417
Views
19
CrossRef citations to date
0
Altmetric
Mini Review

Is indeed the prion protein a Harlequin servant of "many" masters?

Pages 202-205 | Received 31 Jul 2009, Accepted 04 Sep 2009, Published online: 01 Oct 2009

Abstract

Tens of putative interacting partners of the cellular prion protein (PrPC) have been identified, yet the physiologic role of PrPC remains unclear. For the first time, however, a recent paper has demonstrated that the absence of PrPC produces a lethal phenotype. Starting from this evidence, here we discuss the validity of past and more recent literature supporting that, as part of protein platforms at the cell surface, PrPC may bridge extracellular matrix molecules and membrane proteins to intracellular signaling pathways.

Initially, the discovery that the prion protein was the major, if not the unique, component of the prion agent causing transmissible spongiform encephalopathies (TSE)Citation1 has placed the protein in an extremely unfavorable light. Thereafter, however, a wealth of evidence has supported the notion that the protein positively influences several aspects of the cell physiology, and that its duality—in harboring both lethal and beneficial potentials—could be rationalized in terms of a structural switch. Indeed, the protein exists in at least two conformational states: the cellular, α helix-rich isoform, PrPC, and the prion-associated β sheet-rich isoform, PrPSc.Citation2 If it is now unquestionable that the presence of PrPC in the cell is mandatory for prion replication and neurotoxicity to occur,Citation3,Citation4 nonetheless its physiologic function is still debatable, despite the long lasting effort, and the numerous, frequently genetically advanced, animal and cell model systems dedicated to the issue. From these studies the picture of an extremely versatile protein has emerged, whereby PrPC acts in the cell defense against oxidative and apoptotic challenges, but also in cell adhesion, proliferation and differentiation, and in synaptic plasticity.Citation5,Citation6 In an effort to converge these multiple propositions in an unifying functional model, different murine lines devoid of PrPC have been studied. These animals, however, displayed no obvious phenotype,Citation7Citation9 suggesting that either PrPC is dispensable during development and adult life or that compensative mechanisms mask the loss of PrPC function in these paradigms. Thus, identifying the exact role of PrPC in the cell would not only resolve an important biological question, but would also help elucidate the cellular steps of prion pathogenesis necessary for designing early diagnostic tools and therapeutic strategies for TSE.

As is often the case, the employment of a model system unprecedented in prion research has recently disclosed a most interesting scenario with regards to PrPC physiology, having unravelled, for the first time, a lethal phenotype linked to the absence of the protein.Citation10 The paradigm is the zebrafish, which expresses two PrPC isoforms (PrP1 and PrP2). Similarly to mammalian PrPC, they are glycosylated and attached to the external side of the plasma membrane through a glycolipid anchor. PrP1 and PrP2 are, however, expressed in distinct time frames of the zebrafish embryogenesis. Accordingly, the knockdown of the PrP1, or PrP2, gene very early in embryogenesis impaired development at different stages, bypassing putative compensatory mechanisms. By focusing on PrP1, Malaga-Trillo et al. showed that the protein was essential for cell adhesion, and that this event occurred through PrP1 homophilic trans-interactions and signaling. This comprised activation of the Src-related tyrosine (Tyr) kinase p59fyn, and, possibly, Ca2+ metabolism, leading to the regulation of the trafficking of E-cadherin, a member of surface-expressed cell adhesion molecules (CAMs) responsible for cell growth and differentiation.Citation11 It was also reported that overlapping PrP1 functions were performed by PrPCs from other species, while the murine PrPC was capable to replace PrP1 in rescuing, at least in part, the knockdown developmental phenotype. Apart from providing the long-sought proof for a vital role of PrPC, the demonstration that a mammalian isoform corrected the lethal zebrafish phenotype strongly reinforces previous results—mainly obtained in a variety of mammalian primary neurons and cell lines—pointing to a functional interplay of PrPC with CAMs, or extra cellular matrix (ECM) proteins, and cell signaling, to promote neuritogenesis and neuronal survival. A revisit of these data is the main topic of the present minireview.

As mentioned, the capacity of PrPC to act as a cell adhesion, or recognition, molecule, and to entertain interactions with proteins implicated in growth and survival, has already been reported for the mammalian PrPC. A case in point is the interaction, both in cis- and trans-configurations, with the neuronal adhesion protein N-CAM12 that led to neurite outgrowth.Citation13 Like cadherins, N-CAM belongs to the CAM superfamily. Following homo- or heterophylic interactions, it can not only mediate adhesion of cells, or link ECM proteins to the cytoskeleton, but also act as a receptor to transduce signals ultimately resulting in modulating neurite outgrowth, neuronal survival and synaptic plasticity.Citation11 Another example is the binding of PrPC to laminin, an ECM heterotrimeric glycoprotein, which induced neuritogenesis together with neurite adhesion and maintenance,Citation14,Citation15 but also learning and memory consolidation.Citation16 Further, it has been described that PrPC interacted with the mature 67 kDa-receptor (67LR) (and its 37 kDa-precursor) for laminin, and with glycosamminoglycans (GAGs), each of which is involved in neuronal differentiation and axon growth.Citation17Citation21 More recently, Hajj et al.Citation22 have reported that the direct interaction of PrPC with another ECM protein, vitronectin, could accomplish the same process, and that the absence of PrPC could be functionally compensated by the overexpression of integrin, another laminin receptor.Citation23 Incidentally, the latter finding may provide a plausible explanation for the absence of clear phenotypes in mammalian PrP-null paradigms. By exposing primary cultured neurons to recombinant PrPs, others have shown that trans-interactions of PrPC are equally important for neuronal outgrowth,Citation24,Citation25 including the formation of synaptic contacts.Citation25 Finally, it has been demonstrated that the binding of PrPC with the secreted co-chaperone stress-inducible protein 1 (STI1) stimulated neuritogenesis.Citation26 This same interaction had also a pro-survival effect, as did the interaction of PrPC with its recombinant form.Citation24 Notably, the involvement of PrPC in cell protection has been heightened by experiments with whole animals. By applying transient or permanent focal cerebral ischemia to the animals, it was found that their reduced brain damage correlated with spontaneous or adenoviral-mediated, upregulation of PrPC,Citation27Citation29 (reviewed in ref. Citation30), and that PrPC deficiency aggravated their ischemic brain injury.Citation30,Citation31 Thus, now that data are available from phylogenetically distant paradigms (zebrafish and mammalian model systems), it acquires more solid grounds the advocated engagement of PrPC in homo/heterophilic cis/trans interactions to trigger signaling events aiming at neuronal—or, in more general terms, cell—survival and neuritogenesis. The latter notion is consistent with the delayed maturation of different types of PrPC-less neurons, observed both in vitro and in vivo.Citation32,Citation33

If one assumes that the interaction of PrPC with multiple partners (45 for PrPC and PrPSc, as reviewed in Aguzzi et al.,Citation5 or 46 considering the homophylic interaction) are all functionally significant, the most immediate interpretation of this “sticky” behavior entails that PrPC acts as a scaffolding protein in different membrane protein complexes.Citation5,Citation6 Each complex could then activate a specific signaling pathway depending on the type and maturation of cells, the expression and glycosylation of PrPC, and availability of extra- and intra-cellular signaling partners. At large, all these signals have been shown to be advantageous to the cell. However, because in a cell only a subtle line divides the “good” from the “bad,” instances can be envisioned in which a pro-life signal turns into a pro-death signal. A typical example of this possibility is glutamate excitotoxicity resulting in dangerous, glutamate receptor-linked, Ca2+ overload. Likewise, an excessive or over-stimulated signal elicited by PrPC, or by the putative complex housing the protein could become noxious to the cell. This possibility may explain why the massive expression of PrPC caused degeneration of the nervous system,Citation34 and of skeletal muscles,Citation34,Citation35 in transgenic animals. More intriguing is the finding that—in a mouse line expressing anchorless PrPC—PrPSc was capable to replicate without threatening the integrity of neurons.Citation36 This may suggest that native membrane-bound PrPC acts as, or takes part into, a “receptor for PrPSc”, and that lasting PrPSc-PrPC interactions distort the otherwise beneficial signal of the protein/complex and cause neurodegeneration.Citation37 Consistent with this hypothesis is the finding that the in vivo antibody-mediated ligation of PrPC provoked apoptosis of the antibody-injected brain area.Citation38 Speculatively, the action of N-terminally, or N-proximally truncated PrPs whose expression in PrP-less transgenic mice induced extensive neurodegeneration,Citation39Citation41 may be traced back to the same hyper-activation of PrPC signaling. Possibly, this may hold true also for the synaptic impairment that, recorded only in PrPC-expressing neurons, was attributed to the binding of amyloid beta (Aβ) peptide oligomers implicated in Alzheimer disease, to PrPC.Citation42,Citation43

But which is (are) the cellular signaling pathway(s) conveyed by the engagement of PrPC in different signaling complexes? In line with its multifaceted behavior, several intracellular effectors have been proposed, including p59fyn, mitogen-activated kinases (MAPK) Erk1/2, PI3K/Akt and cAMP-PKA. p59fyn is the most reported downstream effector, suggesting that, in accordance with its behavior, p59fyn could serve as the sorting point for multiple incoming and outgoing signals also in the case of PrPC. The initial evidence of the PrPC-p59fyn connection came from cells subjected to antibody-mediated cross-linking of PrPC.Citation44 Later, it was shown that the PrPC-p59fyn signal converged to Erk1/2 through a pathway dependent on (but also independent of) reactive oxygen species generated by NADPH oxidase.Citation45 A PrPC-dependent activation of p59fynCitation13,Citation25 and Erk1/2 (but also of PI3K and cAMP-PKA)Citation24 was evident in other neuronal cell paradigms and consistent with the almost ubiquitous expression of PrPC, in non-neuronal cells such as Jurkat and T cells.Citation46 Not to forget that in zebrafish embryonic cells activated p59fyn was found in the same focal adhesion sites harboring PrP1.Citation10 Regarding the activation of the ERK1/2 pathway promoted by the PrPC-STI1 complex, and leading to neuritogenesis, the role of p59fyn was not investigated.Citation26 The same holds true for the transduction of a neuroprotective signal by the PrPC-STI1 complex involving the cAMP-PKA pathway.Citation26 Interestingly, this is not the only example reporting that engagement of PrPC activates simultaneously two independent pathways. In fact, possibly after transactivating the receptor for the epidermal growth factor, the antibody-mediated clustering of PrPC was shown to impinge on both the Erk1/2 pathway, and on a protein (stathmin) involved in controlling microtubule dynamics.Citation47

Yet, if p59fyn is implicated in mammalian PrPC-activated signaling cascade, a protein linking extracellular PrPC to p59fyn is needed, given the attachment of the enzyme to the inner leaflet of the plasma membrane through palmitoylated/myristoylated anchors. In this, the PrPC partner N-CAM (isoform 140) seems ideal to fulfill the task, given that p59fyn is part of N-CAM-mediated signaling. Indeed, after recruitment of N-CAM to lipid rafts—which may also depend on PrPC,Citation13—together with the receptor protein Tyr phosphatase α (RPTPα), the Tyr-phosphate removing activity of RPTPα allows the subsequent activation of p59fyn through an autophosphorylation step.Citation48 This event recruits and activates the focal adhesion kinase (FAK),Citation11 another non-receptor Tyr kinase. Finally, formation of the FAK-p59fyn complex triggers neuritogenesis through both Erk1/2 and PI3K/Akt pathways.Citation49,Citation50 Parenthetically, the FAK-p59fyn and PI3K/Akt connection would be suitable to explain why aggravation of ischemic brain injury in PrP-deficient brains was linked to a depressed Akt activation.Citation31 FAK-p59fyn complex, however, may be also involved in the signal triggered by the still mysterious PrPC partner, 67LR. This protein was reported not only to act as a laminin receptor but also to facilitate the interaction of laminin with integrins,Citation51 thereby possibly activating (through integrins) FAK-p59fyn-regulated pathways.Citation49 Conversely, other data have supported the candidature of caveolin-1 for coordinating the signal that from PrPC reaches Erk1/2 through p59fyn.Citation44,Citation45,Citation52 Further scrutiny of this route has shown that it comprised players such as laminin and integrins (upstream), FAK-p59fyn, paxillin and the Src-homology-2 domain containing adaptor protein (downstream), and that caveolin-1, a substrate of the FAK-p59fyn complex, facilitated the interaction of these signaling partners by recruiting them in caveolae-like membrane domains.Citation53

For the relevance they bear, we need to acknowledge recent propositions supporting the commitment of PrPC with proteins whose function is unrelated from the above-mentioned cell adhesion or ECM molecules; namely, the β-site amyloid precursor protein (APP) cleaving enzime (BACE1) and the N-methyl-D-aspartate (NMDA)-receptor. BACE1 is a proteolytic enzyme involved in Aβ production. It has been shown that overexpressed PrPC restricted, while depletion of PrPC increased the access of BACE1 to APP, possibly because PrPC interacts with BACE1 via GAGs.Citation54 Thus, native PrPC reduces the production of Aβ peptides. A beneficial effect of PrPC was also highlighted by Khosravani et al.Citation55 showing that, by physically associating with the subunit 2D of the NMDA-receptor, PrPC attenuated neuronal Ca2+ entry and its possible excitotoxic effect. This clear example for the control of PrPC on Ca2+ metabolism is particularly intriguing in light of previous reports linking Ca2+ homeostasis to PrPC pathophysiology (reviewed in ref. Citation56). Also, it is important to mention that a few partners of PrPC or downstream effectors may initiate signals that increase intracellular Ca2+, and that, in turn, local Ca2+ fluctuations regulate some of the afore-mentioned pathways.Citation11,Citation49,Citation57,Citation58

In conclusion, although still somehow speculative, the implication of Ca2+ in PrPC-dependent pathways raises the possibility that the different input signals originating from the interaction of PrPC with diverse partners may all converge to the universal, highly versatile Ca2+ signaling. Were indeed this the case, then clearly the acting of PrPC as Harlequin, the famous character of the 18th century Venetian playwright Carlo Goldoni, who struggles to fill the orders of two masters, would be merely circumstantial.

Acknowledgements

This work was supported by grants by the Italian Ministero dell'Università e della Ricerca (Prin 2006, to M.C.S.), and by the University of Padova (Progetto d'Ateneo CPDA089551, to A.B.).

References

  • Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science 1982; 216:136 - 144
  • Prusiner SB. Prions. Proc Natl Acad Sci USA 1998; 95:13363 - 13383
  • Büeler H, Aguzzi A, Sailer A, Greiner RA, Autenried P, Aguet M, et al. Mice devoid of PrP are resistant to scrapie. Cell 1993; 73:1339 - 1347
  • Mallucci G, Dickinson A, Linehan J, Klöhn PC, Brandner S, Collinge J. Depleting neuronal PrP in prion infection prevents disease and reverses spongiosis. Science 2003; 302:871 - 874
  • Aguzzi A, Baumann F, Bremer J. The prion's elusive reason for being. Annu Rev Neurosci 2008; 31:439 - 477
  • Linden R, Martins VR, Prado MA, Cammarota M, Izquierdo I, Brentani RR. Physiology of the prion protein. Physiol Rev 2008; 88:673 - 728
  • Büeler H, Fischer M, Lang Y, Bluethmann H, Lipp HP, DeArmond SJ, et al. Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature 1992; 356:577 - 582
  • Manson JC, Clarke AR, Hooper ML, Aitchison L, McConnell I, Hope J. 129/Ola mice carrying a null mutation in PrP that abolishes mRNA production are developmentally normal. Mol Neurobiol 1994; 8:121 - 127
  • Mallucci GR, Ratté S, Asante EA, Linehan J, Gowland I, Jefferys JG. Post-natal knockout of prion protein alters hippocampal CA1 properties, but does not result in neurodegeneration. EMBO J 2002; 21:202 - 210
  • Málaga-Trillo E, Solis GP, Schrock Y, Geiss C, Luncz L, Thomanetz V, et al. Regulation of embryonic cell adhesion by the prion protein. PLoS Biol 2009; 7:55
  • Hansen SM, Berezin V, Bock E. Signaling mechanisms of neurite outgrowth induced by the cell adhesion molecules NCAM and N-cadherin. Cell Mol Life Sci 2008; 65:3809 - 3821
  • Schmitt-Ulms G, Legname G, Baldwin MA, Ball HL, Bradon N, Bosque PJ, et al. Binding of neural cell adhesion molecules (N-CAMs) to the cellular prion protein. J Mol Biol 2001; 314:1209 - 1225
  • Santuccione A, Sytnyk V, Leshchyns'ka I, Schachner M. Prion protein recruits its neuronal receptor NCAM to lipid rafts to activate p59fyn and to enhance neurite outgrowth. J Cell Biol 2005; 169:341 - 354
  • Graner E, Mercadante AF, Zanata SM, Forlenza OV, Cabral AL, Veiga SS, et al. Cellular prion protein binds laminin and mediates neuritogenesis. Brain Res Mol Brain Res 2000; 76:85 - 92
  • Graner E, Mercadante AF, Zanata SM, Martins VR, Jay DG, Brentani RR. Laminin-induced PC-12 cell differentiation is inhibited following laser inactivation of cellular prion protein. FEBS Lett 2000; 482:257 - 260
  • Coitinho AS, Freitas AR, Lopes MH, Hajj GN, Roesler R, Walz R, et al. The interaction between prion protein and laminin modulates memory consolidation. Eur J Neurosci 2006; 24:3255 - 3264
  • Caughey B, Brown K, Raymond GJ, Katzenstein GE, Thresher W. Binding of the protease-sensitive form of PrP (prion protein) to sulfated glycosaminoglycan and congo red. J Virol 1994; 68:2135 - 2141
  • Rieger R, Edenhofer F, Lasmézas CI, Weiss S. The human 37-kDa laminin receptor precursor interacts with the prion protein in eukaryotic cells. Nat Med 1997; 3:1383 - 1388
  • Gauczynski S, Peyrin JM, Haïk S, Leucht C, Hundt C, Rieger R, et al. The 37-kDa/67-kDa laminin receptor acts as the cell-surface receptor for the cellular prion protein. EMBO J 2001; 20:5863 - 5875
  • Hundt C, Peyrin JM, Haïk S, Gauczynski S, Leucht C, Rieger R, et al. Identification of interaction domains of the prion protein with its 37-kDa/67-kDa laminin receptor. EMBO J 2001; 20:5876 - 5886
  • Pan T, Wong BS, Liu T, Li R, Petersen RB, Sy MS. Cell-surface prion protein interacts with glycosaminoglycans. Biochem J 2002; 368:81 - 90
  • Hajj GN, Lopes MH, Mercadante AF, Veiga SS, da Silveira RB, Santos TG, et al. Cellular prion protein interaction with vitronectin supports axonal growth and is compensated by integrins. J Cell Sci 2007; 120:1915 - 1926
  • McKerracher L, Chamoux M, Arregui CO. Role of laminin and integrin interactions in growth cone guidance. Mol Neurobiol 1996; 12:95 - 116
  • Chen S, Mangé A, Dong L, Lehmann S, Schachner M. Prion protein as trans-interacting partner for neurons is involved in neurite outgrowth and neuronal survival. Mol Cell Neurosci 2003; 22:227 - 233
  • Kanaani J, Prusiner SB, Diacovo J, Baekkeskov S, Legname G. Recombinant prion protein induces rapid polarization and development of synapses in embryonic rat hippocampal neurons in vitro. J Neurochem 2005; 95:1373 - 1386
  • Lopes MH, Hajj GN, Muras AG, Mancini GL, Castro RM, Ribeiro KC, 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
  • McLennan NF, Brennan PM, McNeill A, Davies I, Fotheringham A, Rennison KA, et al. Prion protein accumulation and neuroprotection in hypoxic brain damage. Am J Pathol 2004; 165:227 - 235
  • Weise J, Crome O, Sandau R, Schulz-Schaeffer W, Bähr M, Zerr I. Upregulation of cellular prion protein (PrPc) after focal cerebral ischemia and influence of lesion severity. Neurosci Lett 2004; 372:146 - 150
  • Shyu WC, Lin SZ, Chiang MF, Ding DC, Li KW, Chen SF, et al. Overexpression of PrPC by adenovirus-mediated gene targeting reduces ischemic injury in a stroke rat model. J Neurosci 2005; 25:8967 - 8977
  • Spudich A, Frigg R, Kilic E, Kilic U, Oesch B, Raeber A, et al. Aggravation of ischemic brain injury by prion protein deficiency: role of ERK-1/-2 and STAT-1. Neurobiol Dis 2005; 20:442 - 449
  • Weise J, Sandau R, Schwarting S, Crome O, Wrede A, Schulz-Schaeffer W, et al. Deletion of cellular prion protein results in reduced Akt activation, enhanced postischemic caspase-3 activation, and exacerbation of ischemic brain injury. Stroke 2006; 37:1296 - 1300
  • Steele AD, Emsley JG, Ozdinler PH, Lindquist S, Macklis JD. Prion protein (PrPc) positively regulates neural precursor proliferation during developmental and adult mammalian neurogenesis. Proc Natl Acad Sci USA 2006; 103:3416 - 3421
  • Prestori F, Rossi P, Bearzatto B, Lainé J, Necchi D, Diwakar S, et al. Altered neuron excitability and synaptic plasticity in the cerebellar granular layer of juvenile prion protein knock-out mice with impaired motor control. J Neurosci 2008; 28:7091 - 7103
  • Westaway D, DeArmond SJ, Cayetano-Canlas J, Groth D, Foster D, Yang SL, et al. Degeneration of skeletal muscle, peripheral nerves, and the central nervous system in transgenic mice overexpressing wild-type prion proteins. Cell 1994; 76:117 - 129
  • Huang S, Liang J, Zheng M, Li X, Wang M, Wang P, et al. Inducible overexpression of wild-type prion protein in the muscles leads to a primary myopathy in transgenic mice. Proc Natl Acad Sci USA 2007; 104:6800 - 6805
  • Chesebro B, Trifilo M, Race R, Meade-White K, Teng C, LaCasse R, et al. Anchorless prion protein results in infectious amyloid disease without clinical scrapie. Science 2005; 308:1435 - 1439
  • Aguzzi A. Cell biology. Prion toxicity: all sail and no anchor. Science 2005; 308:1420 - 1421
  • Solforosi L, Criado JR, McGavern DB, Wirz S, Sánchez-Alavez M, Sugama S, et al. Cross-linking cellular prion protein triggers neuronal apoptosis in vivo. Science 2004; 303:1514 - 1546
  • Shmerling D, Hegyi I, Fischer M, Blättler T, Brandner S, Götz J, et al. Expression of amino-terminally truncated PrP in the mouse leading to ataxia and specific cerebellar lesions. Cell 1998; 93:203 - 214
  • Baumann F, Tolnay M, Brabeck C, Pahnke J, Kloz U, Niemann HH, et al. Lethal recessive myelin toxicity of prion protein lacking its central domain. EMBO J 2007; 26:538 - 547
  • Li A, Christensen HM, Stewart LR, Roth KA, Chiesa R, Harris DA. Neonatal lethality in transgenic mice expressing prion protein with a deletion of residues 105–125. EMBO J 2007; 26:548 - 558
  • Laurén J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature 2009; 457:1128 - 1132
  • Kim D, Tsai LH. Bridging physiology and pathology in AD. Cell 2009; 137:997 - 1000
  • Mouillet-Richard S, Ermonval M, Chebassier C, Laplanche JL, Lehmann S, Launay JM, et al. Signal transduction through prion protein. Science 2000; 289:1925 - 1928
  • Schneider B, Mutel V, Pietri M, Ermonval M, Mouillet-Richard S, Kellermann O. NADPH oxidase and extracellular regulated kinases 1/2 are targets of prion protein signaling in neuronal and nonneuronal cells. Proc Natl Acad Sci USA 2003; 100:13326 - 13331
  • Stuermer CA, Langhorst MF, Wiechers MF, Legler DF, Von Hanwehr SH, Guse AH, et al. PrPc capping in T cells promotes its association with the lipid raft proteins reggie-1 and reggie-2 and leads to signal transduction. FASEB J 2004; 18:1731 - 1733
  • Monnet C, Gavard J, Mège RM, Sobel A. Clustering of cellular prion protein induces ERK1/2 and stathmin phosphorylation in GT1-7 neuronal cells. FEBS Lett 2004; 576:114 - 118
  • Bodrikov V, Leshchyns'ka I, Sytnyk V, Overvoorde J, den Hertog J, Schachner M. RPTPalpha is essential for NCAM-mediated p59fyn activation and neurite elongation. J Cell Biol 2005; 168:127 - 139
  • Parsons JT. Focal adhesion kinase: the first ten years. J Cell Sci 2003; 116:1409 - 1416
  • Tucker BA, Rahimtula M, Mearow KM. Src and FAK are key early signalling intermediates required for neurite growth in NGF-responsive adult DRG neurons. Cell Signal 2008; 20:241 - 257
  • Nelson J, McFerran NV, Pivato G, Chambers E, Doherty C, Steele D. The 67 kDa laminin receptor: structure, function and role in disease. Biosci Rep 2008; 28:33 - 48
  • Toni M, Spisni E, Griffoni C, Santi S, Riccio M, Lenaz P, et al. Cellular prion protein and caveolin-1 interaction in a neuronal cell line precedes fyn/erk 1/2 signal transduction. J Biomed Biotechnol 2006; 2006:69469
  • Pantera B, Bini C, Cirri P, Paoli P, Camici G, Manao G, et al. PrPc activation induces neurite outgrowth and differentiation in PC12 cells: role for caveolin-1 in the signal transduction pathway. J Neurochem 2009; 110:194 - 207
  • Parkin ET, Watt NT, Hussain I, Eckman EA, Eckman CB, Manson JC, et al. Cellular prion protein regulates beta-secretase cleavage of the Alzheimer's amyloid precursor protein. Proc Natl Acad Sci USA 2007; 104:11062 - 11067
  • Khosravani H, Zhang Y, Tsutsui S, Hameed S, Altier C, Hamid J, et al. Prion protein attenuates excitotoxicity by inhibiting NMDA receptors. J Cell Biol 2008; 181:551 - 565
  • Sorgato MC, Bertoli A. From cell protection to death: may Ca2+ signals explain the chameleonic attributes of the mammalian prion protein?. Biochem Biophys Res Commun 2009; 379:171 - 174
  • Cullen J. Decoding complex Ca2+ signals through the modulation of Ras signaling. Curr Opin Cell Biol 2006; 18:157 - 161
  • Putney JW Jr. SOC: now also store-operated cyclase. Nat Cell Biol 2009; 11:381 - 382

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.