Skip to main content
Cellular and Molecular Neurobiology logoLink to Cellular and Molecular Neurobiology
. 2009 Dec 30;30(5):653–666. doi: 10.1007/s10571-009-9491-2

Interactions of Prion Protein With Intracellular Proteins: So Many Partners and no Consequences?

Krzysztof Nieznanski 1,
PMCID: PMC11498852  PMID: 20041289

Abstract

Prion protein (PrP) plays a key role in the pathogenesis of transmissible spongiform encephalopathies (TSEs)—fatal diseases of the central nervous system. Its physiological function as well as exact role in neurodegeneration remain unclear, hence screens for proteins interacting with PrP seem to be the most promising approach to elucidating these issues. PrP is mostly a plasma membrane-anchored extracellular glycoprotein and only a small fraction resides inside the cell, yet the number of identified intracellular partners of PrP is comparable to that of its membranal or extracellular interactors. Since some TSEs are accompanied by significantly increased levels of cytoplasmic PrP and this fraction of the protein has been found to be neurotoxic, it is of particular interest to characterize the intracellular interactome of PrP. It seems reasonable that at elevated cytoplasmic levels, PrP may exert cytotoxic effect by affecting the physiological functions of its intracellular interactors. This review is focused on the cytoplasmic partners of PrP along with possible consequences of their binding.

Keywords: Prion protein, Protein–protein interactions, Protein aggregation, Transmissible spongiform encephalopathies, Neurotoxicity

Introduction

Transmissible spongiform encephalopathies (TSEs) are a group of fatal neurodegenerative diseases including scrapie, bovine spongiform encephalopathy (BSE), and chronic wasting disease (CWD) in animals and Creutzfeldt–Jakob disease (CJD), Gerstmann–Sträussler–Scheinker disease (GSS), and fatal familial insomnia (FFI) in humans. More than 25 years ago these diseases were linked to prion protein (PrP) (McKinley et al. 1983; Prusiner et al. 1984; rev. in Prusiner 1998). Susceptibility to TSEs requires expression of the cellular prion protein (PrPC), since organisms experimentally devoid of the Prnp gene encoding this protein are resistant to the infection (Büeler et al. 1993). It is also widely accepted that a misfolded form of PrPC, termed PrPSc (the scrapie form of PrP), is the main or even sole component of the TSE infectious agent (rev. in Prusiner 1998). This pathogenic folding, characterized by an increased beta-strand content, leads to the reduction of PrP solubility accompanied by a gain of substantial resistance to proteinase K (PK) digestion. Despite many efforts, the molecular mechanisms of the PrPC conversion into PrPSc and of the PrP-linked neurodegeneration remain elusive. Furthermore, although PrP is highly conserved in mammals, its physiological function is still unclear. PrPC is ubiquitously expressed in many cell types with the highest level reached in the nervous system (Bendheim et al. 1992). This protein is translated as a 253-aa polypeptide (human PrP) containing at the both termini signal sequences of ca. 22 amino acids each (Fig. 1). The N-terminal signal peptide (ER SP) is removed during entry into the lumen of the endoplasmic reticulum (ER) whereas the C-terminal signal (GPI SP) is cleaved off upon attachment of a glycosylphosphatidylinositol (GPI) moiety (Basler et al. 1986; Stahl et al. 1987; Turk et al. 1988). Additionally, inside the ER, PrPC may be variably glycosylated at two asparagine residues (N181 and N197) giving rise to di-, mono- and un-glycosylated species (Haraguchi et al. 1989). The N-terminal part of mature PrPC is basic and unstructured, whereas the C-terminal part forms a slightly acidic globular domain stabilized by a single disulfide bridge (Riek et al. 1997). The central region of the polypeptide, encompassing residues 112–135, is hydrophobic and may function as a transmembrane domain (TM) (Lopez et al. 1990). The N-terminal sequence is flanked (residues 23–28 and 101–106) by cryptic nuclear localization signals (NLS) (Gu et al. 2003). In the N-terminal part, five octapeptide repeats (OR) are localized which constitute the major Cu2+-binding site (Hornshaw et al. 1995). Additional copper-binding sites are localized C-terminally to the ORs (Jackson et al. 2001; Jones et al. 2004). Normal proteolytic processing at residues 110–112 generates fragments termed N1 and C1 corresponding to the above-mentioned unstructured and globular parts of the PrPC molecule, respectively (Chen et al. 1995; Jiménez-Huete et al. 1998). Additional fragments called N2 and C2 are produced in the brain of TSE-infected organisms by proteolytic cleavage at residues 90/91 (Chen et al. 1995).

Fig. 1.

Fig. 1

Scheme of human PrP. Signal peptides governing entry into ER lumen (SP ER) and attachment of GPI (SP GPI), octarepeats (OR), transmembrane domain (TM), two nuclear localization signals (NLS), disulfide bridge (S–S), GPI attachment site, two glycosylated asparagine residues (N-Glyc), three α-helices and two β-strands are indicated. Sites of proteolytic processing related to the maturation of PrP and generation of two shortened forms C1 and C2 are marked with scissors

Numerous studies have focused on the identification of molecules interacting with PrPC or PrPSc. Upon the discovery of Cu2+ binding by PrPC accompanied by a superoxide dismutase-like activity it was proposed that this protein may be involved in the homeostasis of copper ions and protection against oxidative stress (Brown et al. 1999). The basic N-terminal region of PrPC has also been shown to interact with glycosaminoglycans (Caughey et al. 1994; Pan et al. 2002; Warner et al. 2002). Interestingly, PrPC is a nucleic acid-binding protein capable of interacting with DNA as well as RNA (Nandi 1997; Weiss et al. 1997; Gabus et al. 2001). As a cell-surface molecule, PrP interacts with extracellular or plasma-membrane proteins such as, e.g., dystroglycan, vitronectin, caveolin-1, laminin, laminin receptor/precursor, apolipoprotein, and neural cell adhesion molecules (Keshet et al. 2000; Hajj et al. 2007; rev. in Vana et al. 2007). Basing on some of these observations, PrPC has been proposed to take part in neuronal differentiation, signal transduction, cytoprotection, and cell adhesion. Surprisingly, it has also been demonstrated that PrP may interact with numerous intracellular proteins, of which the majority are found in the cytosol (summarized in Table 1). This issue will be reviewed in detail below.

Table 1.

Intracellular partners of PrP. The table indicates which PrP form was found to bind to the interactor, how the interaction was detected, in which compartment may the interaction occur, and its effect

PrP interactor Interacting form of PrP/technique Localization Effect of interaction Reference
Neuroglobin rPrP/affinity Cytosol Aggregation Lechauve et al. (2009)
Synapsin 1b rPrP, PrPC/Y2H, co-IP, co-fractionation Cytosol ? Spielhaupter and Schätzl (2001)
Grb2 rPrP, PrPC/Y2H, co-IP, co-fractionation Cytosol, nucleus ? Lysek and Wüthrich (2004), Spielhaupter and Schätzl (2001)
GFAP rPrP, PrPC, PrPSc/pull-down, overlay, co-IP Cytosol ? Dong et al. (2008b), Oesch et al. (1990)
Tubulin rPrP, PrPC, PrPSc/pull-down, co-IP, co-fractionation, cross-linking, affinity Cytosol Oligomerization, aggregation, inhibition of microtubule assembly Brown et al. (1998), Brown (2000), Dong et al. (2008a), Giorgi et al. (2009), Hachiya et al. (2004a), Keshet et al. (2000), Nieznanski et al. (2005), Nieznanski et al. (2006), Osiecka et al. (2009)
Tau rPrP, PrPC, PrPSc/pull-down, co-IP Cytosol Reduction of binding to tubulin Han et al. (2005), Han et al. (2006), Wang et al. (2008)
hnRNP A2/B1 rPrP, PrPC/overlay, co-IP Nucleus/cytosol ? Strom et al. (2006)
Aldolase C rPrP, PrPC/overlay Cytosol ? Strom et al. (2006)
Bcl-2 rPrP, cytoPrP/Y2H, co-IP, affinity Cytosol Aggregation, loss of Bcl-2 function, induction of apoptosis Kurschner and Morgan (1995), Rambold et al. (2006)
NRAGE rPrP, cytoPrP, PrPC/pull-down, Y2H, co-IP Cytosol Aggregation, affected mitochondrial membrane potential Bragason and Palsdottir (2005)
Nrf2 rPrP/screen of bacteriophage expression library of brain cDNA Cytosol, nucleus ? Yehiely et al. (1997)
CK2 rPrP, PrPC/pull-down, overlay, co-IP, SPR Cytosol, nucleus, extracellular matrix Increase of enzymatic activity, phosphorylation of PrP Meggio et al. (2000), Negro et al. (2000)
14-3-3 protein rPrP, PrPC, PrPSc ?/pull-down, overlay, co-IP Cytosol ? Mei et al. (2009), Satoh et al. (2005)
Mahogunin cytoPrP, CtmPrP/pull-down, affinity Cytosol Aggregation, loss of mahogunin function, neurodegeneration Chakrabarti and Hegde (2009)
HOXA1 PrPC/microarray, co-IP Nucleus ? Satoh et al. (2009)
MPG PrPC/microarray, co-IP Nucleus ? Satoh et al. (2009)
PLK3 PrPC/microarray, co-IP Cytosol, cell membrane ? Satoh et al. (2009)

The abbreviations used in this table are: co-IP co-immunoprecipitation, SPR surface plasmon resonance, Y2H yeast two-hybrid screen

Intracellular Localization of PrP

Usually most molecules of the PrP are found anchored by the GPI moiety in detergent-resistant raft domains on the cell surface and hence it is commonly considered extracellular (Stahl et al. 1990; Naslavsky et al. 1997). This is, however, an incomplete picture of PrP biology. Subpopulations of neurons in the hippocampus, thalamus, and neocortex have been found where PrP is localized predominantly to the cytosol (Mironov et al. 2003). This fraction of the protein, not associated with membranous structures, was designated cytPrP. Moreover, due to the central hydrophobic sequence and inefficient translocation, PrP may cross the ER membrane only partially and adopt either of two transmembranal topologies termed CtmPrP and NtmPrP (Hegde et al. 1998; Yost et al. 1990; Kim et al. 2001; rev. in Chakrabarti et al. 2009). In the former, the N-terminal part of PrP, retaining its signal sequence, is exposed to the cytosol whereas the C-terminal domain resides in the ER lumen. In the latter, the N-terminal region of PrP is inside the ER while the C-terminal domain faces the cytosol. In the brain of healthy organisms, these forms do not exceed 10% whereas in TSE they may constitute up to 30% of all PrP molecules. Mutations within or near the hydrophobic domain are responsible for increased generation of CtmPrP. Particularly, the GSS-linked mutations P105L and A117V favor production of CtmPrP (Hegde et al. 1998; Kim and Hegde 2002). ER stress and other pathogenic mutations evoking incorrect translocation of PrP may lead to its accumulation in a fully cytosolic form (rev. in Miesbauer et al. 2009). There is no uniform nomenclature for this cytosolically mistargeted protein and in various reports it is termed cyPrP, cytPrP, or cytoPrP, irrespectively of its origin. It was reported that GSS-linked mutants W145Stop and Q160Stop exhibited reduced translocation into the ER and were detected in the cytosol and nucleus (Zanusso et al. 1999; Heske et al. 2004). Associated with FFI and CJD in humans, PrP mutant D177N may be mistargeted to the cytosol as a result of retrograde translocation from the ER (Ma and Lindquist 2001). Usually, in a quality control process, the retro-translocated PrP undergoes degradation by the ubiquitin–proteasome system (Yedidia et al. 2001). Inhibition of the proteasome activity led to accumulation of aggregated PrP in the cytosol (Ma and Lindquist 2001; Ma et al. 2002). Interestingly, proteasome inhibition is observed in TSE-infected cells. Notably, PrPSc has been shown to inhibit the proteasome (Kristiansen et al. 2007). It was also demonstrated in yeast two-hybrid screen that PrP interacted with proteasome subunit alpha type-3 (Lim et al. 2006). Furthermore, as mentioned above, the NLS sequences may target PrP into the nucleus (Gu et al. 2003). In scrapie-infected cells, PrPSc accumulates in the nucleus and this localization requires a microtubule-dependent transport (Mangé et al. 2004). Hence, particularly at pathologic conditions, intracellular proteins may be exposed to interactions with PrP. Importantly, numerous studies have demonstrated that cytosolic PrP may be neurotoxic (Hegde et al. 1998; Ma et al. 2002; Grenier et al. 2006). Therefore, analysis of these interactions may be critical for elucidation of the molecular mechanism of TSE-related neurodegeneration and also the physiological function of PrPC.

Intracellular Partners of PrP

Below are listed intracellular proteins shown to interact with PrP, particularly those partners for which published reports may imply direct binding. Brief description of the physiological functions of the interactors is provided to facilitate prediction of possible consequences of their binding to PrP.

Neuroglobin

Neuroglobin is a heme protein distantly related to myoglobin and hemoglobin, present in the cytosol of retina and brain neurons (rev. in Greenberg et al. 2008). This protein may be involved in neuroprotection in response to ischemia and hypoxia, and against amyloid-beta toxicity in Alzheimer’s disease.

Recently, a direct interaction between neuroglobin and PrP was demonstrated by light-scattering-monitored co-aggregation of purified proteins (Lechauve et al. 2009). Interestingly, the aggregates were formed without substantial changes in the PrP secondary structure, as judged by infrared spectroscopy. The unstructured N-terminal part of PrP was responsible for the binding. The authors did not observe aggregation using the C-terminal domain of PrP or when full-length PrP was mixed with myoglobin. Formation of aggregates with neuroglobin was reduced by increased salt concentration, indicating an ionic character of the binding. The interaction seems to occur in vivo since both proteins were found to colocalize in the perinuclear region of retina cells. It remains to be elucidated whether co-aggregation with PrP may affect the neuroprotective functions of neuroglobin in the cell.

Synapsin

Synapsins are a family of adaptor-like phosphoproteins which can bind to the surface of small synaptic vesicles as well as to the cytoskeletal structures: actin filaments and microtubules (rev. in Ferreira and Rapoport 2002). These proteins are found in the cytoplasm of nerve terminals and play an important role in synaptogenesis, synaptic vesicle turnover, and neurotransmitter release.

Interaction of PrP23–231 with synapsin 1b was discovered by means of a yeast two-hybrid screen of murine brain cDNA library (Spielhaupter and Schätzl 2001). This observation was further confirmed by co-immunoprecipitation from kidney cells transfected with plasmids encoding both proteins. The binding sites for synapsin were localized in the N- and C-terminal parts of PrP. Notably, both proteins are present at high concentrations in the presynaptic nerve terminals. Furthermore, synapsin co-fractionated with PrPC in the microsomal fractions of N2a cells, suggesting that these proteins may interact in vivo at intracellular vesicles. One can speculate that PrP, by interacting with synapsin, may affect synaptic signal transduction. Interestingly, reduced level of synapsin 1 was detected in the brain of scrapie-infected mice (Sisó et al. 2002).

Grb2

Growth factor receptor-bound protein 2 (Grb2) is a key molecule in the transduction of intracellular signal which links activated cell surface receptors to downstream targets of the Ras signaling pathway (rev. in Giubellino et al. 2008). This adapter protein is ubiquitously expressed and found both in cytosolic and nuclear fractions. Grb2-mediated signaling is critical for actin-based cell motility and cell cycle progression; it is also involved in angiogenesis, vasculogenesis, and oncogenesis.

Interaction of PrP with Grb2 was first reported by Spielhaupter and Schätzl (2001), as was the interaction with synapsin (see above). As for synapsin, the interaction with Grb2 was identified in the yeast two-hybrid screen and confirmed by co-immunoprecipitation and co-fractionation. In the immunoprecipitation assays, the binding sites for Grb2 were initially localized both in the N- and C-terminal parts of PrP. Surprisingly, PrP mutations P102L and P105L, linked to GSS and localized within the potential Grb-2-binding motif XPXXP, had no effect on co-immunoprecipitation of the proteins. Subsequently, the binding site for C-terminal SH3 (Src homology domain 3) domain of Grb2 was mapped within the PrP sequence 100–109 by means of NMR, circular dichroism, and fluorescence spectroscopy (Lysek and Wüthrich 2004). For this binding a K d value of 5.5 μM was determined. Contrary to the previous report, in experiments on purified recombinant PrP90–231, the P102L and P105L substitutions abolished the interaction. Since the docking surfaces of Grb2 and PrP bear complementary charges the binding seems to be electrostatic. Notably, Grb2 interacts also with synapsin, which enables formation of a complex composed of all three proteins. The binding to Grb2 and synapsin suggests an influence of PrP on diverse signal transduction pathways.

Tubulin

The recent decade of studies has provided numerous reports evidencing association and even direct interaction of PrP with tubulin—the major component of microtubular cytoskeleton. Heterodimers composed of α- and β-tubulin assemble into tubular polymers called microtubules (rev. in Nogales 2000). Networks of these structures form the cellular cytoskeleton and serve as tracks for active transport. Besides intracellular trafficking and determination of cell morphology, microtubules play a pivotal role in chromosome segregation, cell division, secretion, and cell motility. Microtubules are very dynamic structures that can switch stochastically between assembly and disassembly phases. This property, termed dynamic instability, is crucial to microtubule functions. The assembly and stability of microtubules are regulated by a numerous group of molecules referred to as microtubule associated proteins (MAPs) (rev. in Amos and Schlieper 2005).

The first study implying an association of PrP with tubulin was performed employing a synthetic peptide corresponding to PrP residues 106–126 (Brown et al. 1998). This neurotoxic peptide bound to α- and β-tubulin from brain extracts. Furthermore, it was shown by turbidity measurements that PrP106–126 inhibited tau-dependent microtubule assembly (Brown 2000). The peptide carrying mutation A117V linked to GSS was much more efficient in the inhibition than the wild-type PrP106–126. In the absence of tau neither peptide affected tubulin polymerization. Surprisingly, in subsequent studies (see below) this region of PrP molecule has been shown to lack a tubulin-binding ability. One cannot exclude that in the earlier studies the PrP106–126 peptide interacted in fact with tau protein bound to tubulin. In subsequent studies, complexes of tubulin with native full-length PrP were also reported but interpreted as a result of an indirect interaction (Keshet et al. 2000) or even an artifact of the experimental procedures used (Schmitt-Ulms et al. 2004). Colocalization with the microtubular cytoskeleton has been demonstrated for an N-terminal fragment of PrP produced proteolytically in neuroblastoma N2a cells (Hachiya et al. 2004a). In the same study, co-immunoprecipitation of this part of the PrP molecule with tubulin was observed in homogenates of N2a cells. The PrP region encompassing residues 1–91 was identified as responsible for the colocalization with microtubules. Moreover, nocodazole treatment that disrupts the microtubular network, but not latrunculin A sequestering monomeric actin, affected the intracellular distribution of PrP. The association of PrP with microtubules was proposed to be related to the active transport of this protein in neurons. It was shown that retrograde and anterograde transport of PrP is dependent on microtubule-associated molecular motors belonging to the dynein and kinesin families (Hachiya et al. 2004b). Employing deletion mutants, PrP sequences 23–33 and 53–91 were identified as indispensable for the retrograde and anterograde transport of the molecule, respectively. Also, nuclear targeting of PrPSc has been shown to require an intact microtubule network (Mangé et al. 2004). By covalent cross-linking of purified proteins, we have demonstrated for the first time a direct interaction between PrP and tubulin (Nieznanski et al. 2005). Moreover, we found that PrP co-sedimented with microtubules. A convincing evidence for the in vivo interaction was co-purification of PrP with tubulin from brain homogenate, following a specific multi-step procedure allowing isolation of highly purified tubulin. It is commonly accepted that such a preparation contains only traces of MAPs—molecules of physiological function tightly bound to tubulin. Only the full-length PrP, but not its in vivo-generated proteolytic fragment C1 lacking residues 23–110/112, co-purified with tubulin. In a subsequent study, we showed that the direct binding of PrP to tubulin led to the inhibition of microtubule assembly by inducing tubulin oligomerization (Nieznanski et al. 2006). Furthermore, we observed that the oligomers associated into large aggregates. Employing proteolytic fragments of PrP and a panel of PrP deletion mutants, we identified two microtubule-binding regions within the N-terminal part of the molecule, with residues 23–32 constituting the major binding site and residues 101–110 representing a weak one (Osiecka et al. 2009). Moreover, we found that the sequence 23–32 was responsible for inducing tubulin oligomerization and inhibition of microtubule formation. The effects of PrP were also confirmed at the cellular level (Osiecka et al. 2009). The sequence 23–30, guided into the cytoplasm by the signal peptide 1–22, caused disintegration of the microtubular cytoskeleton and congregation of tubulin in the perinuclear area of human epithelial cells. It is plausible that at low cytosolic concentrations PrP may, similarly to some MAPs, modulate the dynamic instability of microtubules, whereas at high, pathologic levels it may aggregate tubulin causing disassembly of the microtubular cytoskeleton and cell death. Notably, it was demonstrated that tubulin co-immunoprecipitated with PrPSc in brain homogenates of scrapie-infected hamsters (Dong et al. 2008a).

Tau

Tau belongs to a family of structural MAPs that regulate the dynamics of microtubular cytoskeleton (rev. in Avila et al. 2004). Tau is a phosphoprotein mainly expressed in neural cells where it promotes assembly of microtubules and is engaged in their stabilization. Phosphorylation of tau at specific residues negatively regulates its binding to tubulin. Additionally, hyperphosphorylated-tau aggregates into insoluble neurofibrillary tangles which are a hallmark of Alzheimer’s and other neurodegenerative diseases.

Interaction between purified recombinant tau and PrP as well as between the native proteins in brain extracts has been demonstrated by means of pull-down assays and co-immunoprecipitations (Han et al. 2006). It was also shown that tau may bind PrPSc in brain extracts from scrapie-infected hamsters. Furthermore, tau and PrP colocalized in Chinese hamster ovary (CHO) cells co-transfected with plasmids encoding both proteins. The interaction site was roughly mapped within the PrP region 23–91. Deletion of the octarepeats significantly reduced the binding, implying an involvement of this region of the PrP molecule. Surprisingly, the PrP–tau binding was strengthened by introduction of the P102L mutation linked to GSS (Wang et al. 2008). The interaction sites with PrP were mapped within tau regions 1–91 and 186–283 (Wang et al. 2008), of which the latter is also involved in binding to tubulin (rev. in Avila et al. 2004). Therefore, the previously reported reduction in tau–microtubule association in the presence of PrP (Han et al. 2005) may result from blocking of the tubulin-binding region of tau by PrP. Since the tau region 186–279 is also known to promote microtubule assembly (rev. in Avila et al. 2004), its interaction with PrP may affect the physiological function of this MAP and hence cell viability.

GFAP

Glial fibrillary acidic protein (GFAP) is a major component of intermediate filaments (rev. in Messing and Brenner 2003). This cytoskeletal protein is abundantly expressed in astrocytes where it is involved in determination of cell shape.

GFAP was one of the first identified cytosolic interactors of PrP. It was demonstrated in an overlay assay (ligand blotting) that both purified PrPC and PrPSc bound to GFAP from hamster brain homogenates (Oesch et al. 1990). Recombinant GFAP did not bind to PrP in those studies, which was explained by the lack of an N-terminal sequence of GFAP and/or a lack of posttranslational modifications of GFAP. The interaction between PrP and GFAP was subsequently confirmed by co-immunoprecipitations from normal and scrapie-infected brains and in pull-down experiments with purified recombinant proteins (Dong et al. 2008b). The interaction site on PrP was mapped within the C-terminal fragment encompassing residues 91–230. Accordingly, PK-treated PrPSc, corresponding to the globular domain, retained the ability to interact with GFAP. It is not known whether PrP may affect formation of intermediate filaments and what could be the physiological or pathological effect of this interaction. Interestingly, GFAP was found to be more abundant in the brain of scrapie-infected than healthy animals (Dormont et al. 1981; Mackenzie 1983). On the other hand, mice deficient in GFAP did not exhibit changes in TSE susceptibility or production of PrPSc (Tatzelt et al. 1996).

hnRNP A2/B1 and Aldolase C

In an overlay assay, it was also found that PrP may interact with hnRNP A2/B1 and aldolase C (Strom et al. 2006). Interestingly, all those proteins are able to bind RNA. Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) belongs to a family of RNA-binding proteins involved in mRNA nucleo-cytoplasmic transport and splicing (rev. in Weighardt et al. 1996). hnRNP A2/B1 is expressed in various cell types, including neurons. Aldolases are glycolytic enzymes catalyzing the aldol cleavage of fructose-1,6-bisphosphate. Aldolase C/zebrin II is an isoform expressed predominantly in astrocytes and Purkinje cells (Thompson et al. 1982) which also exhibits mRNA binding activity (Cañete-Soler et al. 2005).

On ligand blots, purified recombinant PrP23–231 bound to the both proteins from the cytosolic fraction of brain homogenate obtained from Prnp-knockout mice (Strom et al. 2006). Moreover, hnRNP A2/B1 co-immunoprecipitated with PrPC in the cytosolic fraction of human Hodgkin’s lymphoma cells. It is plausible that the binding to hnRNP may be related to the postulated involvement of PrP in nucleic acid metabolism. Further studies are required to determine the consequences of these interactions. Interestingly, in the brain of BSE-infected mice at the terminal stage of disease, an increased concentration of a long transcript of the aldolase C gene was reported (Dandoy-Dron et al. 2000).

Nrf2

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a member of the family of basic leucine zipper transcription factors, which controls expression of antioxidant and detoxifying genes (rev. in Motohashi and Yamamoto 2004; Nguyen et al. 2009). Nrf2 is sequestered in the cytoplasm by the actin-associated protein Keap 1. In response to oxidative stress, Nrf2 is released from Keap 1, which allows for its translocation from the cytoplasm to the nucleus and activation of target gene expression. Nrf2 is ubiquitously expressed in a wide range of tissues and, notably, is involved in neuroprotection.

Interaction with Nrf2 has been found in a screen of a bacteriophage expression library of mouse brain cDNA using mouse PrP1–232 fused with alkaline phosphatase as a probe (Yehiely et al. 1997). At present, it is equally plausible that PrP may interact with the cytosolic and the nuclear fraction of Nrf2. It would be of particular interest to check whether PrP can tether Nrf2 in an inactive form in the cytosol.

Bcl-2

B-cell lymphoma 2 protein (Bcl-2) belongs to the group of molecules that regulate cell survival and death (rev. in Wong and Puthalakath 2008; Szegezdi et al. 2009). Bcl-2 is an anti-apoptotic protein which binds to pro-apoptotic members of the Bcl-2 family and antagonizes their functions. Bcl-2 prevents the release of cytochrome c into the cytoplasm thereby blocking activation of caspase cascade and apoptotic death. Furthermore, Bcl-2 is involved in the regulation of intracellular calcium homeostasis. This ubiquitously expressed oncoprotein is anchored on the cytoplasmic face of the nuclear envelope, ER, and mitochondrial membrane. Notably, expression of Bcl-2 is induced in the brain following ischemia (rev. in Soane and Fiskum 2005). This observation is related to the Bcl-2 neuroprotective functions that prevent ischemic death of neurons.

Selective binding of PrP to Bcl-2 has been demonstrated in a yeast two-hybrid screen of a murine cerebellar cDNA library (Kurschner and Morgan 1995). In this system, mouse PrP72–254 interacted with Bcl-2 but not with Bax or A1—other members of the Bcl-2 family. Surprisingly, the authors were unable to demonstrate co-immunoprecipitation of Bcl-2 with PrP. Subsequent studies employing PrP constructs directed to different cellular compartments showed that cytosolically localized PrP lacking the N- and C-terminal signal sequences associated with Bcl-2 (Rambold et al. 2006). Bcl-2 and this cytosolic PrP could be co-immunoprecipitated from transiently transfected neuroblastoma cells. Notably, Bcl-2 co-aggregated with cytosolic PrP indicating that this protein may sequester Bcl-2 leading to a loss of its anti-apoptotic function. In fact, PrP aggregated in the cytosol induced apoptosis in neurons. Overexpression of Bcl-2 interfered with the toxicity of the cytosolic fraction of PrP. Binding of cytosolic PrP to Bcl-2, recruitment of Bcl-2 into PrP aggregates, as well as PrP-induced apoptosis were prevented by chaperones Hsp70 and Hsp40, which were also shown to interact with PrP. Notably, aggregation of Bcl-2 may be related to the pathology of TSE since, in contrast to healthy animals, Bcl-2 has been found in a detergent-insoluble fraction of brain extracts of scrapie-infected mice. According to the results of Kurschner and Morgan (1995), Bcl-2 did not co-immunoprecipitate with PrP from cells expressing wild-type PrP. Only PrP missorted to the cytosol was cytotoxic. Inhibition of proteasome significantly enhanced the apoptotic effect of cytosolic PrP. PrP constructs targeted to the nucleus, mitochondria, or ER did not affect cell viability. Experiments on deletion mutants, including the TSE-linked mutants Q160Stop and W145Stop, have revealed that region 115–146 of PrP is responsible for the toxicity of the cytosolic protein. In light of the above observations, it has been postulated that the cytotoxicity of cytosolic PrP results from the co-aggregation of Bcl-2 and loss of Bcl-2 function.

NRAGE

Neurotrophin receptor-interacting MAGE homolog (NRAGE), also known as Dlxin-1 or MAGE-D1, is a pro-apoptotic member of the melanoma-associated antigen family (MAGE) (Salehi et al. 2000). NRAGE interacts with the cytoplasmic domain of the p75 neurothropin receptor, which induces activation of caspases and cell death through the Jun N-terminal kinase (JNK)-dependent mitochondrial pathway (Salehi et al. 2002). Notably, NRAGE is involved in the regulation of the cell cycle and apoptosis during neuronal development.

In a yeast two-hybrid screen of a rat brain cDNA library, Bragason and Palsdottir (2005) have demonstrated that PrP22–231 may interact with NRAGE. The interaction was confirmed in a GST pull-down assay for purified recombinant proteins and by co-immunoprecipitation from COS7 cells co-transfected with plasmids encoding NRAGE and a cytosolic construct of PrP lacking both signal sequences. The docking site for NRAGE was localized to the C-terminal globular domain of PrP (residues 122–231). After proteasome inhibition, PrPC containing the signal sequences and NRAGE co-aggregated together in the cytosol of co-transfected COS7 cells. Importantly, co-expression of NRAGE and the cytosolic construct of PrP in neuroblastoma (N2a) cells affected mitochondrial membrane potential, which is an indicator of the early stages of apoptosis. This is another study demonstrating that interaction of PrP with a cytosolic protein may induce cell death.

CK2

Casein kinase 2 (CK2) is a pleiotropic Ser/Thr protein kinase, under certain conditions able to phosphorylate tyrosyl residues as well (rev. by Pinna 2002). CK2 is found in almost all tissues and nearly every compartment of eukaryotic cells (rev. in Faust and Montenarh 2000). This ubiquitous enzyme is involved in the regulation of numerous crucial processes including signal transduction, apoptosis, transcription, tumorigenesis, cell division, and proliferation. Furthermore, CK2 plays important roles in regulation of actin and tubulin cytoskeletons (rev. in Canton and Litchfield 2006). CK2 is composed of two constitutively active catalytic subunits and two regulatory ones.

Employing an overlay assay and the surface plasmon resonance technique, Meggio et al. (2000) have demonstrated that PrP interacts with the catalytic subunits of CK2. A K d of approx. 0.5 μM was calculated for this binding. The C-terminal part of bovine PrP composed of residues 105–242 was identified as an interaction site. Importantly, PrP binding stimulated the catalytic activity of the kinase. For this stimulation, the 25–116 fragment of PrP was responsible, most probably through its N-terminal basic sequence. PrP did not bind to casein kinase 1, highlighting specificity of the interaction. Moreover, it has been demonstrated that bovine PrP is an in vitro substrate of CK2 (Negro et al. 2000). However, the interaction is not a simple substrate binding by the enzyme, since mouse PrP which lacks a phosphorylatable site still interacts with CK2. The interaction was subsequently confirmed in pull-down assays performed for purified recombinant proteins, as well as in co-immunoprecipitation of native proteins from brain extracts (Chen et al. 2008a). It was also shown that CK2 interacted more strongly with unglycosylated PrP than with its glycosylated forms. Furthermore, changes in the expression pattern of CK2 subunits were observed in the brain of scrapie-infected animals and human TSE cases (Chen et al. 2008b). Since CK2 is also an ectokinase found in the extracellular matrix (Canton and Litchfield 2006), it is possible that it may interact not only with cytoPrP or nuclear PrP, but also with PrPC anchored on the plasma membrane. Further studies are required to check whether PrP enhances CK2 activity also in the cell.

14-3-3 protein

The family of 14-3-3 proteins consists of seven isoforms (β, γ, ε, σ, ζ, τ, and η) expressed most abundantly in neurons and glial cells of the central nervous system (rev. in Berg et al. 2003). These proteins constitute about 1% of the cytosolic proteome of neuronal cells and interact with numerous proteins, including apoptosis regulators, cytoskeletal proteins, signaling proteins, transcription factors, metabolic enzymes, and tumor suppressors.

14-3-3 protein is detected in the cerebrospinal fluid of Creutzfeldt–Jacob-diseased individuals and hence it is used as an ante-mortem marker as well (Hsich et al. 1996). Notably, the ζ isoform of this protein has been found in PrP amyloid deposits of sporadic CJD and variant CJD patients (Richard et al. 2003) whereas isoform ε is a specific component of PrP amyloid plaques found in GSS (Di Fede et al. 2007). These observations suggest that 14-3-3 protein binds to PrPSc and/or PrPC. In fact, 14-3-3 protein has been demonstrated to interact directly with PrP (Satoh et al. 2005). PrPC co-immunoprecipitated with 14-3-3 protein in human brain extract and was bound by purified human recombinant 14-3-3ζ protein in an overlay assay. Additionally, purified recombinant PrP and 14-3-3 protein interacted on an overlay blot, indicating that the binding was direct. The interaction site with 14-3-3 protein was roughly localized within the N-terminal part of the PrP molecule spanning residues 23–137. Furthermore, immunocytochemical analysis showed that PrPC and 14-3-3 protein colocalized in mitochondria of cultured neuronal progenitor cells. In the same study, an interaction between 14-3-3 protein and Hsp60, and a colocalization similar to that with PrPC, were demonstrated suggesting formation of a ternary complex. Recently, the interaction site with 14-3-3 protein has been narrowed to the PrP region 106–126 (Mei et al. 2009). The consequences of PrP binding to 14-3-3 protein remain to be determined.

Mahogunin

Mahogunin (Mgrn) is a cytosolic Really Interesting New Gene (RING) domain-containing protein of ubiquitin ligase activity involved in cell survival signaling (rev. in He et al. 2003a). Although substrates of this ligase are unknown, loss of its function leads to spongiform neurodegeneration of the brain and a change in coat color found in mahoganoid mutant mice (He et al. 2003b).

Very recently, Chakrabarti and Hegde (2009) have reported functional depletion of Mgrn in cultured cells overexpressing cytosolic form of PrP or transmembranal CtmPrP. By performing semipermeabilization of these cells the authors demonstrated co-aggregation of Mgrn with cytosolic PrP. Recombinant Mgrn pulled down PrP from brain lysate confirming the interaction. The octarepeats of PrP were found to be involved in the binding since deletion of this region abrogated the ability to co-aggregate Mgrn. Moreover, a synthetic peptide corresponding to the PrP octarepeat sequence could pull down Mgrn. Although PrP undergoes ubiquitination (Yedidia et al. 2001), it is not a substrate for Mgrn (He et al. 2003b). The interaction with cytosolic PrP leads to Mgrn sequestration and triggers pathological changes resembling the effects of Mgrn depletion. These changes may be reversed or prevented by competition for the PrP–Mgrn binding, overexpression of functional Mgrn or redistribution of cytosolic PrP. Hence, it has been proposed that the molecular mechanism of cytotoxicity of cytosolic PrP is closely related to the interaction with Mgrn resulting in a loss of its function.

Chaperones

The search for a molecular chaperone capable of influencing the pathological conversion of PrPC into PrPSc attracts much attention. To date, interactions with numerous chaperones have been reported which seem to be related to normal cellular folding of PrPC during its biosynthesis. For example, it was demonstrated that PrP bound directly to stress-inducible protein 1 (Martins et al. 1997), αβ-crystallin (Sun et al. 2005), Hsp60 (Edenhofer et al. 1996), and co-immunoprecipitated with protein disulfide isomerase, calnexin, calreticulin, BiP, grp94 (Capellari et al. 1999), Hsp40, and Hsp70 (Rambold et al. 2006). There is no evidence indicating that interaction with PrP may affect function of any of these chaperones.

PrP as Component of Multimolecular Complexes

The number of already identified PrP interactors implies that this protein may be a component of large multimolecular complexes. Some of these complexes seem to contain intracellular proteins. In a proteomic approach, Giorgi et al. (2009) identified proteins co-purifying with the proteinase K-resistant core of PrPSc isolated from scrapie-infected hamster brain. The major components of the PrPSc-enriched preparations, identified by mass spectrometry, were ferritin heavy chain, calcium/calmodulin-dependent protein kinase α type II (CaMKIIα), apolipoprotein E, and tubulin. Among these proteins only apolipoprotein E is an extracellular protein. Since the purification procedure employed PK treatment, the co-purified proteins seem to acquire protease resistance by co-aggregation with PrPSc, with a possible exception of ferritin which is known to be intrinsically PK-resistant (Atkinson et al. 1989). The possible association was confirmed by colocalization of PrPSc with CaMKIIα in the cytoplasm of hippocampal neurons and with apolipoprotein E around lateral ventricles and blood vessels of the brain. Interestingly, an increased expression of CaMKIIα has been observed in the brain of scrapie-infected animals (Jin et al. 1999; Cosseddu et al. 2007). Similarly, expression of ferritin, the intracellular protein responsible for sequestration and storage of iron (rev. in Koorts and Viljoen 2007), was elevated in the brain of TSE-infected mice (Kim et al. 2007). Association of PrPSc with the heavy and light chains of ferritin had been demonstrated earlier by co-purification and co-immunoprecipitation from CJD brain homogenates (Mishra et al. 2004). The complex could be dissociated with salt suggesting an ionic character of the interactions. Furthermore, PrPSc and ferritin were co-endocytosed and co-transported in vesicles across intestinal epithelial cells. It was proposed that ferritin may facilitate intestinal uptake of PrPSc from contaminated food. It remains, however, undetermined whether in the above study all co-purifying proteins interacted directly with PrPSc.

Complexes with proteins residing in PrPC vicinity were covalently conserved in the brain by means of time-controlled transcardiac perfusion cross-linking (Schmitt-Ulms et al. 2004). In these complexes, numerous plasma-membrane and extracellular molecules were found: amyloid precursor protein (APP), amyloid beta precursor-like protein 2 (APLP2), neural cell adhesion molecules (N-CAM), opioid binding protein (OBCAM), neurotrimin, nectin-like 1 protein, calsyntenin 1 (CLSTN1), myelin-associated glycoprotein (MAG), clusterin precursor (CLU), and dipeptidyl aminopeptidase-like protein 6 (DPP6). Interestingly, in this experimental approach an intracellular protein—dynamin-1 (DNM1)—was also identified as a component of the PrPC complexes. Notably, dynamin is an enzyme involved in membrane trafficking which interacts with microtubular cytoskeleton (Shpetner and Vallee 1989). Also in this study, it is unclear which of the above proteins were cross-linked directly with PrPC.

Recently, proteomic profiling of PrPC interactome was performed by means of microarray technology and again revealed the intracellular partners of this protein (Satoh et al. 2009). An array of approximately 5000 purified human proteins was probed with human PrP spanning residues 23–231. Native-like posttranslational modifications were ensured by expression of the target proteins in the baculovirus system and of PrP in human embryonic kidney cells (HEK293). In this study, 47 novel binding partners were identified, which again suggests formation of multimolecular complexes with PrP. Surprisingly, the majority of these interactors are known as proteins involved in the recognition of nucleic acids. The results of the microarray analysis were verified for selected proteins: homeobox A1 protein (HOXA1), polo-like kinase 3 (PLK3), and methylpurine-DNA glycosylase (MPG) in immunoprecipitation and colocalization experiments. HOXA1 is a transcription factor critical for development of the hindbrain (Carpenter et al. 1993), PLK3 is a Ser/Thr kinase involved in regulation of the entry into mitosis (rev. in Myer et al. 2005), whereas MPG is an enzyme repairing damaged DNA (rev. in Bouziane et al. 1998). All of these proteins underwent co-immunoprecipitation with PrP23-231 in HEK293 cells lysates. Interestingly, DsRed-tagged PrP23-231 transiently expressed in HEK293 cells was found predominantly in the nucleus and cytoplasm and less abundantly on the plasma membrane. In this cell culture, PrP coexisted with HOXA1 and MPG in the nucleus, whereas with PLK3 in the cytosol and on the cell surface.

Concluding Remarks

There is growing evidence implying that cytosolically mislocalized PrP may cause neurodegeneration. Experiments on transgenic mice and cell culture models of neurodegenerative conditions have revealed that PrP residing fully or partially (CtmPrP) in the cytosol can be neurotoxic. Transgenic mice that generate CtmPrP above a threshold level develop a TSE-like disease (Hegde et al. 1999). Forced cytosolic expression of PrP in transgenic mice leads to severe degeneration of cerebellar granule neurons and gliosis (Ma et al. 2002). The mistargeted PrP is also toxic to forebrain neurons, which has been demonstrated for inducible transgenic mice expressing cytoPrP in this region of the brain (Wang et al. 2009). The consequences of cytoPrP accumulation seem to depend on the neuronal context in the brain. The neuropathological changes observed in the 1D4 mouse model expressing cytoPrP varied among the brain regions: cerebellum, hippocampus, and neocortex (Faas et al. 2009). Surprisingly, although cytosolic PrP exhibits a toxic potential, it has never been shown to be infectious. Conflicting results have been obtained for cell cultures expressing cytoPrP. In those studies the mislocalized PrP was cytotoxic (Ma et al. 2002; Rane et al. 2004; Grenier et al. 2006), had no effect on cell viability (Fioriti et al. 2005) or even protected neurons against Bax-mediated apoptosis (Roucou et al. 2003). A cell type-selective toxicity of cytoPrP may be an explanation for some of these discrepancies. Plausibly, cytoPrP may be cytotoxic only at particular conditions and/or concentrations.

How might the cytosolically mistargeted PrP be neurotoxic? One can assume that most of the physiological partners of PrPC are extracellular or membrane proteins. It is also plausible that at its normal, low concentration in the cytoplasm PrP may modulate the physiological functions of the interacting proteins whereas at elevated, pathological levels it may dysregulate them leading to neurodegeneration. Notably, as demonstrated by Rambold et al. (2006), only PrP accumulated in the cytosol was neurotoxic. Aggregation of intracellular partners induced by PrP has been observed in numerous studies, e.g., for mahogunin, NRAGE, tubulin, and Bcl-2 (summarized in Table 1). This aggregation/co-aggregation may lead to a loss of function of the interactor and may represent a molecular mechanism of toxicity of the PrP mistargeted into the cytosol. Therefore, the interactions of PrP with some cytosolic proteins may be considered as potential targets for drugs (inhibitors of the interaction) in therapy of TSEs. It should be noted that some physiological interactions with intracellular proteins may be related to biogenesis/modifications of PrP (e.g., interactions with chaperones) and trafficking of PrP within the cell (e.g., endocytosis, microtubule-dependent transport). The consequences of the numerous interactions outlined in this review remain to be elucidated, however, one can suppose that most of them will turn out to be deleterious to the cell.

Acknowledgments

The author thanks Dr. Hanna Nieznanska for critical reading of the manuscript and helpful comments. This work was supported by a statutory grant to the Nencki Institute of Experimental Biology from the Ministry of Science and Higher Education.

Abbreviations

Bcl-2

B-cell lymphoma 2 protein

BSE

Bovine spongiform encephalopathy

C1

N-terminally truncated form of PrP encompassing residues ~110/112–231

C2

N-terminally truncated form of PrP encompassing residues ~90/91–231

CaMKIIα

Calcium/calmodulin-dependent protein kinase α type II

CJD

Creutzfeldt–Jakob disease

CK2

Casein kinase 2

CtmPrP

Transmembrane form of PrP with the C-terminus residing in the lumen of ER

CWD

Chronic wasting disease

cyPrP, cytPrP, cytoPrP

PrP residing entirely in the cytosol

ER

Endoplasmic reticulum

FFI

Fatal familial insomnia

GFAP

Glial fibrillary acidic protein

GPI

Glycosylphosphatidylinositol

Grb2

Growth factor receptor-bound protein 2

GSS

Gerstmann–Sträussler–Scheinker disease

HEK

Human embryonic kidney cells

hnRNP

Heterogeneous nuclear ribonucleoprotein

Hsp

Heat shock protein

MAPs

Microtubule-associated proteins

Mgrn

Mahogunin

NLS

Nuclear localization signal

NRAGE

Neurotrophin receptor-interacting MAGE homolog

Nrf2

Nuclear factor erythroid 2-related factor 2

NtmPrP

Transmembrane form of PrP with the N-terminus residing in the lumen of ER

OR

Octapeptide repeats

PK

Proteinase K

Prnp

Prion protein gene

PrP

Prion protein

PrPC

Cellular form of PrP

PrPSc

Scrapie form of PrP

rPrP

Recombinant PrP

SP

Signal peptide

TM

Transmembrane domain

TSE

Transmissible spongiform encephalopathy

References

  1. Amos LA, Schlieper D (2005) Microtubules and maps. Adv Protein Chem 71:257–298 [DOI] [PubMed] [Google Scholar]
  2. Atkinson BG, Dean RL, Tomlinson J, Blaker TW (1989) Rapid purification of ferritin from lysates of red blood cells using proteinase-K. Biochem Cell Biol 67:52–57 [DOI] [PubMed] [Google Scholar]
  3. Avila J, Lucas JJ, Perez M, Hernandez F (2004) Role of tau protein in both physiological and pathological conditions. Physiol Rev 84:361–384 [DOI] [PubMed] [Google Scholar]
  4. Basler K, Oesch B, Scott M, Westaway D, Wälchli M, Groth DF, McKinley MP, Prusiner SB, Weissmann C (1986) Scrapie and cellular PrP isoforms are encoded by the same chromosomal gene. Cell 46:417–428 [DOI] [PubMed] [Google Scholar]
  5. Bendheim PE, Brown HR, Rudelli RD, Scala LJ, Goller NL, Wen GY, Kascsak RJ, Cashman NR, Bolton DC (1992) Nearly ubiquitous tissue distribution of the scrapie agent precursor protein. Neurology 42:149–156 [DOI] [PubMed] [Google Scholar]
  6. Berg D, Holzmann C, Riess O (2003) 14-3-3 proteins in the nervous system. Nat Rev Neurosci 4:752–762 [DOI] [PubMed] [Google Scholar]
  7. Bouziane M, Miao F, Ye N, Holmquist G, Chyzak G, O’Connor TR (1998) Repair of DNA alkylation damage. Acta Biochim Pol 45:191–202 [PubMed] [Google Scholar]
  8. Bragason BT, Palsdottir A (2005) Interaction of PrP with NRAGE, a protein involved in neuronal apoptosis. Mol Cell Neurosci 29:232–244 [DOI] [PubMed] [Google Scholar]
  9. Brown DR (2000) Altered toxicity of the prion protein peptide PrP106–126 carrying the Ala(117)–>Val mutation. Biochem J 346:785–791 [PMC free article] [PubMed] [Google Scholar]
  10. Brown DR, Schmidt B, Kretzschmar HA (1998) Prion protein fragment interacts with PrP-deficient cells. J Neurosci Res 52:260–267 [DOI] [PubMed] [Google Scholar]
  11. Brown DR, Wong BS, Hafiz F, Clive C, Haswell SJ, Jones IM (1999) Normal prion protein has an activity like that of superoxide dismutase. Biochem J 344:1–5 [PMC free article] [PubMed] [Google Scholar]
  12. Büeler H, Aguzzi A, Sailer A, Greiner RA, Autenried P, Aguet M, Weissmann C (1993) Mice devoid of PrP are resistant to scrapie. Cell 73:1339–1347 [DOI] [PubMed] [Google Scholar]
  13. Cañete-Soler R, Reddy KS, Tolan DR, Zhai J (2005) Aldolases A and C are ribonucleolytic components of a neuronal complex that regulates the stability of the light-neurofilament mRNA. J Neurosci 25:4353–4364 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Canton DA, Litchfield DW (2006) The shape of things to come: an emerging role for protein kinase CK2 in the regulation of cell morphology and the cytoskeleton. Cell Signal 18:267–275 [DOI] [PubMed] [Google Scholar]
  15. Capellari S, Zaidi SI, Urig CB, Perry G, Smith MA, Petersen RB (1999) Prion protein glycosylation is sensitive to redox change. J Biol Chem 274:34846–34850 [DOI] [PubMed] [Google Scholar]
  16. Carpenter EM, Goddard JM, Chisaka O, Manley NR, Capecchi MR (1993) Loss of Hox-A1 (Hox-1.6) function results in the reorganization of the murine hindbrain. Development 118:1063–1075 [DOI] [PubMed] [Google Scholar]
  17. Caughey B, Brown K, Raymond GJ, Katzenstein GE, Thresher W (1994) Binding of the protease-sensitive form of PrP (prion protein) to sulfated glycosaminoglycan and Congo red. J Virol 68:2135–2141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chakrabarti O, Hegde RS (2009) Functional depletion of mahogunin by cytosolically exposed prion protein contributes to neurodegeneration. Cell 137:1136–1147 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chakrabarti O, Ashok A, Hegde RS (2009) Prion protein biosynthesis and its emerging role in neurodegeneration. Trends Biochem Sci 34:287–295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chen SG, Teplow DB, Parchi P, Teller JK, Gambetti P, Autilio-Gambetti L (1995) Truncated forms of the human prion protein in normal brain and in prion diseases. J Biol Chem 270:19173–19180 [DOI] [PubMed] [Google Scholar]
  21. Chen J, Gao C, Shi Q, Wang G, Lei Y, Shan B, Zhang B, Dong C, Shi S, Wang X, Tian C, Han J, Dong X (2008a) Casein kinase II interacts with prion protein in vitro and forms complex with native prion protein in vivo. Acta Biochim Biophys Sin (Shanghai) 40:1039–1047 [DOI] [PubMed] [Google Scholar]
  22. Chen JM, Gao C, Shi Q, Shan B, Lei YJ, Dong CF, An R, Wang GR, Zhang BY, Han J, Dong XP (2008b) Different expression patterns of CK2 subunits in the brains of experimental animals and patients with transmissible spongiform encephalopathies. Arch Virol 153:1013–1020 [DOI] [PubMed] [Google Scholar]
  23. Cosseddu GM, Andréoletti O, Maestrale C, Robert B, Ligios C, Piumi F, Agrimi U, Vaiman D (2007) Gene expression profiling on sheep brain reveals differential transcripts in scrapie-affected/not-affected animals. Brain Res 1142:217–222 [DOI] [PubMed] [Google Scholar]
  24. Dandoy-Dron F, Benboudjema L, Guillo F, Jaegly A, Jasmin C, Dormont D, Tovey MG, Dron M (2000) Enhanced levels of scrapie responsive gene mRNA in BSE-infected mouse brain. Brain Res Mol Brain Res 76:173–179 [DOI] [PubMed] [Google Scholar]
  25. Di Fede G, Giaccone G, Limido L, Mangieri M, Suardi S, Puoti G, Morbin M, Mazzoleni G, Ghetti B, Tagliavini F (2007) The epsilon isoform of 14-3-3 protein is a component of the prion protein amyloid deposits of Gerstmann–Sträussler–Scheinker disease. J Neuropathol Exp Neurol 66:124–130 [DOI] [PubMed] [Google Scholar]
  26. Dong CF, Shi S, Wang XF, An R, Li P, Chen JM, Wang X, Wang GR, Shan B, Zhang BY, Han J, Dong XP (2008a) The N-terminus of PrP is responsible for interacting with tubulin and fCJD related PrP mutants possess stronger inhibitive effect on microtubule assembly in vitro. Arch Biochem Biophys 470:83–92 [DOI] [PubMed] [Google Scholar]
  27. Dong CF, Wang XF, Wang X, Shi S, Wang GR, Shan B, An R, Li XL, Zhang BY, Han J, Dong XP (2008b) Molecular interaction between prion protein and GFAP both in native and recombinant forms in vitro. Med Microbiol Immunol 197:361–368 [DOI] [PubMed] [Google Scholar]
  28. Dormont D, Delpech B, Delpech A, Courcel MN, Viret J, Markovits P, Court L (1981) Hyperproduction of glial fibrillary acidic protein (GFA) during development of experimental scrapie in mice. C R Seances Acad Sci III 293:53–56 [PubMed] [Google Scholar]
  29. Edenhofer F, Rieger R, Famulok M, Wendler W, Weiss S, Winnacker EL (1996) Prion protein PrPc interacts with molecular chaperones of the Hsp60 family. J Virol 70:4724–4728 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Faas H, Jackson WS, Borkowski AW, Wang X, Ma J, Lindquist S, Jasanoff A (2009) Context-dependent perturbation of neural systems in transgenic mice expressing a cytosolic prion protein. Neuroimage. doi:10.1016/j.neuroimage.2009.10.009 [DOI] [PMC free article] [PubMed]
  31. Faust M, Montenarh M (2000) Subcellular localization of protein kinase CK2. A key to its function? Cell Tissue Res 301:329–340 [DOI] [PubMed] [Google Scholar]
  32. Ferreira A, Rapoport M (2002) The synapsins: beyond the regulation of neurotransmitter release. Cell Mol Life Sci 59:589–595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Fioriti L, Dossena S, Stewart LR, Stewart RS, Harris DA, Forloni G, Chiesa R (2005) Cytosolic prion protein (PrP) is not toxic in N2a cells and primary neurons expressing pathogenic PrP mutations. J Biol Chem 280:11320–11328 [DOI] [PubMed] [Google Scholar]
  34. Gabus C, Derrington E, Leblanc P, Chnaiderman J, Dormont D, Swietnicki W, Morillas M, Surewicz WK, Marc D, Nandi P, Darlix JL (2001) The prion protein has RNA binding and chaperoning properties characteristic of nucleocapsid protein NCP7 of HIV-1. J Biol Chem 276:19301–19309 [DOI] [PubMed] [Google Scholar]
  35. Giorgi A, Di Francesco L, Principe S, Mignogna G, Sennels L, Mancone C, Alonzi T, Sbriccoli M, De Pascalis A, Rappsilber J, Cardone F, Pocchiari M, Maras B, Schininà ME (2009) Proteomic profiling of PrP27–30-enriched preparations extracted from the brain of hamsters with experimental scrapie. Proteomics 9:3802–3814 [DOI] [PubMed] [Google Scholar]
  36. Giubellino A, Burke TR Jr, Bottaro DP (2008) Grb2 signaling in cell motility and cancer. Expert Opin Ther Targets 12:1021–1033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Greenberg DA, Jin K, Khan AA (2008) Neuroglobin: an endogenous neuroprotectant. Curr Opin Pharmacol 8:20–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Grenier C, Bissonnette C, Volkov L, Roucou X (2006) Molecular morphology and toxicity of cytoplasmic prion protein aggregates in neuronal and non-neuronal cells. J Neurochem 97:1456–1466 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Gu Y, Hinnerwisch J, Fredricks R, Kalepu S, Mishra RS, Singh N (2003) Identification of cryptic nuclear localization signals in the prion protein. Neurobiol Dis 12:133–149 [DOI] [PubMed] [Google Scholar]
  40. Hachiya NS, Watanabe K, Sakasegawa Y, Kaneko K (2004a) Microtubules-associated intracellular localization of the NH2-terminal cellular prion protein fragment. Biochem Biophys Res Commun 313:818–823 [DOI] [PubMed] [Google Scholar]
  41. Hachiya NS, Watanabe K, Yamada M, Sakasegawa Y, Kaneko K (2004b) Anterograde and retrograde intracellular trafficking of fluorescent cellular prion protein. Biochem Biophys Res Commun 315:802–807 [DOI] [PubMed] [Google Scholar]
  42. Hajj GN, Lopes MH, Mercadante AF, Veiga SS, da Silveira RB, Santos TG, Ribeiro KC, Juliano MA, Jacchieri SG, Zanata SM, Martins VR (2007) Cellular prion protein interaction with vitronectin supports axonal growth and is compensated by integrins. J Cell Sci 120:1915–1926 [DOI] [PubMed] [Google Scholar]
  43. Han J, Wang X, Yao H, Gao C, Li F, Zhang B, Jiang H, Dong X (2005) Tau-mediated microtubule formation inhibited by prion protein. Neurosci Bull 21:398–403 [Google Scholar]
  44. Han J, Zhang J, Yao H, Wang X, Li F, Chen L, Gao C, Gao J, Nie K, Zhou W, Dong X (2006) Study on interaction between microtubule associated protein tau and prion protein. Sci China C Life Sci 49:473–479 [DOI] [PubMed] [Google Scholar]
  45. Haraguchi T, Fisher S, Olofsson S, Endo T, Groth D, Tarentino A, Borchelt DR, Teplow D, Hood L, Burlingame A, Lycke E, Kobata A, Prusiner SB (1989) Asparagine-linked glycosylation of the scrapie and cellular prion proteins. Arch Biochem Biophys 274:1–13 [DOI] [PubMed] [Google Scholar]
  46. He L, Eldridge AG, Jackson PK, Gunn TM, Barsh GS (2003a) Accessory proteins for melanocortin signaling: attractin and mahogunin. Ann N Y Acad Sci 994:288–298 [DOI] [PubMed] [Google Scholar]
  47. He L, Lu XY, Jolly AF, Eldridge AG, Watson SJ, Jackson PK, Barsh GS, Gunn TM (2003b) Spongiform degeneration in mahoganoid mutant mice. Science 299:710–712 [DOI] [PubMed] [Google Scholar]
  48. Hegde RS, Mastrianni JA, Scott MR, DeFea KA, Tremblay P, Torchia M, DeArmond SJ, Prusiner SB, Lingappa VR (1998) A transmembrane form of the prion protein in neurodegenerative disease. Science 279:827–834 [DOI] [PubMed] [Google Scholar]
  49. Hegde RS, Tremblay P, Groth D, DeArmond SJ, Prusiner SB, Lingappa VR (1999) Transmissible and genetic prion diseases share a common pathway of neurodegeneration. Nature 402:822–826 [DOI] [PubMed] [Google Scholar]
  50. Heske J, Heller U, Winklhofer KF, Tatzelt J (2004) The C-terminal globular domain of the prion protein is necessary and sufficient for import into the endoplasmic reticulum. J Biol Chem 279:5435–5443 [DOI] [PubMed] [Google Scholar]
  51. Hornshaw MP, McDermott JR, Candy JM (1995) Copper binding to the N-terminal tandem repeat regions of mammalian and avian prion protein. Biochem Biophys Res Commun 207:621–629 [DOI] [PubMed] [Google Scholar]
  52. Hsich G, Kenney K, Gibbs CJ, Lee KH, Harrington MG (1996) The 14-3-3 brain protein in cerebrospinal fluid as a marker for transmissible spongiform encephalopathies. N Engl J Med 335:924–930 [DOI] [PubMed] [Google Scholar]
  53. Jackson GS, Murray I, Hosszu LL, Gibbs N, Waltho JP, Clarke AR, Collinge J (2001) Location and properties of metal-binding sites on the human prion protein. Proc Natl Acad Sci USA 98:8531–8535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Jiménez-Huete A, Lievens PM, Vidal R, Piccardo P, Ghetti B, Tagliavini F, Frangione B, Prelli F (1998) Endogenous proteolytic cleavage of normal and disease-associated isoforms of the human prion protein in neural and non-neural tissues. Am J Pathol 153:1561–1572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Jin JK, Choi JK, Lee HG, Kim YS, Carp RI, Choi EK (1999) Increased expression of CaM kinase II alpha in the brains of scrapie-infected mice. Neurosci Lett 273:37–40 [DOI] [PubMed] [Google Scholar]
  56. Jones CE, Abdelraheim SR, Brown DR, Viles JH (2004) Preferential Cu2+ coordination by His96 and His111 induces beta-sheet formation in the unstructured amyloidogenic region of the prion protein. J Biol Chem 279:32018–32027 [DOI] [PubMed] [Google Scholar]
  57. Keshet GI, Bar-Peled O, Yaffe D, Nudel U, Gabizon R (2000) The cellular prion protein colocalizes with the dystroglycan complex in the brain. J Neurochem 75:1889–1897 [DOI] [PubMed] [Google Scholar]
  58. Kim SJ, Hegde RS (2002) Cotranslational partitioning of nascent prion protein into multiple populations at the translocation channel. Mol Biol Cell 13:3775–3786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Kim SJ, Rahbar R, Hegde RS (2001) Combinatorial control of prion protein biogenesis by the signal sequence and transmembrane domain. J Biol Chem 276:26132–26140 [DOI] [PubMed] [Google Scholar]
  60. Kim BH, Jun YC, Jin JK, Kim JI, Kim NH, Leibold EA, Connor JR, Choi EK, Carp RI, Kim YS (2007) Alteration of iron regulatory proteins (IRP1 and IRP2) and ferritin in the brains of scrapie-infected mice. Neurosci Lett 422:158–163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Koorts AM, Viljoen M (2007) Ferritin and ferritin isoforms I: structure-function relationships, synthesis, degradation and secretion. Arch Physiol Biochem 113:30–54 [DOI] [PubMed] [Google Scholar]
  62. Kristiansen M, Deriziotis P, Dimcheff DE, Jackson GS, Ovaa H, Naumann H, Clarke AR, van Leeuwen FW, Menéndez-Benito V, Dantuma NP, Portis JL, Collinge J, Tabrizi SJ (2007) Disease-associated prion protein oligomers inhibit the 26S proteasome. Mol Cell 26:175–188 [DOI] [PubMed] [Google Scholar]
  63. Kurschner C, Morgan JI (1995) The cellular prion protein (PrP) selectively binds to Bcl-2 in the yeast two-hybrid system. Brain Res Mol Brain Res 30:165–168 [DOI] [PubMed] [Google Scholar]
  64. Lechauve C, Rezaei H, Celier C, Kiger L, Corral-Debrinski M, Noinville S, Chauvierre C, Hamdane D, Pato C, Marden MC (2009) Neuroglobin and prion cellular localization: investigation of a potential interaction. J Mol Biol 388:968–977 [DOI] [PubMed] [Google Scholar]
  65. Lim J, Hao T, Shaw C, Patel AJ, Szabó G, Rual JF, Fisk CJ, Li N, Smolyar A, Hill DE, Barabási AL, Vidal M, Zoghbi HY (2006) A protein-protein interaction network for human inherited ataxias and disorders of Purkinje cell degeneration. Cell 125:801–814 [DOI] [PubMed] [Google Scholar]
  66. Lopez CD, Yost CS, Prusiner SB, Myers RM, Lingappa VR (1990) Unusual topogenic sequence directs prion protein biogenesis. Science 248:226–229 [DOI] [PubMed] [Google Scholar]
  67. Lysek DA, Wüthrich K (2004) Prion protein interaction with the C-terminal SH3 domain of Grb2 studied using NMR and optical spectroscopy. Biochemistry 43:10393–10399 [DOI] [PubMed] [Google Scholar]
  68. Ma J, Lindquist S (2001) Wild-type PrP and a mutant associated with prion disease are subject to retrograde transport and proteasome degradation. Proc Natl Acad Sci USA 98:14955–14960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Ma J, Wollmann R, Lindquist S (2002) Neurotoxicity and neurodegeneration when PrP accumulates in the cytosol. Science 298:1781–1785 [DOI] [PubMed] [Google Scholar]
  70. Mackenzie A (1983) Immunohistochemical demonstration of glial fibrillary acidic protein in scrapie. J Comp Pathol 93:251–259 [DOI] [PubMed] [Google Scholar]
  71. Mangé A, Crozet C, Lehmann S, Béranger F (2004) Scrapie-like prion protein is translocated to the nuclei of infected cells independently of proteasome inhibition and interacts with chromatin. J Cell Sci 117:2411–2416 [DOI] [PubMed] [Google Scholar]
  72. Martins VR, Graner E, Garcia-Abreu J, de Souza SJ, Mercadante AF, Veiga SS, Zanata SM, Neto VM, Brentani RR (1997) Complementary hydropathy identifies a cellular prion protein receptor. Nat Med 3:1376–1382 [DOI] [PubMed] [Google Scholar]
  73. McKinley MP, Bolton DC, Prusiner SB (1983) A protease-resistant protein is a structural component of the scrapie prion. Cell 35:57–62 [DOI] [PubMed] [Google Scholar]
  74. Meggio F, Negro A, Sarno S, Ruzzene M, Bertoli A, Sorgato MC, Pinna LA (2000) Bovine prion protein as a modulator of protein kinase CK2. Biochem J 352:191–196 [PMC free article] [PubMed] [Google Scholar]
  75. Mei GY, Li Y, Wang GR, Zhang BY, Tian C, Chen C, Zhou RM, Wang X, Li XL, Wang KX, Han J, Dong XP (2009) Molecular interaction between PrP protein and the signal protein 14-3-3 beta. Bing Du Xue Bao 25:208–212 [PubMed] [Google Scholar]
  76. Messing A, Brenner M (2003) GFAP: functional implications gleaned from studies of genetically engineered mice. Glia 43:87–90 [DOI] [PubMed] [Google Scholar]
  77. Miesbauer M, Rambold AS, Winklhofer KF, Tatzelt J (2009) Targeting of the prion protein to the cytosol: mechanisms and consequences. Curr Issues Mol Biol 12:109–118 [PubMed] [Google Scholar]
  78. Mironov A Jr, Latawiec D, Wille H, Bouzamondo-Bernstein E, Legname G, Williamson RA, Burton D, DeArmond SJ, Prusiner SB, Peters PJ (2003) Cytosolic prion protein in neurons. J Neurosci 23:7183–7193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Mishra RS, Basu S, Gu Y, Luo X, Zou WQ, Mishra R, Li R, Chen SG, Gambetti P, Fujioka H, Singh N (2004) Protease-resistant human prion protein and ferritin are cotransported across Caco-2 epithelial cells: implications for species barrier in prion uptake from the intestine. J Neurosci 24:11280–11290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Motohashi H, Yamamoto M (2004) Nrf2-Keap1 defines a physiologically important stress response mechanism. Trends Mol Med 10:549–557 [DOI] [PubMed] [Google Scholar]
  81. Myer DL, Bahassi el M, Stambrook PJ (2005) The Plk3-Cdc25 circuit. Oncogene 24:299–305 [DOI] [PubMed] [Google Scholar]
  82. Nandi PK (1997) Interaction of prion peptide HuPrP106–126 with nucleic acid. Arch Virol 142:2537–2545 [DOI] [PubMed] [Google Scholar]
  83. Naslavsky N, Stein R, Yanai A, Friedlander G, Taraboulos A (1997) Characterization of detergent-insoluble complexes containing the cellular prion protein and its scrapie isoform. J Biol Chem 272:6324–6331 [DOI] [PubMed] [Google Scholar]
  84. Negro A, Meggio F, Bertoli A, Battistutta R, Sorgato MC, Pinna LA (2000) Susceptibility of the prion protein to enzymic phosphorylation. Biochem Biophys Res Commun 271:337–341 [DOI] [PubMed] [Google Scholar]
  85. Nguyen T, Nioi P, Pickett CB (2009) The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem 284:13291–13295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Nieznanski K, Nieznanska H, Skowronek KJ, Osiecka KM, Stepkowski D (2005) Direct interaction between prion protein and tubulin. Biochem Biophys Res Commun 334:403–411 [DOI] [PubMed] [Google Scholar]
  87. Nieznanski K, Podlubnaya ZA, Nieznanska H (2006) Prion protein inhibits microtubule assembly by inducing tubulin oligomerization. Biochem Biophys Res Commun 349:391–399 [DOI] [PubMed] [Google Scholar]
  88. Nogales E (2000) Structural insights into microtubule function. Annu Rev Biochem 69:277–302 [DOI] [PubMed] [Google Scholar]
  89. Oesch B, Teplow DB, Stahl N, Serban D, Hood LE, Prusiner SB (1990) Identification of cellular proteins binding to the scrapie prion protein. Biochemistry 29:5848–5855 [DOI] [PubMed] [Google Scholar]
  90. Osiecka KM, Nieznanska H, Skowronek KJ, Karolczak J, Schneider G, Nieznanski K (2009) Prion protein region 23–32 interacts with tubulin and inhibits microtubule assembly. Proteins 77:279–296 [DOI] [PubMed] [Google Scholar]
  91. Pan T, Wong BS, Liu T, Li R, Petersen RB, Sy MS (2002) Cell-surface prion protein interacts with glycosaminoglycans. Biochem J 368:81–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Pinna LA (2002) Protein kinase CK2: a challenge to canons. J Cell Sci 115:3873–3878 [DOI] [PubMed] [Google Scholar]
  93. Prusiner SB (1998) Prions. Proc Natl Acad Sci USA 95:13363–13383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Prusiner SB, Groth DF, Bolton DC, Kent SB, Hood LE (1984) Purification and structural studies of a major scrapie prion protein. Cell 38:127–134 [DOI] [PubMed] [Google Scholar]
  95. Rambold AS, Miesbauer M, Rapaport D, Bartke T, Baier M, Winklhofer KF, Tatzelt J (2006) Association of Bcl-2 with misfolded prion protein is linked to the toxic potential of cytosolic PrP. Mol Biol Cell 17:3356–3368 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Rane NS, Yonkovich JL, Hegde RS (2004) Protection from cytosolic prion protein toxicity by modulation of protein translocation. EMBO J 23:4550–4559 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Richard M, Biacabe AG, Streichenberger N, Ironside JW, Mohr M, Kopp N, Perret-Liaudet A (2003) Immunohistochemical localization of 14.3.3 zeta protein in amyloid plaques in human spongiform encephalopathies. Acta Neuropathol 105:296–302 [DOI] [PubMed] [Google Scholar]
  98. Riek R, Hornemann S, Wider G, Glockshuber R, Wüthrich K (1997) NMR characterization of the full-length recombinant murine prion protein, mPrP(23–231). FEBS Lett 413:282–288 [DOI] [PubMed] [Google Scholar]
  99. Roucou X, Guo Q, Zhang Y, Goodyer CG, LeBlanc AC (2003) Cytosolic prion protein is not toxic and protects against Bax-mediated cell death in human primary neurons. J Biol Chem 278:40877–40881 [DOI] [PubMed] [Google Scholar]
  100. Salehi AH, Roux PP, Kubu CJ, Zeindler C, Bhakar A, Tannis LL, Verdi JM, Barker PA (2000) NRAGE, a novel MAGE protein, interacts with the p75 neurotrophin receptor and facilitates nerve growth factor-dependent apoptosis. Neuron 27:279–288 [DOI] [PubMed] [Google Scholar]
  101. Salehi AH, Xanthoudakis S, Barker PA (2002) NRAGE, a p75 neurotrophin receptor-interacting protein, induces caspase activation and cell death through a JNK-dependent mitochondrial pathway. J Biol Chem 277:48043–48050 [DOI] [PubMed] [Google Scholar]
  102. Satoh J, Onoue H, Arima K, Yamamura T (2005) The 14-3-3 protein forms a molecular complex with heat shock protein Hsp60 and cellular prion protein. J Neuropathol Exp Neurol 64:858–868 [DOI] [PubMed] [Google Scholar]
  103. Satoh J, Obayashi S, Misawa T, Sumiyoshi K, Oosumi K, Tabunoki H (2009) Protein microarray analysis identifies human cellular prion protein interactors. Neuropathol Appl Neurobiol 35:16–35 [DOI] [PubMed] [Google Scholar]
  104. Schmitt-Ulms G, Hansen K, Liu J, Cowdrey C, Yang J, DeArmond SJ, Cohen FE, Prusiner SB, Baldwin MA (2004) Time-controlled transcardiac perfusion cross-linking for the study of protein interactions in complex tissues. Nat Biotechnol 22:724–731 [DOI] [PubMed] [Google Scholar]
  105. Shpetner HS, Vallee RB (1989) Identification of dynamin, a novel mechanochemical enzyme that mediates interactions between microtubules. Cell 59:421–432 [DOI] [PubMed] [Google Scholar]
  106. Sisó S, Puig B, Varea R, Vidal E, Acín C, Prinz M, Montrasio F, Badiola J, Aguzzi A, Pumarola M, Ferrer I (2002) Abnormal synaptic protein expression and cell death in murine scrapie. Acta Neuropathol 103:615–626 [DOI] [PubMed] [Google Scholar]
  107. Soane L, Fiskum G (2005) Inhibition of mitochondrial neural cell death pathways by protein transduction of Bcl-2 family proteins. J Bioenerg Biomembr 37:179–190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Spielhaupter C, Schätzl HM (2001) PrPC directly interacts with proteins involved in signaling pathways. J Biol Chem 276:44604–44612 [DOI] [PubMed] [Google Scholar]
  109. Stahl N, Borchelt DR, Hsiao K, Prusiner SB (1987) Scrapie prion protein contains a phosphatidylinositol glycolipid. Cell 51:229–240 [DOI] [PubMed] [Google Scholar]
  110. Stahl N, Borchelt DR, Prusiner SB (1990) Differential release of cellular and scrapie prion proteins from cellular membranes by phosphatidylinositol-specific phospholipase C. Biochemistry 29:5405–5412 [DOI] [PubMed] [Google Scholar]
  111. Strom A, Diecke S, Hunsmann G, Stuke AW (2006) Identification of prion protein binding proteins by combined use of far-Western immunoblotting, two dimensional gel electrophoresis and mass spectrometry. Proteomics 6:26–34 [DOI] [PubMed] [Google Scholar]
  112. Sun G, Guo M, Shen A, Mei F, Peng X, Gong R, Guo D, Wu J, Tien P, Xiao G (2005) Bovine PrPC directly interacts with alphaB-crystalline. FEBS Lett 579:5419–5424 [DOI] [PubMed] [Google Scholar]
  113. Szegezdi E, Macdonald DC, Ní Chonghaile T, Gupta S, Samali A (2009) Bcl-2 family on guard at the ER. Am J Physiol Cell Physiol 296:C941–C953 [DOI] [PubMed] [Google Scholar]
  114. Tatzelt J, Maeda N, Pekny M, Yang SL, Betsholtz C, Eliasson C, Cayetano J, Camerino AP, DeArmond SJ, Prusiner SB (1996) Scrapie in mice deficient in apolipoprotein E or glial fibrillary acidic protein. Neurology 47:449–453 [DOI] [PubMed] [Google Scholar]
  115. Thompson RJ, Kynoch PA, Willson VJ (1982) Cellular localization of aldolase C subunits in human brain. Brain Res 232:489–493 [DOI] [PubMed] [Google Scholar]
  116. Turk E, Teplow DB, Hood LE, Prusiner SB (1988) Purification and properties of the cellular and scrapie hamster prion proteins. Eur J Biochem 176:21–30 [DOI] [PubMed] [Google Scholar]
  117. Vana K, Zuber C, Nikles D, Weiss S (2007) Novel aspects of prions, their receptor molecules, and innovative approaches for TSE therapy. Cell Mol Neurobiol 27:107–128 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Wang XF, Dong CF, Zhang J, Wan YZ, Li F, Huang YX, Han L, Shan B, Gao C, Han J, Dong XP (2008) Human tau protein forms complex with PrP and some GSS- and fCJD-related PrP mutants possess stronger binding activities with tau in vitro. Mol Cell Biochem 310:49–55 [DOI] [PubMed] [Google Scholar]
  119. Wang X, Bowers SL, Wang F, Pu XA, Nelson RJ, Ma J (2009) Cytoplasmic prion protein induces forebrain neurotoxicity. Biochim Biophys Acta 1792:555–563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Warner RG, Hundt C, Weiss S, Turnbull JE (2002) Identification of the heparan sulfate binding sites in the cellular prion protein. J Biol Chem 277:18421–18430 [DOI] [PubMed] [Google Scholar]
  121. Weighardt F, Biamonti G, Riva S (1996) The roles of heterogeneous nuclear ribonucleoproteins (hnRNP) in RNA metabolism. Bioessays 18:747–756 [DOI] [PubMed] [Google Scholar]
  122. Weiss S, Proske D, Neumann M, Groschup MH, Kretzschmar HA, Famulok M, Winnacker EL (1997) RNA aptamers specifically interact with the prion protein PrP. J Virol 71:8790–8797 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Wong WW, Puthalakath H (2008) Bcl-2 family proteins: the sentinels of the mitochondrial apoptosis pathway. IUBMB Life 60:390–397 [DOI] [PubMed] [Google Scholar]
  124. Yedidia Y, Horonchik L, Tzaban S, Yanai A, Taraboulos A (2001) Proteasomes and ubiquitin are involved in the turnover of the wild-type prion protein. EMBO J 20:5383–5391 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Yehiely F, Bamborough P, Da Costa M, Perry BJ, Thinakaran G, Cohen FE, Carlson GA, Prusiner SB (1997) Identification of candidate proteins binding to prion protein. Neurobiol Dis 3:339–355 [DOI] [PubMed] [Google Scholar]
  126. Yost CS, Lopez CD, Prusiner SB, Myers RM, Lingappa VR (1990) Non-hydrophobic extracytoplasmic determinant of stop transfer in the prion protein. Nature 343:669–672 [DOI] [PubMed] [Google Scholar]
  127. Zanusso G, Petersen RB, Jin T, Jing Y, Kanoush R, Ferrari S, Gambetti P, Singh N (1999) Proteasomal degradation and N-terminal protease resistance of the codon 145 mutant prion protein. J Biol Chem 274:23396–23404 [DOI] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Neurobiology are provided here courtesy of Springer

RESOURCES