724
Views
3
CrossRef citations to date
0
Altmetric
Extra View

Neuronal pathophysiology featuring PrPC and its control over Ca2+ metabolism

ORCID Icon &
Pages 28-33 | Received 13 Oct 2017, Accepted 28 Nov 2017, Published online: 05 Jan 2018

ABSTRACT

Calcium (Ca2+) is an intracellular second messenger that ubiquitously masters remarkably diverse biological processes, including cell death. Growing evidence substantiates an involvement of the prion protein (PrPC) in regulating neuronal Ca2+ homeostasis, which could rationalize most of the wide range of functions ascribed to the protein. We have recently demonstrated that PrPC controls extracellular Ca2+ fluxes, and mitochondrial Ca2+ uptake, in neurons stimulated with glutamate (De Mario et al., J Cell Sci 2017; 130:2736-46), suggesting that PrPC protects neurons from threatening Ca2+ overloads and excitotoxicity. In light of these results and of recent reports in the literature, here we review the connection of PrPC with Ca2+ metabolism and also provide some speculative hints on the physiologic outcomes of this link. In addition, because PrPC is implicated in neurodegenerative diseases, including prion disorders and Alzheimer's disease, we will also discuss possible ways by which disruption of PrPC-Ca2+ association could be mechanistically connected with these pathologies.

Introduction

Undoubtedly, stringent precautions for safe animal nourishment have now strongly lowered the threat for prion-tainted food that pervaded the last decades of the past century. Based on sporadic events or genetic grounds, prions originate from misfolded β-enriched conformers of an ubiquitously expressed protein, the cellular prion protein (PrPC), and cause neurodegenerative pathologies of humans and animals, named transmissible spongiform encephalopathies or prion diseases [Citation1]. Much advance has been made on the pathological aspects of these diseases. To date, however, secure therapeutic interventions are not available, nor it is established whether neurodegeneration arises from prion toxicity, or lack of the still mysterious PrPC function, or a combination of both. In this view, understanding PrPC cellular behavior would solve a long-standing biological issue, but would concurrently serve as strategic prerequisite to the design of drugs capable to halt the progression of fatal prion disorders. Localization of PrPC to the external cell surface has stimulated investigations in a plethora of experimental paradigms aimed at assessing a possible (co-)receptor function of the protein and its downstream signals. Also this field has witnessed important advances, by proposing a number of PrPC partners that together could trigger beneficial effects by governing particular pathways, and/or the concentration or activity of intracellular signaling mediators (lengthily reviewed in recent publications) [Citation2-4].

In addition to being recognized as the first protein to become infectious, PrPC presents a structure that is per se extremely interesting, being composed of a highly compact (α helix-rich) C-terminus (that which undergoes the α-to-β pathogenic conversion), and an unstructured N-terminus that is typical of “intrinsically disordered proteins” (IDPs) [Citation5,Citation6]. Lack of stable secondary and/or tertiary folding under physiological conditions provides IDPs with the capacity to interact with multiple functional partners, and to serve as central hubs in the coordination and integration of signaling networks [Citation6,Citation7]. In line with this concept, the extended unstructured N-terminus could perfectly tailor to PrPC to explaining its pleiotropicity, in terms of the high number of cell surface partners and regulated signaling pathways that have emerged from decade-long research on the issue. It is also to underline that IDPs are implicated in the pathogenesis of several human diseases, such as cancer, type II diabetes, cardiovascular diseases and – most importantly – different neurodegenerative disorders [Citation8,Citation9]. This aspect is not secondary to the (benign and toxic) dichotomic nature of PrPC. Indeed, it was proposed that dysregulation of intrinsically disordered regions, or genetic mutations, reduce the capacity of IDPs to recognize correct binding partners, thus allowing formation of non-functional complexes and aggregates generating aberrant signalling.

Under the framework of such a multifaceted trait of PrPC, the interest of our laboratory over the last few years has been focused in identifying a signaling factor with similar pleiotropic properties, i.e., serving as target of numerous upstream pathways, and, concurrently, regulating multiple downstream cell events. Our choice fell on calcium (Ca2+), one of the most exploited cofactors and signaling mediators controlling all cell stages (from oocyte fertilization, to cell differentiation and death, to name a few), through rapid and transient concentration changes in restricted cell domains [Citation10]. However, our choice considered also aspects common to PrPC and Ca2+ in neurons; (i), the positive action of both that can switch into a danger for the cell life (aberrant conformation changes of PrPC produce deadly prions; uncontrolled Ca2+ increases trigger cell death); (ii), synapses, whose dysfunction ultimately leads to loss of neurons [Citation11], are the regions in which PrPC is mainly expressed [Citation12], and where the fundamental action of Ca2+ ensures correct synaptic transmissions by spatio-temporally coordinating electric signals and neurotransmitter release [Citation10].

Here, we will briefly summarize our recent findings, along with evidence from the literature, connecting PrPC pathophysiology with Ca2+ metabolism. Speculative functional consequences of such a still enigmatic liaison will also be proposed.

The pathophysiologic connection of PrPC with Ca2+ metabolism

The possible connection of PrPC to Ca2+ metabolism is far from recent. It can be traced back to studies aimed at understanding the toxic mechanism of prions, and to initial attempts designed to identifying PrPC physiology through the comparison of paradigms harboring, or not (PrP-knockout, PrP-KO), PrPC. Although not always was Ca2+ the direct investigation target, altered Ca2+ homeostasis was observed, or could be envisaged, in prion-infected or PrP-KO model cells [Citation13-15]. Ultimately, by proving a direct interaction of PrPC with the N-methyl-D-aspartate (NMDA)-sensitive Na+/Ca2+-permeable glutamate receptor (NMDAR), combination of electrophysiological and biochemical approaches in hippocampal paradigms established that native PrPC behaved as sentinel against Ca2+ overload [Citation16]. This result provided a likely explanation for loss of synapse integrity caused by PrPC misfolding.

Our approach to the field was mainly methodological, having used genetically-encoded Ca2+ indicators (aequorins, AEQs) targeting specific cell regions of primary cultures of neurons from co-isogenic mice expressing, or not, PrPC [Citation17,Citation18]. In particular, we used AEQs sensing Ca2+ movements in microdomains of the cytosolic side of the plasma membrane (PM), in the cytosol, in the mitochondrial matrix and in the lumen of the endoplasmic reticulum (ER). Thus, as opposed to chemical dyes widely used for monitoring cytosolic Ca2+ fluctuations, AEQs allow to construct an integrated picture of how different cell compartments respond to stimuli promoting Ca2+ entry from the extracellular space, or release from intracellular Ca2+-reservoirs (e.g., the sarco-endoplasmic reticulum).

PrPC Governs several pathways for Ca2+ entry at the plasma membrane

Store-operated Ca2+ entry

Using mostly primary cerebellar granule neurons (CGN) expressing, or not, PrPC, we first considered store-operated Ca2+ entry (SOCE), one major PM Ca2+ route distinct from PM receptor-channels. Becoming activated upon a decrease of ER Ca2+ content, SOCE serves to refill intracellular Ca2+ reservoirs and to regulate key cell parameters, from gene expression to tissue development and function [Citation19]. Our work demonstrated that, also in the case of SOCE, PrPC served to oppose dangerous Ca2+ overloads because Ca2+ accumulation in PM subdomains was attenuated compared to PrP-KO CGN, and this reflected in lower cytosolic and mitochondrial Ca2+ fluxes [Citation20,Citation21]. In addition, combined biochemical approaches allowed us to substantiating the alleged involvement of PrPC in signal transduction related to the modulation of phosphorylation cascades, in particular that governed by the Src Tyr-kinase Fyn [Citation2]. In fact, PrPC was found to constitutively downregulate Fyn activation and the Tyr-phosphorylation of the stromal interaction molecule 1 [Citation21], an ER transmembrane Ca2+ sensor which is key to the mechanism linking ER Ca2+ depletion to stimulation of the channel-forming proteins of SOCE [Citation19].

It has been proposed that PrPC acts as a cell surface binding partner for, and transduces the neurotoxic action of, β-enriched soluble protein aggregates such as prions and amyloid-β (Aβ) oligomers related to Alzheimer's disease (AD) [Citation22,Citation23]. It was also reported that PrPC-mediated Aβ effects imply the engagement of Fyn [Citation12]. On this, we added another important piece of information by showing that Aβ oligomers subvert the tripartite connection between PrPC, Fyn and SOCE in both CGN and cortical neurons [Citation21]. This enabled us to suggest that disruption of SOCE-mediated Ca2+ signaling could contribute to PrPC-dependent effects in AD.

Glutamate-sensitive receptors

Because hippocampal neurons are protected from excessive extracellular Ca2+ influx owing to PrPC-NMDAR interactions restricting the receptor Ca2+ permeability [Citation16], we took advantage of AEQs to expand analysis of Ca2+ movements in different domains of ionotropic glutamate receptor (iGluR)-stimulated primary CGN and cortical neurons. In particular, we either activated each iGluR sub-type separately [by adding the specific agonist NMDA, or α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA), or kainate], or all iGluRs together (with the physiologic agonist, glutamate). Our findings corroborated in these neurons what had been previously observed for hippocampal NMDAR activity. However, data showing that PrPC equally downregulated AMPAR- and kainateR-mediated Ca2+ entry suggested a much wider action of PrPC in diminishing neuronal vulnerability by glutamate over-stimulation [Citation24], in accord with results obtained in PrP-KO animals exposed to different excitotoxic insults [Citation25-29].

As to the mechanism allowing PrPC to regulate AMPAR-dependent Ca2+ entry, we focused on AMPAR trafficking based on the notion that regulation of this process goes through Ser845 phosphorylation on the GluR1 subunit [Citation30]. Surface biotinylation of CGN with the two PrP genotypes clearly demonstrated that the surface expression of GluR1 was more abundant (∼70%) in PrP-KO neurons than in PrPC-expressing counterparts, a result highly suggestive that the diminished presence of the receptor in PrPC-expressing CGNs could have contributed to the restricted AMPAR-mediated Ca2+ entry[Citation24]. Ser845 is phosphorylated by protein kinase A (PKA) [Citation30], whose activity depends on the interplay between the formation and hydrolysis of cyclic adenosine monophosphate (cAMP). In line with our expectations, extracellular signal-regulated kinase 1 and 2, which potentiates PKA activity by its inhibitory phosphorylation of (cAMP-hydrolyzing) phosphodiesterase [Citation31], was less active (∼50%) in PrPC-expressing than in PrP-KO CGN [Citation24]. This finding adds a further molecular tile for explaining why the surface expression of AMPAR GluR1 was diminished in the presence of PrPC.

PrPC governs movements of, and Ca2+ uptake by, mitochondria

Altogether, works from our and others' laboratory substantiate association of PrPC with the cell apparatus deputed to maintain a correct Ca2+ homeostasis. Specifically, by controlling PM pathways PrPC contributes to prevent potentially noxious high Ca2+ entry [Citation27]. Intriguingly, however, we also found that, despite PrPC sits on the cell surface, its beneficial action strategically extends beyond the PM by protecting mitochondria from an equally deleterious Ca2+ overload [Citation24].

Mitochondria are crucial organelles for the cell life, which physiologically take up Ca2+ from different sources both in their Ca2+-buffering task and to stimulate enzymes enhancing the production of ATP. Likewise, it is well established that excessive Ca2+ accumulation may alter the inner membrane permeability, which ultimately undermines mitochondrial integrity and triggers apoptosis [Citation32]. During the morphological inspection of the two CGN types, somehow surprisingly we observed that, on average, mitochondria of PrP-KO CGN were around 30% more distant from the PM than in PrPC-expressing neurons (an issue further discussed below). However, if this displacement protected PrP-KO mitochondria from increased Ca2+ uptake following the selective stimulation of a single iGluR sub-type, this behavior no longer held when neurons were exposed to glutamate. Such an apparent paradox was eventually resolved by demonstrating that the higher glutamate-mediated Ca2+ influx in PrP-KO neurons was capable to better stimulate the process of Ca2+-induced Ca2+ release via ryanodine receptor channels [Citation10], which, in turn, remarkably increased mitochondrial Ca2+ uptake [Citation24]. This set of data suggests that (at least in CGN) the protection by PrPC against Ca2+ overload is integrated towards a few regions of the cell, which may have a crucial relevance under pathologic settings.

Open questions on PrPC role in neuronal pathophysiology

If what hitherto reported clearly underlines the capacity of PrPC to regulate multiple aspects of Ca2+ metabolism and to protect neurons from abnormal Ca2+ accumulations, these actions need further investigations as exemplified in the following open questions.

(I) How can PrPC perform such a complex task, i.e., which are the events engaging the protein in regulating multiple Ca2+-mobilizing systems?

With regards to Ca2+-permeable pathways at the neuronal PM, data are now available pointing to the capacity of PrPC to reducing Ca2+ entry by acting at different levels. On the one hand, PrPC prevents glutamate potential excitotoxicity because the protein downregulates the channel activity of the NMDAR by interacting directly with one of its subunits [Citation16], and by reducing the affinity for the co-agonist glycine in a copper-dependent manner [Citation33]. On the other hand, in line with suggestions that PrPC affects phosphorylation-based signaling pathways [Citation2,Citation34], we have also shown that PrPC downregulates both SOCE and AMPAR by controlling specific phosphorylation events on the respective molecular machineries (see above) [Citation21,Citation24]. Yet, if the phenomenology of the PrPC-Ca2+ connection in the above instances is quite clear, mechanistic clues of these links still warrant a better definition.

(II) Which are the downstream events of the PrPC-Ca2+ liaison?

Because Ca2+ is a key player in almost all aspects of cell physiology, particularly in excitable cells, it is hard to draw a unique picture rationalizing the various reports of PrPC control over Ca2+ homeostasis and the entire range of outcomes of these regulations. It thus appears more sensible to envision that the protein acts in a context-dependent manner – in terms of type and actual status of the cell, and kind of stimulus (or combination of stimuli) the cell is exposed to – and/or in PrPC capacity to integrating diverse signaling pathways. This view stems from the previously introduced concept, proposing that the intrinsically disordered PrPC N-terminus could interact with multiple functional partners, and behave as a central hub in the coordination and integration of signaling networks [Citation6,Citation7]. A few speculative consequences of the PrPC-Ca2+ interplay, for which we have now collected some evidence, may fall in the following scenarios.

(a)

One of the primary Ca2+-regulated cell functions is gene transcription [Citation35]. On this, we have provided evidence that PrPC could regulate the expression of different neuronal membrane proteins, following a large-scale proteomic study of membrane proteins in primary CGN with the two PrP genotypes [Citation36]. We have also reported that, compared to control neurons, cultured PrP-KO CGN display a significant downregulation of PM and sarco-endoplasmic reticulum Ca2+ ATPases [Citation20], two major cytosolic Ca2+ removal systems [Citation10]. More recently, analysis of the whole proteome has shown a significant downregulation in PrP-KO CGN of a set of proteins functionally related to vesicular trafficking (among which some members of the Rab family of monomeric GTPases), resulting in reduced glutamate release and impaired synaptic vesicle re-uptake (Peggion et al., unpublished observations). These results are very intriguing although, here also, molecular details of a likely PrPC-Ca2+-gene transcription axis need a more refined characterization.

(b)

Another possible outcome of PrPC control over Ca2+ refers to SOCE. Following the notion that SOCE controls the differentiation of non-neuronal cell types [Citation37], and neurogenesis [Citation19], we assessed whether this was applicable to CGN differentiation. To this end, we analyzed the impact of SOCE on the in-vitro maturation of wild-type CGN using specific inhibitors, and post-transcriptional silencing of molecular constituents, of SOCE. Preliminary data collected in this study indicate that such a Ca2+-mobilizing mechanism regulates the differentiation of CGN cultured under basal conditions (Peggion et al., unpublished observations). In light of this information, and the alleged role of PrPC in (neuronal) cell differentiation [Citation2,Citation38], and in SOCE modulation [Citation20,Citation21], it seems plausible that PrPC regulates neurogenesis via SOCE. Although we did not observe any apparent alteration of PrP-KO CGN differentiation compared to the PrP-expressing counterpart in our experimental constraints, still an involvement of the PrPC-SOCE connection in neuronal development under different in vitro and/or in vivo settings cannot yet be excluded.

(c)

The final framework refers to our finding that mitochondria are displaced from the PM in cultured PrP-KO CGN (see above) [Citation24]. The cytoskeletal-mediated movement of mitochondria is of fundamental importance for cells, and for neurons in particular, since it couples the biogenesis of mitochondria to their transport to specific locations such as synapses. This complex and energy-dependent process, which is not yet entirely defined, is based on molecular motors involving several proteins, including those belonging to the Miro family that contains both GTPase and Ca2+-binding EF-hand domains [Citation39]. The suggestion that mitochondria transport along axons may depend on local variations of Ca2+ concentration, and the by now-established role of PrPC in Ca2+ homeostasis, should therefore stimulate investigations on the possible connection between PrPC-Ca2+ coupling and the neuronal transport of mitochondria.

(III) Is the PrPC-Ca2+ relationship important in neuropathology?

It is good to underline that the “gold standard” of our experimental paradigms to tackle the issue of PrPC physiology was the use of PrP-KO mouse models, and primary neuronal cultures thereof. It may thus be questionable if and how our findings can be directly translated into neuropathological contexts, e.g., prion diseases, primarily because all generated PrP-KO mouse lines do not display overt phenotypes, with the exception of a chronic peripheral demyelination associated to late-onset polyneuropathy [Citation2,Citation4]. On the other hand, one cannot rule out that under specific circumstances, such as those in which neurons are subjected to neurotoxic challenges altering PrPC structure and function, the Ca2+-related phenotypes reported by us in in-vitro PrP-KO models may participate in relevant pathogenic routes. This concept has been significantly reinforced by the finding that PrPC acts as a high affinity receptor for neurotoxic β-rich protein aggregates, such as prions and Aβ oligomers [Citation22,Citation23], opening the possibility that these interactions may (at least) exacerbate oligomers' neurotoxic effects by ultimately displacing PrPC from its native protective functions, including control of local Ca2+ homeostasis.

Various mechanisms can be hypothesized relating an impaired control of PrPC on Ca2+ metabolism and alterations of neuronal function and survival, the most obvious of which is harmful Ca2+ overload associated to glutamate excitotoxicity [Citation27]. Excessive mitochondrial Ca2+ uptake can also be a fatal conveyor of doom for neurons by leading to increased mitochondrial permeability, loss of mitochondrial functions and release of pro-apoptotic factors [Citation32]. Our suggestion that PrPC defends against perilous Ca2+ transients not only in the cytosol but also in the mitochondrial matrix, supports the possibility that loss of this function of PrPC contributes to neuronal demise under neuropathological conditions. The latter line of reasoning may apply as well to alterations of neurotransmitter release, already recognized as central in different neurodegenerative disorders and model systems [Citation11], which we observed in PrP-KO CGN as a consequence of the perturbed protein expression pattern and defective synaptic vesicle trafficking (see above). Within the same hypothesis, our observation that mitochondria in PrP-KO neurons are displaced from the PM [Citation24], in a likely Ca2+-dependent way, could also be pathologically relevant, since mitochondria localization near the synaptic membrane is undoubtedly strategic to satisfying the high energy demand necessary to ensure intact synaptic functions.

Disclosure of potential conflicts of interest

Authors declare no potential conflict of interest.

Acknowledgments

Our gratitude goes to all collaborators whose work over the years has been essential to shed light on the PrPC-Ca2+ connection.

Funding

Università degli Studi di Padova, PRAT CPDA158035/15 to A.B.

Università degli Studi di Padova, PRAT CPDA121988/12 to M.C.S.

References

  • Prusiner SB. Prions. Proc Natl Acad Sci USA. 1998;95(23):13363–83. doi:10.1073/pnas.95.23.13363.
  • Castle AR, Gill AC. Physiological Functions of the Cellular Prion Protein. Front Mol Biosci. 2017;4:19. doi:10.3389/fmolb.2017.00019.
  • Peggion C, Bertoli A, Sorgato MC. Almost a century of prion protein(s): From pathology to physiology, and back to pathology. Biochem Biophys Res Commun. 2017;483(4):1148–55. doi:10.1016/j.bbrc.2016.07.118.
  • Wulf MA, Senatore A, Aguzzi A. The biological function of the cellular prion protein: an update. BMC Biol. 2017;15(1):34. doi:10.1186/s12915-017-0375-5.
  • Zahn R, Liu A, Lührs T, et al. NMR solution structure of the human prion protein. Proc Natl Acad Sci USA. 2000;97(1):145–50. doi:10.1073/pnas.97.1.145.
  • van der Lee R, Buljan M, Lang B, et al. et al. Classification of intrinsically disordered regions and proteins. Chem Rev. 2014;114(13):6589–631. doi:10.1021/cr400525m.
  • Tompa P, Schad E, Tantos A, et al. Intrinsically disordered proteins: emerging interaction specialists. Curr Opin Struct Biol. 2015;35:49–59. doi:10.1016/j.sbi.2015.08.009.
  • Uversky VN, Oldfield CJ, Dunker AK. Intrinsically disordered proteins in human diseases: introducing the D2 concept. Annu Rev Biophys. 2008;37:215–46. doi:10.1146/annurev.biophys.37.032807.125924.
  • Uversky VN. Intrinsically disordered proteins and their (disordered) proteomes in neurodegenerative disorders. Front Aging Neurosci. 2015;7:18. doi:10.3389/fnagi.2015.00018.
  • Berridge MJ. Calcium microdomains: organization and function. Cell Calcium. 2006;40(5–6):405–12. doi:10.1016/j.ceca.2006.09.002.
  • Mattson MP. Calcium and neurodegeneration. Aging Cell. 2007;6(3):337–50. doi:10.1111/j.1474-9726.2007.00275.x.
  • 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):1227–35. doi:10.1038/nn.3178.
  • Wong K, Qiu Y, Hyun W, et al. Decreased receptor-mediated calcium response in prion-infected cells correlates with decreased membrane fluidity and IP3 release. Neurology. 1996;47(3):741–50. doi:10.1212/WNL.47.3.741.
  • Fuhrmann M, Bittner T, Mitteregger G, et al. Loss of the cellular prion protein affects the Ca2+ homeostasis in hippocampal CA1 neurons. J Neurochem. 2006;98(6):1876–85. doi:10.1111/j.1471-4159.2006.04011.x.
  • Peggion C, Bertoli A, Sorgato MC. Possible role for Ca2+ in the pathophysiology of the prion protein? Biofactors. 2011;37(3):241–9. doi:10.1002/biof.161.
  • Khosravani H, Zhang Y, Tsutsui S, et al. et al. Prion protein attenuates excitotoxicity by inhibiting NMDA receptors. J Cell Biol. 2008;181(3):551–65. doi:10.1083/jcb.200711002.
  • Lim D, Bertoli A, Sorgato MC, et al. Generation and usage of aequorin lentiviral vectors for Ca(2+) measurement in sub-cellular compartments of hard-to-transfect cells. Cell Calcium. 2016;59(5):228–39. doi:10.1016/j.ceca.2016.03.001.
  • Suzuki J, Kanemaru K, Iino M. Genetically Encoded Fluorescent Indicators for Organellar Calcium Imaging. Biophys J. 2016;111(6):1119–31. doi:10.1016/j.bpj.2016.04.054.
  • Prakriya M, Lewis RS. Store-Operated Calcium Channels. Physiol Rev. 2015;95(4):1383–436. doi:10.1152/physrev.00020.2014.
  • Lazzari C, Peggion C, Stella R, et al. Cellular prion protein is implicated in the regulation of local Ca2+ movements in cerebellar granule neurons. J Neurochem. 2011;116(5):881–90. doi:10.1111/j.1471-4159.2010.07015.x.
  • De Mario A, Castellani A, Peggion C, Massimino ML, et al. The prion protein constitutively controls neuronal store-operated Ca(2+) entry through Fyn kinase. Front Cell Neurosci. 2015;9:416. doi:10.3389/fncel.2015.00416.
  • Resenberger UK, Harmeier A, Woerner AC, et al. et al. The cellular prion protein mediates neurotoxic signalling of β-sheet-rich conformers independent of prion replication. EMBO J. 2011;30(10):2057–70. doi:10.1038/emboj.2011.86.
  • Laurén J, Gimbel DA, Nygaard HB, et al. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature. 2009;457(7233):1128–32. doi:10.1038/nature07761.
  • De Mario A, Peggion C, Massimino ML, Viviani F, et al. The prion protein regulates glutamate-mediated Ca(2+) entry and mitochondrial Ca(2+) accumulation in neurons. J Cell Sci. 2017;130(16):2736–46. doi:10.1242/jcs.196972.
  • Rangel A, Burgaya F, Gavín R, et al. Enhanced susceptibility of Prnp-deficient mice to kainate-induced seizures, neuronal apoptosis, and death: Role of AMPA/kainate receptors. J Neurosci Res. 2007;85(12):2741–55. doi:10.1002/jnr.21215.
  • 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(17):3041–54. doi:10.1091/mbc.E11-04-0321.
  • Black SA, Stys PK, Zamponi GW, et al. Cellular prion protein and NMDA receptor modulation: protecting against excitotoxicity. Front Cell Dev Biol. 2014;2:45. doi:10.3389/fcell.2014.00045.
  • Carulla P, Llorens F, Matamoros-Angles A, et al. Involvement of PrP(C) in kainate-induced excitotoxicity in several mouse strains. Sci Rep. 2015;5:11971. doi:10.1038/srep11971.
  • Bertani I, Iori V, Trusel M, et al. Inhibition of IL-1β Signaling Normalizes NMDA-Dependent Neurotransmission and Reduces Seizure Susceptibility in a Mouse Model of Creutzfeldt-Jakob Disease. J Neurosci. 2017;37(43):10278–89. doi:10.1523/JNEUROSCI.1301-17.2017.
  • Diering GH, Heo S, Hussain NK, et al. Extensive phosphorylation of AMPA receptors in neurons. Proc Natl Acad Sci USA. 2016;113(33):E4920–7. doi:10.1073/pnas.1610631113.
  • Song RS, Massenburg B, Wenderski W, et al. ERK regulation of phosphodiesterase 4 enhances dopamine-stimulated AMPA receptor membrane insertion. Proc Natl Acad Sci USA. 2013;110(38):15437–42. doi:10.1073/pnas.1311783110.
  • Bernardi P, Rasola A, Forte M, et al. The Mitochondrial Permeability Transition Pore: Channel Formation by F-ATP Synthase, Integration in Signal Transduction, and Role in Pathophysiology. Physiol Rev. 2015;95(4):1111–55. doi:10.1152/physrev.00001.2015.
  • You H, Tsutsui S, Hameed S, et al. Aβ neurotoxicity depends on interactions between copper ions, prion protein, and N-methyl-D-aspartate receptors. Proc Natl Acad Sci USA. 2012;109(5):1737–42. doi:10.1073/pnas.1110789109.
  • Sorgato MC, Peggion C, Bertoli A. Is, indeed, the prion protein a Harlequin servant of “many” masters? Prion. 2009;3(4):202–5. PMID: 19887913.
  • West AE, Chen WG, Dalva MB, et al. Calcium regulation of neuronal gene expression. Proc Natl Acad Sci USA. 2001;98(20):11024–31. doi:10.1073/pnas.191352298.
  • Stella R, Cifani P, Peggion C, et al. Relative quantification of membrane proteins in wild-type and prion protein (PrP)-knockout cerebellar granule neurons. J Proteome Res. 2012;11(2):523–36. doi:10.1021/pr200759m.
  • Darbellay B, Arnaudeau S, König S, et al. STIM1- and Orai1-dependent store-operated calcium entry regulates human myoblast differentiation. J Biol Chem. 2009;284(8):5370–80. doi:10.1074/jbc.M806726200.
  • Prodromidou K, Papastefanaki F, Sklaviadis T, et al. Functional cross-talk between the cellular prion protein and the neural cell adhesion molecule is critical for neuronal differentiation of neural stem/precursor cells. Stem Cells. 2014;32(6):1674–87. doi:10.1002/stem.1663.
  • Devine MJ, Birsa N, Kittler JT. Miro sculpts mitochondrial dynamics in neuronal health and disease. Neurobiol Dis. 2016;90:27–34. doi:10.1016/j.nbd.2015.12.008.

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.