555
Views
3
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 II: strategies for therapeutics development

&
Pages 983-991 | Received 26 May 2021, Accepted 05 Aug 2021, Published online: 02 Sep 2021

References

  • Riek R, Hornemann S, Wider G, et al. NMR structure of the mouse prion protein domain PrP(121–231). Nature. 1996;382(6587):180–182.
  • 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):145–501227–1235.
  • Adle-Biassette H, Verney C, Peoc’h K, et al. Immunohistochemical Expression of Prion Protein (PrPC) in the Human Forebrain During Development. J Neuropathol Exp Neurol. 2006;65(7):698–706.
  • Peralta OA, Eyestone WH. Quantitative and qualitative analysis of cellular prion protein (PrP(C)) PrP C) expression in bovine somatic tissues. Prion. 2009;3(3):161–170.
  • Castle AR, Gill AC. Physiological Functions of the Cellular Prion Protein. Front Mol Biosci. 2017;6;4:19.
  • Linden R. The Biological Function of the Prion Protein: a Cell Surface Scaffold of Signaling Modules. Front Mol Neurosci. 2017;10:77.
  • Aguzzi A, Heikenwalder M. Pathogenesis of prion diseases: current status and future outlook. Nat Rev Microbiol. 2006;4(10):765–775.
  • Colby DW, Prusiner SB. Prions. Cold Spring Harb Perspect Biol. 2011;3(1):a006833.
  • Sigurdson CJ, Bartz JC, Cellular GM. Molecular Mechanisms of Prion Disease. Annu Rev Pathol. 2019 Jan 24;14(1):497–516.
  • Laurén J, Gimbel DA, Nygaard HB, et al. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature. 2009;457(7233): 1128–1132.
  • 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.
  • 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.
  • 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.
  • Salazar SV, Strittmatter SM. Cellular prion protein as a receptor for amyloid-β oligomers in Alzheimer’s disease. Biochem Biophys Res Commun. 2017;483(4):1143–1147.
  • Shafiei SS, Guerrero-Muñoz MJ, Castillo-Carranza DL. Tau Oligomers: cytotoxicity, Propagation, and Mitochondrial Damage. Front Aging Neurosci. 2017;9:83.
  • Urrea L, Segura-Feliu M, Masuda-Suzukake M, et al. Involvement of Cellular Prion Protein in α-Synuclein Transport in Neurons. Mol Neurobiol. 2018;55:60–1847.
  • Corbett GT, Wang Z, Hong W, et al. Young-Pearse TL, Billinton A, Walsh DM. PrP is a central player in toxicity mediated by soluble aggregates of neurodegeneration-causing proteins. Acta Neuropathol. 2020;139(3): 503–526.
  • 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(3):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.
  • Bremer J, Baumann F, Tiberi C, et al. Axonal prion protein is required for peripheral myelin maintenance. Nat Neurosci. 2010;13(3):310–83204–3210.
  • Chiarini LB, Freitas AR, Zanata SM, et al. Cellular prion protein transduces neuroprotective signals. EMBO J. 2002;21(13):3317–263101–3103.
  • 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(2):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(49):11330–11339.
  • Dexter E, Kong Q. Neuroprotective effect and potential of cellular prion protein and its cleavage products for treatment of neurodegenerative disorders. Part I. A literature review. Expert Rev Neurother. 2021;21(9):969–982.
  • 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(32):19173–8010238–10243.
  • 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(49):40624–40631.
  • 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.
  • 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.
  • 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, Kong Q. α-Cleavage of cellular prion protein. Prion. 2012a;6(5):453–460.
  • 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, 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. 2014b;289(2):803–813.
  • 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(51): 35973–35986.
  • 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(7):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. 2014a;35(7):1537–481886–1888.
  • 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.
  • Westergard L, Turnbaugh JA, Harris DA. 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;286(51):44234–44242.
  • 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(16):6800–6805.
  • Liang J, Parchaliuk D, Medina S, et al. Activation of p53-regulated pro-apoptotic signaling pathways in PrP-mediated myopathy. BMC Genomics. 2009;10(1):201.
  • 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, Xuan TA, Nguyen 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(40):33104–811330–11339.
  • Calella AM, Farinelli M, Nuvolone M, et al. Prion protein and Abeta-related synaptic toxicity impairment. EMBO Mol Med. 2010;2(8):306–1419173–19180.
  • 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(48):44627–44632.
  • McDonald AJ, Millhauser GL. PrP overdrive: does inhibition of α-cleavage contribute to PrP(C) toxicity and prion disease? Prion. 2014;8(2):183–191
  • Heiseke A, Schöbel S, Lichtenthaler SF, et al. The Novel Sorting Nexin SNX33 Interferes with Cellular PrPSc PrP Sc Formation by Modulation of PrPc PrPc Shedding. Traffic. 2008;9(7):1116–1129.
  • 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):233–4137743–37746.
  • Watts JC, Giles K, Patel S, et al. Evidence that bank vole PrP is a universal acceptor for prions. PLoS Pathog. 2014;10:e1003990.
  • Altmeppen HC, Prox J, Krasemann S, et al. The sheddase ADAM10 is a potent modulator of prion disease. eLife. 2015;4:e04260.
  • An K, Klyubin I, Kim YJ, 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(1): 88–100.
  • 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. 2014;289:24488–24498.
  • Legname G, Scialò C. On the role of the cellular prion protein in the uptake and signaling of pathological aggregates in neurodegenerative diseases. Prion. 2020;14(1):257–270.
  • Béland M, Roucou X. Taking advantage of physiological proteolytic processing of the prion protein for a therapeutic perspective in prion and Alzheimer diseases. Prion. 2014;8(1):106–110.
  • Nieznanska H, Bandyszewska M, Surewicz K, et al. Identification of prion protein-derived peptides of potential use in Alzheimer’s disease therapy. Biochim Biophys Acta. 2018;1864(6):2143–2153.
  • Mohammadi B, Linsenmeier L, Shafiq M, et al. Transgenic overexpression of the disordered prion Protein N1 fragment in mice does not protect against neurodegenerative diseases due to impaired er translocation. Mol Neurobiol. 2020;57(6):2812–2829.
  • Mohammadi B, Glatzel M, Altmeppen HC. Disordered structure and flexible roles: using the prion protein N1 fragment for neuroprotective and regenerative therapy. Neural Regen Res. 2021;16(7):1431–1432.
  • Müller MM. Post-Translational Modifications of Protein Backbones: unique Functions, Mechanisms, and Challenges. Biochemistry. 2018;57:177–185.
  • Fisher E, Pavlenko K, Vlasov A, et al. Peptide-Based Therapeutics for Oncology. Pharmaceut Med. 2019;33:9–20.
  • Ding Y, Jp T, Liu J, et al. Impact of non-proteinogenic amino acids in the discovery and development of peptide therapeutics. Amino Acids. 2020;52(9):1207–1226.
  • San Sebastian W, Samaranch L, Kells AP, et al. Gene therapy for misfolding protein diseases of the central nervous system. Neurotherapeutics. 2013;10(3):498–510.
  • Naidoo J, Stanek LM, Ohno K, et al. Extensive Transduction and Enhanced Spread of a Modified AAV2 Capsid in the Non-human Primate CNS. Mol Ther. 2018;26(10):2418–2430.
  • Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 2019;18:358–378.
  • 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.
  • Lammich S, Kojro E, Postina R, et al. Constitutive and regulated alpha-secretase cleavage of Alzheimer’s amyloid precursor protein by a disintegrin metalloprotease. Proc Natl Acad Sci USA. 1999;96(7): 3922–3927.
  • Kuhn PH, Wang H, Dislich B, et al. ADAM10 is the physiologically relevant, constitutive alpha-secretase of the amyloid precursor protein in primary neurons. EMBO J. 2010;29(17):3020–3032.
  • Postina R, Schroeder A, Dewachter I, et al. A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J Clin Invest. 2004;113(10): 1456–643922–3927.
  • Prinzen C, Trümbach D, Wurst W, et al. Differential gene expression in ADAM10 and mutant ADAM10 transgenic mice. BMC Genomics. 2009;10(1):66.
  • Clement AB, Hanstein R, Schröder A, et al. Effects of neuron-specific ADAM10 modulation in an in vivo model of acute excitotoxic stress. Neuroscience. 2008;152(2):459–468.
  • Mullooly M, McGowan PM, Crown J, et al. The ADAMs family of proteases as targets for the treatment of cancer. Cancer Biol Ther. 2016;17(8):870–801456–1464.
  • Smith TM Jr, Tharakan A, Martin RK. Targeting ADAM10 in Cancer and Autoimmunity. Front Immunol. 2020;11:499.
  • Dreymueller D, Pruessmeyer J, Groth E, et al. The role of ADAM-mediated shedding in vascular biology. Eur J Cell Biol. 2012;91(6–7):472–485.
  • Conrad C, Benzel J, Dorzweiler K, et al. ADAM8 in invasive cancers: links to tumor progression, metastasis, and chemoresistance. Clin Sci (Lond). 2019;133(1):83–99.
  • Tousseyn T, Thathiah A, Jorissen E, et al. ADAM10, the rate-limiting protease of regulated intramembrane proteolysis of Notch and other proteins, is processed by ADAMS-9, ADAMS-15, and the gamma-secretase. J Biol Chem. 2009;284(17):11738–11747.
  • Matthews AL, Noy PJ, Reyat JS, et al. Regulation of A disintegrin and metalloproteinase (ADAM) family sheddases ADAM10 and ADAM17: the emerging role of tetraspanins and rhomboids. Platelets. 2017;28(4):333–341.
  • 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(38):13255–13263.
  • Meier P, Genoud N, Prinz M, et al. Soluble dimeric prion protein binds PrP(Sc) in vivo and antagonizes prion disease. Cell. 2003;113(1):49–60.
  • 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.
  • Dai J, Liu ZQ, Wang XQ, et al. Discovery of Small Molecules for Up-Regulating the Translation of Antiamyloidogenic Secretase, a Disintegrin and Metalloproteinase 10 (ADAM10), by Binding to the G-Quadruplex-Forming Sequence in the 5’ Untranslated Region (UTR) of Its mRNA. J Med Chem. 2015;58(9):3875–3891.
  • Wetzel S, Seipold L, Saftig P. The metalloproteinase ADAM10: a useful therapeutic target? Biochim Biophys Acta Mol Cell Res. 2017;1864(11):2071–2081.
  • Yuyama K, Sun H, Usuki S, et al. A potential function for neuronal exosomes: sequestering intracerebral amyloid-beta peptide. FEBS Lett. 2015;589(1): 84–88.
  • Yang T, Martin P, Fogarty B, et al. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in danio rerio. Pharm Res. 2015;32(6):2003–2014.
  • Chen C, Liu L, Ma F, et al. Elucidation of Exosome Migration Across the Blood–Brain Barrier Model In Vitro. Cell Mol Bioeng. 2016;9(4):509–529.
  • 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(1):011503.
  • Jafari D, Shajari S, Jafari R, et al. Designer Exosomes: a New Platform for Biotechnology Therapeutics. BioDrugs. 2020;34(5):567–586.
  • Song H, Liu B, Dong B, et al. Exosome-Based Delivery of Natural Products in Cancer Therapy. Front Cell Dev Biol. 2021;9:650426.
  • Alvarez-Erviti L, Seow Y, Yin H, et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–345.
  • Xiong Y, Mahmood A, Chopp M. Emerging potential of exosomes for treatment of traumatic brain injury. Neural Regen Res. 2017;12(1):19–22.
  • Stam J, Bartel S, Bischoff R, et al. Isolation of extracellular vesicles with combined enrichment methods. J Chromatogr B Analyt Technol Biomed Life Sci. 2021;1169:122604.
  • Kurian TK, Banik S, Gopal D, et al. Elucidating Methods for Isolation and Quantification of Exosomes: a Review. Mol Biotechnol. 2021;63(4):249–266.
  • Gao B, Zhou S, Sun C, et al. Brain Endothelial Cell-Derived Exosomes Induce Neuroplasticity in Rats with Ischemia/Reperfusion Injury. ACS Chem Neurosci. 2020;11(15):2201–2213.
  • Tu YK, Hsueh YH. Extracellular vesicles isolated from human olfactory ensheathing cells enhance the viability of neural progenitor cells. Neurol Res. 2020;42(11):959–672201–2213.
  • Maia J, Caja S, Strano Moraes MC, et al. Exosome-based cell-cell communication in the tumor microenvironment. Front Cell Dev Biol. 2018;6:18.
  • 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(20):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.
  • 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 intoexosomes is associated with a novel pathway of PrP processing. J Pathol. 2007;211(5):582–590.
  • Cervenakova L, Saa P, Yakovleva O, et al. Are prions transported by plasma exosomes? Transfus Apher Sci. 2016;55(1):70–83.
  • Chesebro B, Trifilo M, Race R, et al. Anchorless prion protein results in infectious amyloid disease without clinical scrapie. Science. 2005;308(5727):1435–1439.
  • 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.
  • 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. The Role of Shed PrPcin the Neuropathogenesis of HIV InfectionInfection. J Immunol. 2017;199(1):224–232.
  • Chou CW, Huang YK, Kuo TT, et al. An Overview of ADAM9: structure, activation, and regulation in human diseases. Int J Mol Sci. 2020;21(20):7790.
  • Daude N. Specific inhibition of pathological prion protein accumulation by small interfering RNAs. J Cell Sci. 2003;116(13):2775–2779.
  • 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.
  • Kong Q. RNAi: a novel strategy for the treatment of prion diseases. J Clin Invest. 2006;116(12):3101–3103.
  • White MD, Farmer M, Mirabile I, et al. Single treatment with RNAi against prion protein rescues early neuronal dysfunction and prolongs survival in mice with prion disease. Proc Natl Acad Sci. 2008;105(29):10238–10243.
  • 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.
  • Onodera T, Sakudo A, Tsubone H, et al. Review of studies that have used knockout mice to assess normal function of prion protein under immunological or pathophysiological stress. Microbiol Immunol. 2014;58(7):361–374.
  • Bender H, Noyes N, Annis JL. 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;54(16):131175.

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.