SlideShare a Scribd company logo
The FASEB Journal article fj.11-201772. Published online June 1, 2012. 
The FASEB Journal • Research Communication 
Endogenous prion protein conversion is required for 
prion-induced neuritic alterations and 
neuronal death 
Sabrina Cronier,*,1 Julie Carimalo,‡,§ Brigitte Schaeffer,† Emilie Jaumain,* 
Vincent Béringue,* Marie-Christine Miquel,‡ Hubert Laude,* and Jean-Michel Peyrin*,‡,1 
*UR892, Virologie et Immunologie Moléculaires, and †UR0341, Mathématiques et Informatique 
Appliquées, Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France, 
‡Neurobiologie des Processus Adaptatifs, Unité Mixte de Recherche (UMR) Centre National de la 
Recherche Scientifique (CNRS)-7102, Université Pierre et Marie Curie, Paris, France; and §National 
Reference Centre for Transmissible Spongiform Encephalopathy Surveillance, Department of 
Neurology, Georg August University, Göttingen, Germany 
ABSTRACT Prions cause fatal neurodegenerative 
conditions and result from the conversion of host-encoded 
cellular prion protein (PrPC) into abnormally 
folded scrapie PrP (PrPSc). Prions can propagate both 
in neurons and astrocytes, yet neurotoxicity mecha-nisms 
remain unclear. Recently, PrPC was proposed to 
mediate neurotoxic signaling of -sheet-rich PrP and 
non-PrP conformers independently of conversion. To 
investigate the role of astrocytes and neuronal PrPC in 
prion-induced neurodegeneration, we set up neuron 
and astrocyte primary cocultures derived from PrP 
transgenic mice. In this system, prion-infected astro-cytes 
delivered ovine PrPSc to neurons lacking PrPC 
(prion-resistant), or expressing a PrPC convertible 
(sheep) or not (mouse, human). We show that interac-tion 
between neuronal PrPC and exogenous PrPSc was 
not sufficient to induce neuronal death but that effi-cient 
PrPC conversion was required for prion-associ-ated 
neurotoxicity. Prion-infected astrocytes markedly 
accelerated neurodegeneration in homologous cocul-tures 
compared to infected single neuronal cultures, 
despite no detectable neurotoxin release. Finally, PrPSc 
accumulation in neurons led to neuritic damages and 
cell death, both potentiated by glutamate and reactive 
oxygen species. Thus, conversion of neuronal PrPC 
rather than PrPC-mediated neurotoxic signaling ap-pears 
as the main culprit in prion-induced neurodegen-eration. 
We suggest that active prion replication in 
neurons sensitizes them to environmental stress regu-lated 
by neighboring cells, including astrocytes.— 
Cronier, S., Carimalo, J., Schaeffer, B., Jaumain, E., 
Béringue, V., Miquel, M.-C., Laude, H., Peyrin, J.-M. 
Endogenous prion protein conversion is required for 
prion-induced neuritic alterations and neuronal death. 
FASEB J. 26, 000–000 (2012). www.fasebj.org 
Key Words: neurodegeneration  primary culture  astrocyte  
scrapie 
Transmissible spongiform encephalopathies (TSEs) 
include Creutzfeldt–Jakob disease in humans, bovine 
spongiform encephalopathy, sheep scrapie, and chronic 
wasting disease in cervids. They feature neuronal loss, 
spongiosis, and pronounced astrogliosis. These fatal 
disorders are caused by prions, a class of unconven-tional 
agents that preferentially target the central ner-vous 
system (CNS). Prions are essentially composed of 
aggregated misfolded scrapie prion protein (PrPSc) 
derived from the host-encoded cellular PrP (PrPC). 
Prion propagation within and between species is be-lieved 
to stem from the ability of PrPSc seeds to promote 
the conformational transition from PrPC to PrPSc, 
through a nucleated polymerization process (for re-view, 
see refs. 1, 2). At the molecular level, cell-free 
assays have suggested 2 sequential steps: initial binding 
of PrPC to pathological PrP species and conversion into 
PrPSc (3). 
Although the essential role of PrPC in prion propa-gation 
is beyond any doubt (4, 5), its involvement in 
neurotoxicity mechanisms remains controversial, and 
the nature of PrP toxic species themselves are still 
unknown (6). Exposure of primary neuronal cultures 
expressing or lacking PrPC to purified PrPSc, aggre- 
1 Correspondence: S.C., INRA, UR892, Virologie et Immunolo-gie 
Moléculaires, 78352 Jouy-en-Josas, France. E-mail: sabrina. 
cronier@jouy.inra.fr; J.-M.P., Neurobiologie des Processus Adapta-tifs, 
UMR CNRS-7102, Université Pierre et Marie Curie–Paris 6, 
75005 Paris, France. E-mail: jean-michel.peyrin@snv.jussieu.fr 
doi: 10.1096/fj.11-201772 
Abbreviations: CAS, cerebellar astrocyte; CGN, cerebellar 
granule neuron; DAPI, 4=,6-diamidino-2-phenylindole; DIV, 
day in vitro; DPE, days postexposure; GFAP, glial fibrillary 
acidic protein; GndSCN, guanidine thiocyanate; MAP2, mi-crotubule- 
associated protein 2; NeuN, neuronal nuclei; PK, 
proteinase K; PrP, prion protein; PrPC, cellular prion protein; 
PrPres, proteinase K-resistant prion protein; PrPSc, scrapie 
prion protein; ScCAS, scrapie-exposed cerebellar astrocyte; 
ScCGN, scrapie-exposed CGN; TSE, transmissible spongiform 
encephalopathy 
0892-6638/-1900/0026-0001 © FASEB 1
gated full-length recombinant PrP or PrP-derived syn-thetic 
fragments provided a number of elements 
toward this end (7–8). They were sometimes contradic-tory 
(9–11), possibly because supraphysiological doses 
used in these acute paradigms did not reflect the “slow” 
natural infection process, or because endogenous PrP 
conversion is needed to produce toxic species (as we 
demonstrate here). In vivo, transmission studies in 
transgenic mice have shown that PrP expression at the 
neuronal cell surface was necessary to the development 
of typical TSE neuropathological changes (4, 12–13). 
However, mice expressing PrP specifically in astrocytes 
(14) appeared to propagate prion and develop TSE-specific 
neuronal lesions, suggesting that astrocytic 
prion replication might be sufficient to induce some 
neuronal damage (15). 
Finally, if PrPC plays a critical role in prion-mediated 
neuronal death following interaction with PrPSc, it is 
still unclear whether the conversion step is involved. 
Indeed, PrPC was recently proposed to mediate toxic 
signaling of -sheet-rich conformers, including amyloid 
 (16–17), and following antibody cross-linking (18– 
19), therefore, pointing at a subversion of its putative 
neuroprotective functions (20), although these obser-vations 
were not consistently reproduced (21–23). 
Sheep scrapie prions could efficiently propagate in 
primary cultures of either neurons or astrocytes and 
induce a late apoptotic neuronal death (24). Here, we 
have established a coculture system in which sheep 
scrapie-infected astrocytes are in contact with neurons 
devoid of PrPC or expressing homologous (i.e., convert-ible) 
or heterologous (i.e., nonconvertible) PrPC spe-cies. 
We report that prion neurotoxicity dramatically 
depends on expression and efficient conversion of 
neuronal PrPC into PrPSc. We further show that a 
dysfunction of prion-infected astrocytes is unlikely to be 
a major determinant of neuronal death initiation but 
rather that infected neurons become more susceptible 
to various subtoxic stimuli, such as oxidation and 
glutamate. 
MATERIALS AND METHODS 
Transgenic mouse lines 
Care of mice was performed according to Institut National de 
la Recherche Agronomique and French animal care commit-tee 
guidelines. Primary cultures were derived from the follow-ing 
homozygous transgenic mouse lines: PrP0/0 (PrP-knock-out 
mice; Zurich I; ref. 25), tg338 (ovine PrPVRQ; ref. 26), 
tga20 (mouse Prnp-a allele; ref. 27) and tg650 (human Met129 
PrP; ref. 28). These PrP-expressing lines were all established 
on the same Zurich I mouse PrP0/0 background, thus ensur-ing 
a consistent genetic background in the cultures. 
Primary cell cultures 
Primary cultures of cerebellar granule neurons (CGNs) were 
established as described previously (24, 29). Briefly, CGNs 
extracted from cerebellum of 6-d-old mice by enzymatic and 
mechanical dissociation were plated (400,000 cells/well) in 
24-well plates coated with poly-d-lysine (10 g/ml, Sigma; St. 
Louis, MO, USA). CGN cultures were maintained at 37°C 
with 6% CO2 in DMEM containing glutamax I (Life Technol-ogies, 
Paisley, UK), 10% FCS (BioWhittaker, Walkersville, 
MD, USA), 20 mM KCl, penicillin, streptomycin (Life Tech-nologies) 
and complemented with N-2 and antioxidant-de-pleted 
B27 supplements (Life Technologies, Grand Island, 
NY, USA). Glucose was maintained at 1 mg/ml by weekly 
supplementation, along with antimitotics uridine and fluoro-deoxyuridine 
(10 M; Sigma). 
Pure cerebellar astrocytes from ovine PrPC-expressing 
tg338 mice (CASOv cells) were dissociated similarly to CGNs, 
seeded on poly-d-lysine-coated plates (1 g/ml) and cultured 
in DMEM-glutamax I containing 10% FCS with antibiotics. 
The cell population was fully astrocytic within a week. Astro-cytes 
were grown to 70% confluence before use, and the 
medium was changed weekly. 
Prion infection of cultured cells 
Infectious 10% (w/v) brain homogenates were prepared in 
PBS from terminally ill tg338 mice inoculated with the 127S 
sheep scrapie strain (24, 30). Confluent astrocytes or day in 
vitro 2 (DIV2) CGNs were exposed to a final concentration of 
0.01% (w/v) infected brain homogenate unless specified, or 
not infected for mock infections. Infectivity of astrocyte-conditioned 
medium was tested by replacing half of the 
culture medium by medium conditioned for 7 d on 21-d 
prion-infected astrocytes, supplemented with KCl and N-2. 
Following infection, the medium was either left unchanged 
for CGN cultures during the whole duration of the experi-ment, 
or changed after 7 d and then 1/wk for CAS cultures. 
PrP immunoblot 
Cell lysates or brain homogenates were treated with protei-nase 
K (PK; 7.5 g/mg or 50 g/ml, respectively; Eurome-dex, 
Mundolsheim, France), as described previously (29). 
After methanol precipitation (1 h, 20°C) and centrifugation 
(16,000 g, 10 min), pellets were resuspended in sample buffer 
and boiled, and proteins were subjected to SDS/PAGE and 
electrotransferred onto nitrocellulose membranes. PK-resis-tant 
PrP (PrPres) was detected with anti-PrP monoclonal 
antibodies ICSM18 or biotinylated Sha31. 
Immunofluorescence 
Cells were fixed for 10 min at room temperature in PBS 
containing 4% paraformaldehyde and 4% sucrose, and sub-sequently 
permeabilized for 5 min with PBS containing 0.1% 
Triton X-100. Following treatment for 5 min with 3 M 
guanidine thiocyanate (GdnSCN), PrPSc was immunode-tected 
with ICSM35 or ICSM33 anti-PrP monoclonal antibod-ies 
(31). Neurons were labeled with monoclonal anti-neuro-nal 
nuclei (NeuN; 1:100; Chemicon; Temecula, CA, USA) 
antibody and astrocytes with polyclonal anti-glial fibrillary 
acidic protein (GFAP; 1:400; Dako, Glostrup, Denmark) 
antibody. Neuronal processes were stained with either mono-clonal 
(1:250; Sigma) or polyclonal (1:500; Chemicon) anti-microtubule- 
associated protein 2 (MAP2) antibodies. Cells 
were then incubated with appropriate FITC- or Alexa Fluor-conjugated 
secondary antibodies and nuclear marker 4=,6- 
diamidino-2-phenylindole (DAPI; 2 g/ml; Sigma) and 
mounted in Fluoromount (Sigma). Cells were observed un-der 
an Axiovert 200 M epifluorescence or an AxioObserver 
Z1 microscope (Zeiss, Oberkochen, Germany), images were 
acquired with Metaview (Universal Imaging, Downingtown, 
PA, USA) and AxioVision (Zeiss) software, respectively, and 
2 Vol. 26 September 2012 The FASEB Journal  www.fasebj.org CRONIER ET AL.
analysis was performed with ImageJ (U.S. National Institutes 
of Health; http://rsbweb.nih.gov/ij/). 
Neuron/astrocyte cocultures 
Mock- or 127S-infected CASOv cultures were maintained for 
21 d. Culture medium was then removed, and freshly disso-ciated 
CGNs from tg338, tga20, tg650 or PrP0/0 mice sus-pended 
in complete neuronal medium were seeded at a 
density of 2000 cells/mm2 on top of CAS cultures. After 7 d, 
cells were processed for immunofluorescence. Neuronal sur-vival 
rate was determined by counting NeuN-positive cells 
with nonpyknotic nuclei. For each experiment, PrPSc immu-nostaining 
was performed to confirm that neurons were in 
contact with heavily infected astrocytes. 
Quantification of astrocyte-conditioned medium toxicity 
Fresh culture medium was conditioned for 7 d on mock- or 
scrapie-infected astrocytes at 21 days postexposure (DPE) to 
brain homogenate, collected, and supplemented with KCl 
and N-2 to obtain neuronal medium. At d 7, 25, 50, or 75% of 
the culture volume of tg338 CGN (CGNOv) cultures was 
replaced by astrocyte-conditioned medium. Cells were moni-tored 
daily and processed for immunofluorescence after 4 d. 
Neuronal survival rate was determined by counting NeuN-positive 
cells with nonpyknotic nuclei. 
Glutamate and free-radical treatments 
After 14 d exposure to mock- or 127S-infected tg338 brain 
homogenate, CGNOv cultures were treated with glutamate 
(1–100 M) or hydrogen peroxide (1–10 M) (Sigma) for 48 
h. Cells were then processed for immunofluorescence. Neu-ronal 
survival was estimated by counting NeuN-positive cells 
with nonpyknotic nuclei, and dendritic area was quantified by 
measuring MAP2 labeling. 
Statistical analysis 
Each data set corresponding to glutamate or hydrogen per-oxide 
treatment was analyzed using the MIXED procedure of 
SAS 1999 (SAS Institute, Cary, NC, USA). The ANOVA model 
included a random factor “day” and fixed factors correspond-ing 
to the cell state (mock- or scrapie-infected), the treatment 
dose, and their interaction. To satisfy homoscedasticity as-sumptions, 
logarithm of neuronal survival and square root of 
dendritic area were considered. LS means were computed, 
and all pair-wise differences were evaluated using Tukey- 
Kramer adjustment for P values. For the other sets of exper-iments, 
data were analyzed by Student’s t test (2-tailed distri-bution; 
2-sample equal variance). 
RESULTS 
Neurons cocultured with prion-infected astrocytes 
rapidly degenerate 
First, we investigated the contribution of prion-infected 
astrocytes in neurodegeneration. Exposure of CASOv 
primary cultures to brain homogenate infected with 
127S sheep scrapie strain led to the detection of 
neosynthesized PrPres from 7 DPE onward by immuno-blot 
(Fig. 1A). PrPres amounts then increased 4-fold by 
28 DPE. These data were consistent with those obtained 
using cell culture medium instead of brain homogenate 
as an infectious source (24). At 21 DPE, a large majority 
(80%) of astrocytes exposed to 127S scrapie inocu-lum 
(ScCASOv) showed a GdnSCN-dependent, bright 
punctated PrP immunostaining specific for prion infec-tion 
(Fig. 1B, C), therefore confirming that astrocyte 
cultures were heavily infected and laden with PrPSc. At 
this time point, we seeded freshly dissociated CGNs 
expressing ovine PrPC (CGNOv) on ScCASOv cells or on 
mock-infected CASOv cells in a medium depleted of 
antioxidants. After 7 d of CGNOv/ScCASOv coculture, 
neuronal survival significantly decreased by 40% as 
compared to CGNOv/CASOv cocultures (n4 indepen-dent 
experiments, Fig. 2A). Colocalization of terminal 
deoxynucleotidyltransferase-mediated dUTP nick-end 
labeling (TUNEL)-positive cells and pyknotic nuclei 
indicated that apoptosis was a prominent feature in 
dying neurons (data not shown). Surviving neurons 
showed massive dendritic fragmentation (Fig. 2D, top 
Figure 1. Efficient prion infection of cerebellar astrocyte cultures. Cerebellar astrocyte cultures from tg338 mice (CASOv) were 
exposed to 127S scrapie-infected tg338 brain homogenate. A) Immunoblot of PK-treated lysates probed with anti-PrP mAb 
ICSM18, showing PrPres accumulation between 7 and 28 DPE in prion-infected CASOv cultures (ScCASOv). As a control, 
nonpermissive CAS0/0 cells were exposed in parallel (ScCAS0/0). B–D) CASOv cells were either mock-infected (B) or 
prion-infected (scrapie; C, D), and immunofluorescence was performed 3 wk after infection. PrPSc was detected with mAb 
ICSM33 (green) after permeabilization (B, C) or not (D) and GdnSCN denaturation. Red, GFAP; blue, DAPI. In infected 
cultures, 80% of cells show a punctate fluorescence assumed to reflect PrPSc accumulation. Scale bars  10 m. 
PRION NEURODEGENERATION REQUIRES PRP CONVERSION 3
panels), indicating widespread ongoing neurodegen-eration. 
Conditioned medium of prion-infected astrocytes is 
not neurotoxic and is poorly infectious 
In other proteinopathies, it has been shown that astro-cytes 
could contribute to neurodegeneration by releas-ing 
toxic factors (32–34). Astrocytes provide trophic 
support to neurons and regulate neurotransmitters and 
toxin concentrations in the extracellular environment. 
To examine whether a possible alteration of such 
functions by astrocyte prion replication could contrib-ute 
to the neuronal death observed in our 7-d cocul-ture, 
the culture medium from mock- or prion-infected 
astrocytes was conditioned between 21 and 28 DPE and 
subsequently added to differentiated CGNOv cells. No 
significant difference in neuronal survival was observed 
after up to 4 d of exposure (Fig. 3A). This suggested 
Figure 3. Conditioned medium of prion-infected astrocytes is 
not toxic to neurons and contains little infectivity. A) Differ-entiated 
CGNOv cells were cultured for 4 d with 25% (open 
bars), 50% (shaded bars), or 75% (solid bars) of mock-infected 
(CASOv) or prion-infected (ScCASOv) astrocyte-con-ditioned 
medium. Neuronal survival is expressed as a percent-age 
of total living neurons (NeuN-positive cells) in 
nontreated cultures. Values are means  se (n3). B) PrPres 
accumulation in CGNOv cells exposed to a serial dilution of 
scrapie-infected tg338 brain homogenate or to ScCASOv-conditioned 
medium for 28 d. As a control, CGN0/0 cells were 
exposed to conditioned medium. C) Comparison of PrPres 
amounts between CASOv cells exposed or not to scrapie-infected 
tg338 brain homogenate (0.01% final concentra-tion) 
at 21 DPE, and the inoculum used. For quantification 
purposes, a serial dilution of the inoculum, ranging from 0.5- 
to 2-fold the amount of brain homogenate inoculated per well, 
was loaded along with lysates of whole CASOv culture wells. B, C) 
Immunoblots were probed with biotinylated mAb Sha31. 
Figure 2. Influence of neuronal PrPC expression and primary 
sequence in prion-induced neuronal death. Freshly dissoci-ated 
neurons expressing ovine (CGNOv), murine (CGNMo), 
or human PrPC (CGNHu), or devoid of PrP (CGN0/0), were 
seeded on top of mock- or sheep scrapie-infected astrocytes 
(CASOv or ScCASOv, respectively) and cocultured for a week. 
A, B) Neuronal survival is expressed as a percentage of total 
living neurons (NeuN-positive cells) in mock-infected cocul-tures 
(control). Values are means  se; n  4 (A); n3 (B). 
In infected cocultures, survival significantly decreased for 
CGNOv, but not for CGN0/0, CGNMo, and CGNHu (N.S., not 
significant). **P  0.01; ***P  0.001; Student’s t test. C) 
PrPSc was quantified by immunofluorescence in CGNOv, 
CGN0/0, and CGNMo cells cocultured with ScCASOv cells 
using mAb ICSM33. For each experiment, PrPSc levels in 
CGN0/0/ScCASOv cultures were normalized to 1. Mean  se 
(n4). D) CGNOv or CGN0/0 cells were cocultured with 
CASOv or ScCASOv cells and stained with anti-MAP2 mAb 
(dendrites, green) and DAPI (blue). Note the intense den-dritic 
fragmentation in CGNOv cells cocultured with prion-infected 
astrocytes (arrows). Scale bars  10 m. 
4 Vol. 26 September 2012 The FASEB Journal  www.fasebj.org CRONIER ET AL.
that ScCASOv culture medium does not contain neuro-toxins, 
including possibly neurotoxic PrPSc species. 
Relative efficacy of infection of CGNOv cells with ScCASOv-conditioned 
medium in comparison with serial dilu-tions 
of infectious brain homogenate indicated a low 
infectivity titer equivalent to 0.0001% brain homoge-nate 
or 103 ID50/ml (Fig. 3B). In contrast, after 21 
DPE, astrocyte cell fraction had an infectivity titer 
100-fold higher (data not shown), and a PrPres con-tent 
equivalent to 0.01% brain homogenate (Fig. 3C). 
Immunostaining of nonpermeabilized GndSCN-treated 
ScCASOv cells showed intense PrPSc labeling, presum-ably 
at the cell surface (Fig. 1D). Together, these results 
indicate that prion-infected astrocytes do not exhibit 
major dysfunctions and rather suggest that, given the 
low infectivity level of the conditioned medium, the 
abundant cell-associated PrPSc could be responsible for 
neuronal death through direct astrocyte contact in 
cocultures. 
Expression and efficient conversion of neuronal PrPC 
are required for neurotoxicity mediated by 
prion-infected astrocytes 
To assess whether the rapid death observed in neurons 
cocultured with prion-infected astrocytes was PrP me-diated, 
neurons derived from mice with identical ge-netic 
background but devoid of PrPC (CGN0/0) were 
cocultured for 1 wk with either CASOv or ScCASOv cells 
in the same culture conditions as CGNOv cells. No 
alteration in neuronal survival was observed in prion-infected 
cocultures (Fig. 2B), in striking contrast with 
the CGNOv situation (Fig. 2A). Moreover, CGN0/0 
neurite immunostaining failed to reveal any ongoing 
neurodegeneration (Fig. 2D, bottom panels). 
As neuronal PrPC expression appeared necessary for 
neuronal cell death in our cocultures, we next exam-ined 
whether its efficient conversion into PrPSc was 
equally crucial. Thus, mock- or scrapie-infected CASOv 
cells were cocultured with CGNs expressing either 
mouse (CGNMo) or human (CGNHu) PrPC, i.e., heter-ologous 
PrP species assumed to be inefficiently con-verted 
by ovine PrPSc (35). Indeed, in vivo, 127S prions 
exhibit a substantial transmission barrier on transmis-sion 
to human and mouse PrP transgenic mice (unpub-lished 
results). As a result, neuronal survival slightly 
decreased, but not to statistically significant levels, 
when CGNHu (P0.29) or CGNMo (P0.10) cells were 
cocultured with ScCASOv cells for 7 d (n3 indepen-dent 
experiments; Fig. 2B). Abnormal PrP levels in the 
cocultures did not vary significantly whether ScCASOv 
cells were cocultured with CGN0/0 or CGNMo cells, as 
assessed by immunostaining and immunoblot (Fig. 2C 
and data not shown). Remarkably, and in contrast, 
PrPSc levels were more than doubled on coculture with 
CGNOv cells, suggesting active prion replication in 
these cells (Fig. 2C). 
Altogether, our coculture system shows that neurons 
apposed to PrPSc-producing astrocytes undergo a rapid 
and significant death only when they express PrPC 
molecules that are efficiently converted by nearby PrPSc 
into new PrPSc species. 
Prion infection of neurons increases their sensitivity 
to glutamate and free radical insults 
Finally, we investigated whether neuronal neosynthe-sized 
PrPSc alone was responsible for CGNOv cell death 
observed in cocultures with ScCASOv cells. Infection of 
“pure” CGNOv cells with 127S-infected brain homoge-nate 
led to specific accumulation of PrPres as early as 7 
DPE. Amounts steadily increased 5-fold by 28 DPE 
(Fig. 4A). By 14 DPE, ScCGNOv cells accumulated 
substantial levels of PrPSc (Fig. 4A), distributed in the 
soma and processes (24). However, at this stage, neu-ronal 
survival was unaltered (Fig. 4B, see dose 0), and a 
Figure 4. Prion-infected neurons display an increased sensi-tivity 
to glutamate and H2O2. A) Immunoblot probed with 
biotinylated mAb Sha31 showing PrPres accumulation kinetics 
in CGNOv cells exposed to 127S scrapie-infected tg338 brain 
homogenate along with CGN0/0 (ScCGN0/0). B) CGNOv cells 
were either mock-treated (open bars) or prion-infected (solid 
bars). At 14 DPE, cells were exposed to increasing concentra-tions 
of glutamate or hydrogen peroxide (H2O2) for 48 h. 
Neuronal survival is expressed as a percentage of total living 
neurons in nontreated cultures. Dendritic area represents the 
extent of MAP2 labeling, expressed in pixels. Quantifications 
correspond to means  se of 3 wells in 1 experiment and are 
representative of n  3–4 independent experiments. Statisti-cal 
analysis was performed using MIXED procedure in SAS 
(see Materials and Methods). **P  0.01; ***P  0.001. 
PRION NEURODEGENERATION REQUIRES PRP CONVERSION 5
significant increase in neuronal death (40%) actually 
occurred at 21 DPE (data not shown). 
Thus, the presence of nearby astrocytes seemed to 
accelerate neurodegeneration in CGNOv/ScCASOv co-cultures. 
Because astrocytes are assumed to regulate 
extracellular signals, such as reactive oxygen species 
and glutamate, we next subjected 14-d CGNOv or Sc- 
CGNOv cultures to subtoxic concentrations of gluta-mate 
or hydrogen peroxide for 48 h (Fig. 4B). Prion 
infection significantly decreased both viability and den-dritic 
area when neurons were exposed to 100 M 
glutamate (P0.001). It also significantly decreased 
dendritic area following treatment with 10 M H2O2 
(P0.01), indicating that ScCGNOv neurons underwent 
a degenerative process. Altogether, these data suggest 
that PrPSc accumulation in neurons increases their 
sensitivity to oxidative stress and glutamate. 
DISCUSSION 
Our study aimed at deciphering the relative impor-tance 
of infected astrocytes and neuronal PrPC expres-sion 
and conversion in prion-mediated neurodegenera-tion, 
by exposing primary differentiated neurons 
expressing or lacking PrPC to a continuous and physi-ological 
source of PrPSc delivered by neighboring 
prion-infected astrocytes. As a main result, we found 
that neuronal PrPC must not only be present but also be 
efficiently converted into PrPSc for neurotoxicity to 
occur, therefore further extending at the cell level 
earlier in vivo observations (4, 13) with another prion 
model. We also provide evidence that prion-infected 
astrocytes are not detrimental per se but that they 
accelerate neurodegeneration. 
No significant increase in neuronal cell death was 
observed on coculture of ovine PrPSc-producing astro-cytes 
with neurons genetically devoid of PrPC or ex-pressing 
mouse or human PrPC. In striking contrast, 
when neurons expressed ovine PrPC, their survival was 
decreased by 40% within a week, the surviving cells 
showing widespread ongoing neurodegeneration con-comitantly 
with detectable PrPSc accumulation. As dif-ferences 
in primary sequence appear to have little 
effect on PrPC-PrPSc binding (36, 37), it is likely that 
ovine PrPSc bound similarly to sheep, mouse, and 
human PrPC, thus dismissing a subversion of PrPC 
neuroprotective function by infecting PrPSc as the 
major cause of the prominent neuronal death observed 
here (38). Instead, the delayed or absence of conver-sion 
of mouse and human PrPC by 127S prions in 
transgenic mice and the significant PrPSc increase 
observed only in homologous cocultures strongly sup-port 
the view that the conversion step is critical in the 
initiation of neurodegeneration. This sharply contrasts 
with the emerging notion that PrP could indifferently 
convey toxicity of prions or other abnormally folded 
proteic conformers, such as those involved in Alzhei-mer’s 
disease. Notably, our results are in apparent 
contradiction with the recent findings of Resenberger 
et al. (17), in which short-term (16 h) coculture with 
minimal cell contact between chronically prion-in-fected 
mouse neuroblastoma (ScN2a) cells and immor-talized 
SH-SY5Y cells transiently expressing various het-erologous 
PrP induced an increased apoptosis rate in 
the latter, independent of PrP sequence and with no 
apparent conversion. However, in these conditions, the 
apoptosis increase appeared marginal (5–10%), and 
when they subjected mouse primary cortical neurons to 
a 5-d coculture with ScN2a cells, a 40% viability de-crease 
was observed, similar to our findings, except that 
bona fide prion replication was not assessed, thus recon-ciling 
their data with our observation that PrP conver-sion 
and accumulation are the major contributors to 
neuronal death. 
Neurons devoid of PrPC have been reported to 
exhibit an increased sensitivity to neurotoxins (39). 
The absence of neurodegeneration when they were 
cocultured with heavily infected astrocytes indicates 
that the latter did not misfunction in a way that would 
affect cocultured neurons, nor produce labile non-PrP-related 
neurotoxins that could have been diluted or 
lacking in astrocyte-conditioned medium. Moreover, 
this indicates that extraneuronal, infectious PrPSc par-ticles, 
either released (at low levels) by astrocytes in the 
extracellular medium or membrane bound, were not 
significantly neurotoxic per se. Intriguingly, however, 
apoptosis and neuritic alterations appeared consider-ably 
earlier in ovine PrPC-expressing neurons infected 
on coculture with prion-infected astrocytes than by 
scrapie inoculum [respectively, at 7 d (this study) vs. 28 
d (24) and 21 d when using antioxidant-depleted 
medium (unpublished results)]. As infectivity and 
PrPres levels of both infectious sources seemed compa-rable, 
this indicates that astrocyte-mediated infection 
was able to potentiate neurodegeneration. Neuron/ 
glia close contacts along with nearly maximal PrPSc 
levels—presumably at the astrocyte cell surface—are 
likely to favor ignition of prion infection in neurons 
through efficient cell-to-cell transfer of PrPSc particles, 
as previously shown in immortalized cell models (31). 
This is further supported by findings that prion infec-tion 
of cells occurs very rapidly and that cell mem-branes 
probably constitute the primary site of prion 
conversion (40). Whether PrPSc produced by long-term 
infected astrocytes not only promotes conversion but 
also catalyzes the formation of neurotoxic species from 
neuronal PrPC, as recently suggested in mice (41), will 
be examined in the future. 
Interestingly, we show here that PrPSc-accumulating 
neurons are more sensitive to exogenous stimuli, such 
as glutamate and hydrogen peroxide, suggesting an 
altered response to excitotoxicity and oxidative stress, 
as reported with other misfolded proteins and peptides 
(42–44). Although our results suggest that infected 
astrocytes do not release toxic species, we cannot 
exclude that they might no longer help neurons cope 
with stresses in their environment, including intracel-lular 
protein aggregates. In other neurodegenerative 
diseases, accumulation of misfolded protein aggregates 
6 Vol. 26 September 2012 The FASEB Journal  www.fasebj.org CRONIER ET AL.
and/or changes in lipid raft content or interactions 
have both been shown to be detrimental to neurons, by 
inducing massive dendritic degeneration and axonal 
damage due to microtubule disruption (45–46). 
Whether similar alterations are responsible for the 
toxicity observed in our study remains to be deter-mined. 
Overall the massive neurodegeneration ob-served 
in our cocultures would, therefore, result from 
both a particularly efficient initiation of prion infection 
in neurons by infected astrocytes and a subsequent 
increased neuronal vulnerability. 
In neurons, the cellular form of the prion protein 
PrPC appears to be involved in a number of possibly 
independent neurodegenerative pathways, some being 
acute following exposure to extraneuronal PrPSc or 
other -sheet-rich conformers (17, 47) and some ne-cessitating 
efficient PrPC conversion, acting putatively 
on a downstream cascade or physiological process (48). 
In this regard, our ex vivo model may help to further 
dissect these neurodegenerative mechanisms that 
could be relevant to other brain disorders and devel-opment 
of rational therapies. 
This project was supported by grants from the French 
government (GIS-Infections a` Prion) and from the European 
Union (Neurodegeneration–QLG3CT2001). S.C. was a recip-ient 
of a French Ministry of Research and Education (MRE) 
fellowship. J.C. was funded by an MRE fellowship and the 
French Foundation France Alzheimer. The authors thank S. 
Hawke (Imperial College, London, UK; now at University of 
Sydney, Sydney, NSW, Australia) and G. S. Jackson (Medical 
Research Council Prion Unit, London, UK) for kindly pro-viding 
the antibodies ICSM18 and ICSM35, and ICSM33, 
respectively; J. Grassi and S. Simon (Commissariat a` l’énergie 
Atomique et aux énergies Alternatives, Saclay, France) for 
Sha31 antibody; C. Weissmann (Scripps Research Institute, 
Jupiter, FL, USA) for authorizing the inclusion of tga20 and 
PrP0/0 mice in this study. The authors thank T. Szeto for 
careful reading of the manuscript and R. Young for prepara-tion 
of the figures. 
REFERENCES 
1. Collinge, J. (2001) Prion diseases of humans and animals: their 
causes and molecular basis. Annu. Rev. Neurosci. 24, 519–550 
2. Prusiner, S. B. (1998) Prions. Proc. Natl. Acad. Sci. U. S. A. 95, 
13363–13383 
3. Silveira, J. R., Caughey, B., and Baron, G. S. (2004) Prion 
protein and the molecular features of transmissible spongiform 
encephalopathy agents. Curr. Top. Microbiol. Immunol. 284, 1–50 
4. Brandner, S., Isenmann, S., Raeber, A., Fischer, M., Sailer, A., 
Kobayashi, Y., Marino, S., Weissmann, C., and Aguzzi, A. (1996) 
Normal host prion protein necessary for scrapie-induced neu-rotoxicity. 
Nature 379, 339–343 
5. Bueler, H., Aguzzi, A., Sailer, A., Greiner, R. A., Autenried, P., 
Aguet, M., and Weissmann, C. (1993) Mice devoid of PrP are 
resistant to scrapie. Cell 73, 1339–1347 
6. Caughey, B., and Lansbury, P. T. (2003) Protofibrils, pores, 
fibrils, and neurodegeneration: separating the responsible pro-tein 
aggregates from the innocent bystanders. Annu. Rev. Neu-rosci. 
26, 267–298 
7. Brown, D. R. (2002) Mayhem of the multiple mechanisms: 
modelling neurodegeneration in prion disease. J. Neurochem. 82, 
209–215 
8. Gavin, R., Braun, N., Nicolas, O., Parra, B., Urena, J. M., 
Mingorance, A., Soriano, E., Torres, J. M., Aguzzi, A., and del 
Rio, J. A. (2005) PrP(106-126) activates neuronal intracellular 
kinases and Egr1 synthesis through activation of NADPH-oxi-dase 
independently of PrPc. FEBS Lett. 579, 4099–4106 
9. Brown, D. R., Herms, J., and Kretzschmar, H. A. (1994) Mouse 
cortical cells lacking cellular PrP survive in culture with a 
neurotoxic PrP fragment. Neuroreport 5, 2057–2060 
10. Kunz, B., Sandmeier, E., and Christen, P. (1999) Neurotoxicity 
of prion peptide 106-126 not confirmed. FEBS Lett. 458, 65–68 
11. Simoneau, S., Rezaei, H., Sales, N., Kaiser-Schulz, G., Lefebvre- 
Roque, M., Vidal, C., Fournier, J. G., Comte, J., Wopfner, F., 
Grosclaude, J., Schatzl, H., and Lasmezas, C. I. (2007) In vitro 
and in vivo neurotoxicity of prion protein oligomers. PLoS 
Pathog. 3, e125 
12. Chesebro, B., Trifilo, M., Race, R., Meade-White, K., Teng, C., 
LaCasse, R., Raymond, L., Favara, C., Baron, G., Priola, S., 
Caughey, B., Masliah, E., and Oldstone, M. (2005) Anchorless 
prion protein results in infectious amyloid disease without 
clinical scrapie. Science 308, 1435–1439 
13. Mallucci, G., Dickinson, A., Linehan, J., Klohn, P. C., Brandner, 
S., and Collinge, J. (2003) Depleting neuronal PrP in prion 
infection prevents disease and reverses spongiosis. Science 302, 
871–874 
14. Raeber, A. J., Race, R. E., Brandner, S., Priola, S. A., Sailer, A., 
Bessen, R. A., Mucke, L., Manson, J., Aguzzi, A., Oldstone, M. B., 
Weissmann, C., and Chesebro, B. (1997) Astrocyte-specific 
expression of hamster prion protein (PrP) renders PrP knock-out 
mice susceptible to hamster scrapie. EMBO J. 16, 6057–6065 
15. Jeffrey, M., Goodsir, C. M., Race, R. E., and Chesebro, B. (2004) 
Scrapie-specific neuronal lesions are independent of neuronal 
PrP expression. Ann. Neurol. 55, 781–792 
16. Lauren, J., Gimbel, D. A., Nygaard, H. B., Gilbert, J. W., and 
Strittmatter, S. M. (2009) Cellular prion protein mediates 
impairment of synaptic plasticity by amyloid-beta oligomers. 
Nature 457, 1128–1132 
17. Resenberger, U. K., Harmeier, A., Woerner, A. C., Goodman, 
J. L., Muller, V., Krishnan, R., Vabulas, R. M., Kretzschmar, 
H. A., Lindquist, S., Hartl, F. U., Multhaup, G., Winklhofer, 
K. F., and Tatzelt, J. (2011) The cellular prion protein mediates 
neurotoxic signalling of beta-sheet-rich conformers indepen-dent 
of prion replication. EMBO J. 30, 2057–2070 
18. Lefebvre-Roque, M., Kremmer, E., Gilch, S., Zou, W. Q., Ferau-det, 
C., Gilles, C. M., Sales, N., Grassi, J., Gambetti, P., Baron, T., 
Schatzl, H., and Lasmezas, C. I. (2007) Toxic effects of intrace-rebral 
PrP antibody administration during the course of BSE 
infection in mice. Prion 1, 198–206 
19. Solforosi, L., Criado, J. R., McGavern, D. B., Wirz, S., Sanchez- 
Alavez, M., Sugama, S., DeGiorgio, L. A., Volpe, B. T., Wiseman, 
E., Abalos, G., Masliah, E., Gilden, D., Oldstone, M. B., Conti, B., 
and Williamson, R. A. (2004) Cross-linking cellular prion pro-tein 
triggers neuronal apoptosis in vivo. Science 303, 1514–1516 
20. Schneider, B., Pietri, M., Pradines, E., Loubet, D., Launay, J. M., 
Kellermann, O., and Mouillet-Richard, S. (2011) Understand-ing 
the neurospecificity of Prion protein signaling. Front. Biosci. 
16, 169–186 
21. Balducci, C., Beeg, M., Stravalaci, M., Bastone, A., Sclip, A., 
Biasini, E., Tapella, L., Colombo, L., Manzoni, C., Borsello, T., 
Chiesa, R., Gobbi, M., Salmona, M., and Forloni, G. (2010) 
Synthetic amyloid-beta oligomers impair long-term memory 
independently of cellular prion protein. Proc. Natl. Acad. Sci. 
U. S. A. 107, 2295–2300 
22. Calella, A. M., Farinelli, M., Nuvolone, M., Mirante, O., Moos, 
R., Falsig, J., Mansuy, I. M., and Aguzzi, A. (2010) Prion protein 
and Abeta-related synaptic toxicity impairment. EMBO Mol. Med. 
2, 306–314 
23. Klohn, P. C., Farmer, M., Linehan, J. M., O’Malley, C., Fernan-dez 
de Marco, M., Taylor, W., Farrow, M., Khalili-Shirazi, A., 
Brandner, S., and Collinge, J. (2012) PrP antibodies do not 
trigger mouse hippocampal neuron apoptosis. Science 335, 52 
24. Cronier, S., Laude, H., and Peyrin, J. M. (2004) Prions can infect 
primary cultured neurons and astrocytes and promote neuronal 
cell death. Proc. Natl. Acad. Sci. U. S. A. 101, 12271–12276 
25. Bueler, H., Fischer, M., Lang, Y., Bluethmann, H., Lipp, H. P., 
DeArmond, S. J., Prusiner, S. B., Aguet, M., and Weissmann, C. 
(1992) Normal development and behaviour of mice lacking the 
neuronal cell-surface PrP protein. Nature 356, 577–582 
26. Laude, H., Vilette, D., Le Dur, A., Archer, F., Soulier, S., 
Besnard, N., Essalmani, R., and Vilotte, J. L. (2002) New in vivo 
PRION NEURODEGENERATION REQUIRES PRP CONVERSION 7
and ex vivo models for the experimental study of sheep scrapie: 
development and perspectives. C. R. Biol. 325, 49–57 
27. Fischer, M., Rulicke, T., Raeber, A., Sailer, A., Moser, M., Oesch, 
B., Brandner, S., Aguzzi, A., and Weissmann, C. (1996) Prion 
protein (PrP) with amino-proximal deletions restoring suscep-tibility 
of PrP knockout mice to scrapie. EMBO J. 15, 1255–1264 
28. Beringue, V., Le Dur, A., Tixador, P., Reine, F., Lepourry, L., 
Perret-Liaudet, A., Haik, S., Vilotte, J. L., Fontes, M., and Laude, 
H. (2008) Prominent and persistent extraneural infection in 
human PrP transgenic mice infected with variant CJD. PLoS One 
3, e1419 
29. Cronier, S., Beringue, V., Bellon, A., Peyrin, J. M., and Laude, H. 
(2007) Prion strain- and species-dependent effects of antiprion 
molecules in primary neuronal cultures. J. Virol. 81, 13794– 
13800 
30. Langevin, C., Andreoletti, O., Le Dur, A., Laude, H., and 
Beringue, V. (2011) Marked influence of the route of infection 
on prion strain apparent phenotype in a scrapie transgenic 
mouse model. Neurobiol. Dis. 41, 219–225 
31. Paquet, S., Langevin, C., Chapuis, J., Jackson, G. S., Laude, H., 
and Vilette, D. (2007) Efficient dissemination of prions through 
preferential transmission to nearby cells. J. Gen. Virol. 88, 
706–713 
32. Custer, S. K., Garden, G. A., Gill, N., Rueb, U., Libby, R. T., 
Schultz, C., Guyenet, S. J., Deller, T., Westrum, L. E., Sopher, 
B. L., and La Spada, A. R. (2006) Bergmann glia expression of 
polyglutamine-expanded ataxin-7 produces neurodegeneration 
by impairing glutamate transport. Nat. Neurosci. 9, 1302–1311 
33. Haidet-Phillips, A. M., Hester, M. E., Miranda, C. J., Meyer, K., 
Braun, L., Frakes, A., Song, S., Likhite, S., Murtha, M. J., Foust, 
K. D., Rao, M., Eagle, A., Kammesheidt, A., Christensen, A., 
Mendell, J. R., Burghes, A. H., and Kaspar, B. K. (2011) 
Astrocytes from familial and sporadic ALS patients are toxic to 
motor neurons. Nat. Biotechnol. 29, 824–828 
34. Nagai, M., Re, D. B., Nagata, T., Chalazonitis, A., Jessell, T. M., 
Wichterle, H., and Przedborski, S. (2007) Astrocytes expressing 
ALS-linked mutated SOD1 release factors selectively toxic to 
motor neurons. Nat. Neurosci. 10, 615–622 
35. Raymond, G. J., Hope, J., Kocisko, D. A., Priola, S. A., Raymond, 
L. D., Bossers, A., Ironside, J., Will, R. G., Chen, S. G., Petersen, 
R. B., Gambetti, P., Rubenstein, R., Smits, M. A., Lansbury, P. T., 
Jr., and Caughey, B. (1997) Molecular assessment of the poten-tial 
transmissibilities of BSE and scrapie to humans. Nature 388, 
285–288 
36. Horiuchi, M., Priola, S. A., Chabry, J., and Caughey, B. (2000) 
Interactions between heterologous forms of prion protein: 
binding, inhibition of conversion, and species barriers. Proc. 
Natl. Acad. Sci. U. S. A. 97, 5836–5841 
37. Priola, S. A., Caughey, B., Race, R. E., and Chesebro, B. (1994) 
Heterologous PrP molecules interfere with accumulation of 
protease-resistant PrP in scrapie-infected murine neuroblas-toma 
cells. J. Virol. 68, 4873–4878 
38. Westergard, L., Christensen, H. M., and Harris, D. A. (2007) 
The cellular prion protein (PrP(C)): its physiological function 
and role in disease. Biochim. Biophys. Acta 1772, 629–644 
39. Sakudo, A., and Ikuta, K. (2009) Prion protein functions and 
dysfunction in prion diseases. Curr. Med. Chem. 16, 380–389 
40. Goold, R., Rabbanian, S., Sutton, L., Andre, R., Arora, P., 
Moonga, J., Clarke, A. R., Schiavo, G., Jat, P., Collinge, J., and 
Tabrizi, S. J. (2011) Rapid cell-surface prion protein conversion 
revealed using a novel cell system. Nat. Commun. 2, 281 
41. Sandberg, M. K., Al-Doujaily, H., Sharps, B., Clarke, A. R., and 
Collinge, J. (2011) Prion propagation and toxicity in vivo occur 
in two distinct mechanistic phases. Nature 470, 540–542 
42. Dong, X. X., Wang, Y., and Qin, Z. H. (2009) Molecular 
mechanisms of excitotoxicity and their relevance to pathogen-esis 
of neurodegenerative diseases. Acta Pharmacol. Sin. 30, 
379–387 
43. Haass, C., and Selkoe, D. J. (2007) Soluble protein oligomers in 
neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. 
Nat. Rev. Mol. Cell Biol. 8, 101–112 
44. Milhavet, O., McMahon, H. E., Rachidi, W., Nishida, N., Kat-amine, 
S., Mange, A., Arlotto, M., Casanova, D., Riondel, J., 
Favier, A., and Lehmann, S. (2000) Prion infection impairs the 
cellular response to oxidative stress. Proc. Natl. Acad. Sci. U. S. A. 
97, 13937–13942 
45. Jin, M., Shepardson, N., Yang, T., Chen, G., Walsh, D., and 
Selkoe, D. J. (2011) Soluble amyloid -protein dimers isolated 
from Alzheimer cortex directly induce Tau hyperphosphoryla-tion 
and neuritic degeneration. Proc. Natl. Acad. Sci. U. S. A. 108, 
5819–5824 
46. Schengrund, C. L. (2010) Lipid rafts: keys to neurodegenera-tion. 
Brain Res. Bull. 82, 7–17 
47. Nygaard, H. B., and Strittmatter, S. M. (2009) Cellular prion 
protein mediates the toxicity of beta-amyloid oligomers: impli-cations 
for Alzheimer disease. Arch. Neurol. 66, 1325–1328 
48. Kristiansen, M., Deriziotis, P., Dimcheff, D. E., Jackson, G. S., 
Ovaa, H., Naumann, H., Clarke, A. R., van Leeuwen, F. W., 
Menendez-Benito, V., Dantuma, N. P., Portis, J. L., Collinge, J., 
and Tabrizi, S. J. (2007) Disease-associated prion protein oli-gomers 
inhibit the 26S proteasome. Mol. Cell 26, 175–188 
Received for publication February 28, 2012. 
Accepted for publication May 21, 2012. 
8 Vol. 26 September 2012 The FASEB Journal  www.fasebj.org CRONIER ET AL.

More Related Content

What's hot

Christian Haas Webinar July 2014
Christian Haas Webinar July 2014Christian Haas Webinar July 2014
Christian Haas Webinar July 2014
Alzforum
 
J Neurosci 2013 Kaur S
J Neurosci 2013 Kaur SJ Neurosci 2013 Kaur S
J Neurosci 2013 Kaur SSatvinder Kaur
 
Kneusel - Neuroinflammation
Kneusel - NeuroinflammationKneusel - Neuroinflammation
Kneusel - Neuroinflammation
Alzforum
 
Boronated Cetuximab CCR tumor targeting in BNCT
Boronated Cetuximab CCR tumor targeting in BNCTBoronated Cetuximab CCR tumor targeting in BNCT
Boronated Cetuximab CCR tumor targeting in BNCTkent.riley
 
My research poster
My research posterMy research poster
My research poster
IonaThelwall
 
TDP-43 shapeshifts to encipher FTD severity
TDP-43 shapeshifts to encipher FTD severityTDP-43 shapeshifts to encipher FTD severity
TDP-43 shapeshifts to encipher FTD severity
Motac Neuroscience Ltd.
 
Cholestasis induces reversible accumulation of periplakin in mouse liver
Cholestasis induces reversible accumulation of periplakin in mouse liverCholestasis induces reversible accumulation of periplakin in mouse liver
Cholestasis induces reversible accumulation of periplakin in mouse liver
Enrique Moreno Gonzalez
 
Hsiao-DevNeurobiol2014
Hsiao-DevNeurobiol2014Hsiao-DevNeurobiol2014
Hsiao-DevNeurobiol2014Katie K. Hsiao
 
Graduate Seminar - Halloween 2009
Graduate Seminar - Halloween 2009Graduate Seminar - Halloween 2009
Graduate Seminar - Halloween 2009jreifert
 
Shapero.ResearchProject.FinalDocument
Shapero.ResearchProject.FinalDocumentShapero.ResearchProject.FinalDocument
Shapero.ResearchProject.FinalDocumentHannah Shapero
 
Vandermeeren1993
Vandermeeren1993Vandermeeren1993
Vandermeeren1993
Rùa Con Con Rùa
 
Activity-dependent transcriptional dynamics in mouse primary cortical and hum...
Activity-dependent transcriptional dynamics in mouse primary cortical and hum...Activity-dependent transcriptional dynamics in mouse primary cortical and hum...
Activity-dependent transcriptional dynamics in mouse primary cortical and hum...
Darya Vanichkina
 
Caffeine, Through Adenosine A3 Receptor-Mediated Actions, Suppresses Amyloid-...
Caffeine, Through Adenosine A3 Receptor-Mediated Actions, Suppresses Amyloid-...Caffeine, Through Adenosine A3 Receptor-Mediated Actions, Suppresses Amyloid-...
Caffeine, Through Adenosine A3 Receptor-Mediated Actions, Suppresses Amyloid-...Mahmoud Lotfy Soliman
 
GAPDH, a well-known glycolytic enzyme, mediates
GAPDH, a well-known glycolytic enzyme, mediatesGAPDH, a well-known glycolytic enzyme, mediates
GAPDH, a well-known glycolytic enzyme, mediatesPei-Ju Chin
 
Research Report- p53
Research Report- p53Research Report- p53
Research Report- p53Ankur Gupta
 
In vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic repliconIn vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic replicon
BinodGupta27
 
Scientifi c Journal of Multiple Sclerosis
Scientifi c Journal of Multiple SclerosisScientifi c Journal of Multiple Sclerosis
Scientifi c Journal of Multiple Sclerosis
SciRes Literature LLC. | Open Access Journals
 

What's hot (20)

Christian Haas Webinar July 2014
Christian Haas Webinar July 2014Christian Haas Webinar July 2014
Christian Haas Webinar July 2014
 
J Neurosci 2013 Kaur S
J Neurosci 2013 Kaur SJ Neurosci 2013 Kaur S
J Neurosci 2013 Kaur S
 
Kneusel - Neuroinflammation
Kneusel - NeuroinflammationKneusel - Neuroinflammation
Kneusel - Neuroinflammation
 
HyperoxiaPaper
HyperoxiaPaperHyperoxiaPaper
HyperoxiaPaper
 
Boronated Cetuximab CCR tumor targeting in BNCT
Boronated Cetuximab CCR tumor targeting in BNCTBoronated Cetuximab CCR tumor targeting in BNCT
Boronated Cetuximab CCR tumor targeting in BNCT
 
Ablooglu, AJ (2014) JBC
Ablooglu, AJ (2014) JBCAblooglu, AJ (2014) JBC
Ablooglu, AJ (2014) JBC
 
My research poster
My research posterMy research poster
My research poster
 
TDP-43 shapeshifts to encipher FTD severity
TDP-43 shapeshifts to encipher FTD severityTDP-43 shapeshifts to encipher FTD severity
TDP-43 shapeshifts to encipher FTD severity
 
Cholestasis induces reversible accumulation of periplakin in mouse liver
Cholestasis induces reversible accumulation of periplakin in mouse liverCholestasis induces reversible accumulation of periplakin in mouse liver
Cholestasis induces reversible accumulation of periplakin in mouse liver
 
Hsiao-DevNeurobiol2014
Hsiao-DevNeurobiol2014Hsiao-DevNeurobiol2014
Hsiao-DevNeurobiol2014
 
Graduate Seminar - Halloween 2009
Graduate Seminar - Halloween 2009Graduate Seminar - Halloween 2009
Graduate Seminar - Halloween 2009
 
Shapero.ResearchProject.FinalDocument
Shapero.ResearchProject.FinalDocumentShapero.ResearchProject.FinalDocument
Shapero.ResearchProject.FinalDocument
 
Vandermeeren1993
Vandermeeren1993Vandermeeren1993
Vandermeeren1993
 
Activity-dependent transcriptional dynamics in mouse primary cortical and hum...
Activity-dependent transcriptional dynamics in mouse primary cortical and hum...Activity-dependent transcriptional dynamics in mouse primary cortical and hum...
Activity-dependent transcriptional dynamics in mouse primary cortical and hum...
 
Caffeine, Through Adenosine A3 Receptor-Mediated Actions, Suppresses Amyloid-...
Caffeine, Through Adenosine A3 Receptor-Mediated Actions, Suppresses Amyloid-...Caffeine, Through Adenosine A3 Receptor-Mediated Actions, Suppresses Amyloid-...
Caffeine, Through Adenosine A3 Receptor-Mediated Actions, Suppresses Amyloid-...
 
Faseb poster2007b
Faseb poster2007bFaseb poster2007b
Faseb poster2007b
 
GAPDH, a well-known glycolytic enzyme, mediates
GAPDH, a well-known glycolytic enzyme, mediatesGAPDH, a well-known glycolytic enzyme, mediates
GAPDH, a well-known glycolytic enzyme, mediates
 
Research Report- p53
Research Report- p53Research Report- p53
Research Report- p53
 
In vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic repliconIn vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic replicon
 
Scientifi c Journal of Multiple Sclerosis
Scientifi c Journal of Multiple SclerosisScientifi c Journal of Multiple Sclerosis
Scientifi c Journal of Multiple Sclerosis
 

Similar to cronier 2012

Prnp
PrnpPrnp
Prion proteins
Prion proteinsPrion proteins
Prion proteins
ArchuKakur
 
DTwohig-LiteratureReview-Prions_TheUPR_&MetalIons
DTwohig-LiteratureReview-Prions_TheUPR_&MetalIonsDTwohig-LiteratureReview-Prions_TheUPR_&MetalIons
DTwohig-LiteratureReview-Prions_TheUPR_&MetalIonsDaniel Twohig
 
140514 - HD synaptic characterization
140514 - HD synaptic characterization140514 - HD synaptic characterization
140514 - HD synaptic characterizationAJ Keefe
 
Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...
Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...
Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...
ANALYTICAL AND QUANTITATIVE CYTOPATHOLOGY AND HISTOPATHOLOGY
 
International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)
inventionjournals
 
PrionModelAbstract
PrionModelAbstractPrionModelAbstract
PrionModelAbstractAri Mandler
 
When proteins misbehave (part 3)
When proteins misbehave (part 3)When proteins misbehave (part 3)
When proteins misbehave (part 3)Adonis Sfera, MD
 
Prions , prions diseases & pcr
Prions , prions diseases & pcrPrions , prions diseases & pcr
Prions , prions diseases & pcr
Dr. Waqas Nawaz
 
Poster PlantBio Conference Tromsø 2013
Poster PlantBio Conference Tromsø 2013Poster PlantBio Conference Tromsø 2013
Poster PlantBio Conference Tromsø 2013Marit Larsen
 
Prion diseases
Prion diseasesPrion diseases
Prion diseases
Swarnendu Pal
 
Temporal-Spatial Expressions of Spy1 in Rat Sciatic Nerve After Crush
Temporal-Spatial Expressions of Spy1 in Rat Sciatic Nerve After CrushTemporal-Spatial Expressions of Spy1 in Rat Sciatic Nerve After Crush
Temporal-Spatial Expressions of Spy1 in Rat Sciatic Nerve After CrushJiao Yang
 
Antimicrobial Activity Of Human Prion Protein Is Mediated By Its N Terminal R...
Antimicrobial Activity Of Human Prion Protein Is Mediated By Its N Terminal R...Antimicrobial Activity Of Human Prion Protein Is Mediated By Its N Terminal R...
Antimicrobial Activity Of Human Prion Protein Is Mediated By Its N Terminal R...
Alfonso Enrique Islas Rodríguez
 
Prions
PrionsPrions
Cupid Peptides presentation wjr
Cupid Peptides presentation wjrCupid Peptides presentation wjr
Cupid Peptides presentation wjr
Cupid Peptides
 

Similar to cronier 2012 (20)

cronier 2004
cronier 2004cronier 2004
cronier 2004
 
Prnp
PrnpPrnp
Prnp
 
Prion proteins
Prion proteinsPrion proteins
Prion proteins
 
DTwohig-LiteratureReview-Prions_TheUPR_&MetalIons
DTwohig-LiteratureReview-Prions_TheUPR_&MetalIonsDTwohig-LiteratureReview-Prions_TheUPR_&MetalIons
DTwohig-LiteratureReview-Prions_TheUPR_&MetalIons
 
1506.full
1506.full1506.full
1506.full
 
140514 - HD synaptic characterization
140514 - HD synaptic characterization140514 - HD synaptic characterization
140514 - HD synaptic characterization
 
Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...
Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...
Gasdermin D Open Sepsis-Induced Acute Kidney Injury via Cell Pyroptosis by NL...
 
International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)
 
PrionModelAbstract
PrionModelAbstractPrionModelAbstract
PrionModelAbstract
 
bbrc aghdam 2013
bbrc aghdam 2013bbrc aghdam 2013
bbrc aghdam 2013
 
When proteins misbehave (part 3)
When proteins misbehave (part 3)When proteins misbehave (part 3)
When proteins misbehave (part 3)
 
Microbiology.
Microbiology.Microbiology.
Microbiology.
 
Prions , prions diseases & pcr
Prions , prions diseases & pcrPrions , prions diseases & pcr
Prions , prions diseases & pcr
 
Poster PlantBio Conference Tromsø 2013
Poster PlantBio Conference Tromsø 2013Poster PlantBio Conference Tromsø 2013
Poster PlantBio Conference Tromsø 2013
 
Prion diseases
Prion diseasesPrion diseases
Prion diseases
 
Temporal-Spatial Expressions of Spy1 in Rat Sciatic Nerve After Crush
Temporal-Spatial Expressions of Spy1 in Rat Sciatic Nerve After CrushTemporal-Spatial Expressions of Spy1 in Rat Sciatic Nerve After Crush
Temporal-Spatial Expressions of Spy1 in Rat Sciatic Nerve After Crush
 
mtm201513
mtm201513mtm201513
mtm201513
 
Antimicrobial Activity Of Human Prion Protein Is Mediated By Its N Terminal R...
Antimicrobial Activity Of Human Prion Protein Is Mediated By Its N Terminal R...Antimicrobial Activity Of Human Prion Protein Is Mediated By Its N Terminal R...
Antimicrobial Activity Of Human Prion Protein Is Mediated By Its N Terminal R...
 
Prions
PrionsPrions
Prions
 
Cupid Peptides presentation wjr
Cupid Peptides presentation wjrCupid Peptides presentation wjr
Cupid Peptides presentation wjr
 

cronier 2012

  • 1. The FASEB Journal article fj.11-201772. Published online June 1, 2012. The FASEB Journal • Research Communication Endogenous prion protein conversion is required for prion-induced neuritic alterations and neuronal death Sabrina Cronier,*,1 Julie Carimalo,‡,§ Brigitte Schaeffer,† Emilie Jaumain,* Vincent Béringue,* Marie-Christine Miquel,‡ Hubert Laude,* and Jean-Michel Peyrin*,‡,1 *UR892, Virologie et Immunologie Moléculaires, and †UR0341, Mathématiques et Informatique Appliquées, Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France, ‡Neurobiologie des Processus Adaptatifs, Unité Mixte de Recherche (UMR) Centre National de la Recherche Scientifique (CNRS)-7102, Université Pierre et Marie Curie, Paris, France; and §National Reference Centre for Transmissible Spongiform Encephalopathy Surveillance, Department of Neurology, Georg August University, Göttingen, Germany ABSTRACT Prions cause fatal neurodegenerative conditions and result from the conversion of host-encoded cellular prion protein (PrPC) into abnormally folded scrapie PrP (PrPSc). Prions can propagate both in neurons and astrocytes, yet neurotoxicity mecha-nisms remain unclear. Recently, PrPC was proposed to mediate neurotoxic signaling of -sheet-rich PrP and non-PrP conformers independently of conversion. To investigate the role of astrocytes and neuronal PrPC in prion-induced neurodegeneration, we set up neuron and astrocyte primary cocultures derived from PrP transgenic mice. In this system, prion-infected astro-cytes delivered ovine PrPSc to neurons lacking PrPC (prion-resistant), or expressing a PrPC convertible (sheep) or not (mouse, human). We show that interac-tion between neuronal PrPC and exogenous PrPSc was not sufficient to induce neuronal death but that effi-cient PrPC conversion was required for prion-associ-ated neurotoxicity. Prion-infected astrocytes markedly accelerated neurodegeneration in homologous cocul-tures compared to infected single neuronal cultures, despite no detectable neurotoxin release. Finally, PrPSc accumulation in neurons led to neuritic damages and cell death, both potentiated by glutamate and reactive oxygen species. Thus, conversion of neuronal PrPC rather than PrPC-mediated neurotoxic signaling ap-pears as the main culprit in prion-induced neurodegen-eration. We suggest that active prion replication in neurons sensitizes them to environmental stress regu-lated by neighboring cells, including astrocytes.— Cronier, S., Carimalo, J., Schaeffer, B., Jaumain, E., Béringue, V., Miquel, M.-C., Laude, H., Peyrin, J.-M. Endogenous prion protein conversion is required for prion-induced neuritic alterations and neuronal death. FASEB J. 26, 000–000 (2012). www.fasebj.org Key Words: neurodegeneration primary culture astrocyte scrapie Transmissible spongiform encephalopathies (TSEs) include Creutzfeldt–Jakob disease in humans, bovine spongiform encephalopathy, sheep scrapie, and chronic wasting disease in cervids. They feature neuronal loss, spongiosis, and pronounced astrogliosis. These fatal disorders are caused by prions, a class of unconven-tional agents that preferentially target the central ner-vous system (CNS). Prions are essentially composed of aggregated misfolded scrapie prion protein (PrPSc) derived from the host-encoded cellular PrP (PrPC). Prion propagation within and between species is be-lieved to stem from the ability of PrPSc seeds to promote the conformational transition from PrPC to PrPSc, through a nucleated polymerization process (for re-view, see refs. 1, 2). At the molecular level, cell-free assays have suggested 2 sequential steps: initial binding of PrPC to pathological PrP species and conversion into PrPSc (3). Although the essential role of PrPC in prion propa-gation is beyond any doubt (4, 5), its involvement in neurotoxicity mechanisms remains controversial, and the nature of PrP toxic species themselves are still unknown (6). Exposure of primary neuronal cultures expressing or lacking PrPC to purified PrPSc, aggre- 1 Correspondence: S.C., INRA, UR892, Virologie et Immunolo-gie Moléculaires, 78352 Jouy-en-Josas, France. E-mail: sabrina. cronier@jouy.inra.fr; J.-M.P., Neurobiologie des Processus Adapta-tifs, UMR CNRS-7102, Université Pierre et Marie Curie–Paris 6, 75005 Paris, France. E-mail: jean-michel.peyrin@snv.jussieu.fr doi: 10.1096/fj.11-201772 Abbreviations: CAS, cerebellar astrocyte; CGN, cerebellar granule neuron; DAPI, 4=,6-diamidino-2-phenylindole; DIV, day in vitro; DPE, days postexposure; GFAP, glial fibrillary acidic protein; GndSCN, guanidine thiocyanate; MAP2, mi-crotubule- associated protein 2; NeuN, neuronal nuclei; PK, proteinase K; PrP, prion protein; PrPC, cellular prion protein; PrPres, proteinase K-resistant prion protein; PrPSc, scrapie prion protein; ScCAS, scrapie-exposed cerebellar astrocyte; ScCGN, scrapie-exposed CGN; TSE, transmissible spongiform encephalopathy 0892-6638/-1900/0026-0001 © FASEB 1
  • 2. gated full-length recombinant PrP or PrP-derived syn-thetic fragments provided a number of elements toward this end (7–8). They were sometimes contradic-tory (9–11), possibly because supraphysiological doses used in these acute paradigms did not reflect the “slow” natural infection process, or because endogenous PrP conversion is needed to produce toxic species (as we demonstrate here). In vivo, transmission studies in transgenic mice have shown that PrP expression at the neuronal cell surface was necessary to the development of typical TSE neuropathological changes (4, 12–13). However, mice expressing PrP specifically in astrocytes (14) appeared to propagate prion and develop TSE-specific neuronal lesions, suggesting that astrocytic prion replication might be sufficient to induce some neuronal damage (15). Finally, if PrPC plays a critical role in prion-mediated neuronal death following interaction with PrPSc, it is still unclear whether the conversion step is involved. Indeed, PrPC was recently proposed to mediate toxic signaling of -sheet-rich conformers, including amyloid (16–17), and following antibody cross-linking (18– 19), therefore, pointing at a subversion of its putative neuroprotective functions (20), although these obser-vations were not consistently reproduced (21–23). Sheep scrapie prions could efficiently propagate in primary cultures of either neurons or astrocytes and induce a late apoptotic neuronal death (24). Here, we have established a coculture system in which sheep scrapie-infected astrocytes are in contact with neurons devoid of PrPC or expressing homologous (i.e., convert-ible) or heterologous (i.e., nonconvertible) PrPC spe-cies. We report that prion neurotoxicity dramatically depends on expression and efficient conversion of neuronal PrPC into PrPSc. We further show that a dysfunction of prion-infected astrocytes is unlikely to be a major determinant of neuronal death initiation but rather that infected neurons become more susceptible to various subtoxic stimuli, such as oxidation and glutamate. MATERIALS AND METHODS Transgenic mouse lines Care of mice was performed according to Institut National de la Recherche Agronomique and French animal care commit-tee guidelines. Primary cultures were derived from the follow-ing homozygous transgenic mouse lines: PrP0/0 (PrP-knock-out mice; Zurich I; ref. 25), tg338 (ovine PrPVRQ; ref. 26), tga20 (mouse Prnp-a allele; ref. 27) and tg650 (human Met129 PrP; ref. 28). These PrP-expressing lines were all established on the same Zurich I mouse PrP0/0 background, thus ensur-ing a consistent genetic background in the cultures. Primary cell cultures Primary cultures of cerebellar granule neurons (CGNs) were established as described previously (24, 29). Briefly, CGNs extracted from cerebellum of 6-d-old mice by enzymatic and mechanical dissociation were plated (400,000 cells/well) in 24-well plates coated with poly-d-lysine (10 g/ml, Sigma; St. Louis, MO, USA). CGN cultures were maintained at 37°C with 6% CO2 in DMEM containing glutamax I (Life Technol-ogies, Paisley, UK), 10% FCS (BioWhittaker, Walkersville, MD, USA), 20 mM KCl, penicillin, streptomycin (Life Tech-nologies) and complemented with N-2 and antioxidant-de-pleted B27 supplements (Life Technologies, Grand Island, NY, USA). Glucose was maintained at 1 mg/ml by weekly supplementation, along with antimitotics uridine and fluoro-deoxyuridine (10 M; Sigma). Pure cerebellar astrocytes from ovine PrPC-expressing tg338 mice (CASOv cells) were dissociated similarly to CGNs, seeded on poly-d-lysine-coated plates (1 g/ml) and cultured in DMEM-glutamax I containing 10% FCS with antibiotics. The cell population was fully astrocytic within a week. Astro-cytes were grown to 70% confluence before use, and the medium was changed weekly. Prion infection of cultured cells Infectious 10% (w/v) brain homogenates were prepared in PBS from terminally ill tg338 mice inoculated with the 127S sheep scrapie strain (24, 30). Confluent astrocytes or day in vitro 2 (DIV2) CGNs were exposed to a final concentration of 0.01% (w/v) infected brain homogenate unless specified, or not infected for mock infections. Infectivity of astrocyte-conditioned medium was tested by replacing half of the culture medium by medium conditioned for 7 d on 21-d prion-infected astrocytes, supplemented with KCl and N-2. Following infection, the medium was either left unchanged for CGN cultures during the whole duration of the experi-ment, or changed after 7 d and then 1/wk for CAS cultures. PrP immunoblot Cell lysates or brain homogenates were treated with protei-nase K (PK; 7.5 g/mg or 50 g/ml, respectively; Eurome-dex, Mundolsheim, France), as described previously (29). After methanol precipitation (1 h, 20°C) and centrifugation (16,000 g, 10 min), pellets were resuspended in sample buffer and boiled, and proteins were subjected to SDS/PAGE and electrotransferred onto nitrocellulose membranes. PK-resis-tant PrP (PrPres) was detected with anti-PrP monoclonal antibodies ICSM18 or biotinylated Sha31. Immunofluorescence Cells were fixed for 10 min at room temperature in PBS containing 4% paraformaldehyde and 4% sucrose, and sub-sequently permeabilized for 5 min with PBS containing 0.1% Triton X-100. Following treatment for 5 min with 3 M guanidine thiocyanate (GdnSCN), PrPSc was immunode-tected with ICSM35 or ICSM33 anti-PrP monoclonal antibod-ies (31). Neurons were labeled with monoclonal anti-neuro-nal nuclei (NeuN; 1:100; Chemicon; Temecula, CA, USA) antibody and astrocytes with polyclonal anti-glial fibrillary acidic protein (GFAP; 1:400; Dako, Glostrup, Denmark) antibody. Neuronal processes were stained with either mono-clonal (1:250; Sigma) or polyclonal (1:500; Chemicon) anti-microtubule- associated protein 2 (MAP2) antibodies. Cells were then incubated with appropriate FITC- or Alexa Fluor-conjugated secondary antibodies and nuclear marker 4=,6- diamidino-2-phenylindole (DAPI; 2 g/ml; Sigma) and mounted in Fluoromount (Sigma). Cells were observed un-der an Axiovert 200 M epifluorescence or an AxioObserver Z1 microscope (Zeiss, Oberkochen, Germany), images were acquired with Metaview (Universal Imaging, Downingtown, PA, USA) and AxioVision (Zeiss) software, respectively, and 2 Vol. 26 September 2012 The FASEB Journal www.fasebj.org CRONIER ET AL.
  • 3. analysis was performed with ImageJ (U.S. National Institutes of Health; http://rsbweb.nih.gov/ij/). Neuron/astrocyte cocultures Mock- or 127S-infected CASOv cultures were maintained for 21 d. Culture medium was then removed, and freshly disso-ciated CGNs from tg338, tga20, tg650 or PrP0/0 mice sus-pended in complete neuronal medium were seeded at a density of 2000 cells/mm2 on top of CAS cultures. After 7 d, cells were processed for immunofluorescence. Neuronal sur-vival rate was determined by counting NeuN-positive cells with nonpyknotic nuclei. For each experiment, PrPSc immu-nostaining was performed to confirm that neurons were in contact with heavily infected astrocytes. Quantification of astrocyte-conditioned medium toxicity Fresh culture medium was conditioned for 7 d on mock- or scrapie-infected astrocytes at 21 days postexposure (DPE) to brain homogenate, collected, and supplemented with KCl and N-2 to obtain neuronal medium. At d 7, 25, 50, or 75% of the culture volume of tg338 CGN (CGNOv) cultures was replaced by astrocyte-conditioned medium. Cells were moni-tored daily and processed for immunofluorescence after 4 d. Neuronal survival rate was determined by counting NeuN-positive cells with nonpyknotic nuclei. Glutamate and free-radical treatments After 14 d exposure to mock- or 127S-infected tg338 brain homogenate, CGNOv cultures were treated with glutamate (1–100 M) or hydrogen peroxide (1–10 M) (Sigma) for 48 h. Cells were then processed for immunofluorescence. Neu-ronal survival was estimated by counting NeuN-positive cells with nonpyknotic nuclei, and dendritic area was quantified by measuring MAP2 labeling. Statistical analysis Each data set corresponding to glutamate or hydrogen per-oxide treatment was analyzed using the MIXED procedure of SAS 1999 (SAS Institute, Cary, NC, USA). The ANOVA model included a random factor “day” and fixed factors correspond-ing to the cell state (mock- or scrapie-infected), the treatment dose, and their interaction. To satisfy homoscedasticity as-sumptions, logarithm of neuronal survival and square root of dendritic area were considered. LS means were computed, and all pair-wise differences were evaluated using Tukey- Kramer adjustment for P values. For the other sets of exper-iments, data were analyzed by Student’s t test (2-tailed distri-bution; 2-sample equal variance). RESULTS Neurons cocultured with prion-infected astrocytes rapidly degenerate First, we investigated the contribution of prion-infected astrocytes in neurodegeneration. Exposure of CASOv primary cultures to brain homogenate infected with 127S sheep scrapie strain led to the detection of neosynthesized PrPres from 7 DPE onward by immuno-blot (Fig. 1A). PrPres amounts then increased 4-fold by 28 DPE. These data were consistent with those obtained using cell culture medium instead of brain homogenate as an infectious source (24). At 21 DPE, a large majority (80%) of astrocytes exposed to 127S scrapie inocu-lum (ScCASOv) showed a GdnSCN-dependent, bright punctated PrP immunostaining specific for prion infec-tion (Fig. 1B, C), therefore confirming that astrocyte cultures were heavily infected and laden with PrPSc. At this time point, we seeded freshly dissociated CGNs expressing ovine PrPC (CGNOv) on ScCASOv cells or on mock-infected CASOv cells in a medium depleted of antioxidants. After 7 d of CGNOv/ScCASOv coculture, neuronal survival significantly decreased by 40% as compared to CGNOv/CASOv cocultures (n4 indepen-dent experiments, Fig. 2A). Colocalization of terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling (TUNEL)-positive cells and pyknotic nuclei indicated that apoptosis was a prominent feature in dying neurons (data not shown). Surviving neurons showed massive dendritic fragmentation (Fig. 2D, top Figure 1. Efficient prion infection of cerebellar astrocyte cultures. Cerebellar astrocyte cultures from tg338 mice (CASOv) were exposed to 127S scrapie-infected tg338 brain homogenate. A) Immunoblot of PK-treated lysates probed with anti-PrP mAb ICSM18, showing PrPres accumulation between 7 and 28 DPE in prion-infected CASOv cultures (ScCASOv). As a control, nonpermissive CAS0/0 cells were exposed in parallel (ScCAS0/0). B–D) CASOv cells were either mock-infected (B) or prion-infected (scrapie; C, D), and immunofluorescence was performed 3 wk after infection. PrPSc was detected with mAb ICSM33 (green) after permeabilization (B, C) or not (D) and GdnSCN denaturation. Red, GFAP; blue, DAPI. In infected cultures, 80% of cells show a punctate fluorescence assumed to reflect PrPSc accumulation. Scale bars 10 m. PRION NEURODEGENERATION REQUIRES PRP CONVERSION 3
  • 4. panels), indicating widespread ongoing neurodegen-eration. Conditioned medium of prion-infected astrocytes is not neurotoxic and is poorly infectious In other proteinopathies, it has been shown that astro-cytes could contribute to neurodegeneration by releas-ing toxic factors (32–34). Astrocytes provide trophic support to neurons and regulate neurotransmitters and toxin concentrations in the extracellular environment. To examine whether a possible alteration of such functions by astrocyte prion replication could contrib-ute to the neuronal death observed in our 7-d cocul-ture, the culture medium from mock- or prion-infected astrocytes was conditioned between 21 and 28 DPE and subsequently added to differentiated CGNOv cells. No significant difference in neuronal survival was observed after up to 4 d of exposure (Fig. 3A). This suggested Figure 3. Conditioned medium of prion-infected astrocytes is not toxic to neurons and contains little infectivity. A) Differ-entiated CGNOv cells were cultured for 4 d with 25% (open bars), 50% (shaded bars), or 75% (solid bars) of mock-infected (CASOv) or prion-infected (ScCASOv) astrocyte-con-ditioned medium. Neuronal survival is expressed as a percent-age of total living neurons (NeuN-positive cells) in nontreated cultures. Values are means se (n3). B) PrPres accumulation in CGNOv cells exposed to a serial dilution of scrapie-infected tg338 brain homogenate or to ScCASOv-conditioned medium for 28 d. As a control, CGN0/0 cells were exposed to conditioned medium. C) Comparison of PrPres amounts between CASOv cells exposed or not to scrapie-infected tg338 brain homogenate (0.01% final concentra-tion) at 21 DPE, and the inoculum used. For quantification purposes, a serial dilution of the inoculum, ranging from 0.5- to 2-fold the amount of brain homogenate inoculated per well, was loaded along with lysates of whole CASOv culture wells. B, C) Immunoblots were probed with biotinylated mAb Sha31. Figure 2. Influence of neuronal PrPC expression and primary sequence in prion-induced neuronal death. Freshly dissoci-ated neurons expressing ovine (CGNOv), murine (CGNMo), or human PrPC (CGNHu), or devoid of PrP (CGN0/0), were seeded on top of mock- or sheep scrapie-infected astrocytes (CASOv or ScCASOv, respectively) and cocultured for a week. A, B) Neuronal survival is expressed as a percentage of total living neurons (NeuN-positive cells) in mock-infected cocul-tures (control). Values are means se; n 4 (A); n3 (B). In infected cocultures, survival significantly decreased for CGNOv, but not for CGN0/0, CGNMo, and CGNHu (N.S., not significant). **P 0.01; ***P 0.001; Student’s t test. C) PrPSc was quantified by immunofluorescence in CGNOv, CGN0/0, and CGNMo cells cocultured with ScCASOv cells using mAb ICSM33. For each experiment, PrPSc levels in CGN0/0/ScCASOv cultures were normalized to 1. Mean se (n4). D) CGNOv or CGN0/0 cells were cocultured with CASOv or ScCASOv cells and stained with anti-MAP2 mAb (dendrites, green) and DAPI (blue). Note the intense den-dritic fragmentation in CGNOv cells cocultured with prion-infected astrocytes (arrows). Scale bars 10 m. 4 Vol. 26 September 2012 The FASEB Journal www.fasebj.org CRONIER ET AL.
  • 5. that ScCASOv culture medium does not contain neuro-toxins, including possibly neurotoxic PrPSc species. Relative efficacy of infection of CGNOv cells with ScCASOv-conditioned medium in comparison with serial dilu-tions of infectious brain homogenate indicated a low infectivity titer equivalent to 0.0001% brain homoge-nate or 103 ID50/ml (Fig. 3B). In contrast, after 21 DPE, astrocyte cell fraction had an infectivity titer 100-fold higher (data not shown), and a PrPres con-tent equivalent to 0.01% brain homogenate (Fig. 3C). Immunostaining of nonpermeabilized GndSCN-treated ScCASOv cells showed intense PrPSc labeling, presum-ably at the cell surface (Fig. 1D). Together, these results indicate that prion-infected astrocytes do not exhibit major dysfunctions and rather suggest that, given the low infectivity level of the conditioned medium, the abundant cell-associated PrPSc could be responsible for neuronal death through direct astrocyte contact in cocultures. Expression and efficient conversion of neuronal PrPC are required for neurotoxicity mediated by prion-infected astrocytes To assess whether the rapid death observed in neurons cocultured with prion-infected astrocytes was PrP me-diated, neurons derived from mice with identical ge-netic background but devoid of PrPC (CGN0/0) were cocultured for 1 wk with either CASOv or ScCASOv cells in the same culture conditions as CGNOv cells. No alteration in neuronal survival was observed in prion-infected cocultures (Fig. 2B), in striking contrast with the CGNOv situation (Fig. 2A). Moreover, CGN0/0 neurite immunostaining failed to reveal any ongoing neurodegeneration (Fig. 2D, bottom panels). As neuronal PrPC expression appeared necessary for neuronal cell death in our cocultures, we next exam-ined whether its efficient conversion into PrPSc was equally crucial. Thus, mock- or scrapie-infected CASOv cells were cocultured with CGNs expressing either mouse (CGNMo) or human (CGNHu) PrPC, i.e., heter-ologous PrP species assumed to be inefficiently con-verted by ovine PrPSc (35). Indeed, in vivo, 127S prions exhibit a substantial transmission barrier on transmis-sion to human and mouse PrP transgenic mice (unpub-lished results). As a result, neuronal survival slightly decreased, but not to statistically significant levels, when CGNHu (P0.29) or CGNMo (P0.10) cells were cocultured with ScCASOv cells for 7 d (n3 indepen-dent experiments; Fig. 2B). Abnormal PrP levels in the cocultures did not vary significantly whether ScCASOv cells were cocultured with CGN0/0 or CGNMo cells, as assessed by immunostaining and immunoblot (Fig. 2C and data not shown). Remarkably, and in contrast, PrPSc levels were more than doubled on coculture with CGNOv cells, suggesting active prion replication in these cells (Fig. 2C). Altogether, our coculture system shows that neurons apposed to PrPSc-producing astrocytes undergo a rapid and significant death only when they express PrPC molecules that are efficiently converted by nearby PrPSc into new PrPSc species. Prion infection of neurons increases their sensitivity to glutamate and free radical insults Finally, we investigated whether neuronal neosynthe-sized PrPSc alone was responsible for CGNOv cell death observed in cocultures with ScCASOv cells. Infection of “pure” CGNOv cells with 127S-infected brain homoge-nate led to specific accumulation of PrPres as early as 7 DPE. Amounts steadily increased 5-fold by 28 DPE (Fig. 4A). By 14 DPE, ScCGNOv cells accumulated substantial levels of PrPSc (Fig. 4A), distributed in the soma and processes (24). However, at this stage, neu-ronal survival was unaltered (Fig. 4B, see dose 0), and a Figure 4. Prion-infected neurons display an increased sensi-tivity to glutamate and H2O2. A) Immunoblot probed with biotinylated mAb Sha31 showing PrPres accumulation kinetics in CGNOv cells exposed to 127S scrapie-infected tg338 brain homogenate along with CGN0/0 (ScCGN0/0). B) CGNOv cells were either mock-treated (open bars) or prion-infected (solid bars). At 14 DPE, cells were exposed to increasing concentra-tions of glutamate or hydrogen peroxide (H2O2) for 48 h. Neuronal survival is expressed as a percentage of total living neurons in nontreated cultures. Dendritic area represents the extent of MAP2 labeling, expressed in pixels. Quantifications correspond to means se of 3 wells in 1 experiment and are representative of n 3–4 independent experiments. Statisti-cal analysis was performed using MIXED procedure in SAS (see Materials and Methods). **P 0.01; ***P 0.001. PRION NEURODEGENERATION REQUIRES PRP CONVERSION 5
  • 6. significant increase in neuronal death (40%) actually occurred at 21 DPE (data not shown). Thus, the presence of nearby astrocytes seemed to accelerate neurodegeneration in CGNOv/ScCASOv co-cultures. Because astrocytes are assumed to regulate extracellular signals, such as reactive oxygen species and glutamate, we next subjected 14-d CGNOv or Sc- CGNOv cultures to subtoxic concentrations of gluta-mate or hydrogen peroxide for 48 h (Fig. 4B). Prion infection significantly decreased both viability and den-dritic area when neurons were exposed to 100 M glutamate (P0.001). It also significantly decreased dendritic area following treatment with 10 M H2O2 (P0.01), indicating that ScCGNOv neurons underwent a degenerative process. Altogether, these data suggest that PrPSc accumulation in neurons increases their sensitivity to oxidative stress and glutamate. DISCUSSION Our study aimed at deciphering the relative impor-tance of infected astrocytes and neuronal PrPC expres-sion and conversion in prion-mediated neurodegenera-tion, by exposing primary differentiated neurons expressing or lacking PrPC to a continuous and physi-ological source of PrPSc delivered by neighboring prion-infected astrocytes. As a main result, we found that neuronal PrPC must not only be present but also be efficiently converted into PrPSc for neurotoxicity to occur, therefore further extending at the cell level earlier in vivo observations (4, 13) with another prion model. We also provide evidence that prion-infected astrocytes are not detrimental per se but that they accelerate neurodegeneration. No significant increase in neuronal cell death was observed on coculture of ovine PrPSc-producing astro-cytes with neurons genetically devoid of PrPC or ex-pressing mouse or human PrPC. In striking contrast, when neurons expressed ovine PrPC, their survival was decreased by 40% within a week, the surviving cells showing widespread ongoing neurodegeneration con-comitantly with detectable PrPSc accumulation. As dif-ferences in primary sequence appear to have little effect on PrPC-PrPSc binding (36, 37), it is likely that ovine PrPSc bound similarly to sheep, mouse, and human PrPC, thus dismissing a subversion of PrPC neuroprotective function by infecting PrPSc as the major cause of the prominent neuronal death observed here (38). Instead, the delayed or absence of conver-sion of mouse and human PrPC by 127S prions in transgenic mice and the significant PrPSc increase observed only in homologous cocultures strongly sup-port the view that the conversion step is critical in the initiation of neurodegeneration. This sharply contrasts with the emerging notion that PrP could indifferently convey toxicity of prions or other abnormally folded proteic conformers, such as those involved in Alzhei-mer’s disease. Notably, our results are in apparent contradiction with the recent findings of Resenberger et al. (17), in which short-term (16 h) coculture with minimal cell contact between chronically prion-in-fected mouse neuroblastoma (ScN2a) cells and immor-talized SH-SY5Y cells transiently expressing various het-erologous PrP induced an increased apoptosis rate in the latter, independent of PrP sequence and with no apparent conversion. However, in these conditions, the apoptosis increase appeared marginal (5–10%), and when they subjected mouse primary cortical neurons to a 5-d coculture with ScN2a cells, a 40% viability de-crease was observed, similar to our findings, except that bona fide prion replication was not assessed, thus recon-ciling their data with our observation that PrP conver-sion and accumulation are the major contributors to neuronal death. Neurons devoid of PrPC have been reported to exhibit an increased sensitivity to neurotoxins (39). The absence of neurodegeneration when they were cocultured with heavily infected astrocytes indicates that the latter did not misfunction in a way that would affect cocultured neurons, nor produce labile non-PrP-related neurotoxins that could have been diluted or lacking in astrocyte-conditioned medium. Moreover, this indicates that extraneuronal, infectious PrPSc par-ticles, either released (at low levels) by astrocytes in the extracellular medium or membrane bound, were not significantly neurotoxic per se. Intriguingly, however, apoptosis and neuritic alterations appeared consider-ably earlier in ovine PrPC-expressing neurons infected on coculture with prion-infected astrocytes than by scrapie inoculum [respectively, at 7 d (this study) vs. 28 d (24) and 21 d when using antioxidant-depleted medium (unpublished results)]. As infectivity and PrPres levels of both infectious sources seemed compa-rable, this indicates that astrocyte-mediated infection was able to potentiate neurodegeneration. Neuron/ glia close contacts along with nearly maximal PrPSc levels—presumably at the astrocyte cell surface—are likely to favor ignition of prion infection in neurons through efficient cell-to-cell transfer of PrPSc particles, as previously shown in immortalized cell models (31). This is further supported by findings that prion infec-tion of cells occurs very rapidly and that cell mem-branes probably constitute the primary site of prion conversion (40). Whether PrPSc produced by long-term infected astrocytes not only promotes conversion but also catalyzes the formation of neurotoxic species from neuronal PrPC, as recently suggested in mice (41), will be examined in the future. Interestingly, we show here that PrPSc-accumulating neurons are more sensitive to exogenous stimuli, such as glutamate and hydrogen peroxide, suggesting an altered response to excitotoxicity and oxidative stress, as reported with other misfolded proteins and peptides (42–44). Although our results suggest that infected astrocytes do not release toxic species, we cannot exclude that they might no longer help neurons cope with stresses in their environment, including intracel-lular protein aggregates. In other neurodegenerative diseases, accumulation of misfolded protein aggregates 6 Vol. 26 September 2012 The FASEB Journal www.fasebj.org CRONIER ET AL.
  • 7. and/or changes in lipid raft content or interactions have both been shown to be detrimental to neurons, by inducing massive dendritic degeneration and axonal damage due to microtubule disruption (45–46). Whether similar alterations are responsible for the toxicity observed in our study remains to be deter-mined. Overall the massive neurodegeneration ob-served in our cocultures would, therefore, result from both a particularly efficient initiation of prion infection in neurons by infected astrocytes and a subsequent increased neuronal vulnerability. In neurons, the cellular form of the prion protein PrPC appears to be involved in a number of possibly independent neurodegenerative pathways, some being acute following exposure to extraneuronal PrPSc or other -sheet-rich conformers (17, 47) and some ne-cessitating efficient PrPC conversion, acting putatively on a downstream cascade or physiological process (48). In this regard, our ex vivo model may help to further dissect these neurodegenerative mechanisms that could be relevant to other brain disorders and devel-opment of rational therapies. This project was supported by grants from the French government (GIS-Infections a` Prion) and from the European Union (Neurodegeneration–QLG3CT2001). S.C. was a recip-ient of a French Ministry of Research and Education (MRE) fellowship. J.C. was funded by an MRE fellowship and the French Foundation France Alzheimer. The authors thank S. Hawke (Imperial College, London, UK; now at University of Sydney, Sydney, NSW, Australia) and G. S. Jackson (Medical Research Council Prion Unit, London, UK) for kindly pro-viding the antibodies ICSM18 and ICSM35, and ICSM33, respectively; J. Grassi and S. Simon (Commissariat a` l’énergie Atomique et aux énergies Alternatives, Saclay, France) for Sha31 antibody; C. Weissmann (Scripps Research Institute, Jupiter, FL, USA) for authorizing the inclusion of tga20 and PrP0/0 mice in this study. The authors thank T. Szeto for careful reading of the manuscript and R. Young for prepara-tion of the figures. REFERENCES 1. Collinge, J. (2001) Prion diseases of humans and animals: their causes and molecular basis. Annu. Rev. Neurosci. 24, 519–550 2. Prusiner, S. B. (1998) Prions. Proc. Natl. Acad. Sci. U. S. A. 95, 13363–13383 3. Silveira, J. R., Caughey, B., and Baron, G. S. (2004) Prion protein and the molecular features of transmissible spongiform encephalopathy agents. Curr. Top. Microbiol. Immunol. 284, 1–50 4. Brandner, S., Isenmann, S., Raeber, A., Fischer, M., Sailer, A., Kobayashi, Y., Marino, S., Weissmann, C., and Aguzzi, A. (1996) Normal host prion protein necessary for scrapie-induced neu-rotoxicity. Nature 379, 339–343 5. Bueler, H., Aguzzi, A., Sailer, A., Greiner, R. A., Autenried, P., Aguet, M., and Weissmann, C. (1993) Mice devoid of PrP are resistant to scrapie. Cell 73, 1339–1347 6. Caughey, B., and Lansbury, P. T. (2003) Protofibrils, pores, fibrils, and neurodegeneration: separating the responsible pro-tein aggregates from the innocent bystanders. Annu. Rev. Neu-rosci. 26, 267–298 7. Brown, D. R. (2002) Mayhem of the multiple mechanisms: modelling neurodegeneration in prion disease. J. Neurochem. 82, 209–215 8. Gavin, R., Braun, N., Nicolas, O., Parra, B., Urena, J. M., Mingorance, A., Soriano, E., Torres, J. M., Aguzzi, A., and del Rio, J. A. (2005) PrP(106-126) activates neuronal intracellular kinases and Egr1 synthesis through activation of NADPH-oxi-dase independently of PrPc. FEBS Lett. 579, 4099–4106 9. Brown, D. R., Herms, J., and Kretzschmar, H. A. (1994) Mouse cortical cells lacking cellular PrP survive in culture with a neurotoxic PrP fragment. Neuroreport 5, 2057–2060 10. Kunz, B., Sandmeier, E., and Christen, P. (1999) Neurotoxicity of prion peptide 106-126 not confirmed. FEBS Lett. 458, 65–68 11. Simoneau, S., Rezaei, H., Sales, N., Kaiser-Schulz, G., Lefebvre- Roque, M., Vidal, C., Fournier, J. G., Comte, J., Wopfner, F., Grosclaude, J., Schatzl, H., and Lasmezas, C. I. (2007) In vitro and in vivo neurotoxicity of prion protein oligomers. PLoS Pathog. 3, e125 12. Chesebro, B., Trifilo, M., Race, R., Meade-White, K., Teng, C., LaCasse, R., Raymond, L., Favara, C., Baron, G., Priola, S., Caughey, B., Masliah, E., and Oldstone, M. (2005) Anchorless prion protein results in infectious amyloid disease without clinical scrapie. Science 308, 1435–1439 13. Mallucci, G., Dickinson, A., Linehan, J., Klohn, P. C., Brandner, S., and Collinge, J. (2003) Depleting neuronal PrP in prion infection prevents disease and reverses spongiosis. Science 302, 871–874 14. Raeber, A. J., Race, R. E., Brandner, S., Priola, S. A., Sailer, A., Bessen, R. A., Mucke, L., Manson, J., Aguzzi, A., Oldstone, M. B., Weissmann, C., and Chesebro, B. (1997) Astrocyte-specific expression of hamster prion protein (PrP) renders PrP knock-out mice susceptible to hamster scrapie. EMBO J. 16, 6057–6065 15. Jeffrey, M., Goodsir, C. M., Race, R. E., and Chesebro, B. (2004) Scrapie-specific neuronal lesions are independent of neuronal PrP expression. Ann. Neurol. 55, 781–792 16. Lauren, J., Gimbel, D. A., Nygaard, H. B., Gilbert, J. W., and Strittmatter, S. M. (2009) Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature 457, 1128–1132 17. Resenberger, U. K., Harmeier, A., Woerner, A. C., Goodman, J. L., Muller, V., Krishnan, R., Vabulas, R. M., Kretzschmar, H. A., Lindquist, S., Hartl, F. U., Multhaup, G., Winklhofer, K. F., and Tatzelt, J. (2011) The cellular prion protein mediates neurotoxic signalling of beta-sheet-rich conformers indepen-dent of prion replication. EMBO J. 30, 2057–2070 18. Lefebvre-Roque, M., Kremmer, E., Gilch, S., Zou, W. Q., Ferau-det, C., Gilles, C. M., Sales, N., Grassi, J., Gambetti, P., Baron, T., Schatzl, H., and Lasmezas, C. I. (2007) Toxic effects of intrace-rebral PrP antibody administration during the course of BSE infection in mice. Prion 1, 198–206 19. Solforosi, L., Criado, J. R., McGavern, D. B., Wirz, S., Sanchez- Alavez, M., Sugama, S., DeGiorgio, L. A., Volpe, B. T., Wiseman, E., Abalos, G., Masliah, E., Gilden, D., Oldstone, M. B., Conti, B., and Williamson, R. A. (2004) Cross-linking cellular prion pro-tein triggers neuronal apoptosis in vivo. Science 303, 1514–1516 20. Schneider, B., Pietri, M., Pradines, E., Loubet, D., Launay, J. M., Kellermann, O., and Mouillet-Richard, S. (2011) Understand-ing the neurospecificity of Prion protein signaling. Front. Biosci. 16, 169–186 21. Balducci, C., Beeg, M., Stravalaci, M., Bastone, A., Sclip, A., Biasini, E., Tapella, L., Colombo, L., Manzoni, C., Borsello, T., Chiesa, R., Gobbi, M., Salmona, M., and Forloni, G. (2010) Synthetic amyloid-beta oligomers impair long-term memory independently of cellular prion protein. Proc. Natl. Acad. Sci. U. S. A. 107, 2295–2300 22. Calella, A. M., Farinelli, M., Nuvolone, M., Mirante, O., Moos, R., Falsig, J., Mansuy, I. M., and Aguzzi, A. (2010) Prion protein and Abeta-related synaptic toxicity impairment. EMBO Mol. Med. 2, 306–314 23. Klohn, P. C., Farmer, M., Linehan, J. M., O’Malley, C., Fernan-dez de Marco, M., Taylor, W., Farrow, M., Khalili-Shirazi, A., Brandner, S., and Collinge, J. (2012) PrP antibodies do not trigger mouse hippocampal neuron apoptosis. Science 335, 52 24. Cronier, S., Laude, H., and Peyrin, J. M. (2004) Prions can infect primary cultured neurons and astrocytes and promote neuronal cell death. Proc. Natl. Acad. Sci. U. S. A. 101, 12271–12276 25. Bueler, H., Fischer, M., Lang, Y., Bluethmann, H., Lipp, H. P., DeArmond, S. J., Prusiner, S. B., Aguet, M., and Weissmann, C. (1992) Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature 356, 577–582 26. Laude, H., Vilette, D., Le Dur, A., Archer, F., Soulier, S., Besnard, N., Essalmani, R., and Vilotte, J. L. (2002) New in vivo PRION NEURODEGENERATION REQUIRES PRP CONVERSION 7
  • 8. and ex vivo models for the experimental study of sheep scrapie: development and perspectives. C. R. Biol. 325, 49–57 27. Fischer, M., Rulicke, T., Raeber, A., Sailer, A., Moser, M., Oesch, B., Brandner, S., Aguzzi, A., and Weissmann, C. (1996) Prion protein (PrP) with amino-proximal deletions restoring suscep-tibility of PrP knockout mice to scrapie. EMBO J. 15, 1255–1264 28. Beringue, V., Le Dur, A., Tixador, P., Reine, F., Lepourry, L., Perret-Liaudet, A., Haik, S., Vilotte, J. L., Fontes, M., and Laude, H. (2008) Prominent and persistent extraneural infection in human PrP transgenic mice infected with variant CJD. PLoS One 3, e1419 29. Cronier, S., Beringue, V., Bellon, A., Peyrin, J. M., and Laude, H. (2007) Prion strain- and species-dependent effects of antiprion molecules in primary neuronal cultures. J. Virol. 81, 13794– 13800 30. Langevin, C., Andreoletti, O., Le Dur, A., Laude, H., and Beringue, V. (2011) Marked influence of the route of infection on prion strain apparent phenotype in a scrapie transgenic mouse model. Neurobiol. Dis. 41, 219–225 31. Paquet, S., Langevin, C., Chapuis, J., Jackson, G. S., Laude, H., and Vilette, D. (2007) Efficient dissemination of prions through preferential transmission to nearby cells. J. Gen. Virol. 88, 706–713 32. Custer, S. K., Garden, G. A., Gill, N., Rueb, U., Libby, R. T., Schultz, C., Guyenet, S. J., Deller, T., Westrum, L. E., Sopher, B. L., and La Spada, A. R. (2006) Bergmann glia expression of polyglutamine-expanded ataxin-7 produces neurodegeneration by impairing glutamate transport. Nat. Neurosci. 9, 1302–1311 33. Haidet-Phillips, A. M., Hester, M. E., Miranda, C. J., Meyer, K., Braun, L., Frakes, A., Song, S., Likhite, S., Murtha, M. J., Foust, K. D., Rao, M., Eagle, A., Kammesheidt, A., Christensen, A., Mendell, J. R., Burghes, A. H., and Kaspar, B. K. (2011) Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nat. Biotechnol. 29, 824–828 34. Nagai, M., Re, D. B., Nagata, T., Chalazonitis, A., Jessell, T. M., Wichterle, H., and Przedborski, S. (2007) Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat. Neurosci. 10, 615–622 35. Raymond, G. J., Hope, J., Kocisko, D. A., Priola, S. A., Raymond, L. D., Bossers, A., Ironside, J., Will, R. G., Chen, S. G., Petersen, R. B., Gambetti, P., Rubenstein, R., Smits, M. A., Lansbury, P. T., Jr., and Caughey, B. (1997) Molecular assessment of the poten-tial transmissibilities of BSE and scrapie to humans. Nature 388, 285–288 36. Horiuchi, M., Priola, S. A., Chabry, J., and Caughey, B. (2000) Interactions between heterologous forms of prion protein: binding, inhibition of conversion, and species barriers. Proc. Natl. Acad. Sci. U. S. A. 97, 5836–5841 37. Priola, S. A., Caughey, B., Race, R. E., and Chesebro, B. (1994) Heterologous PrP molecules interfere with accumulation of protease-resistant PrP in scrapie-infected murine neuroblas-toma cells. J. Virol. 68, 4873–4878 38. Westergard, L., Christensen, H. M., and Harris, D. A. (2007) The cellular prion protein (PrP(C)): its physiological function and role in disease. Biochim. Biophys. Acta 1772, 629–644 39. Sakudo, A., and Ikuta, K. (2009) Prion protein functions and dysfunction in prion diseases. Curr. Med. Chem. 16, 380–389 40. Goold, R., Rabbanian, S., Sutton, L., Andre, R., Arora, P., Moonga, J., Clarke, A. R., Schiavo, G., Jat, P., Collinge, J., and Tabrizi, S. J. (2011) Rapid cell-surface prion protein conversion revealed using a novel cell system. Nat. Commun. 2, 281 41. Sandberg, M. K., Al-Doujaily, H., Sharps, B., Clarke, A. R., and Collinge, J. (2011) Prion propagation and toxicity in vivo occur in two distinct mechanistic phases. Nature 470, 540–542 42. Dong, X. X., Wang, Y., and Qin, Z. H. (2009) Molecular mechanisms of excitotoxicity and their relevance to pathogen-esis of neurodegenerative diseases. Acta Pharmacol. Sin. 30, 379–387 43. Haass, C., and Selkoe, D. J. (2007) Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat. Rev. Mol. Cell Biol. 8, 101–112 44. Milhavet, O., McMahon, H. E., Rachidi, W., Nishida, N., Kat-amine, S., Mange, A., Arlotto, M., Casanova, D., Riondel, J., Favier, A., and Lehmann, S. (2000) Prion infection impairs the cellular response to oxidative stress. Proc. Natl. Acad. Sci. U. S. A. 97, 13937–13942 45. Jin, M., Shepardson, N., Yang, T., Chen, G., Walsh, D., and Selkoe, D. J. (2011) Soluble amyloid -protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphoryla-tion and neuritic degeneration. Proc. Natl. Acad. Sci. U. S. A. 108, 5819–5824 46. Schengrund, C. L. (2010) Lipid rafts: keys to neurodegenera-tion. Brain Res. Bull. 82, 7–17 47. Nygaard, H. B., and Strittmatter, S. M. (2009) Cellular prion protein mediates the toxicity of beta-amyloid oligomers: impli-cations for Alzheimer disease. Arch. Neurol. 66, 1325–1328 48. Kristiansen, M., Deriziotis, P., Dimcheff, D. E., Jackson, G. S., Ovaa, H., Naumann, H., Clarke, A. R., van Leeuwen, F. W., Menendez-Benito, V., Dantuma, N. P., Portis, J. L., Collinge, J., and Tabrizi, S. J. (2007) Disease-associated prion protein oli-gomers inhibit the 26S proteasome. Mol. Cell 26, 175–188 Received for publication February 28, 2012. Accepted for publication May 21, 2012. 8 Vol. 26 September 2012 The FASEB Journal www.fasebj.org CRONIER ET AL.