Skip to main content
Top
Published in:

Open Access 01-12-2017 | Review

Protein misfolding in neurodegenerative diseases: implications and strategies

Authors: Patrick Sweeney, Hyunsun Park, Marc Baumann, John Dunlop, Judith Frydman, Ron Kopito, Alexander McCampbell, Gabrielle Leblanc, Anjli Venkateswaran, Antti Nurmi, Robert Hodgson

Published in: Translational Neurodegeneration | Issue 1/2017

Login to get access

Abstract

A hallmark of neurodegenerative proteinopathies is the formation of misfolded protein aggregates that cause cellular toxicity and contribute to cellular proteostatic collapse. Therapeutic options are currently being explored that target different steps in the production and processing of proteins implicated in neurodegenerative disease, including synthesis, chaperone-assisted folding and trafficking, and degradation via the proteasome and autophagy pathways. Other therapies, like mTOR inhibitors and activators of the heat shock response, can rebalance the entire proteostatic network. However, there are major challenges that impact the development of novel therapies, including incomplete knowledge of druggable disease targets and their mechanism of action as well as a lack of biomarkers to monitor disease progression and therapeutic response. A notable development is the creation of collaborative ecosystems that include patients, clinicians, basic and translational researchers, foundations and regulatory agencies to promote scientific rigor and clinical data to accelerate the development of therapies that prevent, reverse or delay the progression of neurodegenerative proteinopathies.
Footnotes
1
This review is based in part on an open scientific satellite symposium entitled “Protein Misfolding – Implications and Strategies” held in conjunction with the 45th Annual Society for Neuroscience meeting in 2015, sponsored and organized by Charles River Laboratories. The meeting panelists included: Hyunsun Park (Health & Life Science Consulting), Judith Frydman (Stanford University), John Dunlop (Astra Zeneca), Marc Baumann (University of Helsinki), Alexander McCampbell (Biogen), Ron Kopito (Stanford University), Patrick Sweeney (Royal Veterinary College, University of London, Charles River Laboratories) and Robert Hodgson (Charles River Laboratories).
 
Literature
2.
4.
go back to reference Knowles TP, Vendruscolo M, Dobson CM. The amyloid state and its association with protein misfolding diseases. Nature reviews. Mol Cell Biol. 2014;15:384–96. doi:10.1038/nrm3810. Knowles TP, Vendruscolo M, Dobson CM. The amyloid state and its association with protein misfolding diseases. Nature reviews. Mol Cell Biol. 2014;15:384–96. doi:10.​1038/​nrm3810.
5.
go back to reference Kallijarvi J, Haltia M, Baumann MH. Amphoterin includes a sequence motif which is homologous to the Alzheimer’s beta-amyloid peptide (Abeta), forms amyloid fibrils in vitro, and binds avidly to Abeta. Biochemistry. 2001;40:10032–7.PubMedCrossRef Kallijarvi J, Haltia M, Baumann MH. Amphoterin includes a sequence motif which is homologous to the Alzheimer’s beta-amyloid peptide (Abeta), forms amyloid fibrils in vitro, and binds avidly to Abeta. Biochemistry. 2001;40:10032–7.PubMedCrossRef
21.
go back to reference Myeku N, Clelland CL, Emrani S, Kukushkin NV, Yu WH, Goldberg AL, Duff KE. 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. doi:10.1038/nm.4011.PubMedCrossRef Myeku N, Clelland CL, Emrani S, Kukushkin NV, Yu WH, Goldberg AL, Duff KE. 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. doi:10.​1038/​nm.​4011.PubMedCrossRef
27.
go back to reference Wu JW, Hussaini SA, Bastille IM, Rodriguez GA, Mrejeru A, Rilett K, Sanders DW, Cook C, Fu H, Boonen RA, Herman M, Nahmani E, Emrani S, Figueroa YH, Diamond MI, Clelland CL, Wray S, Duff KE. Neuronal activity enhances tau propagation and tau pathology in vivo. Nat Neurosci. 2016;19(8):1085–92. doi:10.1038/nn.4328.PubMedPubMedCentralCrossRef Wu JW, Hussaini SA, Bastille IM, Rodriguez GA, Mrejeru A, Rilett K, Sanders DW, Cook C, Fu H, Boonen RA, Herman M, Nahmani E, Emrani S, Figueroa YH, Diamond MI, Clelland CL, Wray S, Duff KE. Neuronal activity enhances tau propagation and tau pathology in vivo. Nat Neurosci. 2016;19(8):1085–92. doi:10.​1038/​nn.​4328.PubMedPubMedCentralCrossRef
28.
go back to reference Fontaine SN, Zheng D, Sabbagh JJ, Martin MD, Chaput D, Darling A, Trotter JH, Stothert AR, Nordhues BA, Lussier A, Baker J, Shelton L, Kahn M, Blair LJ, Stevens Jr SM, Dickey CA. DnaJ/Hsc70 chaperone complexes control the extracellular release of neurodegenerative-associated proteins. EMBO J. 2016;35(14):1537–49. doi:10.15252/embj.201593489.PubMedCrossRef Fontaine SN, Zheng D, Sabbagh JJ, Martin MD, Chaput D, Darling A, Trotter JH, Stothert AR, Nordhues BA, Lussier A, Baker J, Shelton L, Kahn M, Blair LJ, Stevens Jr SM, Dickey CA. DnaJ/Hsc70 chaperone complexes control the extracellular release of neurodegenerative-associated proteins. EMBO J. 2016;35(14):1537–49. doi:10.​15252/​embj.​201593489.PubMedCrossRef
29.
go back to reference Lee JG, Takahama S, Zhang G, Tomarev SI, Ye Y. Unconventional secretion of misfolded proteins promotes adaptation to proteasome dysfunction in mammalian cells. Nat Cell Biol. 2016;18(7):765–76. doi:10.1038/ncb3372.PubMedCrossRef Lee JG, Takahama S, Zhang G, Tomarev SI, Ye Y. Unconventional secretion of misfolded proteins promotes adaptation to proteasome dysfunction in mammalian cells. Nat Cell Biol. 2016;18(7):765–76. doi:10.​1038/​ncb3372.PubMedCrossRef
30.
go back to reference Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, Brett FM, Farrell MA, Rowan MJ, Lemere CA, Regan CM, Walsh DM, Sabatini BL, Selkoe DJ. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837–42. doi:10.1038/nm1782.PubMedPubMedCentralCrossRef Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, Brett FM, Farrell MA, Rowan MJ, Lemere CA, Regan CM, Walsh DM, Sabatini BL, Selkoe DJ. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837–42. doi:10.​1038/​nm1782.PubMedPubMedCentralCrossRef
33.
go back to reference Thies E, Mandelkow EM. Missorting of tau in neurons causes degeneration of synapses that can be rescued by the kinase MARK2/Par-1. J Neurosci. 2007;27(11):2896–907.PubMedCrossRef Thies E, Mandelkow EM. Missorting of tau in neurons causes degeneration of synapses that can be rescued by the kinase MARK2/Par-1. J Neurosci. 2007;27(11):2896–907.PubMedCrossRef
35.
go back to reference Olzscha H, Schermann SM, Woerner AC, Pinkert S, Hecht MH, Tartaglia GG, Vendruscolo M, Hayer-Hartl M, Hartl FU, Vabular RM. Amyloid-like aggregates sequester numerous metastable proteins with essential cellular functions. Cell. 2011;144:67–78. doi:10.1016/j.cell.2010.11.050.PubMedCrossRef Olzscha H, Schermann SM, Woerner AC, Pinkert S, Hecht MH, Tartaglia GG, Vendruscolo M, Hayer-Hartl M, Hartl FU, Vabular RM. Amyloid-like aggregates sequester numerous metastable proteins with essential cellular functions. Cell. 2011;144:67–78. doi:10.​1016/​j.​cell.​2010.​11.​050.PubMedCrossRef
36.
go back to reference Woerner AC, Frottin F, Hornburg D, Feng LR, Meissner F, Patra M, Tatzelt J, Mann M, Winklhofer KF, Hartl FU, Hipp MS. Cytoplasmic protein aggregates interfere with nucleocytoplasmic transport of protein and RNA. Science. 2016;351(6269):173–6. doi:10.1126/science.aad2033.PubMedCrossRef Woerner AC, Frottin F, Hornburg D, Feng LR, Meissner F, Patra M, Tatzelt J, Mann M, Winklhofer KF, Hartl FU, Hipp MS. Cytoplasmic protein aggregates interfere with nucleocytoplasmic transport of protein and RNA. Science. 2016;351(6269):173–6. doi:10.​1126/​science.​aad2033.PubMedCrossRef
40.
44.
go back to reference Chakrabarty P, Li A, Ceballos-Diaz C, Eddy JA, Funk CC, Moore B, DiNunno N, Rosario AM, Cruz PE, Verbeeck C, Sacino A, Nix S, Janus C, Price ND, Das P, Golde TE. IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron. 2015;85(3):519–33. doi:10.1016/j.neuron.2014.11.020.PubMedPubMedCentralCrossRef Chakrabarty P, Li A, Ceballos-Diaz C, Eddy JA, Funk CC, Moore B, DiNunno N, Rosario AM, Cruz PE, Verbeeck C, Sacino A, Nix S, Janus C, Price ND, Das P, Golde TE. IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron. 2015;85(3):519–33. doi:10.​1016/​j.​neuron.​2014.​11.​020.PubMedPubMedCentralCrossRef
52.
go back to reference Pujol-Pina R, Vilaprinyo-Pascual S, Mazzucato R, Arcella A, Vilaseca M, Orozco M, Carulla N. SDS-PAGE analysis of Abeta oligomers is disserving research into Alzheimer s disease: appealing for ESI-IM-MS. Scientific reports. 2015;5:14809. doi:10.1038/srep14809.PubMedPubMedCentralCrossRef Pujol-Pina R, Vilaprinyo-Pascual S, Mazzucato R, Arcella A, Vilaseca M, Orozco M, Carulla N. SDS-PAGE analysis of Abeta oligomers is disserving research into Alzheimer s disease: appealing for ESI-IM-MS. Scientific reports. 2015;5:14809. doi:10.​1038/​srep14809.PubMedPubMedCentralCrossRef
54.
go back to reference Bleiholder C, Do TD, Wu C, Economou NJ, Bernstein SS, BUratto SK, Shea JE, Bowers MT. Ion mobility spectrometry reveals the mechanism of amyloid formation of Abeta (25-35) and its modulation by inhibitors at the molecular level: epigallocatechin gallate and scyllo-inositol. J Am Chem Soc. 2013;135:16926–37. doi:10.1021/ja406197f.PubMedCrossRef Bleiholder C, Do TD, Wu C, Economou NJ, Bernstein SS, BUratto SK, Shea JE, Bowers MT. Ion mobility spectrometry reveals the mechanism of amyloid formation of Abeta (25-35) and its modulation by inhibitors at the molecular level: epigallocatechin gallate and scyllo-inositol. J Am Chem Soc. 2013;135:16926–37. doi:10.​1021/​ja406197f.PubMedCrossRef
55.
go back to reference Neudecker P, Robustelli P, Cavalli A, Walsh P, Lundstrom P, Zarrine-Afsar A, Sharpe S, Vendruscolo M, Kay LE. Structure of an intermediate state in protein folding and aggregation. Science. 2012;336:362–6. doi:10.1126/science.1214203.PubMedCrossRef Neudecker P, Robustelli P, Cavalli A, Walsh P, Lundstrom P, Zarrine-Afsar A, Sharpe S, Vendruscolo M, Kay LE. Structure of an intermediate state in protein folding and aggregation. Science. 2012;336:362–6. doi:10.​1126/​science.​1214203.PubMedCrossRef
57.
go back to reference Kennedy ME, Stamford AW, Chen X, Cox K, Cumming JN, Dockendorf MF, Egan M, Ereshefsky L, Hodgson RA, Hyde LA, Jhee S, Kleijn HJ, Kuvelkar R, Li W, Mattson BA, Mei H, Palcza J, Scott JD, Tanen M, Troyer MD, Tseng JL, Stone JA, Parker EM, Forman MS. The BACE1 inhibitor verubecestat (MK-8931) reduces CNS β-amyloid in animal models and in Alzheimer’s disease patients. Sci Transl Med. 2016;8(363):363ra. 150.CrossRef Kennedy ME, Stamford AW, Chen X, Cox K, Cumming JN, Dockendorf MF, Egan M, Ereshefsky L, Hodgson RA, Hyde LA, Jhee S, Kleijn HJ, Kuvelkar R, Li W, Mattson BA, Mei H, Palcza J, Scott JD, Tanen M, Troyer MD, Tseng JL, Stone JA, Parker EM, Forman MS. The BACE1 inhibitor verubecestat (MK-8931) reduces CNS β-amyloid in animal models and in Alzheimer’s disease patients. Sci Transl Med. 2016;8(363):363ra. 150.CrossRef
58.
go back to reference Llorens-Marítin M, Jurado J, Hernández F, Avila J. GSK-3β, a pivotal kinase in Alzheimer disease. Front Mol Neurosci. 2014;7:46.PubMedCentral Llorens-Marítin M, Jurado J, Hernández F, Avila J. GSK-3β, a pivotal kinase in Alzheimer disease. Front Mol Neurosci. 2014;7:46.PubMedCentral
59.
go back to reference Roy SM, Grum-Tokars VL, Schavocky JP, Saeed F, Staniszewski A, Teich AF, Arancio O, Bachstetter AD, Webster SJ, Van Eldik LJ, Minasov G, Anderson WF, Pelletier JC, Watterson DM. Targeting human central nervous system protein kinases: An isoform selective p38αMAPK inhibitor that attenuates disease progression in Alzheimer’s disease mouse models. ACS Chem Neurosci. 2015;6(4):666–80. Epub 2015 Feb 23.PubMedPubMedCentralCrossRef Roy SM, Grum-Tokars VL, Schavocky JP, Saeed F, Staniszewski A, Teich AF, Arancio O, Bachstetter AD, Webster SJ, Van Eldik LJ, Minasov G, Anderson WF, Pelletier JC, Watterson DM. Targeting human central nervous system protein kinases: An isoform selective p38αMAPK inhibitor that attenuates disease progression in Alzheimer’s disease mouse models. ACS Chem Neurosci. 2015;6(4):666–80. Epub 2015 Feb 23.PubMedPubMedCentralCrossRef
60.
go back to reference Min SW, Chen X, Tracy TE, Li Y, Zhou Y, Wang C, Shirakawa K, Minami SS, Defensor E, Mok SA, Sohn PD, Schilling B, Cong X, Ellerby L, Gibson BW, Johnson J, Krogan N, Shamloo M, Gestwicki J, Masliah E, Verdin E, Gan L. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med. 2015;21(10):1154–62. doi:10.1038/nm.3951.PubMedPubMedCentralCrossRef Min SW, Chen X, Tracy TE, Li Y, Zhou Y, Wang C, Shirakawa K, Minami SS, Defensor E, Mok SA, Sohn PD, Schilling B, Cong X, Ellerby L, Gibson BW, Johnson J, Krogan N, Shamloo M, Gestwicki J, Masliah E, Verdin E, Gan L. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med. 2015;21(10):1154–62. doi:10.​1038/​nm.​3951.PubMedPubMedCentralCrossRef
64.
go back to reference Vilarino-Guell C, Rajput A, Milnerwood AJ, Shah B, Szu-Tu C, Trinh J, Yu I, Encarnacion M, Munsie LN, Tapia L, Gustavsson EK, Chou P, Tatarnikov I, Evans DM, Pishotta FT, Volta M, Beccano-Kelly D, Thompson C, Lin MK, Sherman HE, Han HJ, Guenther BL, Wasserman WW, Bernard V, Ross CJ, Appel-Cresswell S, Stoessl AJ, Robinson CA, Dickson DW, Ross OA, Wszolek ZK, Aasly JO, Wu RM, Hentati F, Gibson RA, McPherson PS, Girard M, Rajput M, Rajput AH, Farrer MJ. DNAJC13 mutations in Parkinson disease. Hum Mol Genet. 2014;23:1794–801. doi:10.1093/hmg/ddt570.PubMedCrossRef Vilarino-Guell C, Rajput A, Milnerwood AJ, Shah B, Szu-Tu C, Trinh J, Yu I, Encarnacion M, Munsie LN, Tapia L, Gustavsson EK, Chou P, Tatarnikov I, Evans DM, Pishotta FT, Volta M, Beccano-Kelly D, Thompson C, Lin MK, Sherman HE, Han HJ, Guenther BL, Wasserman WW, Bernard V, Ross CJ, Appel-Cresswell S, Stoessl AJ, Robinson CA, Dickson DW, Ross OA, Wszolek ZK, Aasly JO, Wu RM, Hentati F, Gibson RA, McPherson PS, Girard M, Rajput M, Rajput AH, Farrer MJ. DNAJC13 mutations in Parkinson disease. Hum Mol Genet. 2014;23:1794–801. doi:10.​1093/​hmg/​ddt570.PubMedCrossRef
66.
go back to reference Johnson JO, Mandrioli J, Benatar M, Abramzon Y, Van Deerlin VM, Trojanowski JQ, Gibbs JR, Brunetti M, Gronka S, Wuu J, Ding J, McCluskey L, Martinez-Lage M, Falcone D, Hernandez DG, Arepalli S, Chong S, Schymick JC, Rothstein J, Landi F, Wang YD, Calvo A, Mora G, Sabatelli M, Monsurrò MR, Battistini S, Salvi F, Spataro R, Sola P, Borghero G, Consortium ITALSGEN, Galassi G, Scholz SW, Taylor JP, Restagno G, Chiò A, Traynor BJ. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron. 2010;68:857–64. doi:10.1016/j.neuron.2010.11.036.PubMedPubMedCentralCrossRef Johnson JO, Mandrioli J, Benatar M, Abramzon Y, Van Deerlin VM, Trojanowski JQ, Gibbs JR, Brunetti M, Gronka S, Wuu J, Ding J, McCluskey L, Martinez-Lage M, Falcone D, Hernandez DG, Arepalli S, Chong S, Schymick JC, Rothstein J, Landi F, Wang YD, Calvo A, Mora G, Sabatelli M, Monsurrò MR, Battistini S, Salvi F, Spataro R, Sola P, Borghero G, Consortium ITALSGEN, Galassi G, Scholz SW, Taylor JP, Restagno G, Chiò A, Traynor BJ. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron. 2010;68:857–64. doi:10.​1016/​j.​neuron.​2010.​11.​036.PubMedPubMedCentralCrossRef
68.
71.
go back to reference Bose S, Cho J. Targeting chaperones, heat shock factor-1, and unfolded protein response: Promising therapeutic approaches for neurodegenerative disorders. Mol Psychiatry. 2016. doi:10.1038/mp.2016.104. Bose S, Cho J. Targeting chaperones, heat shock factor-1, and unfolded protein response: Promising therapeutic approaches for neurodegenerative disorders. Mol Psychiatry. 2016. doi:10.​1038/​mp.​2016.​104.
72.
go back to reference Fontaine SN, Martin MD, Dickey CA. Neurodegeneration and the Heat Shock Protein 70 Machinery: Implications for Therapeutic Development. Curr Top Med Chem. 2016;16(25):2741–52.PubMedCrossRef Fontaine SN, Martin MD, Dickey CA. Neurodegeneration and the Heat Shock Protein 70 Machinery: Implications for Therapeutic Development. Curr Top Med Chem. 2016;16(25):2741–52.PubMedCrossRef
73.
go back to reference Wang B, Liu Y, Huang L, Chen J, Li JJ, Wang R, Kim E, Chen Y, Justicia C, Sakata K, Chen H, Planas A, Ostrom RS, Li W, Yang G, McDonald MP, Chen R, Heck DH, Liao FF. A CNS-permeable Hsp90 inhibitor rescues synaptic dysfunction and memory loss in APP-overexpressing Alzheimer’s mouse model via an HSF1-mediated mechanism. Ageing Res Rev. 2016. doi:10.1016/j.arr.2016.09.004. Wang B, Liu Y, Huang L, Chen J, Li JJ, Wang R, Kim E, Chen Y, Justicia C, Sakata K, Chen H, Planas A, Ostrom RS, Li W, Yang G, McDonald MP, Chen R, Heck DH, Liao FF. A CNS-permeable Hsp90 inhibitor rescues synaptic dysfunction and memory loss in APP-overexpressing Alzheimer’s mouse model via an HSF1-mediated mechanism. Ageing Res Rev. 2016. doi:10.​1016/​j.​arr.​2016.​09.​004.
74.
go back to reference Martin MD, Baker JD, Suntharalingam A, Nordhues BA, Shelton LB, Zheng D, Sabbagh JJ, Haystead TA, Gestwicki JE, Dickey CA. Inhibition of Both Hsp70 Activity and Tau Aggregation in Vitro Best Predicts Tau Lowering Activity of Small Molecules. ACS Chem Biol. 2016;11(7):2041–8. doi:10.1021/acschembio.6b00223.PubMedCrossRef Martin MD, Baker JD, Suntharalingam A, Nordhues BA, Shelton LB, Zheng D, Sabbagh JJ, Haystead TA, Gestwicki JE, Dickey CA. Inhibition of Both Hsp70 Activity and Tau Aggregation in Vitro Best Predicts Tau Lowering Activity of Small Molecules. ACS Chem Biol. 2016;11(7):2041–8. doi:10.​1021/​acschembio.​6b00223.PubMedCrossRef
76.
go back to reference Liu YH, Han YL, Song J, Wang Y, Jing YY, Shi Q, Tian C, Zang ZY, Li CP, Han J, Dong XP. 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(5):768–74. doi:10.1016/j.biocel.2011.01.022. Liu YH, Han YL, Song J, Wang Y, Jing YY, Shi Q, Tian C, Zang ZY, Li CP, Han J, Dong XP. 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(5):768–74. doi:10.​1016/​j.​biocel.​2011.​01.​022.
82.
84.
go back to reference Opattova A, Cente M, Novak M, Filipcik P. The ubiquitin proteasome system as a potential therapeutic target for treatment of neurodegenerative diseases. Gen Physiol Biophys. 2015;34(4):337–52.PubMed Opattova A, Cente M, Novak M, Filipcik P. The ubiquitin proteasome system as a potential therapeutic target for treatment of neurodegenerative diseases. Gen Physiol Biophys. 2015;34(4):337–52.PubMed
91.
go back to reference Manzoni C, Mamais A, Roosen DA, Dihanich S, Soutar MP, Plun-Favreau H, Bandopadhyay R, Hardy J, Tooze SA, Cookson MR, Lewis PA. mTOR independent regulation of macroautophagy by Leucine Rich Repeat Kinase 2 via Beclin-1. Sci Rep. 2016;6:35106. doi:10.1038/srep35106.PubMedPubMedCentralCrossRef Manzoni C, Mamais A, Roosen DA, Dihanich S, Soutar MP, Plun-Favreau H, Bandopadhyay R, Hardy J, Tooze SA, Cookson MR, Lewis PA. mTOR independent regulation of macroautophagy by Leucine Rich Repeat Kinase 2 via Beclin-1. Sci Rep. 2016;6:35106. doi:10.​1038/​srep35106.PubMedPubMedCentralCrossRef
94.
go back to reference Savage MJ, Kalinina J, Wolfe A, Tugusheva K, Korn R, Cash-Mason T, Maxwell JW, Hatcher NG, Haugabook SJ, Wu G, Howell BJ, Renger JJ, Shughrue PJ, McCampbell A. A sensitive abeta oligomer assay discriminates Alzheimer’s and aged control cerebrospinal fluid. J Neurosci. 2014;34:2884–97. doi:10.1523/JNEUROSCI.1675-13.2014.PubMedCrossRef Savage MJ, Kalinina J, Wolfe A, Tugusheva K, Korn R, Cash-Mason T, Maxwell JW, Hatcher NG, Haugabook SJ, Wu G, Howell BJ, Renger JJ, Shughrue PJ, McCampbell A. A sensitive abeta oligomer assay discriminates Alzheimer’s and aged control cerebrospinal fluid. J Neurosci. 2014;34:2884–97. doi:10.​1523/​JNEUROSCI.​1675-13.​2014.PubMedCrossRef
96.
go back to reference Vaikath NN, Majbour NK, Paleologou KE, Ardah MT, van Dam E, van de Berg WD, Forrest SL, Parkkinen L, Gai WP, Hattori N, Takanashi M, Lee SJ, Mann DM, Imai Y, Halliday GM, Li JY, El-Agnaf OM. Generation and characterization of novel conformation-specific monoclonal antibodies for alpha-synuclein pathology. Neurobiol Dis. 2015;79:81–99. doi:10.1016/j.nbd.2015.04.009.PubMedCrossRef Vaikath NN, Majbour NK, Paleologou KE, Ardah MT, van Dam E, van de Berg WD, Forrest SL, Parkkinen L, Gai WP, Hattori N, Takanashi M, Lee SJ, Mann DM, Imai Y, Halliday GM, Li JY, El-Agnaf OM. Generation and characterization of novel conformation-specific monoclonal antibodies for alpha-synuclein pathology. Neurobiol Dis. 2015;79:81–99. doi:10.​1016/​j.​nbd.​2015.​04.​009.PubMedCrossRef
97.
go back to reference Deane R, Bell RD, Sagare R, Zlokovic BV. Clearance of amyloid-beta peptide across the blood-brain barrier: implication for therapies in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2009;8(1):16–30.PubMedPubMedCentralCrossRef Deane R, Bell RD, Sagare R, Zlokovic BV. Clearance of amyloid-beta peptide across the blood-brain barrier: implication for therapies in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2009;8(1):16–30.PubMedPubMedCentralCrossRef
98.
go back to reference Watt AD, Crespi GA, Down RA, Ascher DB, Gunn A, Perez KA, McLean CA, Villemagne VL, Parker MW, Barnham KJ, Miles LA. Do current therapeutic anti-Aβ antibodies for Alzheimer’s disease engage the target? Acta Neuropathol. 2014;127(6):803–10. doi:10.1007/s00401-014-1290-2.PubMedCrossRef Watt AD, Crespi GA, Down RA, Ascher DB, Gunn A, Perez KA, McLean CA, Villemagne VL, Parker MW, Barnham KJ, Miles LA. Do current therapeutic anti-Aβ antibodies for Alzheimer’s disease engage the target? Acta Neuropathol. 2014;127(6):803–10. doi:10.​1007/​s00401-014-1290-2.PubMedCrossRef
99.
go back to reference Bouter Y, Lopez Noguerola JS, Tucholla P, Crespi GA, Parker MW, Wiltfang J, Miles LA, Bayer TA. Abeta targets of the biosimilar antibodies of Bapineuzumab, Crenezumab, Solanezumab in comparison to an antibody against N-truncated Abeta in sporadic Alzheimer disease cases and mouse models. Acta Neuropathol. 2015;130(5):713–29.PubMedCrossRef Bouter Y, Lopez Noguerola JS, Tucholla P, Crespi GA, Parker MW, Wiltfang J, Miles LA, Bayer TA. Abeta targets of the biosimilar antibodies of Bapineuzumab, Crenezumab, Solanezumab in comparison to an antibody against N-truncated Abeta in sporadic Alzheimer disease cases and mouse models. Acta Neuropathol. 2015;130(5):713–29.PubMedCrossRef
102.
go back to reference Cramer PE, Cirrito JR, Wesson DW, Lee CY, Karlo JC, Zinn AE, Casali BT, Restivo JL, Goebel WD, James MJ, Brunden KR, Wilson DA, Landreth GE. ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models. Science. 2012;335:1503–6. doi:10.1126/science.1217697.PubMedPubMedCentralCrossRef Cramer PE, Cirrito JR, Wesson DW, Lee CY, Karlo JC, Zinn AE, Casali BT, Restivo JL, Goebel WD, James MJ, Brunden KR, Wilson DA, Landreth GE. ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models. Science. 2012;335:1503–6. doi:10.​1126/​science.​1217697.PubMedPubMedCentralCrossRef
107.
go back to reference Neef DW, Jaeger AM, Thiele DJ. Heat shock transcription factor 1 as a therapeutic target in neurodegenerative diseases. Nat Rev Drug Discov. 2011;10(12):930–44.PubMedPubMedCentralCrossRef Neef DW, Jaeger AM, Thiele DJ. Heat shock transcription factor 1 as a therapeutic target in neurodegenerative diseases. Nat Rev Drug Discov. 2011;10(12):930–44.PubMedPubMedCentralCrossRef
109.
go back to reference Kondo N, Katsuno M, Adachi H, Minamiyama M, Doi H, Matsumoto S, Miyazaki Y, Iida M, Tohnai G, Nakatsuji H, Ishigaki S, Fujioka Y, Watanabe H, Tanaka F, Nakai A, Sobue G. Heat shock factor-1 influences pathological lesion distribution of polyglutamine-induced neurodegeneration. Nat Commun. 2013;4:1405. doi:10.1038/ncomms2417.PubMedCrossRef Kondo N, Katsuno M, Adachi H, Minamiyama M, Doi H, Matsumoto S, Miyazaki Y, Iida M, Tohnai G, Nakatsuji H, Ishigaki S, Fujioka Y, Watanabe H, Tanaka F, Nakai A, Sobue G. Heat shock factor-1 influences pathological lesion distribution of polyglutamine-induced neurodegeneration. Nat Commun. 2013;4:1405. doi:10.​1038/​ncomms2417.PubMedCrossRef
112.
go back to reference Labbadia J, Cunliffe H, Weiss A, Katsyuba E, Sathasivam K, Seredenina T, Woodman B, Moussaoui S, Frentzel S, Luthi-Carter R, Paganetti P, Bates GP. Altered chromatin architecture underlies progressive impairment of the heat shock response in mouse models of Huntington disease. J Clin Invest. 2011;121:3306–19. doi:10.1172/JCI57413.PubMedPubMedCentralCrossRef Labbadia J, Cunliffe H, Weiss A, Katsyuba E, Sathasivam K, Seredenina T, Woodman B, Moussaoui S, Frentzel S, Luthi-Carter R, Paganetti P, Bates GP. Altered chromatin architecture underlies progressive impairment of the heat shock response in mouse models of Huntington disease. J Clin Invest. 2011;121:3306–19. doi:10.​1172/​JCI57413.PubMedPubMedCentralCrossRef
115.
go back to reference Shineman DW, Basi GS, Bizon JL, Colton CA, Greenberg BD, Hollister BA, Lincecum J, Leblanc GG, Lee LB, Luo F, Morgan D, Morse I, Refolo LM, Riddell DR, Scearce-Levie K, Sweeney P, Yrjänheikki J, Fillit HM. Accelerating drug discovery for Alzheimer’s disease: best practices for preclinical animal studies. Alzheimers Res Ther. 2011;3:28. doi:10.1186/alzrt90.PubMedPubMedCentralCrossRef Shineman DW, Basi GS, Bizon JL, Colton CA, Greenberg BD, Hollister BA, Lincecum J, Leblanc GG, Lee LB, Luo F, Morgan D, Morse I, Refolo LM, Riddell DR, Scearce-Levie K, Sweeney P, Yrjänheikki J, Fillit HM. Accelerating drug discovery for Alzheimer’s disease: best practices for preclinical animal studies. Alzheimers Res Ther. 2011;3:28. doi:10.​1186/​alzrt90.PubMedPubMedCentralCrossRef
118.
go back to reference Langfelder P, Cantle JP, Chatzopoulou D, Wang N, Gao F, Al-Ramahi I, Lu XH, Ramos EM, El-Zein K, Zhao Y, Deverasetty S, Tebbe A, Schaab C, Lavery DJ, Howland D, Kwak S, Botas J, Aaronson JS, Rosinski J, Coppola G, Horvath S, Yang XW. Integrated genomics and proteomics define huntingtin CAG length-dependent networks in mice. Nat Neurosci. 2016;19(4):623–33. doi:10.1038/nn.4256.PubMedCrossRef Langfelder P, Cantle JP, Chatzopoulou D, Wang N, Gao F, Al-Ramahi I, Lu XH, Ramos EM, El-Zein K, Zhao Y, Deverasetty S, Tebbe A, Schaab C, Lavery DJ, Howland D, Kwak S, Botas J, Aaronson JS, Rosinski J, Coppola G, Horvath S, Yang XW. Integrated genomics and proteomics define huntingtin CAG length-dependent networks in mice. Nat Neurosci. 2016;19(4):623–33. doi:10.​1038/​nn.​4256.PubMedCrossRef
119.
go back to reference Yamanaka T, Tosaki A, Miyazaki H, Kurosawa M, Koike M, Uchiyama Y, Maity SN, Misawa H, Takahashi R, Shimogori T, Hattori N, Nukina N. Differential roles of NF-Y transcription factor in ER chaperone expression and neuronal maintenance in the CNS. Sci Rep. 2016;6:34575. doi:10.1038/srep34575.PubMedPubMedCentralCrossRef Yamanaka T, Tosaki A, Miyazaki H, Kurosawa M, Koike M, Uchiyama Y, Maity SN, Misawa H, Takahashi R, Shimogori T, Hattori N, Nukina N. Differential roles of NF-Y transcription factor in ER chaperone expression and neuronal maintenance in the CNS. Sci Rep. 2016;6:34575. doi:10.​1038/​srep34575.PubMedPubMedCentralCrossRef
121.
go back to reference Benraiss A, Wang S, Herrlinger S, Li X, Chandler-Militello D, Mauceri J, Burm HB, Toner M, Osipovitch M, Jim Xu Q, Ding F, Wang F, Kang N, Kang J, Curtin PC, Brunner D, Windrem MS, Munoz-Sanjuan I, Nedergaard M, Goldman SA. Human glia can both induce and rescue aspects of disease phenotype in Huntington disease. Nat Commun. 2016;7:11758. doi:10.1038/ncomms11758.PubMedPubMedCentralCrossRef Benraiss A, Wang S, Herrlinger S, Li X, Chandler-Militello D, Mauceri J, Burm HB, Toner M, Osipovitch M, Jim Xu Q, Ding F, Wang F, Kang N, Kang J, Curtin PC, Brunner D, Windrem MS, Munoz-Sanjuan I, Nedergaard M, Goldman SA. Human glia can both induce and rescue aspects of disease phenotype in Huntington disease. Nat Commun. 2016;7:11758. doi:10.​1038/​ncomms11758.PubMedPubMedCentralCrossRef
122.
123.
go back to reference Catafau AM, Bullich S. Non-amyloid PET imaging biomarkers for neurodegeneration: Focus on tau, alpha-synuclein and neuroinflamation. Curr Alzheimer Res. 2016. [Epub ahead of print]. Catafau AM, Bullich S. Non-amyloid PET imaging biomarkers for neurodegeneration: Focus on tau, alpha-synuclein and neuroinflamation. Curr Alzheimer Res. 2016. [Epub ahead of print].
127.
go back to reference Finneman SJ, Nabulsi NB, Eid T, Detyniecki K, Lin S, Chen M, Dhaher R, Matuskey D, Baum E, Holden D, Spencer DD, Mercier K, Hannestad J, Huang Y, Carson RE. Imaging synaptic density in the living human brain. Sci Transl Med. 2016;8(348):348ra96. doi:10.1126/scitranslmed.aaf6667.CrossRef Finneman SJ, Nabulsi NB, Eid T, Detyniecki K, Lin S, Chen M, Dhaher R, Matuskey D, Baum E, Holden D, Spencer DD, Mercier K, Hannestad J, Huang Y, Carson RE. Imaging synaptic density in the living human brain. Sci Transl Med. 2016;8(348):348ra96. doi:10.​1126/​scitranslmed.​aaf6667.CrossRef
128.
go back to reference Morris M, Sanchez PE, Verret L, Beagle AJ, Guo W, Dubal D, Ranasinghe KG, Koyama A, Ho K, Yu GQ, Vossel KA, Mucke L. Network dysfunction in α-synuclein transgenic mice and human Lewy body dementia. Ann Clin Transl Neurol. 2015;2(11):1012–28. doi:10.1002/acn3.257.PubMedPubMedCentralCrossRef Morris M, Sanchez PE, Verret L, Beagle AJ, Guo W, Dubal D, Ranasinghe KG, Koyama A, Ho K, Yu GQ, Vossel KA, Mucke L. Network dysfunction in α-synuclein transgenic mice and human Lewy body dementia. Ann Clin Transl Neurol. 2015;2(11):1012–28. doi:10.​1002/​acn3.​257.PubMedPubMedCentralCrossRef
129.
Metadata
Title
Protein misfolding in neurodegenerative diseases: implications and strategies
Authors
Patrick Sweeney
Hyunsun Park
Marc Baumann
John Dunlop
Judith Frydman
Ron Kopito
Alexander McCampbell
Gabrielle Leblanc
Anjli Venkateswaran
Antti Nurmi
Robert Hodgson
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Translational Neurodegeneration / Issue 1/2017
Electronic ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-017-0077-5