You are on page 1of 11

Neuron

Article

Targeting Cellular Prion Protein Reverses Early


Cognitive Deficits and Neurophysiological
Dysfunction in Prion-Infected Mice
Giovanna R. Mallucci,1,* Melanie D. White,1 Michael Farmer,1 Andrew Dickinson,1 Husna Khatun,2
Andrew D. Powell,2 Sebastian Brandner,1 John G.R. Jefferys,2 and John Collinge1
1
MRC Prion Unit and Department of Neurodegenerative Disease, Institute of Neurology, Queen Square, London WC1N 3BG,
United Kingdom
2
Department of Neurophysiology, Division of Neuroscience, University of Birmingham, Birmingham, B15 2TT, United Kingdom
*Correspondence: g.mallucci@prion.ucl.ac.uk
DOI 10.1016/j.neuron.2007.01.005

SUMMARY advanced disease and correspond with irreversible neuro-


nal loss. However, there are earlier pathological and phe-
Currently, no treatment can prevent the cogni- notypic changes. Spongiform change and synapse loss
tive and motor decline associated with wide- precede neuronal loss (Jeffrey et al., 2000), and changes
spread neurodegeneration in prion disease. in species-typical behaviors also occur long before typical
However, we previously showed that targeting motor symptoms (Betmouni et al., 1999; Cunningham
endogenous neuronal prion protein (PrPC) (the et al., 2005; Deacon et al., 2001; Guenther et al., 2001)
and correlate with early loss of presynaptic terminals in
precursor of its disease-associated isoform,
the dorsal hippocampus (Cunningham et al., 2003).
PrPSc) in mice with early prion infection re-
We previously showed that early spongiform degenera-
versed spongiform change and prevented clin- tion in the hippocampus of prion-infected mice reverses
ical symptoms and neuronal loss. We now show when PrPC is depleted in neurons (Mallucci et al., 2003).
that cognitive and behavioral deficits and im- We now ask whether this early pathological change pro-
paired neurophysiological function accompany duces functional deficits and whether its reversal is
early hippocampal spongiform pathology. Re- reflected in functional recovery. As the hippocampus is
markably, these behavioral and synaptic im- particularly targeted by several strains of murine prions
pairments recover when neuronal PrPC is de- and is easily accessible for neurophysiological measure-
pleted, in parallel with reversal of spongiosis. ments, we focused on tests of hippocampal function.
Thus, early functional impairments precede We used prion-infected transgenic mice with and without
‘‘induced’’ PrP depletion for our experiments. Thus, tg37
neuronal loss in prion disease and can be res-
mice express PrP from ‘‘floxed’’ PrP sequences (MloxP
cued. Further, they occur before extensive
transgenes) and succumb to Rocky Mountain Laboratory
PrPSc deposits accumulate and recover rapidly (RML) prion infection 13 weeks postinoculation (wpi)
after PrPC depletion, supporting the concept (Mallucci et al., 2002). In these mice, earliest prion patho-
that they are caused by a transient neurotoxic logical changes, including spongiosis, gliosis, and PrPSc
species, distinct from aggregated PrPSc. These deposition, appear by 8 wpi (Mallucci et al., 2003). In
data suggest that early intervention in human double-transgenic NFH-Cre/tg37 mice, PrP expression
prion disease may lead to recovery of cognitive is the same as in tg37 mice until floxed PrP sequences
and behavioral symptoms. are excised by the DNA recombinase, Cre (Sauer and
Henderson, 1989), at 9–10 weeks of age, when neuronal
PrP is depleted (Mallucci et al., 2002). NFH-Cre/tg37 mice
INTRODUCTION infected with prions at 1 week of age develop early hippo-
campal pathology in parallel with control tg37 mice, but
Changes in motivation, mood, and behavior are common spongiosis reverses soon after Cre-mediated PrP deple-
early symptoms in human prion disorders, especially var- tion 8–9 wpi, and the animals survive long term (Mallucci
iant Creutzfeldt-Jakob disease (vCJD), often occurring et al., 2003).
long before diagnosis is made. By the time characteristic We tested memory function and spontaneous etholog-
dementia and motor deficits are established, there is typ- ical behaviors in vivo and measured synaptic responses
ically advanced neuronal loss, with no realistic potential neurophysiologically in vitro. We used the novel object
for curative treatment or recovery. In experimentally in- recognition task, a nonspatial learning task based on the
fected mice, clinical disease is diagnosed by the presence spontaneous preference of both mice and rats for novelty
of locomotor changes, but again, these occur during and their ability to remember previously encountered

Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc. 325


Neuron
Reversal of Prion-Induced Cognitive Defects

objects, that is rapidly learned (Clark et al., 2000; Dodart


et al., 1997; Ennaceur and Delacour, 1988; Messier,
1997). In rodents, the hippocampus is thought to be in-
volved in delay-dependent object-recognition memory
(i.e., when recognition intervals are long, >24 hr after learn-
ing phase), whereas recall over shorter retention intervals
is thought to involve parahippocampal structures (Clark
et al., 2000; Hammond et al., 2004; Mumby et al., 2005).
Novel-object recognition is extensively used for testing
declarative memory in mice, and, critically, performance
is independent of mouse strain or genetic background,
unlike most other memory tasks for which performance
is highly strain dependent (Sik et al., 2003). We also tested
the spontaneous behaviors of burrowing and nesting,
which have a robust association with early prion pathology
(Betmouni et al., 1999; Cunningham et al., 2005; Deacon
et al., 2001; Guenther et al., 2001) and also localize to
the dorsal hippocampus. These behaviors have been pro-
posed as powerful tools for elucidating brain function
(Gerlai and Clayton, 1999), requiring a high degree of orga-
nization and executive function, and are thought to reflect
motivational aspects of spontaneous behavior in rodents.

RESULTS

We infected tg37 and NFH-Cre/tg37 mice with RML prions


at 1 week of age and tested groups of eight to ten mice of
each genotype at each time point behaviorally and neuro-
physiologically from 7 wpi. Tg37 mice were tested up to
12 wpi, after which they developed clinical signs of prion
disease and were culled. NFH-Cre/tg37 mice were tested
up to 30 wpi. Groups of six mice infected in parallel were
examined neuropathologically every 2 weeks postinocula-
tion. Equal numbers of uninfected mice were tested at
three representative time points (pre-, during-, and post-
PrP depletion) to exclude unforeseen effects of loss of
PrP expression itself.

Object Memory Is Impaired Early in Prion Infection


Figure 1. The Ability to Discriminate Novel Objects Is Lost in
We first assessed animals in the novel object recognition
Prion-Infected Mice but Recovers when Neuronal PrPC Is
Depleted, in Parallel with Reversal of Pathological Change
task (Figure 1). We used male mice, as they are generally
(A) Mice are allowed to explore two objects in an arena during the considered superior to females at localizing and recogniz-
learning phase of the test; then, after a retention interval, they are ex- ing objects (Frick and Gresack, 2003). All animals received
posed to a novel object (test phase). Time spent actively exploring the intensive training in an arena containing two objects that
novel object compared to the familiar one is a measure of object- they could explore freely (learning phase). Forty-eight
recognition memory and is expressed as the ratio of exploratory pref-
hours later, they were re-exposed to two objects, one of
erence. (B) Prion-infected tg37 (blue bars) and NFH-Cre/tg37 mice (red
which was familiar and one new (test phase) according
bars) showed normal object-recognition memory at 7 wpi, with prefer-
ential exploration of the novel object. This was significantly impaired in
all mice by 8 wpi, but recovered in mice with Cre-mediated PrP deple-
tion at 9 wpi. Recovery was sustained up to 30 wpi. (C) Hematoxylin mice were significantly more active than NFH-Cre/tg37 mice, consis-
and eosin-stained sections of hippocampi from prion-infected tg37 tent with previous findings in prion infection. n = 8–10 mice for all
and NFH-Cre/tg37 mice have normal appearance at 6 wpi (panels groups except at 8, 9, and 10 wpi when two groups of 8–10 mice
[A] and [D]). Spongiosis develops in all animals (panels [B] and [E]) by each were tested. N = 7 at 12 wpi for tg37, and at 20–30 wpi for
8 wpi when memory is impaired, but by 10 wpi this has reversed in NFH-Cre/tg37 mice. Error bars represent standard error of the mean
mice with Cre-mediated depletion (panel [F]), in parallel with recovery (SEM). Dashed line represents random exploration (exploratory prefer-
of novel-object memory. (D) Reduced exploration of novel object is not ence = 1). *p < 0.05; **p < 0.01; ***p < 0.001 (Student’s t test; two tails).
due to prion-induced reduction in motor activity. Mice were tested for Open arrow indicates onset of earliest signs of scrapie in tg37 mice
activity in the open field by measuring the number of squares crossed (ss). Closed arrow indicates onset of Cre-mediated knockout. Scale
in an arena in 3 min. There was no decline in activity, but at 12 wpi tg37 bar represents 160 mm.

326 Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc.


Neuron
Reversal of Prion-Induced Cognitive Defects

to established protocols (Bozon et al., 2003) (Figure 1A). because group-housed males that were intermittently
The same animals were tested sequentially at different separated for testing fought over time and had to be singly
time points throughout the course of infection, using dif- housed, excluding them from further testing. The differ-
ferent sets of objects at each time point. The percentage ence in exploratory preference ratios, or memory, for all
of time spent exploring the novel compared to the familiar time points after PrP depletion compared to performance
object was expressed as a ratio of exploratory preference at 8 wpi prior to depletion is highly significant: p < 0.00001.
and used as a measure of memory. Random exploration of ANOVA analysis of all data sets for group effects also re-
the two objects results in a ratio of 1; preferential explora- sulted in p < 0.0001. Uninfected mice examined at repre-
tion of the novel object compared to the other gives a ratio sentative time points retained exploratory capacity
greater than 1. Statistical significance of data was calcu- (Figure S2), ruling out the possibility that PrPC depletion it-
lated using parametric tests (Student’s t test; two-tailed) self might alter object-recognition memory.
after confirmation that data were normally distributed. The loss of preferential exploration of a novel object in
All mice tested showed random exploration of the tg37 mice was not due to prion-related changes in motor
equally unfamiliar objects during the learning phase of function. Indeed, we found that activity in the open field in-
the test, with a baseline exploratory ratio of 1 (see Fig- creased, rather than decreased, with disease progression
ure S1 in the Supplemental Data available online). At 7 up to 12 wpi in tg37 animals (Figure 1D) (p = 0.013 for
wpi, all prion-infected animals displayed normal novel- activity of tg37 mice at 12 wpi compared to 8 wpi, and
object discrimination during the test phase (Figure 1B), p = 0.015 compared to 10 wpi), but not in NFH-Cre/tg37
with mean exploratory preference ratios of 1.70 ± 0.19 mice or in uninfected animals (Figure S3). This is consis-
SEM (tg37 controls) and 1.81 ± 0.07 SEM (NFH-Cre/tg37 tent with previous observations in prion-infected mice of
mice). But at 8 wpi, both groups of infected mice showed several strains (Cunningham et al., 2005; Deacon et al.,
significant inability to discriminate a novel object during 2001; Dell’Omo et al., 2002; Guenther et al., 2001). Limb
the test phase, with mean exploratory preference ratios power, assessed by testing grip strength, was unaffected
of 1.05 6 0.05 SEM and 0.90 6 0.03 SEM, respectively, in all mice, and coordinated movement, assessed here by
for the novel object: i.e., no preference. These differences the ability of the mice to reorientate themselves on a verti-
in exploration at 7 wpi versus 8 wpi were significant for cal wire grid after inversion, was also maintained (data not
mice of both genotypes: p = 0.0001 for NFH-Cre/tg37 shown).
mice and p = 0.0498 for tg37 mice, and p = 0.0001 when Loss of PrP expression due to Cre-mediated recombi-
mice of both groups are pooled at 7 and 8 wpi and com- nation in the hippocampi of NFH-Cre/tg37 mice after
pared. The loss in novel-object discriminatory capacity 8 wpi (or 9 weeks of age for uninfected mice) was con-
coincided with the appearance of early spongiform firmed by immunohistochemistry and RT-PCR for PrP
change in all prion-infected mice (Figure 1C, center mRNA expression at representative time points (Figure 2).
panels) and with reduction of synaptic responses (see be-
low and Figures 5A–5C). In prion-diseased tg37 mice, this Burrowing Declines in Early Prion Infection
impairment of memory persisted throughout the course of We also tested the mice for changes in burrowing and
infection (Figure 1B). By 12 wpi, when they were beginning nesting previously described in early prion infection. In
to show early signs of prion disease (such as reduced the burrowing task, mice actively burrow in a container
grooming, but not overt motor signs), the exploratory pref- that is filled with objects, pushing or carrying them out
erence ratio was inverted at 0.64 ± 0.05 SEM (p = 0.0012 into the cage until the container is almost emptied
for exploration compared to 7 wpi), possibly reflecting (Figure 3A). We found that all mice burrowed actively, dis-
preference for the familiar object and neophobia (anxiety placing 70%–80% of the pellets in a 24 hr period at 6
induced by novel objects) at this stage in tg37 mice. (mean 78% ± 5 SEM) and 7 wpi (73% ± 4 SEM), respec-
tively (Figure 3B). Mice burrowed less from 8 wpi (to
PrP Depletion Reverses Early Cognitive Deficits a mean of 66% ± 6 for tg37 and 54% ± 7 for NFH-Cre/
In contrast to tg37 mice, NFH-Cre/tg37 mice recovered tg37 mice, falling to 42% ± 6 and 45% ± 6, respectively,
the ability to discriminate novel objects after neuronal at 9 wpi). The timing of onset of the impairment is consis-
PrPC depletion. The exploratory preference for the novel tent with observations in other prion-infected mouse
object returned at 9 wpi (mean ratio of 1.40 ± 0.05 SEM; strains (Cunningham et al., 2003, 2005; Deacon et al.,
Figure 1B), very soon after PrP depletion, (p = 0.0004 for 2001; Guenther et al., 2001). The decline was significant
NFH-Cre/tg37 mice at 9 wpi compared to 8 wpi). The dif- for burrowing at 8 and 9 wpi compared to activity at 6
ference between mice with and without PrP depletion at and 7 wpi for both tg37 mice and NFH-Cre/tg37 mice
9 wpi is also significant (p = 0.0451). This functional recov- (p = 0.001 in each case). In tg37 mice, burrowing con-
ery occurred in parallel with reversal of spongiform pathol- tinued to decline until the mice reached onset of clinical
ogy seen in NFH-Cre/tg37 mice examined histologically disease, when they displaced just 21%–28% of pellets.
over this period (Figure 1C, panels E and F). Further, the
recovery in memory was sustained up to 20+ wpi, with PrP Depletion Reverses Burrowing Deficits
a mean exploratory ratio of 1.67 ± 0.25 SEM. Numbers In contrast, in mice with Cre-mediated PrP knockout,
available for testing after 20 weeks were diminished there was recovery of burrowing behavior by 10 wpi,

Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc. 327


Neuron
Reversal of Prion-Induced Cognitive Defects

Figure 2. Cre-Mediated PrP Depletion Occurs after 8 wpi or


9 Weeks of Age in NFH-Cre/tg37 Mice
(A) Immunohistochemical detection of PrP using ICSM 35 antibody
shows loss of PrP expression after 8 wpi (panels [b] and [c]). Box indi-
cates CA1 region where neurophysiological recordings were made. (B)
RT-PCR confirms reduction of PrP mRNA in hippocampal mRNA
extracts from NFH-Cre/tg37 mice after 8 wpi (or 9 weeks of age in Figure 3. Burrowing Behavior Declines Early in Prion Infec-
uninfected mice). Total transcript is reduced by 30%–40%, consis- tion but Recovers in Mice following Neuronal PrP Depletion
tent with its absence within neurons in the hippocampus at this (A) Healthy rodents ‘‘burrow’’ food pellets, carrying them out of a con-
stage. The reduction is equivalent to the fraction of recombined tainer and into their cage. (B) Early in prion infection, all mice burrowed
DNA detected by Southern blotting previously described (Mallucci equally. Burrowing then declined significantly in both tg37 and
et al., 2002, 2003). RNA was pooled from five mice at each time NFH-Cre/tg37 mice, respectively, at 9 wpi and continued to decline
point, and each reaction was performed in triplicate. Error bars repre- in tg37 mice. In mice with Cre-mediated PrP depletion, burrowing
sent standard deviation. Arrow indicates onset of Cre-mediated behavior resumed by 10 wpi and remained normal up to 20–30 wpi.
knockout. Numbers and symbols are as for Figure 1 legend.

which was significant when compared to control tg37 relatively later than burrowing behavior in the disease
mice at this time point (p = 0.004) and highly significant process, when Cre-mediated recombination and reversal
at 12 wpi (p = 0.0001). The increase in burrowing activity of early pathology has already occurred.
in mice 10–14 wpi (post-PrP depletion) compared with As motor impairment is not a factor here, these data
the reduction seen in mice at the earlier time points of support a motivational—rather than a motor—impairment
8 and 9 wpi (prior to/around time of PrP depletion) was as a basis for loss of burrowing activity, as proposed by
also significant (p = 0.001). Uninfected mice of both geno- Deacon et al. (Deacon et al., 2001).
types showed sustained burrowing activity when tested at
representative time points pre- and post-PrP depletion Behavioral Recovery Parallels Reversal of Pathology
(Figure S4). ANOVA analysis of all data to account for Histological examination of age-matched RML prion-
group effects confirmed significance of data; p < 0.0001. infected mice confirmed absence of detectable prion
The later recovery of burrowing (at 10 wpi) compared pathological change at 6 and 7 wpi, when all mice showed
to that of novel-object recognition (at 9 wpi) likely reflects normal learning, memory, and burrowing behavior (Fig-
the fact the animals were tested at the beginning of ures 1C, panels A and D and Figures 4A–4C and 4J–4L).
the week for burrowing and at the end for novel-object Loss of memory and burrowing correlated with the devel-
recognition, so that the actual difference in age at testing opment of spongiform change in neurons at 8 wpi (Fig-
is 2–3 days. ure 1C, panels B and E), and recovery of these behaviors
All mice constructed nests from nestlets up to 10 wpi, correlated with the reversal of spongiosis by 10 wpi in
when tg37 mice then stopped shredding their nestlets. mice with PrP depletion (Figure 1C, panel F). Reduced
Mice with Cre-mediated PrP depletion did not show any levels of synaptophysin 1 in the dorsal hippocampus
change in this behavior at any stage. This is probably have been correlated with reduced burrowing behavior
due to the fact that in this model nesting is affected in prion-infected mice (Cunningham et al., 2003). We

328 Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc.


Neuron
Reversal of Prion-Induced Cognitive Defects

Figure 4. Prion-Infected Mice with Neu-


ronal PrP Depletion Have Preserved
Synaptic Density in the Hippocampus
Despite PrPSc Accumulation
Hippocampal sections of prion-infected mice
were stained for synaptophysin, PrPSc, and
glial fibrillary acidic protein (GFAP) immunore-
activity. There are no pathological signs of
prion infection in any mice at 6 wpi when learn-
ing and behavior are normal. By 10 wpi, tg37
mice have marked loss of synaptophysin
immunoreactivity in the dorsal hippocampus,
PrPSc accumulation, and gliosis, but mice
with Cre-mediated PrP depletion have pre-
served synaptic density despite accumulating
extraneuronal PrPSc and gliosis up to 40 wpi.

found marked loss of synaptophysin immunoreactivity infected tg37 and NFH-Cre/tg37 mice showed similar
throughout the hippocampus in tg37 mice from 10 wpi, significant reductions in evoked excitatory postsynaptic
most notable in regions CA1-3 (Figure 4, panel I). We field potentials (EPSPs) at 50% of values seen in both
also found reduction in total synaptophysin levels in scra- uninfected mice and in recovered NFH-Cre/tg37 animals
pie sick tg37 animals on Western blotting of hippocampal (Figures 5A–5C). Cre-mediated neuronal PrP depletion in
homogenates (Figures S5A), but we did not detect NFH-Cre/tg37 mice resulted in rapid recovery of EPSPs
reduced levels at earlier time points in correlation with to control levels, a recovery sustained in mice examined
behavioral change here (Figure S5B), suggesting that up to 18 wpi. In contrast, synaptic responses in the
these behavioral deficits occur before there is significant prion-infected tg37 mice continued to decline, and at
loss of synapses structurally. Importantly, normal levels 9 wpi were 22% of those of uninfected control mice. The
of synaptophysin immunoreactivity were maintained in stimulus-response curves differed significantly between
infected NFH-Cre/tg37 mice up to 40 wpi (Figure 4, panel groups at different time points (p < 0.001, general linear
U), suggesting that synaptic integrity and density remains model [GLM]; pair-wise differences described above as
intact long term despite the extensive accumulation of significant had p < 0.001 with Bonferroni correction for
non-neuronal PrPSc (Figure 4, panel T). multiple comparisons). Action potentials in the presynap-
tic axons give rise to ‘‘fiber volleys’’ (Winegar and MacIver,
CA1 Synaptic Responses Are Impaired but Rapidly 2006) that can be seen immediately after the stimulus
Recover after PrP Depletion artifact. We found that the fiber volleys’ amplitudes were
We found that behavioral changes were paralleled by neu- linearly related to the EPSPs (correlation coefficient 0.97;
rophysiological changes (Figure 5). We tested synaptic Figure 6A), suggesting that the change in EPSP was
responses and plasticity in the stratum radiatum of the directly due to a change in the presynaptic action poten-
CA1 region of hippocampal slices. At 8 wpi, both prion- tials, rather than at a synaptic level.

Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc. 329


Neuron
Reversal of Prion-Induced Cognitive Defects

Figure 5. Synaptic Responses Are Depressed in Prion-


Infected Mice but Recover when Neuronal PrPC Is Depleted
(A) Field excitatory postsynaptic potential (EPSP) slope input-output
Figure 6. Deficits in Synaptic Responses Correlate with
curves show marked differences between the experimental groups.
Reduction in the Presynaptic Axonal Fiber Volley, but Synap-
At 8 wpi, both tg37 (blue squares) and NFH-Cre/tg37 mice (red
tic Plasticity Is Preserved in Prion-Infected Mice, and They
squares) have smaller synaptic responses than age-matched uninoc-
Display Normal Long-Term Potentiation
ulated control mice of both genotypes (gray diamonds). In prion-
(A) The relationship between EPSP slope and fiber volley (FV) ampli-
infected tg37 mice at 9 wpi (blue circles), synaptic responses are fur-
tude is unaltered during scrapie infectivity (linear regression, solid
ther reduced. However, after Cre-mediated PrP depletion, synaptic
line, R2 = 0.97). (B) Representative fiber volleys for each group (indi-
responses return to control levels in NFH-Cre/tg37 mice at 9 wpi (red
cated by arrows) are shown in the sample traces of the synaptic
circles) and are sustained at this level up to 18 wpi (red triangles).
responses. For clarity, the stimulus artifact was removed as indicated
p < 0.001, general linear model analysis (GLM). (B) Means for the
by the break in the traces. The amplitude of response in uninfected
experimental groups in the GLM analysis reveal significantly smaller
control mice is shown at the top (gray diamond). This amplitude is
EPSPs in scrapie-infected tg37 mice (blue bars) than in both NFH-
reduced at 8 wpi in tg37 and NFH-Cre/tg37 mice (blue and red
Cre/tg37 mice (red bars) after PrP depletion at 9 wpi and uninfected
squares, respectively) and is further diminished in tg37 mice at 9 wpi
control mice (gray bar). p < 0.001, pairwise comparisons, using
(blue circles). The response recovers in mice with PrP depletion at
the Bonferroni correction for multiple comparisons; dotted line repre-
9 wpi (red circle), which is sustained in mice up to 18 wpi (red triangle).
sents mean EPSP value and dashed lines represent 95% confidence
(C) Plot of mean population synaptic potential (PSP) slope versus time
intervals of the control group. Error bars represent SEM. (C) Represen-
(LTP). LTP averaged for each experimental group show that all had sig-
tative sample traces for each group. The amplitude of response in
nificant potentiation of EPSP. A theta-burst protocol was applied at
uninfected control mice is shown at the top (gray diamond). This
time 0 (see Experimental Procedures for details) and resulted in signif-
amplitude is reduced at 8 wpi in tg37 and NFH-Cre/tg37 mice (blue
icant potentiation of the PSP slope. Symbols represent mean PSP, er-
and red squares, respectively) and is further diminished in tg37 mice
ror bars represent SEM. n = 7–12 slices from at least four animals per
at 9 wpi (blue circles). The response recovers in mice with PrP deple-
group, except for 14 wpi and 18 wpi mice, for which data were pooled,
tion at 9 wpi (red circle), which is sustained in mice up to 18 wpi (red
thus n = 10 animals for this group.
triangle).

We also measured long-term potentiation (LTP) (Fig- DISCUSSION


ure 6C and Figure S6), a form of synaptic plasticity
linked with some forms of learning and memory. In all Our combined behavioral and neurophysiological analy-
mice, LTP was sustained for at least 30 min despite differ- ses provide the first direct evidence (to our knowledge)
ences in baseline (half-maximal) EPSPs between the ex- for early neuronal dysfunction producing functional cogni-
perimental groups and the particularly low baseline values tive impairment in prion disease. We have shown that early
for tg37 mice at 9 wpi. As the EPSPs are expressed as hippocampal pathology in prion-infected mice is associ-
a percentage of the mean preconditioning control values; ated with impaired synaptic responses and with cognitive
the apparently greater LTP in the 9 wpi tg37 mice and behavioral deficits in hippocampal tasks. These defi-
(Figure 6C) may be a result of this normalization (see cits precede neuronal and synaptic loss, and they are rap-
also Figure S6). Overall, we found no reduction in LTP in idly reversed when PrPC is depleted, demonstrating the
prion-infected mice with or without PrP depletion. potential for recovery of neuronal function at this stage.

330 Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc.


Neuron
Reversal of Prion-Induced Cognitive Defects

Further, both their occurrence and recovery are inde- in effective synaptic integration itself, implicating mecha-
pendent of PrPSc accumulation, with novel implications nisms other than CA1 LTP in deficient memory. Dissocia-
for both mechanisms of neurotoxicity and therapeutic tion of mechanisms underlying the acquisition and storage
possibilities. of information and the endpoint of such changes in
We found that relatively early in prion infection, by 8 wpi, expressed behavior have been reported elsewhere (Frag-
mice lost the capacity for novel-object recognition, a test kouli et al., 2005), and learning and memory have been
of nonspatial memory, and showed significantly reduced found associated with impaired synaptic responses and
burrowing behavior (Figures 1 and 3). These were not intact LTP in other rodent models (Brace et al., 1985).
due to impaired motor function (Figure 1D) and thus reflect However, LTP alterations have been described in prion-
impaired cognitive and motivational function. Further, PrP infected mice, of different mouse strains than those
depletion itself had no effect on behavior and cognition in used here, at a relatively later stage in the disease process
uninfected animals (Figures S1–S4). Neurophysiologically, and infected with different prion strains and using
we observed reduced synaptic responses in the CA1 more intense conditioning stimulation to induce LTP
region at the same time points (Figure 5). Remarkably, (Chiti et al., 2006).
by 9 wpi, soon after neuronal PrP depletion in NFH-Cre/ Both cognitive and neurophysiological impairment and
tg37 mice both synaptic and cognitive/behavioral deficits recovery in our model appeared to be independent of
recovered, and this recovery was sustained up to 30 wpi. PrPSc accumulation. Impaired object-recognition mem-
The development of phenotypic deficits thus paralleled ory, burrowing behavior, and reduced postsynaptic
the development of early spongiform change, and recov- potentials in CA1 occur at 8 wpi in both mouse lines,
ery of these deficits in mice with PrP depletion paralleled before extensive deposits of PrPSc are apparent. Further,
its reversal. We found no objective evidence of synapse both the loss and recovery of memory and species-typical
loss in the early stages of infection associated with early behaviors appear to be independent of aggregated PrPSc
cognitive failure (Figure 4 and Figure S5), in contrast to deposition, up to 20+ wpi, as is synaptic function up to
observations made by others in association with burrow- 18 wpi.
ing decline in prion infection (Cunningham et al., 2003), Indeed, early synaptic pathology and dendritic dysfunc-
which may reflect differences in the strains of mice and tion precede PrPSc accumulation (Jamieson et al., 2001),
of infectious prions used. The relationship between spon- consistent with our observations on behavioral and neuro-
giform change and the functional deficits we observed is physiological changes in the absence of extensive aggre-
not clear. Spongiform degeneration in prion disorders in- gated PrPSc deposits here.
volves the formation of membrane-bound vacuoles within Our data are also consistent with findings in other
neurons and neuropil—predominantly within dendrites— models of neurodegenerative disorders: that neuronal
and intradendritic distensions and dendritic spine loss dysfunction and cognitive defects occur before cellular
colocalize with vacuolar pathology in the hippocampus degeneration and independently of pathological aggrega-
(Jeffrey et al., 1997). The deficits we observe may thus tion of disease-associated proteins (Lambert et al., 1998;
reflect the earliest neuronal and dendritic dysfunction, Santacruz et al., 2005; Yamamoto et al., 2000), and they
although more detailed neurophysiological analysis impli- can result from synaptic dysfunction rather than neuronal
cates impaired function at the level of the presynaptic loss (Buttini et al., 2002; Mucke et al., 2000; Walsh et al.,
axon also. 2002; Wang et al., 2002). In several models of Alzheimer’s
Thus, we found that synaptic responses were de- disease there is increasing evidence that this may be due
pressed in our mouse model, which can be explained to soluble Ab oligomers rather than amyloid (Billings et al.,
by the proportional loss of the presynaptic fiber volley 2005; Dodart et al., 2002; Kotilinek et al., 2002), explaining
(Figure 6B): the correlation between reduced field synap- plaque-independent cognitive failure in these animals.
tic potentials and presynaptic fiber volleys is very strong at Consistent with these studies, our findings support the
0.97, strongly supporting a causal link (Figure 6A). Depres- concept that a transient—as yet unidentified—neurotoxic
sion of presynaptic fiber volleys has been reported late in species is generated within neurons when PrPC is con-
the progression of another scrapie model (Chiti et al., verted to PrPSc, which rapidly impairs neuronal function
2006), where it was tentatively attributed to loss of the and synaptic responses. Depleting neuronal PrPC would
presynaptic neurons (CA3 pyramidal cells). The present halt formation of such an intermediate, whether it be
loss of the fiber volley was rapidly reversible and therefore a new molecular species or an oligomeric form of PrPSc,
cannot be due to neuronal loss; more likely it was due to allowing rapid recovery of neuronal function.
axonal dysfunction. Targeting PrPC therapeutically remains somewhat con-
Synaptic plasticity appears to be maintained, however: troversial, however, as the physiological role of the native
the presence of LTP recordings that we found in all mice at protein is still unclear. While both embryonic (Bueler et al.,
all stages of disease implies that those synapses still 1992; Manson et al., 1994) and postnatal (Mallucci et al.,
remaining in prion-infected tg37 mice have the intact 2002) PrP knockout mice are viable and phenotypically
molecular apparatus both to release transmitter and to essentially normal, there is conflicting evidence for a role
sustain LTP, for review see Collingridge et al., 2004. It for PrPC in some learning tasks (Criado et al., 2005) but
appears that here memory is impaired due to reduction not others (Roesler et al., 1999). Our data suggest that at

Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc. 331


Neuron
Reversal of Prion-Induced Cognitive Defects

least nonspatial memory tasks are independent of PrPC Inoculation with Prions
expression specifically, depending instead on neuronal in- One-week-old mice were anesthetized and inoculated intracerebrally
into the right parietal lobe with 20 ml of 1% brain homogenate of
tegrity and effective synaptic transmission. Also, we know
Chandler/RML mouse-passaged scrapie strain (RML, Rocky Mountain
that PrPC depletion does affect certain intrinsic properties Laboratories) and subsequently examined daily for signs of scrapie, as
of CA1 cells, causing reduction in the late afterhyperpola- described (Mallucci et al., 2003).
rization potential (AHP) (Colling et al., 1996; Mallucci et al.,
2002). However, the only known phenotypic correlate of Behavioral Testing
late AHP reduction in transgenic mice is loss of social All testing took place in a room with sombre lighting and constant
transference of food preference between mice, which background noise. Mice were handled for 10 min a day for several
days and habituated to the test environment prior to testing. Only
can be reversed by environmental enrichment (Giese
group-housed males were used. Groups of eight to ten mice were
et al., 1998). tested sequentially over time for each paradigm, unless otherwise
Recent work implicates PrP expression in neurogenesis stated. Mice were handled, habituated, trained, and tested at the
and in the the proliferation of multipotent neuronal precur- same time for each experiment.
sor cells in the dentate gyrus in vivo. Despite having
reduced numbers of proliferating precursor cells com- Novel Object Recognition Task
This was performed as described (Bozon et al., 2003). Briefly, mice
pared to wild-type or PrP-overexpressing animals, mice
were tested in a dark cylindrical arena (69 cm diameter) mounted
constitutively lacking in PrP nonetheless have equal final with a 96 LED cluster infra-red light source and Watec video camera
numbers of neurons in the dentate gyrus as PrP-express- (both Track-sys). Various plastic objects were constructed from inter-
ing mice (Steele et al., 2006). Consistent with these data, locking plastic building blocks and were used in all experiments. (Pilot
we also found that numbers of granule cell neurons in studies confirmed all objects were of equal inherent interest.) On day 1
the dentate gyrus were unaltered after PrP depletion in in- of each experiment (learning phase), two objects were placed in the
center of two 15 cm diameter circles inside the arena. Each mouse
fected and uninfected NFH-Cre/tg37 mice (Figure S7).
was placed in the arena for two blocks of 10 min for exploration of
Overall, we conclude that the dramatic benefits to neuro-
the objects with an intertrial interval of 5 min. Forty-eight hours later,
nal function and survival in prion-infected mice we have a novel object was randomly exchanged for one of the familiar ones,
shown here support targeting neuronal PrPC directly as and retention was tested by placing the mice back in the arena for
a therapeutic approach. a 5 min session (test phase). The amount of time spent exploring all
Our findings of early reversible neurophysiological and objects was measured for each animal with the examiner blind to
cognitive deficits occurring prior to neuronal loss open genotype and time point for the animals. All objects and arena were
cleansed thoroughly between trials to ensure the absence of olfactory
new avenues in the prion field. To date, prion infection in
cues. Criteria for exploration were based strictly on active exploration,
mice has conventionally been diagnosed when motor where mice had at least both forelimbs within a circle of 15 cm around
deficits reflect advanced neurodegeneration. Now the an object, head oriented toward it or touching it with their noses. Mice
identification of earlier dysfunction helps direct the study with overt motor symptoms were not used. New sets of objects were
of mechanisms of neurotoxicity and therapies to earlier used at each time point.
stages of disease, when rescue is still possible. Eventually
Burrowing and Nesting
it may also enable preclinical testing of therapeutic strate-
Two hours before the start of the dark period, mice (which had not
gies through cognitive endpoints. These data now lead to been food deprived) were placed in individual plastic cages containing
the hope that early intervention in human prion disease, a gray plastic tube 20 cm long 3 6.8 cm diameter, filled with 200 g of
will not only halt clinical progression but allow reversal of normal food pellets as described (Cunningham et al., 2003). The weight
early behavioral and cognitive abnormalities. of pellets remaining in the tube after 24 hr was measured, and the
percentage displaced (burrowed) was calculated. A pressed cotton
square (‘‘Nestlet’’) was also placed in each cage, and the occurrence
of nest building after 24 hr was observed.
EXPERIMENTAL PROCEDURES
Open-Field Activity
Animals Each mouse was placed in the corner of an empty, clear plastic arena
tg37 and NFH-Cre transgenic mice (Mallucci et al., 2002) were bred in- (50 3 32 3 20cm) divided into 10 3 10 cm squares and observed for
house and were housed in a temperature- and light-controlled mouse 3 min. The number of squares entered (all four paws inside) and num-
colony room (12 hr light/dark cycle) in groups of four to six. All mice had ber of rears was recorded.
free access to food and water. All cages contained Perspex boxes
(‘‘igloos’’), cylindrical cardboard tubes, and folded sheets of tissue Inverted Screen Test of Muscular Strength and Reorientation
paper in view of the positive effects of such enrichment on cognition Each mouse was placed onto a horizontal wire mesh of 12 mm squares
in rodents. All animal work conformed to the United Kingdom regula- made from 2 mm diameter wire. The mesh was slowly inverted, and the
tions and institutional guidelines and was performed under Home time for the mouse to fall off was measured up to 1 min. For orientation,
Office project license. Generation and characterization of these the mouse was first placed facing upward on the vertical mesh, which
mice, including onset and timing of recombination and genotyping was then rotated through 180 in the vertical plane so that the mouse
by PCR screening of DNA from tail biopsy samples are described faced the floor. The time taken for the mouse to reorientate itself and
(Mallucci et al., 2002). The mice were generated on a Prnp0/0 back- return to its original position was recorded.
ground so that all PrP expression is from the PrP (MloxP) transgene.
The genetic background is predominantly FVB after ten generations Electrophysiology
of backcrossing. All animals are hemizygous for one or both trans- Mice were anesthetized by intraperitoneal injection of a mixture of
genes (MloxP alone or also with NFH-Cre). medetomide (1 mg/kg) and ketamine (76 mg/kg) prior to being killed

332 Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc.


Neuron
Reversal of Prion-Induced Cognitive Defects

by cervical dislocation, their brains removed for preparation of 400 mm Business-stat/otherapplets/Normality.htm). Student’s t tests were
horizontal slices of ventral hippocampus, using a remote control Vibro- applied to all data sets with two tails (two samples; unequal var-
slice (Campden Instruments, Loughborough, UK) housed in a HEPA- iance). ANOVA testing was performed using one-way analysis for
filtered thin-film isolator to contain contaminated aerosols. Slices group effects (http://www.physics.csbsju.edu/stats/anova.html). Mann
were kept in an interface holding chamber in the isolator until trans- Whitney U tests (Wilcoxon rank sum tests) were performed where
ferred to an interface chamber for recording. ACSF comprised (in data was not normally distributed; notably for motor testing. Analysis
mM) 135 NaCl, 16 NaHCO3, 1.25 NaH2PO4, 3 KCl, 2 CaCl2, 1 MgCl, of neurophysiological data were performed using general linear model
and 10 glucose, pH 7.4, gassed with 95% O2, 5% CO2, maintained (GLM) in SPSS version 13.
at 32 C, and directed to a container containing 2 M NaOH after single
passage through the chamber.
Supplemental Data
Stimuli were applied through a bipolar stimulating electrode placed
The Supplemental Data for this article, including seven figures and
in the stratum radiatum approximately 150 mm from the CA1 pyramidal
Supplemental Experimental Procedures, can be found online at
cell layer. Extracellular recordings of EPSPs were made from the same
http://www.neuron.org/cgi/content/full/53/3/325/DC1/.
layer of stratum radiatum, 500 mm away, using glass microelec-
trodes, an Axoclamp 2B amplifier, and a 1401plus signal acquisition
system running Signal version 2.15 (CED, Cambridge, UK). Stimulus ACKNOWLEDGMENTS
response curves used a fixed set of stimulus strengths, and the field
EPSPs were measured as the initial slope (between 20%–80% of We thank Julie Underwood, Michelle Hainsworth, and Jackie Linehan
maximum). LTP was induced by a high-intensity theta-burst protocol for technical assistance; Ray Young for help with graphics; Sam Cooke
(Morgan and Teyler, 2001) comprising five trains of four pulses at and Tim Bliss (National Institute of Medical Research, Mill Hill) and
100 Hz separated by 200 ms, repeated six times with an interburst in- Colm Cunningham (University of Southampton) for advice and guid-
terval of 10 s, delivered after at least 30 min of recording half-maximal ance on behavioral testing. This work was funded by the Medical Re-
stimuli delivered every 30 s. The same test stimuli were then delivered search Council of Great Britain. The neurophysiological experiments
for a further 45 min. were funded by a Physiological Society summer studentship (H.K.)
and the Biotechnology and Biological Sciences Research Council
Neuropathological Examination of Brain Sections (A.D.P.).
PrPSc in brain tissue was detected after denaturation of fixed sections
in formic acid (Hill et al., 1997) using monoclonal antibody to PrP, ICSM Received: September 28, 2006
35 (Asante et al., 2002), and an automated immunostaining system Revised: December 11, 2006
(www.ventanamed.com) as described (Joiner et al., 2002). Astrocyto- Accepted: January 5, 2007
sis was detected using anti-GFAP polyclonal antiserum (Dako; 1:500 Published: January 31, 2007
dilution), and synaptophysin was detected with polyclonal rabbit anti-
serum (Zymed; prediluted) using the same system. Neu N staining was REFERENCES
performed using a mouse monoclonal antibody (Chemicon Interna-
tional; 1:2000 dilution). Sections of brains from all animals culled at Asante, E.A., Linehan, J.M., Desbruslais, M., Joiner, S., Gowland, I.,
each time point and from those succumbing to scrapie were examined Wood, A., Welch, J., Hill, A.F., Lloyd, S.E., Wadsworth, J.D.F., and Col-
by the same person, blinded to the identity of the animal and time of linge, J. (2002). BSE prions propagate as either variant CJD-like or
culling. Sections were scored for spongiosis, neuronal loss, gliosis, sporadic CJD-like prion strains in transgenic mice expressing human
PrPSc deposition, and synaptophysin-signal intensity. prion protein. EMBO J. 21, 6358–6366.
Betmouni, S., Deacon, R.M.J., Rawlins, J.N.P., and Ferry, V.H. (1999).
Immunohistochemistry in Frozen Sections
Behavioral consequences of prion disease targeted to the hippocam-
This was performed as described (Mallucci et al., 2002). PrP was
pus in a mouse model of scrapie. Psychobiology 27, 63–71.
detected using monoclonal antibody ICSM 35, as above, at 1:100
dilution for 1 hr at room tempertature followed by incubation for Billings, L.M., Oddo, S., Green, K.N., McGaugh, J.L., and LaFerla, F.M.
45 min in HRP-conjugated secondary antibody (Sigma; 1:10,000 dilu- (2005). Intraneuronal Ab causes the onset of early Alzheimer’s disease-
tion). Sections were counterstained with hematoxylin (Harris) for 5 min. related cognitive deficits in transgenic mice. Neuron 45, 675–688.
Bozon, B., Davis, S., and Laroche, S. (2003). A requirement for the
Reverse Transcriptase-PCR immediate early gene zif268 in reconsolidation of recognition memory
Hippocampi were dissected from freshly culled mice and stored after retrieval. Neuron 40, 695–701.
at 20 C in five volumes of RNAlater (QIAGEN). RNA was extracted Brace, H.M., Jefferys, J.G.R., and Mellanby, J. (1985). Long-term
with an RNeasy midi kit according to manufacturer’s instructions changes in hippocampal physiology and in learning ability of rats after
(QIAGEN). One-step RT-PCR amplification of the MloxP transgene intrahippocampal tetanus toxin. J. Physiol. 368, 343–357.
mRNA and b-actin was performed on a PRISM 7000 Taqman machine
Bueler, H., Fischer, M., Lang, Y., Bluethmann, H., Lipp, H.-P.,
(ABI). Primers and probes for the Prnp transgene were as follows.
DeArmond, S.J., Prusiner, S.B., Aguet, M., and Weissmann, C.
Forward primer, 50 -GGCCCATGATCCATTTTGG-30 ; reverse primer,
(1992). Normal development and behaviour of mice lacking the neuro-
50 -GCGGTACATGTTTTCACGGTAGT-30 ; probe, 50 -FAM-AACGACTG
nal cell-surface PrP protein. Nature 356, 577–582.
GGAGGACC-30 . b-actin was amplified using a commercially available
ABI assay: Mouse ACTB Endogenous control, VIC labeled MGB Buttini, M., Yu, G.Q., Shockley, K., Huang, Y., Jones, B., Masliah, E.,
probe. All reactions were performed in triplicate, and negative controls Mallory, M., Yeo, T., Longo, F.M., and Mucke, L. (2002). Modulation
included H2O only and omission of Reverse Transcriptase. PrP expres- of Alzheimer-like synaptic and cholinergic deficits in transgenic mice
sion was normalized to b-actin. Cycling conditions were 50 C for by human apolipoprotein E depends on isoform, aging, and overex-
20 min, 95 C for 15 min, then 40 cycles of 94 C for 45 s followed by pression of amyloid beta peptides but not on plaque formation.
60 C for 45 s. J. Neurosci. 22, 10539–10548.
Chiti, Z., Knutsen, O.M., Betmouni, S., and Greene, J.R. (2006). An
Statistical Analyses integrated, temporal study of the behavioural, electrophysiological
Individual data sets were tested for normality using the goodness of and neuropathological consequences of murine prion disease. Neuro-
fit test, Lilliefor’s test of normality (http://home.ubalt.edu/ntsbarsh/ biol. Dis. 22, 363–373.

Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc. 333


Neuron
Reversal of Prion-Induced Cognitive Defects

Clark, R.E., Zola, S.M., and Squire, L.R. (2000). Impaired recognition Jamieson, E., Jeffrey, M., Ironside, J.W., and Fraser, J.R. (2001). Apo-
memory in rats after damage to the hippocampus. J. Neurosci. 20, ptosis and dendritic dysfunction precede prion protein accumulation in
8853–8860. 87V scrapie. Neuroreport 12, 2147–2153.
Colling, S.B., Collinge, J., and Jefferys, J.G.R. (1996). Hippocampal Jeffrey, M., Goodsir, C.M., Bruce, M.E., McBride, P.A., and Fraser,
slices from prion protein null mice: Disrupted Ca2+-activated K+ cur- J.R. (1997). In vivo toxicity of prion protein in murine scrapie: ultrastruc-
rents. Neurosci. Lett. 209, 49–52. tural and immunogold studies. Neuropathol. Appl. Neurobiol. 23,
93–101.
Collingridge, G.L., Isaac, J.T., and Wang, Y.T. (2004). Receptor
trafficking and synaptic plasticity. Nat. Rev. Neurosci. 5, 952–962. Jeffrey, M., Halliday, W.G., Bell, J., Johnston, A.R., Macleod, N.K.,
Ingham, C., Sayers, A.R., Brown, D.A., and Fraser, J.R. (2000).
Criado, J.R., Sanchez-Alavez, M., Conti, B., Giacchino, J.L., Wills,
Synapse loss associated with abnormal PrP precedes neuronal de-
D.N., Henriksen, S.J., Race, R., Manson, J.C., Chesebro, B., and Old-
generation in the scrapie-infected murine hippocampus. Neuropathol.
stone, M.B. (2005). Mice devoid of prion protein have cognitive deficits
Appl. Neurobiol. 26, 41–54.
that are rescued by reconstitution of PrP in neurons. Neurobiol. Dis. 19,
255–265. Joiner, S., Linehan, J., Brandner, S., Wadsworth, J.D., and Collinge, J.
(2002). Irregular presence of abnormal prion protein in appendix in
Cunningham, C., Deacon, R., Wells, H., Boche, D., Waters, S., Diniz, variant Creutzfeldt-Jakob disease. J. Neurol. Neurosurg. Psychiatry
C.P., Scott, H., Rawlins, J.N., and Perry, V.H. (2003). Synaptic changes 73, 597–598.
characterize early behavioural signs in the ME7 model of murine prion
Kotilinek, L.A., Bacskai, B., Westerman, M., Kawarabayashi, T., Youn-
disease. Eur. J. Neurosci. 17, 2147–2155.
kin, L., Hyman, B.T., Younkin, S., and Ashe, K.H. (2002). Reversible
Cunningham, C., Deacon, R.M., Chan, K., Boche, D., Rawlins, J.N., memory loss in a mouse transgenic model of Alzheimer’s disease.
and Perry, V.H. (2005). Neuropathologically distinct prion strains give J. Neurosci. 22, 6331–6335.
rise to similar temporal profiles of behavioral deficits. Neurobiol. Dis.
Lambert, M.P., Barlow, A.K., Chromy, B.A., Edwards, C., Freed, R.,
18, 258–269.
Liosatos, M., Morgan, T.E., Rozovsky, I., Trommer, B., Viola, K.L.,
Deacon, R.M.J., Raley, J.M., Perry, V.H., and Rawlins, J.N.P. (2001). et al. (1998). Diffusible, nonfibrillar ligands derived from Abeta1–42
Burrowing into prion disease. Neuroreport 12, 2053–2057. are potent central nervous system neurotoxins. Proc. Natl. Acad.
Dell’Omo, G., Vannoni, E., Vyssotski, A.L., Di Bari, M.A., Nonno, R., Sci. USA 95, 6448–6453.
Agrimi, U., and Lipp, H.P. (2002). Early behavioural changes in mice Mallucci, G.R., Ratté, S., Asante, E.A., Linehan, J., Gowland, I.,
infected with BSE and scrapie: automated home cage monitoring Jefferys, J.G.R., and Collinge, J. (2002). Post-natal knockout of prion
reveals prion strain differences. Eur. J. Neurosci. 16, 735–742. protein alters hippocampal CA1 properties, but does not result in
Dodart, J.C., Mathis, C., and Ungerer, A. (1997). Scopolamine-induced neurodegeneration. EMBO J. 21, 202–210.
deficits in a two-trial object recognition task in mice. Neuroreport 8, Mallucci, G., Dickinson, A., Linehan, J., Klohn, P.C., Brandner, S., and
1173–1178. Collinge, J. (2003). Depleting neuronal PrP in prion infection prevents
disease and reverses spongiosis. Science 302, 871–874.
Dodart, J.C., Bales, K.R., Gannon, K.S., Greene, S.J., DeMattos, R.B.,
Mathis, C., DeLong, C.A., Wu, S., Wu, X., Holtzman, D.M., and Paul, Manson, J.C., Clarke, A.R., Hooper, M.L., Aitchison, L., McConnell, I.,
S.M. (2002). Immunization reverses memory deficits without reducing and Hope, J. (1994). 129/Ola mice carrying a null mutation in PrP that
brain Abeta burden in Alzheimer’s disease model. Nat. Neurosci. 5, abolishes mRNA production are developmentally normal. Mol. Neuro-
452–457. biol. 8, 121–127.

Ennaceur, A., and Delacour, J. (1988). A new one-trial test for neurobi- Messier, C. (1997). Object recognition in mice: improvement of mem-
ological studies of memory in rats. 1: Behavioral data. Behav. Brain ory by glucose. Neurobiol. Learn. Mem. 67, 172–175.
Res. 31, 47–59. Morgan, S.L., and Teyler, T.J. (2001). Electrical stimuli patterned after
teh theta-rhythm induce multiple forms of LTP. J. Neurophysiol. 86,
Fragkouli, A., Hearn, C., Errington, M., Cooke, S., Grigoriou, M., Bliss,
1289–1296.
T., Stylianopoulou, F., and Pachnis, V. (2005). Loss of forebrain cholin-
ergic neurons and impairment in spatial learning and memory in LHX7- Mucke, L., Masliah, E., Yu, G.Q., Mallory, M., Rockenstein, E.M., Tat-
deficient mice. Eur. J. Neurosci. 21, 2923–2938. suno, G., Hu, K., Kholodenko, D., Johnson-Wood, K., and McCon-
logue, L. (2000). High-level neuronal expression of abeta 1–42 in
Frick, K.M., and Gresack, J.E. (2003). Sex differences in the behavioral
wild-type human amyloid protein precursor transgenic mice: synapto-
response to spatial and object novelty in adult C57BL/6 mice. Behav.
toxicity without plaque formation. J. Neurosci. 20, 4050–4058.
Neurosci. 117, 1283–1291.
Mumby, D.G., Tremblay, A., Lecluse, V., and Lehmann, H. (2005). Hip-
Gerlai, R., and Clayton, N.S. (1999). Analysing hippocampal function
pocampal damage and anterograde object-recognition in rats after
in transgenic mice: an ethological perspective. Trends Neurosci. 22,
long retention intervals. Hippocampus 15, 1050–1056.
47–51.
Roesler, R., Walz, R., Quevedo, J., De-Paris, F., Zanata, S.M., Graner,
Giese, K.P., Storm, J.F., Reuter, D., Fedorov, N.B., Shao, L.R., Leicher, E., Izquierdo, I., Martins, V.R., and Brentani, R.R. (1999). Normal inhib-
T., Pongs, O., and Silva, A.J. (1998). Reduced K+channel inactivation, itory avoidance learning and anxiety, but increased locomotor activity
spike broadening, and after-hyperpolarization in Kvbetal.1-deficient in mice devoid of PrPC. Brain Res. Mol. Brain Res. 71, 349–353.
mice with impaired learning. Learn. Mem. 5, 257–273.
Santacruz, K., Lewis, J., Spires, T., Paulson, J., Kotilinek, L., Ingelsson,
Guenther, K., Deacon, R.M., Perry, V.H., and Rawlins, J.N. (2001). M., Guimaraes, A., DeTure, M., Ramsden, M., McGowan, E., et al.
Early behavioural changes in scrapie-affected mice and the influence (2005). Tau suppression in a neurodegenerative mouse model
of dapsone. Eur. J. Neurosci. 14, 401–409. improves memory function. Science 309, 476–481.
Hammond, R.S., Tull, L.E., and Stackman, R.W. (2004). On the delay- Sauer, B., and Henderson, N. (1989). Cre-stimulated recombination at
dependent involvement of the hippocampus in object recognition loxP-containing DNA sequences placed into the mammalian genome.
memory. Neurobiol. Learn. Mem. 82, 26–34. Nucleic Acids Res. 17, 147–161.
Hill, A.F., Desbruslais, M., Joiner, S., Sidle, K.C.L., Gowland, I., and Sik, A., van Nieuwehuyzen, P., Prickaerts, J., and Blokland, A. (2003).
Collinge, J. (1997). The same prion strain causes vCJD and BSE. Performance of different mouse strains in an object recognition task.
Nature 389, 448–450. Behav. Brain Res. 147, 49–54.

334 Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc.


Neuron
Reversal of Prion-Induced Cognitive Defects

Steele, A.D., Emsley, J.G., Ozdinler, P.H., Lindquist, S., and Macklis, Trommer, B.L. (2002). Soluble oligomers of beta amyloid (1–42) inhibit
J.D. (2006). Prion protein (PrPc) positively regulates neural precursor long-term potentiation but not long-term depression in rat dentate gy-
proliferation during developmental and adult mammalian neurogene- rus. Brain Res. 924, 133–140.
sis. Proc. Natl. Acad. Sci. USA 103, 3416–3421. Winegar, B.D., and MacIver, M.B. (2006). Isoflurane depresses hippo-
Walsh, D.M., Klyubin, I., Fadeeva, J.V., Cullen, W.K., Anwyl, R., Wolfe, campal CA1 glutamate nerve terminals without inhibiting fiber volleys.
M.S., Rowan, M.J., and Selkoe, D.J. (2002). Naturally secreted oligo- BMC Neurosci. 7, 5. Published online January 12, 2006. 10.1186/
mers of amyloid beta protein potently inhibit hippocampal long-term 1471-2202-7-5.
potentiation in vivo. Nature 416, 535–539. Yamamoto, A., Lucas, J.J., and Hen, R. (2000). Reversal of neuropa-
Wang, H.W., Pasternak, J.F., Kuo, H., Ristic, H., Lambert, M.P., thology and motor dysfunction in a conditional model of Huntington’s
Chromy, B., Viola, K.L., Klein, W.L., Stine, W.B., Krafft, G.A., and disease. Cell 101, 57–66.

Neuron 53, 325–335, February 1, 2007 ª2007 Elsevier Inc. 335

You might also like