1,587
Views
1
CrossRef citations to date
0
Altmetric
Research Paper

Synthesis and anti-prion aggregation activity of acylthiosemicarbazide analogues

, , , , , , , , , , , , , , , & show all
Article: 2191164 | Received 15 Nov 2022, Accepted 10 Mar 2023, Published online: 23 Mar 2023

References

  • Prusiner SB. Prions. Proc Natl Acad Sci USA. 1998;95(23):13363–13383.
  • Ryou C. Prions and prion diseases: fundamentals and mechanistic details. J Microbiol Biotechnol. 2007;17(7):1059–1070.
  • Prusiner SB. The prion diseases. Brain Pathol. 1998;8(3):499–513.
  • Ritchie DL, Ironside JW. Neuropathology of human prion diseases. Prog Mol Biol Transl Sci. 2017;150:319–339.
  • Lakkaraju AKK, Frontzek K, Lemes E, Herrmann U, Losa M, Marpakwar R, Aguzzi A. Loss of PIKfyve drives the spongiform degeneration in prion diseases. EMBO Mol Med. 2021;13(9):e14714.
  • Frontzek K, Bardelli M, Senatore A, Henzi A, Reimann RR, Bedir S, Marino M, Hussain R, Jurt S, Meisl G, et al. A conformational switch controlling the toxicity of the prion protein. Nat Struct Mol Biol. 2022;29(8):831–840.
  • Tuite MF, Serio TR. The prion hypothesis: from biological anomaly to basic regulatory mechanism. Nat Rev Mol Cell Biol. 2010;11(12):823–833.
  • Ryou C, Mays CE. Prion propagation in vitro: are we there yet? Int J Med Sci. 2008;5(6):347–353.
  • Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science. 1982;216(4542):136–144.
  • Kraus A, Hoyt F, Schwartz CL, Hansen B, Artikis E, Hughson AG, Raymond GJ, Race B, Baron GS, Caughey B. High-resolution structure and strain comparison of infectious mammalian prions. Mol Cell. 2021;81(21):4540–4551.e6.
  • Hoyt F, Standke HG, Artikis E, Schwartz CL, Hansen B, Li K, Hughson AG, Manca M, Thomas OR, Raymond GJ, et al. Cryo-EM structure of anchorless RML prion reveals variations in shared motifs between distinct strains. Nat Commun. 2022;13(1):4005.
  • Safar J, Prusiner SB. Molecular studies of prion diseases. Prog Brain Res. 1998;117:421–434.
  • Supattapone S, Nguyen HO, Cohen FE, Prusiner SB, Scott MR. Elimination of prions by branched polyamines and implications for therapeutics. Proc Natl Acad Sci USA. 1999;96(25):14529–14534.
  • Solassol J, Crozet C, Perrier V, Leclaire J, Beranger F, Caminade AM, Meunier B, Dormont D, Majoral JP, Lehmann S. Cationic phosphorus-containing dendrimers reduce prion replication both in cell culture and in mice infected with scrapie. J Gen Virol. 2004;85(Pt 6):1791–1799.
  • Haïk S, Marcon G, Mallet A, Tettamanti M, Welaratne A, Giaccone G, Azimi S, Pietrini V, Fabreguettes J-R, Imperiale D, et al. Doxycycline in Creutzfeldt-Jakob disease: a phase 2, randomized, double-blind, placebo-controlled trial. Lancet Neurol. 2014;13(2):150–158.
  • Lim YB, Mays CE, Kim Y, Titlow WB, Ryou C. The inhibition of prions through blocking prion conversion by permanently charged branched polyamines of low cytotoxicity. Biomaterials. 2010;31(8):2025–2033.
  • Ryou C, Legname G, Peretz D, Craig JC, Baldwin MA, Prusiner SB. Differential inhibition of prion propagation by enantiomers of quinacrine. Lab Invest. 2003;83(6):837–843.
  • Murakami-Kubo I, Doh-Ura K, Ishikawa K, Kawatake S, Sasaki K, Kira JI, Ohta S, Iwaki T. Quinoline derivatives are therapeutic candidates for transmissible spongiform encephalopathies. J Virol. 2004;78(3):1281–1288.
  • Shimizu Y, Kaku-Ushiki Y, Iwamaru Y, Muramoto T, Kitamoto T, Yokoyama T, Mohri S, Tagawa Y. A novel anti-prion protein monoclonal antibody and its single-chain fragment variable derivative with ability to inhibit abnormal prion protein accumulation in cultured cells. Microbiol Immunol. 2010;54(2):112–121.
  • Kim DH, Ren C, Ryou C, Li J. Direct interaction of DNMT inhibitors to PrP(C) suppresses pathogenic process of prion. Acta Pharm Sin B. 2019;9(5):952–959.
  • Kuwata K, Nishida N, Matsumoto T, Kamatari YO, Hosokawa-Muto J, Kodama K, Nakamura HK, Kimura K, Kawasaki M, Takakura Y, et al. Hot spots in prion protein for pathogenic conversion. Proc Natl Acad Sci USA. 2007;104(29):11921–11926.
  • Kimura T, Hosokawa-Muto J, Kamatari YO, Kuwata K. Synthesis of GN8 derivatives and evaluation of their antiprion activity in TSE-infected cells. Bioorg Med Chem Lett. 2011;21(5):1502–1507.
  • Korth C, May BC, Cohen FE, Prusiner SB. Acridine and phenothiazine derivatives as pharmacotherapeutics for prion disease. Proc Natl Acad Sci USA. 2001;98(17):9836–9841.
  • Ishibashi D, Nakagaki T, Ishikawa T, Atarashi R, Watanabe K, Cruz FA, Hamada T, Nishida N. Structure-based drug discovery for prion disease using a novel binding simulation. eBiomedicine. 2016;9:238–249.
  • Li JJ, Ryou C, Kim DH. Effects of SGI-1027 on formation and elimination of PrPSc in prion-infected cells. Mol Biol. 2020;54(3):412–415.
  • Telling GC, Scott M, Mastrianni J, Gabizon R, Torchia M, Cohen FE, DeArmond SJ, Prusiner SB. Prion propagation in mice expressing human and chimeric PrP transgenes implicates the interaction of cellular PrP with another protein. Cell. 1995;83(1):79–90.
  • Kaneko K, Zulianello L, Scott M, Cooper CM, Wallace AC, James TL, Cohen FE, Prusiner SB. Evidence for protein X binding to a discontinuous epitope on the cellular prion protein during scrapie prion propagation. Proc Natl Acad Sci USA. 1997;94(19):10069–10074.
  • Mays CE, Ryou C. Plasminogen stimulates propagation of protease-resistant prion protein in vitro. Faseb J. 2010;24(12):5102–5112.
  • Baldeschi AC, Zattoni M, Vanni S, Nikolic L, Ferracin C, La Sala G, Summa M, Bertorelli R, Bertozzi SM, Giachin G, et al. Innovative non-PrP-targeted drug strategy designed to enhance prion clearance. J Med Chem. 2022;65(13):8998–9010.
  • Ali T, Hannaoui S, Nemani S, Tahir W, Zemlyankina I, Cherry P, Shim SY, Sim V, Schaetzl HM, Gilch S. Oral administration of repurposed drug targeting CYP46A1 increases survival times of prion infected mice. Acta Neuropathol Commun. 2021;9(1):58.
  • Ryou C, Titlow WB, Mays CE, Bae Y, Kim S. The suppression of prion propagation using poly-L-lysine by targeting plasminogen that stimulates prion protein conversion. Biomaterials. 2011;32(11):3141–3149.
  • Barreca ML, Iraci N, Biggi S, Cecchetti V, Biasini E. Pharmacological agents targeting the cellular prion protein. Pathogens. 2018;7(1):27.
  • Trevitt CR, Collinge J. A systemic review of prion therapeutics in experimental models. Brain. 2006;129(9):2241–2265.
  • Choi H, Yun W, Lee JH, Jang S, Park SW, Kim DH, Seon KP, Hyun JM, Jeong K, Ku JM, et al. Synthesis and anti-endoplasmic reticulum stress activity of N-substituted-2-arylcarbonylhydrazinecarbothioamides. Med Chem Res. 2019;28(12):2142–2152.
  • Doyle KM, Kennedy D, Gorman AM, Gupta S, Healy SJ, Samali A. Unfolded proteins and endoplasmic reticulum stress in neurodegenerative disorders. J Cell Mol Med. 2011;15(10):2025–2039.
  • Yoshida H. ER stress and diseases. Febs J. 2007;274(3):630–658.
  • Sano R, Reed JC. ER stress-induced cell death mechanisms. Biochim Biophys Acta. 2013;1833(12):3460–3470.
  • Jeon JH, Im S, Kim HS, Lee D, Jeong K, Ku JM, Nam TG. Chemical chaperones to inhibit endoplasmic reticulum stress: implications in diseases. DDDT. 2022;16:4385–4397.
  • Kim SG, Lee HM, Ryou C. Parameters that affect macromolecular self-assembly of prion protein. Protein J. 2014;33(3):243–252.
  • Bocharova OV, Breydo L, Parfenov AS, Salnikov VV, Baskakov IV. In vitro conversion of full-length mammalian prion protein produces amyloid form with physical properties of PrP(Sc). J Mol Biol. 2005;346(2):645–659.
  • Hwang HG, Kim DH, Lee J, Mo Y, Lee SH, Lee Y, Hyeon JW, Lee SM, Cheon YP, Choi EK, et al. High-level production of high-purity human and murine recombinant prion proteins functionally compatible to in vitro seeding assay. J Microbiol Biotechnol. 2018;28(10):1749–1759.
  • Dean DN, Das PK, Rana P, Burg F, Levites Y, Morgan SE, Ghosh P, Rangachari V. Strain-specific fibril propagation by an Abeta dodecamer. Sci Rep. 2017;7(1):40787.
  • Halfmann R, Lindquist S. Screening for amyloid aggregation by semi-denaturing detergent-agarose gel electrophoresis. J Vis Exp. 2008;17:838.
  • Wilham JM, Orru CD, Bessen RA, Atarashi R, Sano K, Race B, Meade-White KD, Taubner LM, Timmes A, Caughey B. Rapid end-point quantitation of prion seeding activity with sensitivity comparable to bioassays. PLoS Pathog. 2010;6(12):e1001217.
  • Atarashi R, Wilham JM, Christensen L, Hughson AG, Moore RA, Johnson LM, Onwubiko HA, Priola SA, Caughey B. Simplified ultrasensitive prion detection by recombinant PrP conversion with shaking. Nat Methods. 2008;5(3):211–212.
  • Waqas M, Lee HM, Kim J, Telling G, Kim JK, Kim DH, Ryou C. Effect of poly-L-arginine in inhibiting scrapie prion protein of cultured cells. Mol Cell Biochem. 2017;428(1–2):57–66.
  • Lee S, Lee H, Kim J, Kim JH, Gao EM, Lee Y, Yoo M, Trinh THT, Kim J, Kim CY, et al. The effect of Curcuma phaeocaulis Valeton (Zingiberaceae) extract on prion propagation in cell-based and animal models. IJMS. 2022;24(1):182.
  • Biancalana M, Koide S. Molecular mechanism of thioflavin-T binding to amyloid fibrils. Biochim Biophys Acta. 2010;1804(7):1405–1412.
  • Kang HE, Mo Y, Abd Rahim R, Lee HM, Ryou C. Prion diagnosis: application of real-time quaking-induced conversion. Biomed Res Int. 2017;2017:1–8.
  • Cheng K, Sloan A, Waitt B, Vendramelli R, Godal D, Simon SLR, O'Neil J, Carpenter M, Jackson D, Eastlake J, et al. Altered rPrP substrate structures and their influence on real-time quaking induced conversion reactions. Protein Expr Purif. 2018;143:20–27.
  • Abskharon R, Dang J, Elfarash A, Wang Z, Shen P, Zou LS, Hassan S, Wang F, Fujioka H, Steyaert J, et al. Soluble polymorphic bank vole prion proteins induced by co-expression of quiescin sulfhydryl oxidase in E. coli and their aggregation behaviors. Microb Cell Fact. 2017;16(1):170.
  • Yen CF, Harischandra DS, Kanthasamy A, Sivasanka S. Copper-induced structural conversion templates prion protein oligomerization and neurotoxicity. Sci Adv. 2016;2(7):e1600014.
  • Kim YE, Kim DH, Choi A, Jang S, Jeong K, Kim YM, Nam TG. Bi-aryl analogues of salicylic acids: design, synthesis and SAR study to ameliorate endoplasmic reticulum stress. DDDT. 2021;15:3593–3604.
  • Simoneau S, Rezaei H, Salès N, Kaiser-Schulz G, Lefebvre-Roque M, Vidal C, Fournier JG, Comte J, Wopfner F, Grosclaude J, et al. In vitro and in vivo neurotoxicity of prion protein oligomers. PLoS Pathog. 2007;3(8):e125.