.
  119. Franco, A.; Gracia, P.; Colom, A.; Camino, J.D.; Fernández-Higuero, J.Á.; Orozco, N.; Dulebo, A.; Saiz, L.; Cremades, N.; Vilar, J.M.G.; et al. All-or-none amyloid disassembly via chaperone-triggered fibril unzipping favors clearance of α-synuclein toxic species. Proc. Natl. Acad. Sci. USA 2021, 118, e2105548118.
  120. Shorter, J. Hsp104: A weapon to combat diverse neurodegenerative disorders. NeuroSignals 2007, 16, 63–74.
  121. Mosser, D.D.; Ho, S.; Glover, J.R. Saccharomyces cerevisiae Hsp104 enhances the chaperone capacity of human cells and inhibits heat stress-induced proapoptotic signaling. Biochemistry 2004, 43, 8107–8115.
  122. Lo Bianco, C.; Shorter, J.; Régulier, E.; Lashuel, H.; Iwatsubo, T.; Lindquist, S.; Aebischer, P. Hsp104 antagonizes alpha-synuclein aggregation and reduces dopaminergic degeneration in a rat model of Parkinson disease. J. Clin. Invest. 2008, 118, 3087–3097.
  123. Liu, Y.-H.; Han, Y.-L.; Song, J.; Wang, Y.; Jing, Y.-Y.; Shi, Q.; Tian, C.; Wang, Z.-Y.; Li, C.-P.; Han, J.; et al. Heat shock protein 104 inhibited the fibrillization of prion peptide 106-126 and disassembled prion peptide 106-126 fibrils in vitro. Int. J. Biochem. Cell Biol. 2011, 43, 768–774.
  124. Cushman-Nick, M.; Bonini, N.M.; Shorter, J. Hsp104 suppresses polyglutamine-induced degeneration post onset in a drosophila MJD/SCA3 model. PLoS Genet. 2013, 9, e1003781.
  125. Vacher, C.; Garcia-Oroz, L.; Rubinsztein, D.C. Overexpression of yeast hsp104 reduces polyglutamine aggregation and prolongs survival of a transgenic mouse model of Huntington’s disease. Hum. Mol. Genet. 2005, 14, 3425–3433.
  126. Satyal, S.H.; Schmidt, E.; Kitagawa, K.; Sondheimer, N.; Lindquist, S.; Kramer, J.M.; Morimoto, R.I. Polyglutamine aggregates alter protein folding homeostasis in Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 2000, 97, 5750–5755.
  127. Perrin, V.; Régulier, E.; Abbas-Terki, T.; Hassig, R.; Brouillet, E.; Aebischer, P.; Luthi-Carter, R.; Déglon, N. Neuroprotection by Hsp104 and Hsp27 in lentiviral-based rat models of Huntington’s disease. Mol. Ther. 2007, 15, 903–911.
  128. Mack, K.L.; Shorter, J. Engineering and Evolution of Molecular Chaperones and Protein Disaggregases with Enhanced Activity. Front. Mol. Biosci. 2016, 3, 8.
  129. Jackrel, M.E.; Yee, K.; Tariq, A.; Chen, A.I.; Shorter, J. Disparate Mutations Confer Therapeutic Gain of Hsp104 Function. ACS Chem. Biol. 2015, 10, 2672–2679.
  130. Jackrel, M.E.; Desantis, M.E.; Martinez, B.A.; Castellano, L.M.; Stewart, R.M.; Caldwell, K.A.; Caldwell, G.A.; Shorter, J. Potentiated Hsp104 variants antagonize diverse proteotoxic misfolding events. Cell 2014, 156, 170–182.
  131. Sweeny, E.A.; Jackrel, M.E.; Go, M.S.; Sochor, M.A.; Razzo, B.M.; DeSantis, M.E.; Gupta, K.; Shorter, J. The Hsp104 N-terminal domain enables disaggregase plasticity and potentiation. Mol. Cell 2015, 57, 836–849.
  132. Tariq, A.; Lin, J.; Noll, M.M.; Torrente, M.P.; Mack, K.L.; Murillo, O.H.; Jackrel, M.E.; Shorter, J. Potentiating Hsp104 activity via phosphomimetic mutations in the middle domain. FEMS Yeast Res. 2018, 18, foy042.
  133. Tariq, A.; Lin, J.; Jackrel, M.E.; Hesketh, C.D.; Carman, P.J.; Mack, K.L.; Weitzman, R.; Gambogi, C.; Hernandez Murillo, O.A.; Sweeny, E.A.; et al. Mining Disaggregase Sequence Space to Safely Counter TDP-43, FUS, and α-Synuclein Proteotoxicity. Cell Rep. 2019, 28, 2080–2095.e6.
  134. Ryan, J.J.; Bao, A.; Bell, B.; Ling, C.; Jackrel, M.E. Drivers of Hsp104 potentiation revealed by scanning mutagenesis of the middle domain. Protein Sci. 2021, 30, 1667–1685.
  135. Reidy, M.; Miot, M.; Masison, D.C. Prokaryotic Chaperones Support Yeast Prions and Thermoto lerance and Define Disaggregation Machinery Interactions. Genetics 2012, 192, 185–193.
  136. March, Z.M.; Sweeney, K.; Kim, H.; Yan, X.; Castellano, L.M.; Jackrel, M.E.; Lin, J.; Chuang, E.; Gomes, E.; Willicott, C.W.; et al. Therapeutic genetic variation revealed in diverse Hsp104 homologs. Elife 2020, 9, e57457.
  137. Michalska, K.; Zhang, K.; March, Z.M.; Hatzos-Skintges, C.; Pintilie, G.; Bigelow, L.; Castellano, L.M.; Miles, L.J.; Jackrel, M.E.; Chuang, E.; et al. Structure of Calcarisporiella thermophila Hsp104 Disaggregase that Antagonizes Diverse Proteotoxic Misfolding Events. Structure 2019, 27, 449–463.e7.
  138. Cupo, R.R.; Shorter, J. Skd3 (human ClpB) is a potent mitochondrial protein disaggregase that is inactivated by 3-methylglutaconic aciduria-linked mutations. Elife 2020, 9, e55279.
  139. Smith, D.M.; Benaroudj, N.; Goldberg, A. Proteasomes and their associated ATPases: A destructive combination. J. Struct. Biol. 2006, 156, 72–83.
  140. Brooks, C.; Snoberger, A.; Belcastro, M.; Murphy, J.; Kisselev, O.G.; Smith, D.M.; Sokolov, M. Archaeal Unfoldase Counteracts Protein Misfolding Retinopathy in Mice. J. Neurosci. 2018, 38, 7248–7254.
  141. Tittelmeier, J.; Sandhof, C.A.; Ries, H.M.; Druffel-Augustin, S.; Mogk, A.; Bukau, B.; Nussbaum-Krammer, C. The HSP110/HSP70 disaggregation system generates spreading-competent toxic α-synuclein species. EMBO J. 2020, 39, e103954.
  142. Demaegd, K.; Schymkowitz, J.; Rousseau, F. Transcellular Spreading of Tau in Tauopathies. Chembiochem 2018, 19, 2424–2432.
More
Video Production Service