688
Views
4
CrossRef citations to date
0
Altmetric
Review

Neuroprotective effect and potential of cellular prion protein and its cleavage products for treatment of neurodegenerative disorders part I. a literature review

&
Pages 969-982 | Received 26 May 2021, Accepted 05 Aug 2021, Published online: 02 Sep 2021

References

  • Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science. 1982;216(4542):136–144.
  • Colby DW, Prusiner SB. Prions. Cold Spring Harb Perspect Biol. 2011;3(1):a006833.
  • Sigurdson CJ, Bartz JC, Glatzel M, et al. Cellular and molecular mechanisms of prion disease. Annu Rev Pathol. 2019 Jan 24;14(1):497–516.
  • Castle AR, Gill AC. Physiological functions of the cellular prion protein. Front Mol Biosci. 2017;4:19.
  • Linden R. The biological function of the prion protein: a cell surface scaffold of signaling modules. Front Mol Neurosci. 2017;10:77.
  • Chiarini LB, Freitas AR, Zanata SM, et al. Cellular prion protein transduces neuroprotective signals. EMBO J. 2002;21:3317–3326.
  • Chen S, Mangé A, Dong L, et al. Prion protein as trans-interacting partner for neurons is involved in neurite outgrowth and neuronal survival. Mol Cell Neurosci. 2003;22:227–233.
  • Lopes MH, Hajj GN, Muras AG, et al. Interaction of cellular prion and stress-inducible protein 1 promotes neuritogenesis and neuroprotection by distinct signaling pathways. J Neurosci. 2005;25:11330–11339.
  • Brown DR, Nicholas RSJ, Canevari L, et al. Lack of prion protein expression results in a neuronal phenotype sensitive to stress. J Neurosci Res. 2002;67:211–224.
  • McLennan NF, Brennan PM, McNeill A, et al. Prion protein accumulation and neuroprotection in hypoxic brain damage. Am J Pathol. 2004;165(1):227–235.
  • Mitteregger G, Vosko M, Krebs B, et al. The role of the octarepeat region in neuroprotective function of the cellular prion protein. Brain Pathol. 2007 Apr;17(2):174–183.
  • Krebs B, Wiebelitz A, Balitzki-Korte B, et al. Cellular prion protein modulates the intracellular calcium response to hydrogen peroxide. J Neurochem. 2007;100(2):358–367.
  • Rambold AS, Müller V, Ron U, et al. Stress-protective signaling of prion protein is corrupted by scrapie prions. EMBO J. 2008;27:1974–1984.
  • Zhang B, Cowden D, Zhang F, et al. Prion protein protects against renal ischemia/reperfusion injury. PLoS One. 2015;10(9):e0136923.
  • Khosravani H, Zhang Y, Tsutsui S, et al. Prion protein attenuates excitotoxicity by inhibiting NMDA receptors. J Cell Biol. 2008 May 5;181(3):551–565.
  • Beraldo FH, Arantes CP, Santos TG, et al. Role of alpha7 nicotinic acetylcholine receptor in calcium signaling induced by prion protein interaction with stress-inducible protein 1. J Biol Chem. 2019;285:36542–36550.
  • Carulla P, Bribián A, Rangel A, et al. Neuroprotective role of PrPC against kainate-induced epileptic seizures and cell death depends on the modulation of JNK3 activation by GluR6/7-PSD-95 binding. Mol Biol Cell. 2011;22:3041–3054.
  • Alfaidy N, Chauvet S, Donadio-Andrei S, et al. Prion protein expression and functional importance in developmental angiogenesis: role in oxidative stress and copper homeostasis. Antioxid Redox Signal. 2013;18(4):400–411.
  • Gasperini L, Meneghetti E, Pastore B, et al. Prion protein and copper cooperatively protect neurons by modulating NMDA receptor through S-nitrosylation. Antioxid Redox Signal. 2015;22:772–784.
  • Bremer J, Baumann F, Tiberi C, et al. Axonal prion protein is required for peripheral myelin maintenance. Nat Neurosci. 2010;13(3):310–318.
  • Santuccione A, Sytnyk V, Leshchyns’ka I, et al. Prion protein recruits its neuronal receptor NCAM to lipid rafts to activate p59fyn and to enhance neurite outgrowth. J Cell Biol. 2005 Apr 25;169(2):341–354.
  • Mehrabian M, Brethour D, MacIsaac S, et al. CRISPR-Cas9-based knockout of the prion protein and its effect on the proteome. PLoS One. 2014;9(12):e114594.
  • Mehrabian M, Brethour D, Wang H, et al. The prion protein controls polysialylation of neural cell adhesion molecule 1 during cellular morphogenesis. PLoS One. 2015;10(8):e0133741.
  • Mehrabian M, Brethour D, Williams D, et al. Prion protein deficiency causes diverse proteome shifts in Cell models that escape detection in brain tissue. PLoS One. 2016;11(6):e0156779.
  • Haddon DJ, Hughes MR, Antignano F, et al. Prion protein expression and release by mast cells after activation. J Infect Dis. 2009;200:827–831.
  • Chatterjea D, Martinov T. Mast cells: versatile gatekeepers of pain. Mol Immunol. 2015;63:38–44.
  • Tripathi AK, Haldar S, Qian J, et al. Prion protein functions as a ferrireductase partner for ZIP14 and DMT1. Free Radic Biol Med. 2015;84:322–330.
  • Tobler I, Deboer T, Fischer M, et al. Sleep and sleep regulation in normal and prion protein-deficient mice. J Neurosci. 1997;17:1869–1879.
  • Tobler I, Gaus SE, Deboer T, et al. Altered circadian activity rhythms and sleep in mice devoid of prion protein. Nature. 1996;380(6575):639–642.
  • Cagampang FRA, Whatley SA, Mitchell AL, et al. Circadian regulation of prion protein messenger RNA in the rat forebrain: a widespread and synchronous rhythm. Neuroscience. 1999;91(4):1201–1204.
  • Aguzzi A, Heikenwalder M. Pathogenesis of prion diseases: current status and future outlook. Nat Rev Microbiol. 2006;4(10):765–775.
  • Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, et al. Tau oligomers impair memory and induce synaptic and mitochondrial dysfunction in wild-type mice. Mol Neurodegener. 2011;6(1):39.
  • Castillo-Carranza DL, Gerson JE, Sengupta U, et al. Specific targeting of tau oligomers in Htau mice prevents cognitive impairment and tau toxicity following injection with brain-derived tau oligomeric seeds. J Alzheimers Dis. 2014;40:S97–S111.
  • Salazar SV, Strittmatter SM. Cellular prion protein as a receptor for amyloid-β oligomers in Alzheimer’s disease. Biochem Biophys Res Commun. 2017;483:1143–1147.
  • Corbett GT, Wang Z, Hong W, et al. PrP is a central player in toxicity mediated by soluble aggregates of neurodegeneration-causing proteins. Acta Neuropathol. 2020;139(3): 503–526.
  • La Vitola P, Beeg M, Balducci C, et al. Cellular prion protein neither binds to alpha-synuclein oligomers nor mediates their detrimental effects. Brain. 2019;142(2):249–254.
  • Büeler H, Fischer M, Lang Y, et al. Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature. 1992;356(6370): 577–582.
  • Manson JC, Clarke AR, Hooper ML, et al. 129/ola mice carrying a null mutation in PrP that abolishes mRNA production are developmentally normal. Mol Neurobiol. 1994;8(2–3):121–127.
  • Weissmann C, Flechsig E. PrP knock-out and PrP transgenic mice in prion research. Br Med Bull. 2003;66(1):43–60.
  • Steele AD, Lindquist S, Aguzzi A, et al. The prion protein knockout mouse: a phenotype under challenge. Prion. 2007;1:83–93.
  • Richt JA, Kasinathan P, Hamir AN, et al. Production of cattle lacking prion protein. Nat Biotechnol. 2007;25(1): 132–138.
  • Benestad SL, Austbø L, Tranulis MA, et al. Healthy goats naturally devoid of prion protein. Vet Res. 2012;43(1):87.
  • Skedsmo FS, Malachin G, Våge DI, et al. Demyelinating polyneuropathy in goats lacking prion protein. FASEB J. 2020;34(2):2359–2375.
  • Criado JR, Sánchez-Alavez M, Conti B, et al. Mice devoid of prion protein have cognitive deficits that are rescued by reconstitution of PrP in neurons. Neurobiol Dis. 2005;19(1–2):255–265.
  • Wulf MA, Senatore A, Aguzzi A, et al. The biological function of the cellular prion protein: an update. BMC Biol. 2017;15:34.
  • Bendheim PE, Brown HR, Rudelli RD, et al. Nearly ubiquitous tissue distribution of the scrapie agent precursor protein. Neurology. 1992;42:149–156.
  • Lainé J, Marc ME, Sy MS, et al. Cellular and subcellular morphological localization of normal prion protein in rodent cerebellum. Eur J Neurosci. 2001;14:47–56.
  • Liu T, Li R, Wong BS, et al. Normal cellular prion protein is preferentially expressed on subpopulations of murine hemopoietic cells. J Immunol. 2001a;166:3733–3742.
  • Liu T, Zwingman T, Li R, et al. Differential expression of cellular prion protein in mouse brain as detected with multiple anti-PrP monoclonal antibodies. Brain Res. 2001b;896:118–129.
  • Miele G, Alejo Blanco AR, Baybutt H, et al. Embryonic activation and developmental expression of the murine prion protein gene. Gene Expr. 2003;11:1–12.
  • Shmakov AN, McLennan NF, McBride P, et al. Cellular prion protein is expressed in the human enteric nervous system. Nat Med. 2000;6(8):840–841.
  • Tremblay P, Bouzamondo-Bernstein E, Heinrich C, et al. Developmental expression of PrP in the post-implantation embryo. Brain Res. 2007;1139:60–67.
  • Riek R, Hornemann S, Wider G, et al. NMR structure of the mouse prion protein domain PrP(121-231). Nature. 1996;382:180–182.
  • Riek R, Hornemann S, Wider G, et al. NMR characterization of the full-length recombinant murine prion protein, mPrP(23-231). FEBS Lett. 1997;413:282–288.
  • Zahn R, Liu A, Lührs T, et al. NMR solution structure of the human prion protein. Proc Natl Acad Sci U S A. 2000;97(1):145–150.
  • Pan KM, Baldwin M, Nguyen J, et al. Conversion of alpha-helices into beta-sheets features in the formation of the scrapie prion proteins. Proc Natl Acad Sci U S A. 1993;90(23):10962–10966.
  • Marín-Moreno A, Fernández-Borges N, Espinosa JC, et al. Transmission and replication of prions. Prog Mol Biol Transl Sci. 2017;150:181–201.
  • Yim YI, Park BC, Yadavalli R, et al. The multivesicular body is the major internal site of prion conversion. J Cell Sci. 2015;128:1434–1443.
  • Huang P, Lian F, Wen Y, et al. Prion protein oligomer and its neurotoxicity. Acta Biochim Biophys Sin (Shanghai). 2013;45(6):442–451.
  • Benilova I, Reilly M, Terry C, et al. Highly infectious prions are not directly neurotoxic. Proc Natl Acad Sci U S A. 2020;117(38):23815–23822.
  • Shafiq M, Zafar S, Younas N, et al. Prion protein oligomers cause neuronal cytoskeletal damage in rapidly progressive alzheimer’s disease. Mol Neurodegener. 2021;16(1):11.
  • Chen SG, Teplow DB, Parchi P, et al. Truncated forms of the human prion protein in normal brain and in prion diseases. J Biol Chem. 1995;270:19173–19180.
  • Jimenez-Huete A, Lievens PM, Vidal R, et al. Endogenous proteolytic cleavage of normal and disease-associated isoforms of the human prion protein in neural and non-neural tissues. Am J Pathol. 1998;153:1561–1572.
  • Mange A, Beranger F, Peoc’h K, et al. Alpha- and beta- cleavages of the amino-terminus of the cellular prion protein. Biol Cell. 2004;96:125–132.
  • Tagliavini F, Prelli F, Porro M, et al. A soluble form of prion protein in human cerebrospinal fluid: implications for prion-related encephalopathies. Biochem Biophys Res Commun. 1992;184(3):1398–1404.
  • Borchelt DR, Rogers M, Stahl N, et al. Release of the cellular prion protein from cultured cells after loss of its glycoinositol phospholipid anchor. Glycobiology. 1993;3(4):319–329.
  • Parizek P, Roeckl C, Weber J, et al. Similar turnover and shedding of the cellular prion protein in primary lymphoid and neuronal cells. J Biol Chem. 2001;276:44627–44632.
  • Taylor DR, Parkin ET, Cocklin SL, et al. Role of ADAMs in the ectodomain shedding and conformational conversion of the prion protein. J Biol Chem. 2009;284(34):22590–22600.
  • Watt NT, Hooper NM. Reactive oxygen species (ROS)-mediated beta-cleavage of the prion protein in the mechanism of the cellular response to oxidative stress. Biochem Soc Trans. 2005;33:1123–1125.
  • Collins SJ, Tumpach C, Groveman BR, et al. Prion protein cleavage fragments regulate adult neural stem cell quiescence through redox modulation of mitochondrial fission and SOD2 expression. Cell Mol Life Sci. 2018;75(17):3231–3249.
  • McDonald AJ, Dibble JP, Evans EG, et al. A new paradigm for enzymatic control of α-cleavage and β-cleavage of the prion protein. J Biol Chem. 2014a;289:803–813.
  • Wik L, Klingeborn M, Willander H, et al. Separate mechanisms act concurrently to shed and release the prion protein from the cell. Prion. 2012;6(5):498–509.
  • Guillot-Sestier MV, Sunyach C, Druon C, et al. The alpha-secretase-derived N-terminal product of cellular prion, N1, displays neuroprotective function in vitro and in vivo. J Biol Chem. 2009;284:35973–35986.
  • Guillot-Sestier MV, Checler F. Cellular prion and its catabolites in the brain: production and function. Curr Mol Med. 2012;12:304–315.
  • Westergard L, Turnbaugh JA, Harris DA, et al. A naturally occurring C-terminal fragment of the prion protein (PrP) delays disease and acts as a dominant-negative inhibitor of PrPSc formation. J Biol Chem. 2011 Dec 23;286(51):44234–44242.
  • Lewis V, Hill AF, Haigh CL, et al. Increased proportions of C1 truncated prion protein protect against cellular M1000 prion infection. J Neuropathol Exp Neurol. 2009;68(10):1125–1135.
  • Munoz-Montesino C, Larkem D, Barbereau C, et al. A seven-residue deletion in PrP leads to generation of a spontaneous prion formed from C-terminal C1 fragment of PrP. J Biol Chem. 2020;295(41):14025–14039.
  • Sunyach C, Cisse MA, Da Costa CA, et al. The C-terminal products of cellular prion protein processing, C1 and C2, exert distinct influence on p53-dependent sTaurosporine-induced caspase-3 activation. J Biol Chem. 2007;282(3):1956–1963.
  • Huang S, Liang J, Zheng M, et al. Regulated over-expression of PrP in the skeletal muscles leads to myopathy in transgenic mice. Proc Natl Acad Sci USA. 2007;104:6800–6805.
  • Liang J, Parchaliuk D, Medina S, et al. Booth SA. Activation of p53-regulated pro-apoptotic signaling pathways in PrP-mediated myopathy. BMC Genomics. 2009;10(1):201.
  • Liang J, Kong Q. α-cleavage of cellular prion protein. Prion. 2012;6(5):453–460.
  • Vincent B, Paitel E, Frobert Y, et al. Phorbol ester-regulated cleavage of normal prion protein in HEK293 human cells and murine neurons. J Biol Chem. 2000;275(45):35612–35616.
  • Oliveira-Martins JB, Yusa S, Calella AM, et al. Unexpected tolerance of alpha-cleavage of the prion protein to sequence variations. PLoS One. 2010;5:107.
  • Vincent B, Paitel E, Saftig P, et al. The disintegrins ADAM10 and TACE contribute to the constitutive and phorbol ester-regulated normal cleavage of the cellular prion protein. J Biol Chem. 2001;276(41):37743–37746.
  • Cissé MA, Sunyach C, Lefranc-Jullien S, et al. The disintegrin ADAM9 indirectly contributes to the physiological processing of cellular prion by modulating ADAM10 activity. J Biol Chem. 2005;280:40624–40631.
  • Béland M, Motard J, Barbarin A, et al. PrP(C) homodimerization stimulates the production of PrPC cleaved fragments PrPN1 and PrPC1. J Neurosci. 2012;32:13255–13263.
  • Altmeppen HC, Prox J, Puig B, et al. Lack of a-disintegrin-and-metalloproteinase ADAM10 leads to intracellular accumulation and loss of shedding of the cellular prion protein in vivo. Mol Neurodegener. 2011;6(1): 36.
  • Liang J, Wang W, Sorensen D, et al. Cellular prion protein regulates its own α-cleavage through ADAM8 in skeletal muscle. J Biol Chem. 2012b;287(20): 16510–16520.
  • McDonald AJ, Millhauser GL. PrP overdrive: does inhibition of α-cleavage contribute to PrP(C) toxicity and prion disease?. Prion. 2014b;8(2):183–191.
  • Aulić S, Masperone L, Narkiewicz J, et al. α-synuclein amyloids hijack prion protein to gain cell entry, facilitate cell-to-cell spreading and block prion replication. Sci Rep. 2017;7(1):10050.
  • Priola SA, Caughey B, Wehrly K, et al. A 60-kDa prion protein (PrP) with properties of both the normal and scrapie-associated forms of PrP. J Biol Chem. 1995;270(7):3299–3305.
  • Sangeetham SB, Huszár K, Bencsura P, et al. Interrogating the dimerization interface of the prion protein via site-specific mutations to p-Benzoyl-L-Phenylalanine. J Mol Biol. 2018;430(17):2784–2801.
  • Engelke AD, Gonsberg A, Thapa S, et al. Dimerization of the cellular prion protein inhibits propagation of scrapie prions. J Biol Chem. 2018;293(21):8020–8031.
  • Meier P, Genoud N, Prinz M, et al. Soluble dimeric prion protein binds PrP(Sc) in vivo and antagonizes prion disease. Cell. 2003;113:49–60.
  • Laurén J, Gimbel DA, Nygaard HB, et al. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature. 2009;457:1128–1132.
  • Balducci C, Beeg M, Stravalaci M, et al. Synthetic amyloid-beta oligomers impair long-term memory independently of cellular prion protein. Proc Natl Acad Sci U S A. 2010;107:2295–2300.
  • Dohler F, Sepulveda-Falla D, Krasemann S, et al. High molecular mass assemblies of amyloid-β oligomers bind prion protein in patients with alzheimer’s disease. Brain. 2014;137(3):873–886.
  • Chen S, Yadav SP, Surewicz WK, et al. Interaction between human prion protein and amyloid- (A) oligomers. Role of N-terminal residues. J Biol Chem. 2010;285:26377–26383.
  • Um JW, Kaufman AC, Kostylev M, et al. Metabotropic glutamate receptor 5 is a coreceptor for alzheimer aβ oligomer bound to cellular prion protein. Neuron. 2013;79(5):887–902.
  • Haas LT, Salazar SV, Kostylev MA, et al. Metabotropic glutamate receptor 5 couples cellular prion protein to intracellular signalling in alzheimer’s disease. Brain. 2016;139(2):526–546.
  • Um JW, Nygaard HB, Heiss JK, et al. Alzheimer amyloid-β oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat Neurosci. 2012;15(9):1227–1235.
  • Kessels HW, Nguyen LN, Nabavi S, et al. The prion protein as a receptor for amyloid-beta. Nature. 2010;466:E3–E5.
  • Calella AM, Farinelli M, Nuvolone M, et al. Prion protein and abeta-related synaptic toxicity impairment. EMBO Mol Med. 2010;2:306–314.
  • Ferreira DG, Temido-Ferreira M, Vicente Miranda H, et al. alpha-synuclein interacts with PrP(C) to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci. 2017;20:1569–1579.
  • De Cecco E, Legname G. The role of the prion protein in the internalization of α-synuclein amyloids. Prion. 2018;12(1):23–27.
  • Urrea L, Segura-Feliu M, Masuda-Suzukake M, et al. Involvement of Cellular Prion Protein in α-Synuclein Transport in Neurons [published correction appears in Mol Neurobiol. 2017 May 5;:]. Mol Neurobiol. 2018. 55:1847-60.
  • Gomes LA, Hipp SA, Rijal Upadhaya A, et al. Aβ-induced acceleration of alzheimer-related τ-pathology spreading and its association with prion protein. Acta Neuropathol. 2019;138(6):913–941.
  • De Cecco E, Celauro L, Vanni S, et al. The uptake of Tau amyloid fibrils is facilitated by the cellular prion protein and hampers prion propagation in cultured cells [published online ahead of print, 2020 May 11]. J Neurochem. 2020. 10.1111.
  • Ondrejcak T, Klyubin I, Corbett GT, et al. Cellular prion protein mediates the disruption of hippocampal synaptic plasticity by soluble tau in vivo. J Neurosci. 2018;38(50): 595–10606.
  • Pan Y, Zhao L, Liang J, et al. Cellular prion protein promotes invasion and metastasis of gastric cancer. FASEB J. 2006;20(11):1886–1888.
  • Han H, Bearss DJ, Browne LW, et al. Identification of differentially expressed genes in pancreatic cancer cells using cDNA microarray. Cancer Res. 2002;62:2890–2896.
  • Li C, Yu S, Nakamura F, et al. Binding of pro-prion to filamin A disrupts cytoskeleton and correlates with poor prognosis in pancreatic cancer. J Clin Invest. 2009;119(9):2725–2736.
  • Sy MS, Li C, Yu S, et al. The fatal attraction between pro-prion and filamin A: prion as a marker in human cancers. Biomark Med. 2010;4:453–464.
  • Antony H, Wiegmans AP, Wei MQ, et al. Potential roles for prions and protein-only inheritance in cancer. Cancer Metastasis Rev. 2012;31(1–2):1–19.
  • Shafiei SS, Guerrero-Muñoz MJ, Castillo-Carranza DL, et al. Tau oligomers: cytotoxicity, propagation, and mitochondrial damage. Front Aging Neurosci. 2017;9:83.
  • Smith LM, Kostylev MA, Lee S, et al. Systematic and standardized comparison of reported amyloid-β receptors for sufficiency, affinity, and alzheimer’s disease relevance. J Biol Chem. 2019;294:6042–6053.
  • Westaway D, DeArmond SJ, Cayetano-Canlas J, et al. Degeneration of skeletal muscle, peripheral nerves, and the central nervous system in transgenic mice overexpressing wt prion proteins. Cell. 1994;76:117–129.
  • Chiesa R, Piccardo P, Biasini E, et al. Aggregated, wt prion protein causes neurological dysfunction and synaptic abnormalities. J Neurosci. 2008;28:13258–13267.
  • Paitel E, Fahraeus R, Checler F, et al. Cellular prion protein sensitizes neurons to apoptotic stimuli through Mdm2-regulated and p53-dependent caspase 3-like activation. J Biol Chem. 2003;278(12):10061–10066.
  • Guillot-Sestier MV, Sunyach C, Ferreira ST, et al. α-secretase-derived fragment of cellular prion, N1, protects against monomeric and oligomeric amyloid β (Aβ)-associated cell death. J Biol Chem. 2011;287:5021–5032.
  • Fluharty BR, Biasini E, Stravalaci M, et al. An N-terminal fragment of the prion protein binds to amyloid-β oligomers and inhibits their neurotoxicity in vivo. J Biol Chem. 2013;288(11):7857–7866.
  • Béland M, Bédard M, Tremblay G, et al. Aβ induces its own prion protein N-terminal fragment (PrPN1)-mediated neutralization in amorphous aggregates. Neurobiol Aging. 2014;35:1537–1548.
  • Scott-mckean JJ, Surewicz K, Choi JK, et al. Soluble prion protein and its N-terminal fragment prevent impairment of synaptic plasticity by Aβ oligomers: implications for novel therapeutic strategy in alzheimer’s disease. Neurobiol Dis. 2016;91:124–131.
  • Carroll JA, Groveman BR, Williams K, et al. Prion protein N1 cleavage peptides stimulate microglial interaction with surrounding cells. Sci Rep. 2020;10(1):6654.
  • Sonati T, Reimann RR, Falsig J, et al. The toxicity of antiprion antibodies is mediated by the flexible tail of the prion protein. Nature. 2013;501:102–106.
  • Dametto P, Lakkaraju AK, Bridel C, et al. Neurodegeneration and unfolded-protein response in mice expressing a membrane-tethered flexible tail of PrP. PLoS One. 2015;10:e0117412.
  • Wu B, McDonald AJ, Markham K, et al. The N-terminus of the prion protein is a toxic effector regulated by the C-terminus. Elife. 2017;6:e23473.
  • Vincent B, Sunyach C, Orzechowski HD, et al. p53-dependent transcriptional control of cellular prion by presenilins. J Neurosci. 2009;29:6752–6760.
  • Glatzel M, Linsenmeier L, Dohler F, et al. Shedding light on prion disease. Prion. 2015;9(4):244–256.
  • Heiseke A, Schöbel S, Lichtenthaler SF, et al. The novel sorting nexin SNX33 interferes with cellular PrPSc formation by modulation of PrPc shedding. Traffic. 2008;9:1116–1129.
  • Kanaani J, Prusiner SB, Diacovo J, et al. Recombinant prion protein induces rapid polarization and development of synapses in embryonic rat hippocampal neurons in vitro. J Neurochem. 2005;95(5):1373–1386.
  • Amin L, Nguyen XTA, Rolle IG, et al. Characterization of prion protein function by focal neurite stimulation. Cell Sci. 2016;129:3878–3891.
  • Nieznanski K, Choi JK, Chen S, et al. Soluble prion protein inhibits amyloid-β (Aβ) fibrillization and toxicity. J Biol Chem. 2012;287:33104–33108.
  • Endres K, Mitteregger G, Kojro E, et al. Influence of ADAM10 on prion protein processing and scrapie infectiosity in vivo. Neurobiol Dis. 2009;36(2):233–241.
  • Altmeppen HC, Prox J, Krasemann S, et al. The sheddase ADAM10 is a potent modulator of prion disease. eLife. 2015;4:e04260.
  • Watts JC, Giles K, Patel S, et al. Evidence that bank vole PrP is a universal acceptor for prions. PLoS Pathog. 2014;10(4):e1003990.
  • Minikel EV, Zhao HT, Le J, et al. Prion protein lowering is a disease-modifying therapy across prion disease stages, strains and endpoints. Nucleic Acids Res. 2020;48(19):10615–10631.
  • Glatzel M, Aguzzi A. PrP(C) expression in the peripheral nervous system is a determinant of prion neuroinvasion. J Gen Virol. 2000;81:2813–2821.
  • McCulloch L, Brown KL, Bradford BM, et al. Follicular dendritic cell-specific prion protein (PrP) expression alone is sufficient to sustain prion infection in the spleen. PLoS Pathog. 2011;7:e1002402.
  • Klingeborn M, Race B, Meade-White KD, et al. Crucial role for prion protein membrane anchoring in the neuroinvasion and neural spread of prion infection. J Virol. 2011;85(4):1484–1494.
  • Chesebro B, Trifilo M, Race R, et al. Anchorless prion protein results in infectious amyloid disease without clinical scrapie. Science. 2005;308:1435–1439.
  • Rangel A, Race B, Klingeborn M, et al. Unusual cerebral vascular prion protein amyloid distribution in scrapie-infected transgenic mice expressing anchorless prion protein. Acta Neuropathol Commun. 2013;1(1):25.
  • Rangel A, Race B, Phillips K, et al. Distinct patterns of spread of prion infection in brains of mice expressing anchorless or anchored forms of prion protein. Acta Neuropathol Commun. 2014;2(1):8.
  • Chesebro B, Race B, Meade-White K, et al. Fatal transmissible amyloid encephalopathy: a new type of prion disease associated with lack of prion protein membrane anchoring. PLoS Pathog. 2010;6(3):e1000800.
  • Stohr J, Watts JC, Legname G, et al. Spontaneous generation of anchorless prions in transgenic mice. Proc Natl Acad Sci USA. 2011;108(52):21223–21228.
  • Endres K, Fahrenholz F. Upregulation of the alpha-secretase ADAM10--risk or reason for hope?. FEBS J. 2010;277:1585–1596.
  • Schulte M, Reiss K, Lettau M, et al. ADAM10 regulates FasL cell surface expression and modulates FasL‐induced cytotoxicity and activation‐induced cell death. Cell Death Differ. 2007;14:1040–1049.
  • Esselens CW, Malapeira J, Colome N, et al. Metastasis‐associated C4.4A, a GPI‐anchored protein cleaved by ADAM10 and ADAM17. Biol Chem. 2008;389:1075–1084.
  • Waldhauer I, Goehlsdorf D, Gieseke F, et al. Tumor‐associated MICA is shed by ADAM proteases. Cancer Res. 2008;68:6368–6376.
  • You B, Shan Y, Shi S, et al. Effects of ADAM10 upregulation on progression, migration, and prognosis of nasopharyngeal carcinoma. Cancer Sci. 2015;106(11):1506–1514.
  • Pasciuto E, Ahmed T, Wahle T, et al. Dysregulated ADAM10-mediated processing of APP during a critical time window leads to synaptic deficits in fragile X syndrome. Neuron. 2015;87(2):39–382.
  • Lo Sardo V, Zuccato C, Gaudenzi G, et al. An evolutionary recent neuroepithelial cell adhesion function of huntingtin implicates ADAM10-Ncadherin. Nat Neurosci. 2012;15(5):713–721.
  • Roberts TK, Eugenin EA, Morgello S, et al. PrPC, the cellular isoform of the human prion protein, is a novel biomarker of HIV-associated neurocognitive impairment and mediates neuroinflammation. Am J Pathol. 2010;177(4):1848–1860.
  • Megra BW, Eugenin EA, Berman JW, et al. The role of shed PrPc in the neuropathogenesis of HIV infection. J Immunol. 2017;199:224–232.
  • Yuyama K, Sun H, Sakai S, et al. Decreased amyloid-beta pathologies by intracerebral loading of glycosphingolipid-enriched exosomes in alzheimer model mice. J Biochem. 2014a;289:24488–24498.
  • Yuyama K, Sun H, Usuki S, et al. A potential function for neuronal exosomes: sequestering intracerebral amyloid-beta peptide. FEBS Lett. 2014b;589:84–88.
  • An K, Klyubin I, Kim Y, et al. Exosomes neutralize synaptic-plasticity-disrupting activity of Aβ assemblies in vivo. Mol Brain. 2013;6:47.
  • Falker C, Hartmann A, Guett I, et al. Exosomal cellular prion protein drives fibrillization of amyloid beta and counteracts amyloid beta-mediated neurotoxicity. J Neurochem. 2016;137:88–100.
  • Fevrier B, Vilette D, Archer F, et al. Cells release prions in association with exosomes. Proc Natl Acad Sci U S A. 2004;101(26):9683–9688.
  • Vella LJ, Sharples RA, Lawson VA, et al. Packaging of prions into exosomes is associated with a novel pathway of PrP processing. J Pathol. 2007;211:582–590.
  • Cervenakova L, Saa P, Yakovleva O, et al. Are prions transported by plasma exosomes? Transfus Apher. Sci. 2016;55(1):70–83.
  • Emmanouilidou E, Melachroinou K, Roumeliotis T, et al. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J Neurosci. 2010;30:6838–6851.
  • Hu G, Yao H, Chaudhuri AD, et al. Exosome-mediated shuttling of microRNA-29 regulates HIV tat and morphine-mediated neuronal dysfunction. Cell Death Dis. 2012;3(8):e381–10.
  • Saman S, Kim W, Raya M, et al. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early alzheimer disease. J Biol Chem. 2012;287(6):3842–3849.
  • Li X, Corbett AL, Taatizadeh E, et al. Challenges and opportunities in exosome research-perspectives from biology, engineering, and cancer therapy. APL Bioeng. 2019;3:011503.
  • Kong Q. RNAi: a novel strategy for the treatment of prion diseases. J Clin Invest. 2006;116(12):3101–3103.
  • Pfeifer A, Eigenbrod S, Al-Khadra S, et al. Lentivector-mediated RNAi efficiently suppresses prion protein and prolongs survival of scrapie-infected mice. J Clin Invest. 2006;116(12):3204–3210.
  • Pulford B, Reim N, Bell A, et al. Liposome-siRNA-peptide complexes cross the blood-brain barrier and significantly decrease PrPC on neuronal cells and PrPRES in infected cell cultures. PLoS One. 2010;5(6):1–13.
  • Bender H, Noyes N, Annis JL, et al. PrPC knockdown by liposome-siRNA-peptide complexes (LSPCs) prolongs survival and normal behavior of prion-infected mice immunotolerant to treatment. PLoS One. 2019;14(7):e0219995.
  • Raymond GJ, Zhao HT, Race B, et al. Antisense oligonucleotides extend survival of prion-infected mice. JCI Insight. 2019;5:131175.
  • Dexter E, Kong Q. Neuroprotective effect and potential of cellular prion protein and its cleavage products for treatment of neurodegenerative disorders Part II. Strategies for therapeutics development. Expert Rev. Neurother. 2021;21(9):983-991.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.