Anda di halaman 1dari 41

by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

The Prion’s Elusive Reason


Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

ANNUAL
REVIEWS Further
Click here for quick links to
Annual Reviews content online,
for Being
including:
• Other articles in this volume
• Top cited articles Adriano Aguzzi, Frank Baumann, and Juliane Bremer
• Top downloaded articles
• Our comprehensive search Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland,
email: Adriano.Aguzzi@usz.ch

Annu. Rev. Neurosci. 2008. 31:439–77 Key Words


First published online as a Review in Advance on transmissible spongiform encephalopathy, prion diseases, PrPC , PrPSc
April 2, 2008

The Annual Review of Neuroscience is online at Abstract


neuro.annualreviews.org
The protein-only hypothesis posits that the infectious agent causing
This article’s doi: transmissible spongiform encephalopathies consists of protein and lacks
10.1146/annurev.neuro.31.060407.125620
any informational nucleic acids. This agent, termed prion by Stanley
Copyright ! c 2008 by Annual Reviews. Prusiner, is thought to consist partly of PrPSc , a conformational isoform
All rights reserved
of a normal cellular protein termed PrPC . Scientists and lay persons
0147-006X/08/0721-0439$20.00 have been fascinated by the prion concept, and it has been subjected
to passionate critique and intense experimental scrutiny. As a result,
PrPC and its isoforms rank among the most intensively studied proteins
encoded by the mammalian genome. Despite all this research, both the
physiological function of PrPC and the molecular pathways leading to
neurodegeneration in prion disease remain unknown. Here we review
the salient traits of those diseases ascribed to improper behavior of the
prion protein and highlight how the physiological functions of PrPC
may help explain the toxic phenotypes observed in prion disease.

439
living beings, including even the most elemen-
Contents tary infectious particles.
Prion diseases are generally characterized
TRANSMISSIBLE PRION
by widespread neurodegeneration and there-
DISEASES AND
fore exhibit clinical signs and symptoms of cog-
NONTRANSMISSIBLE
nitive and motor dysfunction, in addition to
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

PRION-RELATED DISEASES. . . . 440


propagating infectious prions and, in many in-
Prion Disease in Humans
stances, forming striking amyloid plaques. The
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

and Animals . . . . . . . . . . . . . . . . . . . . 441


latter plaques contain aggregates of PrPSc , a
The Nature of the Prion . . . . . . . . . . . 445
misfolded and beta-sheet-rich isoform of the
Neurotoxicity . . . . . . . . . . . . . . . . . . . . . . 447
protein PrPC encoded by the PRNP gene. Fur-
PHYSIOLOGICAL FUNCTION
ther neuropathological features are neuronal
OF THE CELLULAR PRION
loss, astrocytic activation (gliosis), and spongi-
PROTEIN . . . . . . . . . . . . . . . . . . . . . 447
form change. All prion diseases are progressive,
Prion Protein–Deficient Mice . . . . . . 447
fatal, and presently incurable.
Functional Domains
Although the normal cellular prion protein
of the Prion Protein . . . . . . . . . . . . . 449
PrPC can easily be digested with proteinase K
Point Mutations within
(PK), the beta-sheet-rich, misfolded form PrPSc
the Prion Protein . . . . . . . . . . . . . . . 451
is partially proteinase K (PK) resistant. A cru-
Evolution of the Prion Protein. . . . . . 452
cial piece of evidence demonstrating that PrPC
Cellular Processes Influenced
is a key player in prion disease came from ex-
by PrPC Expression . . . . . . . . . . . . . 452
periments showing that mice lacking the prion
PrP and the Immune System . . . . . . . 457
protein gene are resistant to prions (Bueler et al.
Molecular Mechanisms Mediating
1993).
the Function of PrPC . . . . . . . . . . . 457
Although the formation of PrPSc accompa-
Interaction Partners of PrPC . . . . . . . . 461
nies neurodegeneration in prion disease, many
CONCLUSIONS . . . . . . . . . . . . . . . . . . . . 465
lines of evidence indicate that PrPSc is not in-
trinsically neurotoxic. PrPC needs to be pre-
sented by host neurons for neurodegeneration
to occur. Thus, when neurografts propagat-
TRANSMISSIBLE PRION ing PrPSc were implanted into Prnpo/o mice,
DISEASES AND the host mice did not develop prion disease
NONTRANSMISSIBLE (Brandner et al. 1996). Additionally, trans-
PRION-RELATED DISEASES genic mice expressing only a secreted form of
The origin of the word “prion” stems from the PrPC , lacking its membrane attachment via gly-
anagram of “proteinaceous infectious particle.” cosylphosphatidylinositol (GPI) anchor, have
Naturally, the latter is by no means a qualify- been reported to be refractory to develop clini-
ing attribute because all conventional infectious cal signs of prion diseases, although prion inoc-
agents—including all viruses and bacteria—are ulation induces PrPSc formation and aggrega-
proteinaceous to some degree. What sets prions tion of amyloid plaques (Chesebro et al. 2005).
apart, as proposed by Prusiner, is that the actual This finding indicates that membrane attach-
infectious principle consists merely of protein ment of PrPC is a prerequisite for neurode-
and is capable of replicating and transmitting generation to occur and that the presence of
infections without the need for informational PrPSc alone does not cause disease. The im-
nucleic acids. This postulate counters much of portance of neuronal expression of PrPC for
the established molecular biological evidence, prion disease development has been corrobo-
GPI: glycosylphos-
phatidylinositol which predicates that nucleic acids are the basis rated by the phenotype of mice with neuron-
for self-replicating biological information in all specific ablation of PrPC eight weeks after prion

440 Aguzzi · Baumann · Bremer


inoculation. Early spongiform changes were re- and Kuru in humans; bovine spongiform en-
versed, and clinical disease was prevented. This cephalopathy (BSE) in cattle; chronic wasting
reversal occurred despite the accumulation of disease (CWD) in deer and elk; and trans-
TSE: transmissible
extraneuronal PrPSc (Mallucci et al. 2003). missible mink encephalopathy. BSE has been spongiform
Bona fide prion diseases are character- inadvertently transmitted to a variety of captive encephalopathies
ized by their transmissibility and are there- animals, causing feline spongiform en-
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

CJD: Creutzfeldt-
fore also termed transmissible spongiform en- cephalopathy (FSE) and a plethora of diseases Jakob disease
cephalopathies (TSE). Transmissibility is a in zoo animals including kudus, nyalas, and
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

BSE: bovine
defining, and hence indispensable, trait of all greater cats, for example. spongiform
prion diseases. However, transmissibility has encephalopathy
not been formally proven for all kinds of dis- Creutzfeldt-Jakob disease. CJD was initially CWD: chronic
eases thought to be caused by prions. In ad- described as a sporadic disease occurring for wasting disease
dition, some diseases are genetically associated no known cause (sCJD). The incidence of FSE: feline
with the prion protein, yet they are nontrans- CJD is low in all ethnicities and typically af- spongiform
missible. These diseases are sometimes called fects ∼1 person in one million each year. Very encephalopathy
prionopathies. Among these are rare genetic rapid cognitive decline, causing dementia, is the sCJD: sporadic CJD
syndromes that cosegregate with point muta- main symptom. Cerebellar symptoms includ-
tions in the open reading frame of the PRNP ing ataxia and myoclonus are also frequent pre-
gene. In addition to these naturally occurring senting symptoms. Death often occurs within
prionopathies, several transgenic mice have few weeks of the first signs of disease, and
been used to gain insight into functional do- a fulminant, “apoplectiform” course of dis-
mains of PrPC (Weissmann & Flechsig 2003). ease has been documented in the past. So-
In these mice, deletion of parts of PrPC caused matic mutations in the PRNP gene analogous
prionopathies, characterized by a shortened life to those in the germline of genetic CJD pa-
span and the development of white matter tients (see below) have been hypothesized to
disease in the central nervous system (CNS) underlie sporadic CJD. Alternatively, Aguzzi
as well as neuronal cell death in the cere- & Glatzel (2006) suggested that some cases
bellum (Baumann et al. 2007, Li et al. 2007, of alleged sCJD derive from heretofore un-
Shmerling et al. 1998). Overexpression of wild- recognized infections. Finally, PrPC may pos-
type PrPC also caused disease in transgenic mice sess a finite, albeit extremely low, propensity to
(Westaway et al. 1994). self-assemble into ordered aggregates of PrPSc ,
thereby stochastically initiating prion replica-
tion and, ultimately, a sporadic form of disease.
Prion Disease in Humans and Animals The latter scenario could be regarded as the
Prion diseases have occurred in humans and an- bad-luck hypothesis. However, none of this has
imals for many years. A disease similar to scrapie been proven, and therefore the cause of sCJD
was recorded in the mid eighteenth century, is still unknown.
and scholars heavily debated its origin. A crucial
experiment showing incontrovertible trans- Variant Creutzfeldt-Jakob disease and
missibility of scrapie to goats was performed by Bovine Spongiform Encephalopathy. Pub-
Cuille & Chellè in the 1930s (Cuille & Chellè lic understanding of prion disease remained
1939). The first cases of human prion disease, limited for a long time: For example, we
Creutzfeldt-Jakob disease (CJD), were reported have heard neurologists saying that CJD is
in the 1920s (Creutzfeldt 1920, Jakob 1921). an essentially nonexistent disease. However,
The number of human and animal diseases this mindset changed completely when BSE
recognized as TSEs has increased steadily and was first reported in the early 1980s (Wells
now includes Gerstmann-Sträussler-Scheinker et al. 1987). In the following years and until
syndrome (GSS), fatal familial insomnia (FFI), mid 2007, BSE affected ∼190,000 cows

www.annualreviews.org • The Prion’s Elusive Reason for Being 441


(http://www.oie.int/). Some investigators morphisms (129MM, MV or VV), and hetero-
suggested that BSE could cause a new variant dox modes of infection including blood-borne
form of CJD (vCJD) in humans. A direct transmission. If we account for the time it will
vCJD: variant CJD
experimental proof that vCJD represents take to eradicate these secondary transmissions
transmission of BSE prions to humans cannot in the population, vCJD is not likely to disap-
be produced. However, epidemiological, pear entirely in the coming four decades.
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

biochemical, neuropathological evidence and


transmission studies strongly suggest that BSE Iatrogenic CJD. Iatrogenic CJD is acciden-
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

has transmitted to humans in the form of vCJD tally transmitted during the course of med-
(Aguzzi 1996, Aguzzi & Weissmann 1996, ical or surgical procedures. The first docu-
Bruce et al. 1997, Hill et al. 1997). The inci- mented case of iatrogenic prion transmission
dence of vCJD has been rising between 1994, occurred in 1974 and was caused by corneal
when the first patients suffering from vCJD transplantation of a graft derived from a pa-
presented with their initial symptoms, and tient suffering from sCJD (Duffy et al. 1974).
2001, raising fears that a very large epidemic Iatrogenic CJD is also rare, most often ob-
may be looming. At the time of this writing, served in individuals that have received cadav-
vCJD has killed ∼200 individual victims world- eric dura mater implants and human growth
wide (http://www.cjd.ed.ac.uk/). Most of the hormone; some of these individuals received
affected individuals lived in United Kingdom gonadotrophin extracted from human pitu-
and France. Fortunately, in the United King- itary glands or had stereotactically placed elec-
dom the incidence appears to be decreasing trodes in their brains (Will 2003). Four cases
from the year 2001 to 6 diagnosed cases yearly of vCJD transmission by blood transfusions
in 2005 and 2006. In contrast, in France the have been reported recently in the United
number of probable and definite cases of vCJD Kingdom (Llewelyn et al. 2004, Peden et al.
increased from 0 to 3 diagnosed cases per year 2004, Wroe et al. 2006) (see also http://www.
in 1996–2004 to 6 per year in 2005 and 2006. In cjd.ed.ac.uk/TMER/TMER.htm). The fact
2007, the number of cases was back to 3 again. that preclinically infected individuals can trans-
(http://www.invs.sante.fr/publications/mcj/ mit vCJD underscores the important medical
donnees mcj.html). A 30+-year mean in- need for sensitive diagnostic tools, which could
cubation time of BSE/vCJD in humans is be used for screening blood units prior to trans-
not entirely implausible, and therefore some fusion, for example.
authors have predicted a multiphasic human
BSE endemic with a second increase in the in- Kuru. In the mid 1950s, when the remote
cidence of vCJD affecting people heterozygous parts of Papua New Guinea were first explored
at codon 129 (Collinge et al. 2006). Others, by Australians and Westerners, Kuru was first
these authors included, regard the incidence described in research (Gajdusek & Zigas 1957).
of vCJD as subsiding (Andrews et al. 2003) Kuru was, at that time and at least since 1941,
(Figure 1). an endemic disease among some tribes of
It is important to note, however, that the New Guinea aborigines, especially among the
above considerations apply primarily to the epi- Fore linguistic group and neighboring tribes
demiology of primary transmission from cows (Gajdusek & Reid 1961). Kuru in the Fore lan-
to humans. Although, by now a pool of pre- guage means “to shiver,” and along with other
clinically infected humans may have been built. signs of cerebellar ataxia, shivering is a hallmark
Human-to-human transmission may present of the disease. The ritual consumption of dead
with characteristics very different from those relatives as a symbol of respect and mourning
of primary cow-to-human transmission, in- is the attributed route of transmission. As a
cluding enhanced virulence, shortened incu- consequence, the incidence has steadily fallen
bation times, disrespect of allelic PRNP poly- after cessation of cannibalism in Papua New

442 Aguzzi · Baumann · Bremer


a
40,000
Number of yearly identified infected cows

35,000
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

BSE UK
BSE non-UK
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

30,000

25,000

20,000

15,000

10,000

5000

0
<1988 1988 1989 1990 1991 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007

b
Number of yearly diagnosed cases

30
Secondary
Cases UK vCJD cases vCJD cases vCJD (blood
25 (dead) (still alive) transfusion)
Cases France
UK 160 3 4
20 France 21 2 –
Republic of Ireland 4 0 –
15 Italy 1 – –
USA 3 – –
10 Canada 1 – –
Saudi Arabia – 1 –
Japan 1 – –
5
Netherlands 2 – –
Portugal 1 1 –
0 Spain 2 – –
1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007

Figure 1
BSE and vCJD cases reported worldwide. (a) Reported cases of bovine spongiform encephalopathy (BSE) in the United Kingdom (UK)
(blue), and in countries excluding the UK (red ). Non-UK BSE cases include cases from countries both within and outside of the
European Union (EU). Data are as of December 2006 (http://www.oie.int). (b) Reported cases of variant Creutzfeldt-Jakob disease
(vCJD) in the UK (blue) and in countries outside the UK (red ). Non-UK vCJD cases include those reported in France, Republic of
Ireland, Italy, United States, Canada, Saudi Arabia, Japan, the Netherlands, Portugal, and Spain. Data are as of February 2008 and
include cases of vCJD in patients who resided in the UK in the 1980s or 1990s [see the National Creutzfeldt-Jakob Disease Surveillance
Unit Web site for vCJD data to July 2007 (http://www.cjd.ed.ac.uk/)].

www.annualreviews.org • The Prion’s Elusive Reason for Being 443


Guinea (Collinge et al. 2006). In a concise and found in families with hereditary or genetic
extremely clairvoyant observation published in CJD (gCJD). Figure 2 summarizes known mu-
1959, Bill Hadlow noted the epidemiological, tations causing human TSEs. gCJD occurs
gCJD: genetic CJD
clinical, and neuropathological similarities with point mutations mostly affecting the re-
OR: octarepeat region
between Kuru and scrapie (Hadlow 1959). gion between the second and the third he-
These were taken up by Carleton Gajdusek lix of the carboxy-terminus. However, inser-
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

who, in 1966, succeeded in transmitting Kuru tions in the octarepeat region (OR) in the
to three chimpanzees (Gajdusek et al. 1966). amino-terminus, and even one instance of a
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Soon thereafter, serial passage of Kuru and of premature termination codon at position 145,
several other prion diseases was demonstrated have also been associated with human prion
in chimpanzees and other primates (Gajdusek disease. The inheritance was, in all cases,
et al. 1967, 1968). Investigators have since autosomal dominant, often with very high pen-
transmitted human prion disease to various etrance. The clinicopathological disease phe-
species including laboratory rodents. notype varies depending on the actual muta-
tion, as well as on polymorphisms at codon 129,
Genetic CJD and Gerstmann-Sträussler- and most likely on a plethora of yet uniden-
Scheinker syndrome. Several mutations in tified modifiers and cofactors (Kovacs et al.
the prion protein gene (PRNP) have been 2002).

CC1 OR CC2 HC H1 H2 H3 GPI

S S

Mutations causing GSS Octarepeat insertion P102L-129M A117V-129V H187R-129V Q217R-129M

P105L-129V G131V-129M F189S-129V

Y145*-129M D202N-129V

Q212P

Mutations causing gCJD Octarepeat insertion D178N-129V E208H M232R

V180I E200K

T188K V210I

T188R-129V

E196K E211Q

V203I

Mutations causing FFI D178N-129M

Mutations in PRNP associated with familial dementia G114V Q160*-129M N171S T183A
and/or neuropsychiatric symptoms H187R
(not further classified)

Figure 2
The human PrPC protein and its mutants. The mature human PrPC protein contains 208 amino acid residues. It features two positively
charged amino acid clusters denoted CC1 and CC2 (blue boxes), an octapeptide repeat region (OR) ( green boxes), a hydrophobic core
(HC) ( gray box), three α-helixes (H1-H3) (red boxes), one disulphide bond (S–S) between cysteine residues 179 and 214, and two
potential sites for N-linked glycosylation (red forks) at residues 181 and 197. A glycosylphosphatidylinositol anchor (GPI) ( yellow box) is
attached to the C-terminus of PrP. This figure indicates in black framed boxes point mutations and insertions found in the human
PRNP gene in patients with prion disease. The associated polymorphisms of codon 129 (methionine M or valine V) are indicated.
Amino acids are given in single-letter code. The asterisk indicates a stop codon; therefore, this mutation results in a truncated protein.

444 Aguzzi · Baumann · Bremer


The first descriptions of Gerstmann- disease-causing mutation in the prion protein
Sträussler-Scheinker syndrome (GSS) origi- gene (D178N) was identified, thereby allow-
nate from 1928 and 1936 in an Austrian family ing the classification of FFI as a genetically de-
GSS: Gerstmann-
(Gerstmann 1928, Gerstmann et al. 1936). In termined prion disease (Medori et al. 1992). Sträussler-Scheinker
the following years, analogous disorders have The final proof that FFI is a TSE was achieved syndrome
been described, but its classification as a TSE when FFI was successfully transmitted to mice
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

FFI: fatal familial


lagged until 1981, when Masters and colleagues (Tateishi et al. 1995). FFI typically affects the insomnia
(1981) reported that inoculation of brain tis- thalamus, and accordingly, the core clinical fea-
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

sue from three patients with GSS resulted in tures are disruption of the normal sleep-wake
spongiform encephalopathy in nonhuman pri- cycle, sympathetic overactivity, endocrine ab-
mates. The authors also defined clinical hall- normalities, and impaired attention (Collins
marks of GSS (earlier age at onset, longer et al. 2001). In addition to the pathogenic point
disease duration, and prominent cerebellar mutation D178N, the methionine-valine poly-
ataxia) differentiating the disease from CJD. morphism at codon 129 of the PRNP gene con-
Nowadays GSS is considered an autosomal- trols the disease phenotype. Whereas D178N-
dominantly inherited TSE caused by muta- 129MM (homozygosity for methionine at
tions in the prion protein open reading frame, codon 129) was associated with FFI, heterozy-
manifesting typically with progressive cerebel- gosity at codon 129 (D178N-129MV) segre-
lar ataxia or spastic paraparesis and cognitive gated with the familial CJD subtype (Goldfarb
decline. The known GSS-causing mutations are et al. 1992). However, Zarranz et al. (2005)
summarized in Figure 2. In addition to the re- reported more recently that this genotype-
gions affected in gCJD, mutations altering the phenotype association is not absolute. In one
sequence of the central domain can cause GSS. study, several patients have been identified with
Its distinctive neuropathological feature is the a CJD phenotype and a D178N-129MM geno-
presence of widespread large and multicentric type. The authors concluded that rather than
amyloid plaques (Collins et al. 2001). being separate disease entities, prion disease
GSS is generally transmissible (Hsiao et al. phenotypes such as FFI and CJD represent two
1989, Masters et al. 1981, Tateishi et al. 1988); extreme manifestations of a continuous disease
therefore, its classification as a TSE is widely spectrum (Zarranz et al. 2005).
accepted. However, the overall experimental In addition to the familial form of fatal in-
transmissibility of GSS to nonhuman primates somnia, a sporadic form of the disease, termed
and rodents is low. Only for the most com- sporadic fatal insomnia, was described. Spo-
mon GSS-associated mutations (P102L), and radic FFI is not associated with mutations in
only in approximately one third of the cases, the PRNP gene (Mastrianni et al. 1999, Parchi
were brain homogenates derived from patients et al. 1999).
reproducibly capable of inducing disease upon
transmission (Tateishi et al. 1996a). The less
frequent mutations causing GSS often failed The Nature of the Prion
to induce disease after experimental transmis- Although formulated a century ago, Koch’s pos-
sion to nonhuman primates and rodents, and in tulates remain the bedrock of microbiology. Ac-
many cases transmissibility was never assessed cording to Koch, three conditions must be met
(Brown et al. 1994, Tateishi et al. 1996a). to identify a microbe as the causative agent
of any given infection: (a) The microorganism
Fatal familial insomnia. Fatal familial insom- must be detectable in all diseased tissues, (b) its
nia (FFI) is the descriptive name given to a dis- isolation and growth must be achieved in pure
ease identified in 1986. Five members of an Ital- culture, and (c) the culture-derived microorgan-
ian family presented with insomnia and dysau- isms must be able to induce disease after ex-
tonomia (Lugaresi et al. 1986). In 1992, the perimental infection of a subject, from which

www.annualreviews.org • The Prion’s Elusive Reason for Being 445


a further round of reisolation of the microor- blood and already in a presymptomatic disease
ganism should be possible. Although Koch’s state (Castilla et al. 2005b, Saa et al. 2006).
work was performed long before contemporary The use of purified PrPC instead of brain ho-
PMCA: protein
misfolding cyclic molecular biology, his postulates continue to mogenate as a substrate decreased the efficiency
amplification serve remarkably well in defining conventional of amplification, suggesting that additional co-
viral and bacterial agents. factors may facilitate misfolding (Deleault et al.
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

However, as prions are thought to be in- 2005). For a long time, all attempts to use re-
fectious proteins that amplify in a self-catalytic combinant PrP as a substrate for PMCA failed.
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

misfolding process, their microbiological cul- However, Caughey and coworkers have now
ture sensu strictiori is not possible. Therefore succeeded in carrying out PMCA using bacteri-
whether Koch’s postulates can be meaningfully ally expressed hamster PrP as a substrate. While
applied to prion disease is questionable. Fur- this represents a major advance in many ways,
thermore, Koch’s postulates account for the in- the sensitivity was not quite as high as that of
fluence of host susceptibility, which is of utmost the original PMCA (Aguzzi 2007, Atarashi et al.
importance in prion disease. Prion disease de- 2007).
velopment depends on the presence of PrPC Infectivity may not have been generated
on host cells, and the species-specific amino de novo in PMCA in these studies. Instead,
acid sequence and polymorphism of codon 129 prion-infected brain could have been inad-
are important. Alternate postulates for infec- vertently added in the beginning. In an fas-
tious proteinaceous agents have recently been cinating study, Supattapone and coworkers
suggested (Walker et al. 2006), but it remains identified the minimal components (PrPC , cop-
to be seen whether they will garner universal urified lipids, and single-stranded polyanionic
acceptance. molecules) required for amplification of PK-
In the prion field, researchers generally ac- resistant PrP, and they convincingly showed
cept that a reasonable surrogate for Koch’s sec- that prion infectivity can be generated de novo
ond postulate be fulfilled by the generation of in brain homogenates derived from healthy
synthetic prions in vitro, i.e., the recovery of hamsters using PMCA. Inoculation of further
perpetually transmissible infectivity from prion healthy hamsters with the de novo–formed
protein produced recombinantly or chemically prions caused a transmissible prion disease
from defined constituents. Major progress to- (Deleault et al. 2007). This study might be re-
ward this end has been made in recent days. Pu- garded as the final proof of the prion hypoth-
rified PrPSc was used to generate PK-resistant esis. However, it also acknowledges PMCA’s
PrP (PrPres ) in a cell-free system that could even limitation for diagnostic purposes because PK-
reflect two typical features of prions: species resistant material and infectivity can be formed
barrier and strain specificity (Bessen et al. 1995, in the absence of prions, thereby risking the re-
Kocisko et al. 1995). porting of false positive results.
Another approach used a method called A second approach comprises de novo
PMCA (protein misfolding cyclic amplifica- generation of infectivity by misfolding re-
tion), in which PrPres can be amplified by combinant PrPC and subsequently inoculating
incubating and sonicating PrPres -containing wild-type animals. In one attempt, a 55-residue
brain homogenate diluted in normal brain ho- peptide encompassing the GSS mutation
mogenate. Soto and coworkers amplified PrPres P101L was refolded in vitro to a beta-sheet
derived from scrapie-infected hamsters indefi- rich peptide and could induce disease similar
nitely by using PMCA in serial dilutions. Am- to GSS in transgenic mice expressing PrP
plification of PrPres was accompanied by am- (P101L). Transmission to wild-type mice was
plification of infectivity (Castilla et al. 2005a). not successful, and PrP (P101L) was not resis-
Certainly PMCA is a very sensitive method to tant to PK. Because transgenic mice expressing
detect PrPSc even in complex samples such as PrP (P101L) develop disease spontaneously,

446 Aguzzi · Baumann · Bremer


although later in life than those exposed to the mice expressing deletion mutants of PrPC de-
peptide, Nazor et al. (2005) remarked that the velop severe neurotoxic syndromes and identi-
misfolded peptide may have simply accelerated fies the reasons why we believe that study of
HC: hydrophobic
a spontaneously occurring disease. these syndromes may reveal the mechanisms core
Transmission to wild-type mice of an in operative in prion diseases.
vitro–generated misfolded part of the prion
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

protein (amino acid residues 89–231) was


achieved a few years later. Legname and PHYSIOLOGICAL FUNCTION OF
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

coworkers produced PrP (89–231) recombi- THE CELLULAR PRION PROTEIN


nantly and generated amyloid fibrils in vitro. The cellular prion protein PrPC is a GPI-
These fibrils induced prion disease in trans- linked extracellular membrane protein with two
genic mice overexpressing PrP (89–231), which N-linked complex glycosylation sites. PrPC is
was subsequently transmissible to wild-type highly abundant in the developing and ma-
mice (Legname et al. 2004, 2005). ture nervous system, where it is expressed by
neuronal and glial cells. This mature version
originates from a precursor protein proteolyt-
Neurotoxicity ically processed in the endoplasmic reticulum
Current knowledge about the mechanisms be- and Golgi (Stahl et al. 1987). As revealed by its
hind neurodegeneration in prion disease and atomic structure, the mature PrPC protein con-
prionopathies is limited. Apoptosis and oxida- tains a well-defined carboxy-terminal globular
tive stress certainly contribute to some stages of domain comprising residues 127–231 (murine
TSE pathology (Milhavet & Lehmann 2002), numbering), consisting of three alpha helices
but little is known about damage causing pri- and two beta sheets (Hornemann et al. 1997;
mary events. Early pathologic changes that oc- Riek et al. 1996) and a structurally less-defined
cur during prion disease involve synapses, yet amino proximal region containing a stretch of
the molecular underpinnings of these findings several octapeptide repeats, termed the OR,
remain unknown. and framed by two positively charged charge
It is still unclear whether the toxicity of clusters, CC1 (aa 23–27) and CC2 (aa 95–110).
PrPSc represents a gain of function or whether These domains are linked by a hydrophobic
loss of function of PrPC is responsible for neu- stretch of amino acids [aa 111–134, also termed
ropathological changes induced by prions. Al- hydrophobic core (HC)] (Figure 3).
though some authors belief that the toxicity in
prion disease is explainable simply by a loss-
of function of PrPC (Nazor et al. 2007), we Prion Protein–Deficient Mice
and others (Westergard et al. 2007) believe a An astonishing number of independent lines
gain of function is more likely, particularly be- of mice lacking PrPC have been generated by
cause the phenotypes of PrPC -deficient mice homologous recombination in embryonic stem
are very mild. However, a neuroprotective func- cells in many laboratories. Mice with disrup-
tion that may be physiologically provided by tive modifications restricted to the open reading
PrPC , which would protect neurons during frame are known as Prnpo/o [Zürich I] (Bueler
prion infection, could be reduced following its et al. 1992) or Prnp−/− [Edinburgh] (Manson
conversion to PrPSc . et al. 1994). They developed normally, and no
Our laboratory, and many others, has pur- severe pathologies were observed later in life. As
sued the hypothesis that elucidating the phys- predicted by the protein-only hypothesis, these
iological function of PrPC might help re- mice were entirely resistant to prion infections
searchers understand the mechanism involved (Bueler et al. 1993).
in prion-induced neurodegeneration. The fol- In contrast with these earliest lines, three
lowing discussion centers on the discovery that lines generated afterwards: Prnp−/− [Nagasaki],

www.annualreviews.org • The Prion’s Elusive Reason for Being 447


Rcm0, and Prnp−/− [Zürich II] (Moore et al. transgene, the originators of the Nagasaki
1999, Rossi et al. 2001, Sakaguchi et al. mice concluded that it occurred because of
1996) developed ataxia and Purkinje cell loss the lack of PrPC . This, however, would
later in life. Because the phenotype was run counter to the lack of pathology in
abolished by reintroduction of Prnp as a Prnpo/o Zürich-I mice.
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Pheno- Trans- Prion Rescue


SP CC1 OR CC2 HC H1 H2 H3 GPI type mission propagation by PrP Refs.
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

PrPwt S S

PrP∆32–80 no – yes – a

PrP∆32–93 no – yes – b;c

PrP∆32–106 no – no – b;d

PrP∆23–88 no – yes* – e

PrP∆23–88 no – no – e
∆95–107
PrP∆23–88 no – no – e
∆108–121
PrP∆23–88 no – no – e
C178A A S
PrP∆23–88 no – yes – e
∆141–176
PrP∆114–121 no – no** – f;g

PrP∆104–114 no – yes – h

PrP231 # no – yes – i

PrP∆1–22 cerebellar
disorder no no no j
231 #
cerebellar no no
PrP∆32–121 disorder yes b

cerebellar no no yes b;d


PrP∆32–134 disorder
cerebellar no nd yes f
PrP∆94–134 disorder
cerebellar
PrP∆105–125 disorder no nd yes k

PrP∆23–88 storage
disease no no no e
∆177–200
PrP∆23–88 storage no no no e
∆201–217 disease
PrP∆23–88 cerebellar no no no e
∆141–221 disorder

cerebellar no yes no l
PrP PG 14 disorder

PrP∆23–88 nd – – – e
∆122–140
PrP∆23–88 nd – – – e
144 #

PrP 144 # nd – – – e

0 23 32 80 90 107 121 134 177 200 217 231

448 Aguzzi · Baumann · Bremer


The discrepancy between the different ity (Behrens et al. 2002), suggesting that the pri-
lines of PrP knockout mice was not resolved mary physiological function of Dpl is related to
until a novel gene (Prnd ), encoding a protein sperm maturation.
called Doppel (Dpl), was discovered. Prnd is
localized 16 kb downstream of Prnp. In all
three lines of PrPC -deficient mice developing Functional Domains
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

ataxia and Purkinje cell loss, a splice acceptor of the Prion Protein
site to the third exon of Prnp was deleted. This For all the uncertainties surrounding the physi-
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

placed Prnd under transcriptional control of ological and molecular functions of PrPC , some
the Prnp promoter, resulting in the formation knowledge was generated by expressing a series
of chimeric transcripts and in overexpression of partially deleted Prnp variants in cultured
of Dpl in the brain (Moore et al. 1999, Rossi cells and transgenic mice. Some of these mu-
et al. 2001, Sakaguchi et al. 1996). Precisely tants were made to identify the essential do-
why the overexpression of Dpl is deleterious mains necessary for restoring prion susceptibil-
is still unclear. On the basis of the observation ity. However, investigators found that domain
that Dpl expression induced heme oxygenase expression provoked spontaneous neurodegen-
1 (HO-1) and neuronal and inducible nitric erative disease (Figure 3). In many instances,
oxide synthases (nNOS and iNOS), suggesting these syndromes were partially or fully coun-
an increased oxidative stress in the brains teracted by coexpression of wild-type PrPC . Be-
of the Dpl-expressing Prnpo/o mice, Wong cause the lack of PrPC itself did not induce an
et al. (2001c) proposed that Dpl expression obvious phenotype, the latter pathologies indi-
exacerbates oxidative damage by antagonizing cate pathways in which PrPC is functionally ac-
wild-type PrPC ’s antioxidative function. tive. Hence mice expressing PrP, which lacks
The latency period before the various trans- defined domains, may allow for the identifi-
genic mice overexpressing Dpl develop patho- cation of functionally relevant domains within
logical phenotypes is inversely correlated to the PrPC .
Dpl expression level in the brain, indicating
a rather strict gene-dosage effect (Rossi et al. N-terminal deletion mutants of PrP. The
2001). The Dpl-induced disease can be rescued OR has long been suspected to represent a ma-
by coexpression of wild-type PrPC (Nishida jor mediator of PrPC ’s function, and insertion
et al. 1999, Rossi et al. 2001), indicating that mutations affecting the OR are associated with
toxicity of Dpl and the physiological function hereditary human prion disease. However,
of wild-type PrPC are not independent of each transgenic studies indicate that the OR is
other, but rather are involved in a common not required for PrPC to function or for its
pathway. Dpl-deficient mice suffer from steril- convertibility into PrPSc (Flechsig et al. 2000).
←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−
Figure 3
Murine PrPC protein and transgenic mutant PrP. Schematic drawing of full-length murine PrPC , including
the signal peptide of the precursor protein (SP; brown box). Although amino acid numbering differs between
human and mouse PrP, the organization of domains (including CC1 and CC2 , OR, HC, and H1–H3) is
similar to that of human PrPC (see Figure 2). Mouse PrP also contains a disulphide bond (S–S) and a
GPI-anchor. The left column denotes the individual mutants described in the text. The right columns
indicate presence or absence of phenotypic abnormalities (Phenotype) in transgenic mice when expressed on
a PrP-deficient genetic background, transmissibility of this phenotype to recipient mice (Transmission), and
susceptibility of transgenic mice to prions after intracerebral inoculation with a mouse-adapted strain of
scrapie prions. References: a, Fischer et al. (1996); b, Shmerling et al. (1998); c, Flechsig et al. (2000);
d, E. Flechsig, I. Hegyi, A. Aguzzi, and C. Weissman (unpublished results); e, Muramoto et al. (1997);
f, Baumann et al. (2007); g, Holscher et al. (1998); h, Hegde et al. (1998); i, Chesebro et al. (2005);
j, Ma et al. (2002); k, Li et al. (2007); l, Chiesa et al. (1998).

www.annualreviews.org • The Prion’s Elusive Reason for Being 449


The OR appears to have, at best, a modulating HC (PrP!104−114 ) (Hegde et al. 1998), abla-
influence on PrP conversion. Mice expressing tion of CC2 in combination with a partial or
OR-deficient PrPC mutants do not develop complete deletion of HC elicits severe patholo-
pathologies (Fischer et al. 1996, Muramoto gies in mice. PrP!32−121 and PrP!32−134 trans-
et al. 1997, Shmerling et al. 1998). This was genic mice suffer from ataxia and cerebellar
unexpected because a variety of in vitro data granule cell loss in addition to widespread
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

had identified the OR as being responsible for white matter disease (Radovanovic et al. 2005,
copper binding (Aronoff-Spencer et al. 2000, Shmerling et al. 1998). The latter is also seen
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Chattopadhyay et al. 2005, Furlan et al. 2007, in mice expressing deletions encompassing all
Leclerc et al. 2006, Qin et al. 2002, Stockel (PrP!94−134 ) or part (PrP!105−125 ) of the central
et al. 1998; reviewed in Vassallo & Herms 2003) domain (CD) (Baumann et al. 2007, Li et al.
and for conferring protection against oxidative 2007). These pathologies are radically different
stress (Brown et al. 1999, Fukuuchi et al. 2006, from those seen in prion infections, and none of
White et al. 1999, Wong et al. 2001a). On the them goes along with pathological aggregation
other hand, transgenic mice expressing nine of PrP.
supernumerary octapeptide repeats, for a total Each of these pathologies can be coun-
of 14 proline and glycine-rich repeats (Chiesa teracted by coexpression of wild-type PrPC
et al. 1998)—which models a human familial (Baumann et al. 2007, Li et al. 2007, Shmer-
CJD-linked mutation—develop ataxia and ling et al. 1998), suggesting a competition of
cerebellar atrophy, granule cell loss, gliosis, sorts between PrPC and the toxic mutants. In
progressive myopathy, and PrP deposition. one conceivable scenario, PrPC and its variants
The latter phenotype resembles its human may compete for a common ligand. Binding or
counterpart in some ways (Chiesa et al. 2000), complex assembly may represent the first step
yet transmission to wild-type mice failed in a series of events that also involve the inter-
(Chiesa et al. 2003). action of an effector domain located in or con-
In vitro studies indicate that the CC1 region trolled by the central domain (CC2 and HC),
is involved in recycling and internalizing PrPC eventually resulting in signal transduction.
from the cell surface (Sunyach et al. 2003, A partial deletion of HC (PrP!114−121 )
Taylor et al. 2005). Unfortunately, in vivo (Baumann et al. 2007) is nontoxic, but its po-
little evidence supports the latter contention. tential to counteract the toxicity of PrP!32−134
Lack of CC1 in (PrP!23−88 ) (Muramoto et al. is lower than that of wild-type PrPC . Mice
1997) did not induce pathologies in transgenic with deletion of CC2 and HC (PrP!32−121 and
mice, and convertibility to PrPSc was retained. PrP!32−134 as well as PrP!104−114 , PrP!114−121 )
In PrP!23−88 mice, a second charge cluster did not support prion propagation (Flechsig &
(CC2 ) with several lysine residues around Weissmann 2004; Hegde et al. 1998, Holscher
position 100 may replace the function of CC1 . et al. 1998), indicating an involvement of these
However, mice bearing partial deletions of regions in conversion.
CC2 (PrP!23−88 !95−107 and PrP!23−88 !108−121)
are also healthy (Muramoto et al. 1997). Carboxy-proximal deletion mutants of PrP.
The combination of amino-terminal deletion Mice expressing PrP mutants with deletions
with the elimination of amino acids 141–176 affecting Helix 2 (PrP!23−88 !177−200 ), Helix
(PrP!23−88 !141−176 ) was also innocuous and 3 (PrP!23−88 !201−217 ), or both helices 2 and
restored susceptibility to prion infection 3 (PrP!23−88 !141−221 ) suffer from ataxia and
(Muramoto et al. 1996) despite a large deletion present with features of neuronal storage dis-
within the globular domain of PrPC . ease (Muramoto et al. 1997, Supattapone et al.
The function of PrPC may depend on the 2001) but fail to replicate prions (Muramoto
HC region in concert with CC2 . With the ex- et al. 1996). Obviously at least Helix 2 and Helix
ception of a small deletion between CC2 and 3 are indispensable for stabilizing the structure

450 Aguzzi · Baumann · Bremer


of PrPC . None of these diseases proved to be ataxia with cerebellar degeneration and glio-
transmissible to normal wild-type mice, and sis (Ma et al. 2002). Coexpression of wild-type
they all manifested themselves independently PrPC did not influence the phenotypes of these
of the presence or absence of wild-type PrP. mice. Whether cytoplasmic expression of PrP
This stands in sharp contrast to the group of and its cytotoxicity represent realistic models
deletion mutants affecting CC2 and HC. of the events occurring during prion disease re-
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Several attempts to generate mice express- mains very hotly debated (Fioriti et al. 2005,
ing truncated carboxy-terminal mutants lack- Roucou et al. 2003).
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

ing membrane anchoring (PrP!23−88 144# and


PrP144# ) have failed (Fischer et al. 1996,
Muramoto et al. 1997). As in the case of
Point Mutations within
PrP!23−88 !122−140 (Muramoto et al. 1997), lines
the Prion Protein
expressing high levels of mRNA were gener- As previously described, a considerable set of
ated but protein was never detected. Essential point mutations within PRNP has been linked
signals may have been lost, thereby preventing to various forms of human prion diseases.
correct sorting, processing, or folding of PrP Some of these mutations have been expressed
and resulting in a short-lived polypeptide. in mice. With the possible exception of some
strains of mice expressing the P101L vari-
Mutations affecting the localization of ant of PrPC (Hsiao et al. 1994, Telling et al.
PrPC . The affinity of GPI-linked PrPC for 1996), none of these attempts succeeded in
artificial membranes, as measured by surface reproducing the infectiousness of bona fide
plasmon resonance (Elfrink et al. 2007), sug- prions. Point mutations affecting the two N-
gests extremely strong interactions. One might linked glycosylation sites of PrPC proved, as ex-
therefore expect that most PrPC is attached to pected, to alter its glycosylation (Kiachopoulos
cell membranes, with perhaps traces of PrPC et al. 2005). Point mutations N182T, A198T,
floating in body fluids. Thus it may come as or N182T/A198T prevented glycosylation in
a surprise that plasma contains conspicuous transgenic mice without grossly affecting cellu-
amounts of PrPC (Volkel et al. 2001), and the lar sorting in cell culture. Mice developed nor-
concentration of PrPC in cerebrospinal fluid mally and were readily susceptible to scrapie
is even higher (Castagna et al. 2002). How- or BSE (Neuendorf et al. 2004). Knock-in mu-
ever, it is unclear whether this soluble PrPC tants carrying either N180T or N196T, or
is chemically identical to its membrane-bound both mutations, (Cancellotti et al. 2005) did
isoform. Treatment of cultured cells with phos- not suffer from any constitutive phenotype,
phatidylinositol phospholipase C efficiently re- even if the complete blockade of glycosylation
leases PrPC from cultured cell membranes by the N180T/N196T double mutation led
(Stahl et al. 1987), and a similar mechanism may to a mainly intracellular localization of PrPC .
underlie the physiological shedding of PrPC This finding is somewhat surprising because re-
into body fluids. sults from cultured cells had predicted that un-
Release of full-length secreted PrPC was glycosylated PrPC would be prone to sponta-
forced by deletion of its carboxy terminal hy- neous aggregation (Korth et al. 2000, Priola &
drophobic domain (PrP231# ), which is normally Lawson 2001).
replaced by a GPI-anchor. This manipulation Two sets of point mutations, PrP3AV (ex-
did not induce any pathological phenotype change of alanine to valine at positions 113,
(Chesebro et al. 2005). In contrast, targeting 115, and 118) (Prusiner & Scott 1997) and
PrPC to the cytosol (cyPrP = PrP!1−22 231# ) PrPKHII (exchange of lysine 109 and histidine
by deleting its amino-terminal leader peptide 110 for isoleucine) (Hegde et al. 1998), gener-
(which targets PrPC to the endoplasmic retic- ated PrP with altered topology, termed Ctm PrP
ulum and to the secretory pathway) provoked in a cell free assay. Ctm PrP supposedly spans

www.annualreviews.org • The Prion’s Elusive Reason for Being 451


the membrane via the HC domain (Hegde amino-terminal tail, with a positively charged
et al. 1998). Transgenic mice expressing these CC1 at its far end and repetitive domains
proteins developed a fatal neurological disor- of variable numbers, is hooked to a globu-
der (Hegde et al. 1998). A similar phenotype lar carboxy-terminal domain. The fold of this
was observed in transgenic mice with substi- domain is strongly conserved and stabilized
tution of leucine 9 into arginine in addition by a disulfide bridge, although the primary
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

to this 3AV mutation (Stewart et al. 2005, sequence shows considerable diversity. These
Stewart & Harris 2005). However, research two domains are linked by a highly conserved
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

never formally proved that Ctm PrP exists in vivo. hydrophobic linker having a second positive-
It is still noteworthy that coexpression of wild- charge cluster CC2 at its amino-terminus. This
type PrPC with mutants promoting the Ctm PrP linker region is by far the most conserved se-
topology aggravated their phenotype. Subtle quence motive of PrP in all species.
changes, such as the removal of disulfide bridges
(PrP!23−88 C178A ), are tolerated without induc-
Cellular Processes Influenced
ing a spontaneous phenotype though reduc-
by PrPC Expression
ing the susceptibility for conversion into PrPSc
(Muramoto et al. 1997). Several cellular processes in the nervous system
have been influenced by the Prnp-genotype, in-
cluding neuronal survival; neurite outgrowth;
Evolution of the Prion Protein synapse formation, maintenance, and func-
PrP is present in a broad variety of species tion; and maintenance of myelinated fibers
(Figure 4). Genes with similarities to Prnp exist (Figure 5).
in birds (Gabriel et al. 1992), reptiles (Simonic One of the most frequently suggested cel-
et al. 2000), amphibians (Strumbo et al. 2001), lular functions of PrPC is a survival-promoting
and possibly in fish (Favre-Krey et al. 2007, effect on neuronal and nonneuronal cells, which
Oidtmann et al. 2003, Rivera-Milla et al. 2003, has been observed in vitro as well as in in vivo
Suzuki et al. 2002) in addition to all mammals. studies.
However, more primitive organisms such as in- This neuroprotective function, or cytopro-
sects, cephalopods, and protozoa have not been tective function in general reviewed in Roucou
reported to contain PrP homologs. All PrPs & LeBlanc (2005), has been mediated by anti-
are glycosylated and membrane attached by a apoptotic or antioxidative mechanisms.
GPI anchor. The sequence identity among the
known PrP homologs is limited, and protein Antiapoptotic function. Neurons derived
length can vary between ∼250 amino acids in from Prnp−/− mice were originally reported to
tetrapods to ∼600 amino acids in fish. Fish be more susceptible to the induction of apop-
may have developed additional Prnp-like genes tosis by serum-deprivation than were cells ex-
(Rivera-Milla et al. 2006). The putative fish PrP pressing PrPC (Kuwahara et al. 1999), but this
genes are thus far identified only on the basis of effect may have been brought about by Dpl
rather tenuous sequence similarities. The con- overexpression rather than by PrPC ablation.
tention that these molecules indeed represent However, several studies indicate that PrPC
paralogs of PrPC would be greatly strengthened has a cytoprotective function by decreasing
if knockdown-induced phenotypes of zebrafish the rate of apoptosis after particular apoptotic
would be functionally corrected by mammalian stimuli such as Bax overexpression or TNF-α.
PrPC expression. Such experiments have not Bax overexpression induces apoptosis in hu-
been reported. man neuronal cells. Coexpression of wild-type
Comparisons between the available struc- PrPC , but not of PrP lacking the octarepeats,
tures and molecular models suggest that all reversed the Bax-mediated induction of apop-
PrPs share a common blueprint. A flexible tosis (Bounhar et al. 2001).

452 Aguzzi · Baumann · Bremer


The presence of PrP in the cytosol, be it brain, although the genetic homogeneity of the
due to reverse translocation from the endoplas- mice tested in the latter experiment was not
mic reticulum or through direct cytosolic ex- controlled for.
pression, was virulently neurotoxic (Ma et al.
2002). However, other studies failed to con- Protection against oxidative stress. Besides
firm the toxicity of cytosolic PrP and claimed its possible antiapoptotic function, there are
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

that it can instead protect against Bax-mediated many reports about an antioxidative effect of
apoptosis in human primary neurons (Roucou PrPC . These two effects are not necessarily mu-
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

et al. 2003). In this context, PrPC inhibited tually exclusive. Oxidative stress may be in-
the proapoptotic conformational change of Bax volved in TSE pathogenesis. However, one
and cytochrome c release from mitochondria must remember that oxidative stress is very un-
(Roucou et al. 2005). specific and is seen in different kinds of damage
In a screening approach for proteins pro- to the nervous system with impaired mitochon-
tecting cancer cells from apoptosis, researchers drial function such as defects in the ubiquitin-
investigated the gene-expression profile in an proteasome system, protein aggregation, and
established cell clone of MCF-7 breast cancer inflammation.
cell line resistant to TNFα-induced apoptosis. Many investigators believe that the main
PrPC was overexpressed 17-fold. Conversely, function of PrPC consists of protecting against
overexpression of PrPC converted MCF-7 cells oxidative stress (see Milhavet & Lehmann 2002
sensitive to TNFα-induced apoptosis into re- for a review). First hints came from in vitro
sistant cells (Diarra-Mehrpour et al. 2004). studies of rat pheochromocytoma cells. Those
The neuroprotective function of PrPC in selected for resistance to copper toxicity or
the postischemic rodent brain has been inten- oxidative stress showed higher levels of PrPC
sively studied. Levels of PrPC after ischemia (Brown et al. 1997a). Primary neuronal cells
were increased compared with controls (Shyu lacking PrPC were more susceptible to hydro-
et al. 2005, Weise et al. 2004). Moreover, gen peroxide (H2 O2 ) than were wild-type cells.
adenovirus-mediated overexpression of PrPC The increased peroxide toxicity went along with
reduced infarct size in rat brain and improved a significant decrease in glutathione reductase
neurological behavior after cerebral ischemia activity measured in PrPC -deficient neurons
(Shyu et al. 2005). Conversely, in a mouse (White et al. 1999). Also, PrPC -deficient pri-
model of ischemic brain injury Prnpo/o mice mary neurons were more susceptible to treat-
displayed significantly increased infarct vol- ment with agents inducing oxidative stress com-
umes when compared with wild-type mice pared with wild-type cells, a phenomenon that
(McLennan et al. 2004, Weise et al. 2006). Two was explained by a reduced Cu/Zn superox-
groups of researchers showed that mice lacking ide dismutase (SOD) activity observed in vivo
PrPC had enhanced postischemic caspase-3 ac- (Brown et al. 1997b, 2002). Higher levels of ox-
tivation (Spudich et al. 2005, Weise et al. 2006). idative damage to proteins and lipids were iden-
An increase in Erk-1/-2, STAT-1, and JNK- tified in the brain lysates derived from Prnp−/−
1/-2 phosphorylation and activation was iden- compared with wild-type mice (Klamt et al.
tified, suggesting PrPC ’s possible involvement 2001, Wong et al. 2001b).
in cellular signaling (Spudich et al. 2005). Also, PrPC itself could have SOD activity and
a reduced amount of phospho-Akt in the gray thereby mediate the antioxidative function
matter suggested that PrPC deficiency brings (Brown et al. 1999). However, there is sig-
about an impairment of the antiapoptotic phos- nificant controversy about this alleged SOD
phatidylinositol 3-kinase/Akt pathway (Weise activity. Others, ourselves included, failed to
et al. 2006). Finally, Mitteregger et al. (2007) confirm this proposed SOD activity in vitro
claimed that the OR is required within PrPC (Jones et al. 2005) and in vivo (Hutter et al.
for the neuroprotection in the ischemic mouse 2003). Furthermore, PrPC expression level did

www.annualreviews.org • The Prion’s Elusive Reason for Being 453


a Tetrapod PrP Fish PrP
N N T S R H N N T
S R H Zeb. Prp1
Hum. Prp
253 aa 606 aa
S--S S--S
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

S R H N N T S R H N T
Chi. Prp Fug. Prp1
273 aa 461 aa
S--S S--S
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

S R H N N T S R H N T
Tur. Prp Sal. Prp
270 aa 605 aa
S--S S--S

S R H N N T S R H NN T
Xen. Prp Zeb Prp2
216 aa 567 aa
S--S S--S

S R H H N T
Fug. Prp2
435 aa
S--S

b 4.0 Deer
Sheep
3.2 Multiple alignment Cow
Camel
2.4 Rabbit
Bat
1.6 Chimpanzee
Hydrophobicity

Human
0.8
Mouse
Sigmodon
0
Pigeon
Quail
–0.8
Chicken
Duck
–1.6
Turtle
Zebrafish_prp1
–2.4
Trout_prp1
Carp_prp1
–3.2
Zebrafish_prp2
Frog
–4.0
1 165 330 494 658
Position
Figure 4
PrP structural diversity in vertebrates. (a) Schematic drawing of tetrapod PrPs and long (PrP1 and PrP2) fish PrPs. The species
abbreviations refer to sequences from human (Hum), chicken (Chi), turtle (Tur), Xenopus (Xen), zebrafish (Zeb), salmon (Sal), and
Fugu (Fug). The location and relative size of conserved structural features are indicated. However, these features were physically
determined for the structure of human PrPC and represent mere conjectures in the case of fish. Domains are indicated by different
boxes and/or letters: S, signal peptide sequence; R, repetitive region; H, hydrophobic region; S—S, disulfide bridge; N, glycosylation
site; arrow, GPI anchor residue; and T, hydrophobic tail. (b) Comparison of hydrophobicity plots. Sequences of indicated species were
aligned using DNAMAN software (Lynnon BioSoft, Canada), and a hydrophobicity plot was generated using a window of nine
amino-acid residues. Numbering of residues is according to alignment matrix. (c) 3-dimensional structures of human (hum based on
1QM2.pdb model) chicken (chi based on 1U3M.pdb) turtle (tur based on 1U5L.pdb), and frog (fro based on 1XOU.pdb); pdb files are
from the protein database (Berman et al. 2000). Note the similarity of the carboxy-terminal globular domain. (d ) Evolutionary
relationships among vertebrate PrP sequences are based on distance methods (neighbor-joining). Bootstrap values are shown at relevant
nodes using DNAMAN software (Lynnon BioSoft, Canada).

454 Aguzzi · Baumann · Bremer


not significantly influence SOD activity in Role of PrPC in synapses. Synapses have de-
vivo (Hutter et al. 2003, Waggoner et al. veloped into a sort of hot spot in prion research.
2000). Several immuno-electron microscopy studies
Mitochondria play an important role not could show that PrPC is localized in synap-
only in oxidative stress but also in the induction tic boutons, whereas it is mainly presynaptic
of apoptosis. Morphological alterations in mi- (Fournier et al. 1995, Moya et al. 2000, Sales
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

tochondria have been described in scrapie- et al. 1998, Tateishi et al. 1996b). However,
infected hamsters (Choi et al. 1998) and mice others described a much broader distribution of
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

(Lee et al. 1999) as well as in mice lacking PrP, neuronal PrPC (Laine et al. 2001, Mironov et al.
in which the number of mitochondria was re- 2003). Because PrPC is processed and broken
duced (Miele et al. 2002). down into various fragments, not all of which

PrPhum PrPchi PrPtur PrPfro

Deer
d 92Cow
Sheep
Camel
Bat
0.05 100 Chimpanzee
Human

Tetrapod
95 Mouse
100 Sigmodon

100
Rabbit
98 Pigeon
100 Quail
100 Chicken
92
Duck
Turtle
Frog

100 Zebrafish_prp1
Trout_prp1
Fish

100 Carp_prp1
100 Zebrafish_prp2

Figure 4
(Continued )

www.annualreviews.org • The Prion’s Elusive Reason for Being 455


Synapse
Neurite outgrowth • Formation
• Dendrites • Function
• Axons

Maintenance of
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

myelinated axons
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Neuronal survival
• Protection against apoptosis
• Protection against oxidative stress

Figure 5
Physiological processes involving PrPC . Several processes in the nervous system have been influenced by PrPC . Neurite outgrowth,
including growth of axons and dendrites, was observed to be reduced in neurons lacking PrPC . PrPC has often been reported to
promote neuronal survival, in particular following apoptotic or oxidative stress. Cerebellar granule cell apoptosis was observed in mice
expressing toxic N-terminal deletion mutants of PrP. In addition, the latter transgenic mice show an impaired maintenance of
myelinated axons in the white matter. Another site of PrPC action might be the synapse, which is often affected in the first stage of
prion diseases and whose formation was found to be reduced in neuronal cultures devoid of PrPC . Furthermore, electrophysiological
studies indicate a role of PrPC in synapse function, especially in neurotransmitter release.

are recognized by the antibodies used in these the above evidence, however, it should not go
studies, one might speculate that some PrPC undiscussed that synaptic changes can represent
degradation products acquire distinct subcellu- nonspecific phenomena that are seen in essen-
lar topologies. tially all brain diseases at one stage or another.
Early pathologic changes occurring in prion The generally held view that PrPC is an
diseases involve synapse loss and PrPSc depo- important protein in synapses is supported
sition in synaptic terminals (Grigoriev et al. by electrophysiological studies of CA1 hip-
1999, Jeffrey et al. 2000, Kitamoto et al. 1992, pocampal neurons derived from PrPC -deficient
Matsuda et al. 1999, Roikhel et al. 1983). Synap- mice. Excitatory glutamatergic synaptic trans-
tic vesicle proteins associated with exosomes mission, GABAA receptor–mediated fast inhibi-
and neurotransmission are reduced in brains tion, long-term potentiation, and late afterhy-
of patients with spongiform encephalopathy perpolarization were reduced or absent in mice
(Ferrer et al. 1999). Synaptic disorganization lacking PrPC (Carleton et al. 2001; Colling
and loss are fundamental and constant fea- et al. 1994, 1996; Mallucci et al. 2002). Some of
tures of prion disease, irrespective of the pres- the findings could be explained by alterations
ence or absence of spongiform change, neu- in Ca-activated K+ currents (Colling et al.
ronal loss, and severe gliosis (Clinton et al. 1996, Herms et al. 2001). However, the reader
1993). Abnormal electrophysiological record- should note that alterations in synaptic trans-
ings in scrapie-infected mouse and hamster hip- mission were not confirmed by others (Lledo
pocampal and cortical slices further support et al. 1996), and glutamatergic synaptic trans-
the synaptic dysfunction during the course of mission was even observed to be increased in
prion disease (Barrow et al. 1999, Johnston et al. PrPC -deficient mice by yet another laboratory
1998). In a terminal disease state, PrPSc accu- (Maglio et al. 2004, 2006). Another report in-
mulation in synaptosomes correlated with alter- dicates the impact of aging on these alterations
ations in the GABAergic system (Bouzamondo- describing a reduction in the level of postte-
Bernstein et al. 2004). Despite the wealth of tanic potentiation and long-term potentiation

456 Aguzzi · Baumann · Bremer


only in old PrPC -deficient mice (Curtis et al. dent on the recruitment of neural cell adhesion
2003). In summary, the impact of the loss of molecule (N-CAM) to lipid rafts (Santuccione
PrPC on hippocampal electrophysiological pa- et al. 2005). Recent studies show that PrPC pos-
rameters is still being hotly debated despite a itively regulates neural precursor proliferation
full decade of research efforts. Some of the dis- during development and adult mammalian neu-
crepancies may depend on additional genetic rogenesis (Steele et al. 2006).
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

modifiers for which investigators have not rig-


orously controlled.
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Maintenance of the white matter. Central


Other alterations in PrPC -deficient mice nervous system white matter, composed mainly
might be related to synaptic dysfunction of myelinated axons, might be disrupted in
such as altered circadian rhythms and sleep prion diseases and prionopathies. In some cases
(Tobler et al. 1996) and impaired hippocampal- of GSS, cerebellar and frontal white matter are
dependent spatial learning (Criado et al. 2005). affected (Itoh et al. 1994). In an experimental
The neuromuscular junction is another site model of human TSEs in rodents, vacuolation
where PrPC was concentrated, namely en- in myelinated fibers with splitting of myelin
riched in subsynaptic endosomes (Gohel et al. lamellae was observed (Walis et al. 2003). PrPC
1999). A potentiation of acetylcholine release is present in purified myelin fractions derived
from presynaptic axon terminals was observed from brain homogenates (Radovanovic et al.
after administration of recombinant PrP at 2005). Several transgenic mice expressing dele-
nanomolar concentrations to mouse phrenic- tion mutants of PrPC (Baumann et al. 2007, Li
diaphragm preparations (Re et al. 2006). The et al. 2007, Shmerling et al. 1998) as well as
suggestion of an involvement of PrPC in Prnp−/− mice accidentally overexpressing Dpl
synapse formation originated from in vitro ob- (Nishida et al. 1999) show vacuolation and de-
servations in hippocampal neurons, in which generation of myelinated fibers in the central
synaptic-like contacts were increased after addi- nervous system.
tion of recombinant PrP (Kanaani et al. 2005).
It is unknown whether the role of PrPC in
synapses is related to its above-mentioned an- PrP and the Immune System
tiapoptotic or antioxidative effects or whether The immune system plays a fundamental role in
it is mainly the involvement of PrPC in neu- prion disease and PrPC is expressed on cells of
rotransmitter release (e.g., via direct interac- the immune and hematopoietic system, where it
tion with synapsin1 and synaptophysin). How- might have a physiological function. This topic
ever, an as-yet-unidentified process could also is reviewed in depth in Isaacs et al. (2006). Also,
play a key role, or several processes could work Zhang et al. (2006) reported that PrPC is in-
together. volved in self-renewal of hematopoietic stem
cells.

Neurite outgrowth. Several lines of evidence


indicate PrPC ’s involvement in neuronal de- Molecular Mechanisms Mediating
velopment, differentiation, and neurite out- the Function of PrPC
growth. Axon or dendrite outgrowth was asso- Despite the overwhelming number of re-
ciated with PrPC -dependent activation of sig- ports about alterations in mice and cells lack-
nal transduction pathways including p59Fyn ki- ing PrPC summarized above, little is known
nase, cAMP/protein kinase A (PKA), protein about the molecular mechanisms involved in
kinase C (PKC), and MAP kinase activation these cellular processes. Figure 6 depicts some
(Chen et al. 2003, Kanaani et al. 2005, Lopes theoretical models of how PrPC might influ-
et al. 2005, Santuccione et al. 2005). P59Fyn ence cell signaling, endocytosis, and cell ad-
kinase activation in this context was depen- hesion. Whether these events are mutually

www.annualreviews.org • The Prion’s Elusive Reason for Being 457


exclusive, or whether they occur only under to mediate its function via one or more interac-
specific circumstances in a diversity of tissues, tion partners.
or whether they can act in a combined way, One explanation for the diversity of the sug-
remains speculative. In all cases, PrPC is likely gested physiological functions of this PrPC is
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

a Endocytosis of PrPc via Clathrin-coated pits or caveolae


Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

• Cointernalization of another
cell component
• Modulation of signaling pathways
• Degradation of PrPc
cointernalized TM proteins

b Interaction with TM protein in cis

• Modulation of signal transduction


pathways, e.g.activation of:
– Fyn kinase
– Erk1/2
– cAMP
– PKC

Lipid raft Non-raft region

c Interaction with TM protein in trans

• Modulation of signaling pathways


• Modulation of cellular adhesion

Lipid raft Non-raft region

458 Aguzzi · Baumann · Bremer


that it may exert pleiotropic effects, thereby seen in a hypothalamic cell line (Toni et al.
modulating the function of several cellular 2006).
pathways. Examples for such a general cellu- Several studies indicated PrPC involvement
lar process would be stabilization of protein in neurite outgrowth and neuronal survival.
complexes and the targeting of cell compo- Chen et al. reported increased neuronal survival
nents to certain cellular sites, such as rafts or and neurite outgrowth from neurons when cul-
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

endosomes. tured on Chinese hamster ovary (CHO) cells


transfected to express mouse PrP. Although
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

p59Fyn kinase activity in this context was in-


Signaling. The attachment of PrPC to the
volved mainly in neurite outgrowth, the PI3 ki-
membrane by a GPI anchor, its localization in
nase/Akt pathway as well as regulation of Bcl-2
detergent-resistant membranes, also known as
and Bax expression contributed to the survival
lipid rafts, in many cell types may suggest an in-
effect elicited by PrP. Cyclic AMP/protein ki-
volvement in cellular signaling (Shmerling et al.
nase A (PKA) and Erk signaling pathways con-
1998) as is the case for other raft-associated
tributed to both neurite outgrowth and neu-
proteins. Moreover, as we describe below, PrPC
ronal survival (Chen et al. 2003, Santuccione
could also influence cellular signaling events by
et al. 2005).
its involvement in endocytotic pathways.
Some investigators suggested that engage-
Several signaling pathways or signaling
ment of PrPC with stress-inducible protein
components, such as Akt, Fyn, cAMP, and
I (STI1) induces neuroprotective signals that
Erk1/2, are modulated by PrPC expression, its
rescue cells from apoptosis via cAMP/protein
cross-linking, or its interaction with another
kinase A and the Erk signaling pathways
protein. Antibody-mediated cross-linking of
(Chiarini et al. 2002, Zanata et al. 2002). In-
PrPC induced activation of the p59Fyn ki-
teraction with STI1 induced different signaling
nase, a family member of nonreceptor Src-
pathways, promoting neuroprotection by PKA
related kinases, in neuronal differentiated cells
activation and neuritogenesis by activation of
in a caveolin-1-dependent manner (Mouillet-
the MAPK pathway (Lopes et al. 2005).
Richard et al. 2000). As a downstream event,
the same group claims to have identified Erk1/2
phosphorylation (Schneider et al. 2003). PrPC Endocytosis and internalization of PrPC .
cross-linking additionally modulates seroton- PrPC is rapidly internalized from the cell
ergic receptor activity in differentiated neu- membrane. This internalization of PrPC could
ronal cells (Mouillet-Richard et al. 2005). The be crucial for its function, e.g., by influenc-
finding that PrPC cross-linking modulates ac- ing signal transduction pathways. Endocyto-
tivity of serotonergic receptors in differenti- sis of membrane receptors does not neces-
ated neuronal cells await replication and in sarily downregulate receptor activity. While
vivo confirmation. However, p59Fyn activation being internalized, both tyrosine kinase and
and downstream activation of Erk1/2 were also G protein–coupled receptors may remain

←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−
Figure 6
Models of how PrPC could exert its physiological function. (a) Endocytosis of PrPC via clathrin-
coated pits or caveolae may represent a mechanism for the downregulation of PrPC on the cell surface.
Alternatively, or additionally, endocytosis of PrPC leads to cointernalization of another cell component, e.g.,
a proteinaceous interacting partner, thereby regulating the presence of the latter on the cell surface. This
regulation could positively or negatively modulate the activity of signal transduction pathways, e.g., via
inducing degradation of the cointernalized partner. (b) An interaction with a transmembrane (TM) protein in
cis independent of an internalization process may lead to modulation of signal transduction pathways in the
cell carrying PrPC on its cell surface. (c) Similarly, an interaction with another protein in trans may lead to
modulation of signal-transduction pathways or adhesion to adjacent cells.

www.annualreviews.org • The Prion’s Elusive Reason for Being 459


activated and produce intracellular responses Glypican-1, a GPI-anchored heparan sulfate-
along the endosome-lysosome pathway (Prado containing cell-associated proteoglycan, inter-
et al. 2004). Internalization of tyrosine kinase acts and cointernalizes with PrPC in N2a after
receptors was functionally important in stud- induction with copper ions. In cells express-
ies: TrkA receptors, for example, mediate nerve ing PrP, which lacks the OR, internalization of
growth factor (NGF)-dependent cell survival glypican-1 is reduced, suggesting a possible role
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

while they are located at the cell membrane, for PrPC in the cointernalization of other cel-
whereas internalization is required for induc- lular components (Cheng et al. 2006).
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

tion of neurite outgrowth (York et al. 2000, PrPC might participate in the correct local-
Zhang et al. 2000). ization of some other proteins in lipid rafts.
The mechanism of PrPC internalization is Neuronal nitric oxide synthase (nNOS), for ex-
still controversial because both raft/caveolae or ample, involved in various nervous system pro-
caveolae-like (Kaneko et al. 1997, Marella et al. cesses such as development, synaptic plasticity,
2002, Peters et al. 2003, Vey et al. 1996) as well regeneration, and regulation of transmitter re-
as clathrin-dependent endocytosis may be op- lease was associated with lipid rafts in wild-type
erative (Shyng et al. 1994, Taylor et al. 2005). mice. In contrast, in brains of PrPC -deficient as
These mechanisms might as well be equally well as scrapie-infected mice, nNOS was not as-
important. In addition, internalization of the sociated with rafts, and activity of nNOS was re-
same ligand/receptor complex by distinct endo- duced. Therefore PrPC could be important for
cytotic pathways can result in different signal- the proper cellular localization of other proteins
ing outcomes. TGF-β receptor, for example, is (Keshet et al. 1999). Similarly N-CAM was re-
degraded after endocytosis via caveolae, but in- cruited to lipid rafts by PrPC (Santuccione et al.
ternalization via clathrin-coated pits promotes 2005).
its signaling (Di Guglielmo et al. 2003). How- However, PrPC is involved in a number of
ever, in lymphocytes and neuronal cells that do cellular functions and how endocytosis influ-
not express caveolin, internalization can occur ences them in vivo remains widely unknown; in-
in a lipid raft–associated noncaveolar, clathrin- ternalization of PrPC could contribute to down-
independent process (Kirkham & Parton 2005, regulation of a signaling event but could also be
Parton & Richards 2003). Therefore addi- necessary for signaling. A general involvement
tional, less well-characterized endocytosis path- of PrPC in vesicle formation could be a possible
ways including caveolae-like endocytosis might explanation for most of the suggested molecu-
be involved in internalization of PrPC . lar functions of PrPC because it could regulate
For endocytosis by clathrin-coated pits, signaling and influence synaptic transmission.
PrPC would need to leave the lipid rafts prior
to internalization because the rigid structure of PrP and cell adhesion. Several reports are
raft lipids is unlikely to accommodate the tight consistent with a possible function of PrPC
curvature of coated pits. This phenomenon as a cell adhesion or recognition molecule.
occurred after binding of copper to the OR Some interaction partners of PrPC identified
(Sunyach et al. 2003, Taylor et al. 2005), but so far have a role in adhesion, including laminin
its physiological significance is unknown. Low- (Graner et al. 2000a,b), a structural component
density lipoprotein receptor-related protein 1 of basement membrane, laminin-receptor pre-
(LRP1) was later shown to mediate PrPC en- cursor (Gauczynski et al. 2001, Rieger et al.
docytosis (Taylor & Hooper 2007), and CC1 1997), and N-CAM (Schmitt-Ulms et al. 2001).
region was essential for its internalization in These three molecules are involved in adhe-
neuroblastoma cells (Sunyach et al. 2003, Tay- sion in a diversity of signal transduction path-
lor et al. 2005). Sunyach et al. (2003) suggested ways, in differentiation, and in neurite out-
that heparin sulfate proteoglycans are part growth (Colognato & Yurchenco 2000, Maness
of the endocytotic complex involving PrPC . & Schachner 2007). Interaction of laminin

460 Aguzzi · Baumann · Bremer


or N-CAM with PrPC has been associated dous effort put into the identification of its in-
mainly with its suggested role in neuritogenesis teraction partners by different methods such as
and neurite outgrowth (Graner et al. 2000a,b; yeast-two hybrid, coimmunoprecipitations, and
Santuccione et al. 2005). Sales et al. (1998) cross-linking experiments.
also proposed that PrPC in its synaptic location All PrPC interaction partners identified thus
might stabilize apposing synaptic membranes far are summarized in Table 1. They in-
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

through adhesive mechanisms. clude membrane proteins (receptors, enzymes,


The evidence reported so far could indicate Caveolin-1, Na-K-ATPase, and a potassium
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

that PrPC is involved in adhesive mechanisms, channel), cytoplasmic proteins (components


but this is likely not its sole function. Adhe- of the cytoskeleton, heat-shock proteins, and
sion molecules that interact biochemically with adaptor proteins involved in signaling), and
PrPC could also transduce a PrPC -dependent even the nuclear protein CBP70. Tantalizingly,
signal. The neurite-outgrowth-promoting in- several of these interactions partners are known
teraction between PrPC and N-CAM, for ex- to play a role in synaptic vesicle function. Un-
ample, was associated with activation of the fortunately, the physiological relevance of most
p59Fyn kinase (Santuccione et al. 2005). of the proposed interaction partners remains
unconfirmed. Proteins that are not even lo-
calized to the outer leaflet of the cell mem-
Interaction Partners of PrPC brane, where PrPC is located and believed to
Investigators often hypothesized that PrPC ex- exert its function, would at least require an addi-
erts its function via interaction with other tional interacting cell component, meaning that
cell-surface components. GPI-anchored pro- their interaction with PrPC must be indirect
teins would need to interact with a transmem- at best.
brane adaptor to influence intracellular signal- Another possible explanation for cytosolic
transduction pathways, thereby enabling the interaction partners is the suggested presence
transduction of an extracellular signal. An ex- of transmembranous variants of PrPC , termed
Ntm
ample of such a protein is the urokinase-type PrP and Ctm PrP. However, under normal
plasminogen activator receptor uPAR, which conditions they have been described at best in
is involved in cellular adhesion, differentia- minute amounts only (Hegde et al. 1998, 1999;
tion, proliferation, and migration mediated by Stewart & Harris 2001). Cytosolic PrP was later
the interaction with transmembrane adaptors detected (Ma & Lindquist 2002, Ma et al. 2002).
such as integrins, G protein–coupled receptors, The significance of these findings still remains
and caveolins (Blasi & Carmeliet 2002). Analo- obscure.
gously, PrPC may bind to a transmembrane pro- One must consider methodical bottlenecks.
tein or to a protein complex that mediates func- Because PrPC is exposed to the extracellular
tional association with intracellular pathways. space, it is questionable whether a yeast two-
The toxic deletion mutants of PrPC may hybrid screen that artificially exposes PrPC to
destroy such a complex by competing for the the cytosolic compartment with its different
binding of some complex components yet fail- biochemical composition is the most appropri-
ing to interact with the signal-transducing com- ate method by which to study PrPC interaction
ponents. Indeed, several models for the toxi- partners. A high number of false-positive
city of PrPC deletion mutants have proposed results is to be expected, and therefore it is
that PrPC binds to a transmembrane receptor particularly important that additional methods
and that deletion mutants induce a toxic sig- are used to confirm interactions identified by
nal (Li et al. 2007) or prevent a survival signal yeast-two hybrid.
(Baumann et al. 2007, Shmerling et al. 1998). One item of paramount importance in im-
The commonly shared opinion that PrPC munoprecipitation experiments is the choice
binds to a receptor might explain the tremen- of the detergent conditions to allow for weak

www.annualreviews.org • The Prion’s Elusive Reason for Being 461


Table 1 Molecules identified as interaction partners to PrPC or PrPSc a
Interaction Function of interaction partner and
partner Subcellular localization Method Binding site references
Hsp60 and GoEL Mitochondria Y2H 180–210 Chaperone (Edenhofer et al. 1996)
Laminin receptor Plasma membrane Y2H, Co-IP 144–179 Cell adhesion (Hundt et al. 2001,
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

precursor Rieger et al. 1997)


Laminin Extracellular, basement Binding assay Unknown Signal transduction, cell adhesion,
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

membrane neuritogenesis (Graner et al. 2000a,b)


Synapsin Ib Synaptic vesicles Y2H, Co-IP 23–100 and Synaptic vesicle formation, modulation of
90–231 neurotransmitter release and in synap-
togenesis (Spielhaupter & Schatzl 2001)
Grb2 Cytoplasm Y2H, Co-IP 23–100, Adaptor protein mediating signal transduc-
90–231 tion (Spielhaupter & Schatzl 2001)
Pint1 Unknown Y2H, Co-IP 90–231 Unknown (Spielhaupter & Schatzl 2001)
N-CAM Plasma membrane Formaldehyde 141–176 Cell adhesion, signaling, etc. (Maness &
(transmembrane/ cross-linking Schachner 2007, Schmitt-Ulms et al.
GPI-anchored) 2001)
Stress-inducible Cytoplasm, plasma Complementary 113–128 Heat shock protein (Chiarini et al. 2002,
protein 1 (STI 1) membrane? hydropathy Zanata et al. 2002)
Co-IP
Caveolin-1 Caveolae, plasma membrane Co-IP unknown Caveolae formation and endocytosis;
(transmembrane) cross-linking of PrP induces Fyn
activation (Mouillet-Richard et al. 2000)
Fyn kinase Cytoplasm, associated with Co-IP unknown Signal transducer molecule (Mattei et al.
cytosolic side of plasma 2004, Mouillet-Richard et al. 2000)
membrane
ZAP70 Cytoplasm Co-IP Unknown Signal transduction during T cell
activation (Mattei et al. 2004)
Synaptophysin Synaptic vesicles Co-IP Unknown Presynaptic vesicle protein (Keshet et al.
(transmembrane) 2000)
Neuronal nitric Intracellular, partly Co-IP Unknown Production of NO in neuronal tissue,
oxide synthase membrane bound involved in signaling, neurotransmission,
(nNOS) etc. (Keshet et al. 2000)
β-dystroglycan Plasma membrane Co-IP Unknown Transmembrane protein, binds
(transmembrane, part of the extracellularly to α-dystroglycan (bound
dystroglycan complex) to laminin) and, intracellularly, to
dystrophin (Keshet et al. 2000)
Dp71 (dystrophin) Cytoplasmic face of plasma Co-IP Unknown Cytoskeletal protein (Keshet et al. 2000)
membrane (part of the
dystroglycan complex)
α-syntrophin Cytoplasmic face of Co-IP Unknown Adaptor protein, in sarcolemma and the
membrane (part of the neuromuscular junction, binds dystro-
dystroglycan complex) phins and nNOS (Keshet et al. 2000)
α-tubulin Microtubules, cytoplasm Cross-linking, Unknown Microtubule subunit (Keshet et al. 2000,
Co-IP Nieznanski et al. 2005)
Glutamic acid Intracellular, vesicle Co-IP Unknown Enzyme that synthesizes the
decarboxylase membrane neurotransmitter GABA from glutamate
(GAD) (Keshet et al. 2000)
(Continued )

462 Aguzzi · Baumann · Bremer


Table 1 (Continued )
Interaction Function of interaction partner and
partner Subcellular localization Method Binding site references
β-actin Intracellular cytoskeleton Co-IP, affinity Unknown Subunit of microfilaments of the
protein chromatography cytoskeleton (Keshet et al. 2000)
BACE-1 Plasma membrane Co-IP Unknown APP processing (Parkin et al. 2007)
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

(transmembrane)
TREK-1 Plasma membrane Bacterial two- 128–230 Two-pore potassium channel protein
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

(transmembrane) hybrid, Co-IP (Azzalin et al. 2006)


Bcl-2 Cytoplasmic face of mito- Yeast two hybrid 72–254 Inhibition of apoptosis (Kurschner &
chondrial outer membrane, Morgan 1995)
nuclear envelop, and ER
NRAGE Cytoplasm, plasma Y2H, Co-IP 122–231 Binding to p75NTR, permits
(neurotrophin membrane association NGF/p75NTR-dependent apoptosis
receptor–interacting when NGF is bound to (Bragason & Palsdottir 2005, Salehi
MAGE homolog) p75NTR et al. 2000)
Mouse p45 NF-E2 Cytoplasmic or nuclear cDNA library Unknown Transcription factor regulating
related factor 2 (regulated shuttling) screen, interac- expression of a set of detoxifying and
(Nrf2) tion with AP- antioxidant enzyme genes
tagged soluble PrP
Mouse amyloid Plasma membrane cDNA library Unknown Neuronal survival and neurite
precursor-like (transmembrane) screen, interac- outgrowth (Sakai & Hohjoh 2006,
protein 1 (APLP1) tion with AP Yehiely et al. 1997)
tagged soluble PrP
αB-crystalline Cytoplasm Y2H Unknown Small heat-shock protein, chaperone
Ribosomal protein Ribosomes Affinity Unknown Important for ribosomal activity
P0 chromatography (Sun et al. 2005)
CNPase Myelin, cytoplasmic face, Affinity Unknown Membrane anchor for microtubules,
microtubule-associated chromatography required for maintaining the integrity
of paranodes and axons (Petrakis &
Sklaviadis 2006)
Creatine kinase-B Cytoplasm, intracellular Affinity Unknown Catalyzes transfer of phosphate
enzyme chromatography between ATP and various
phosphogens like creatine phosphate
(Petrakis & Sklaviadis 2006)
NSE (neuron- Cytoplasm, intracellular Affinity Unknown Enolase is a glycolytic enzyme; NSE
specific enolase) enzyme chromatography contains γ-subunit, expressed
primarily in neurons and in
neuroendocrine cells (Petrakis &
Sklaviadis 2006)
Clathrin heavy Cytosolic face of coated Affinity Unknown Clathrin: major protein component of
chain 1 vesicles and coated pits chromatography the cytoplasmic face of coated vesicles
and coated pits, involved in intracellu-
lar trafficking of receptors and endo-
cytosis (Petrakis & Sklaviadis 2006)
α-spectrin Cytoskeletal protein, lines Affinity Unknown Maintenance of plasma membrane
intracellular side of the chromatography integrity and cytoskeletal structure
plasma membrane (Petrakis & Sklaviadis 2006)
(Continued )

www.annualreviews.org • The Prion’s Elusive Reason for Being 463


Table 1 (Continued )
Interaction Function of interaction partner and
partner Subcellular localization Method Binding site references
Glial fibrillary Intracellular, intermediate Affinity Unknown Different cellular processes, like cell
acidic protein filament in glial cells such as chromatography structure and movement, cell
(GFAP) astrocytes communication (Petrakis & Sklaviadis
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

2006)
Na+/K+ ATPase Plasma membrane Affinity Unknown Catalytic subunit of P-type ATPase,
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

α3 chromatography active transport of cations across cell


membranes, maintain ionic gradients
(Petrakis & Sklaviadis 2006)
PLP (proteolipid Myelin (transmembrane) Cross-linking Unknown Major component of central nervous
protein) system myelin (Petrakis & Sklaviadis
2006)
STXBP1 (Syntaxin Membrane-associated in Cross-linking Unknown Participance in regulation of synaptic
binding protein 1) presence of syntaxin vesicle docking and fusion (Petrakis &
Sklaviadis 2006)
ζ14–3–3 Intracellular Cross-linking Unknown Phosphoserine or phosphothreonine
binding protein binds diverse signaling
proteins, including kinases,
phosphatases, and transmembrane
receptors, thereby regulating most
cellular processes (Petrakis &
Sklaviadis 2006)
BASP1 Bound to plasma membrane, Cross-linking Unknown Accumulation mainly in axon endings
calmodulin, and actin (growth cones and presynaptic area of
synapses) (Petrakis & Sklaviadis 2006)
Vitronectin Extracellular matrix Binding and 105–119 Axonal growth in the peripheral
glycoprotein competition nervous system (Hajj et al. 2007)
assay
CBP70 Nuclear [and cytoplasmatic Co-IP Lectin (Rybner et al. 2002)
(Rousseau et al. 1997)]
Glycosaminoglycans Connective tissues, ELISA, SPR, 23–35, Various functions (Caughey et al. 1994,
(GAG) covalently linked to proteins heparin-agarose 23–52, Pan et al. 2002, Warner et al. 2002)
to form proteoglycans binding assay 53–93,
110–128
ApoE Secreted Co-IP, 23–90 Main apolipoprotein of chylomicrons,
cross-linking 109–141 involved in neurodegenerative diseases
such as Alzheimer disease (Baumann
et al. 2000, Gao et al. 2006,
Schmitt-Ulms et al. 2004)
Plasminogen Secreted Immobilized Unknown Inactive zymogen form of plasmin,
serum proteins which participates in thrombolysis or
probed with extracellular matrix degradation (Ellis
PrPC or PrPres et al. 2002, Fischer et al. 2000)
Rdj2 Cytoplasmic face of Co-IP with GST Unknown Chaperone (Beck et al. 2006)
membranes fusion proteins
Casein kinase 2 Cytoplasmic Far-Western, SPR 105–242 Protein kinase (Meggio et al. 2000)
α/α’
a
Co-IP, coimmunoprecipitation; Y2H, yeast-two hybrid; SPR, surface plasmon resonance.

464 Aguzzi · Baumann · Bremer


and transient protein-protein interactions but One of the most frequently cited cellular func-
destroy artificial or unspecific interactions. The tions of PrPC is a survival-promoting effect on
influence of the detergent used was exemplified neuronal and nonneuronal cells. This cytopro-
in one study in which the interactions between tective function has been mediated by antiapop-
PrPC and the dystroglycan complex were stud- totic or antioxidative mechanisms, but nothing
ied as a function of the detergent used and on is known about the proximal mechanisms me-
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

the integrity of lipid rafts (Keshet et al. 2000). diating such effects.
Some of the interactions identified by immuno- The frequently voiced opinion that PrPC
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

precipitation may be artifactual—a result that binds to a receptor has driven a large ef-
could be avoided by using stringent controls in- fort toward the identification of its interac-
cluding knockout tissues and specific antibody tion partners by different methods such as
competition experiments. yeast-two hybrid, coimmunoprecipitations, and
cross-linking experiments. A rather large num-
ber of interaction partners have been identified,
CONCLUSIONS yet a functional interaction was unambiguously
Despite the progress discussed above, several discovered for none of them.
important issues in the prion field remain un- Some of the latter results may need to be
resolved. Most conspicuously, both the phys- reconsidered critically. Several reports suffer
iological function of PrPC and the molecular from intrinsic methodological shortcomings,
pathways leading to the fatal neurodegenera- some of which were unavoidable at the time of
tion in prion diseases remain unknown. These publication. Furthermore, the likelihood of in-
two issues may be linked, and elucidation of the teractions between membrane-attached extra-
physiological function of PrPC has the potential cellular PrPC with cytosolic or mitochondrial
to help researchers understand the mechanisms molecule is counterintuitive and may point to
involved in prion-induced neurodegeneration. artifactual effects.
Studies of mice carrying targeted disrup- Although it is highly plausible in our opin-
tions of any given gene have often provided ion, the connection between the normal func-
researchers with useful tools to identify the tion of PrPC and the neurotoxicity of prions
respective gene product’s function. However, remains admittedly hypothetical and lacks ex-
the many lines of mice lacking PrPC that have perimental confirmation. Alternative scenarios
been generated independently by homologous are thinkable, and it is not impossible that the
recombination have failed to uncover a clear cascade of events outlined above will prove in-
molecular physiological function of PrPC . The correct. However, the depth of the knowledge
most obvious phenotype was their resistance to gaps in prion biology, along with the medi-
prion infection. Nevertheless, an overwhelm- cal importance of the neurodegeneration prob-
ing number of molecular, structural, or func- lem, indicates that many exciting discoveries
tional alterations have been reported in Prnp−/− still lie ahead. Therefore, despite the disap-
mice. pearance of “mad cow disease” from the me-
Cellular processes in the nervous system that dia and from public awareness, the field of
have been influenced by the Prnp genotype prion science remains an excellent choice for
include neuronal survival, neurite outgrowth, ambitious PhD students and postdocs willing
synapse formation, maintenance, and function, to make an impact in current experimental
as well as maintenance of myelinated fibers. biology.

DISCLOSURE STATEMENT
The authors are not aware of any biases that might be perceived as affecting the objectivity of this
review.

www.annualreviews.org • The Prion’s Elusive Reason for Being 465


LITERATURE CITED
Aguzzi A. 1996. Between cows and monkeys. Nature 381:734–55
Aguzzi A. 2007. Prion biology: the quest for the test. Nat. Methods 4:614–16
Aguzzi A, Glatzel M. 2006. Prion infections, blood and transfusions. Nat. Clin. Pract. Neurol.
2:321–29
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Aguzzi A, Weissmann C. 1996. Spongiform encephalopathies: a suspicious signature. Nature


383:666–67
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Andrews NJ, Farrington CP, Ward HJ, Cousens SN, Smith PG, et al. 2003. Deaths from variant
Creutzfeldt-Jakob disease in the UK. Lancet 361:751–52
Aronoff-Spencer E, Burns CS, Avdievich NI, Gerfen GJ, Peisach J, et al. 2000. Identification
of the Cu2+ binding sites in the N-terminal domain of the prion protein by EPR and CD
spectroscopy. Biochemistry 39:13760–71
Atarashi R, Moore RA, Sim VL, Hughson AG, Dorward DW, et al. 2007. Ultrasensitive detection
of scrapie prion protein using seeded conversion of recombinant prion protein. Nat. Methods
4:645–50
Azzalin A, Ferrara V, Arias A, Cerri S, Avella D, et al. 2006. Interaction between the cellular prion
(PrPC ) and the 2P domain K+ channel TREK-1 protein. Biochem. Biophys. Res. Commun.
346:108–15
Barrow PA, Holmgren CD, Tapper AJ, Jefferys JG. 1999. Intrinsic physiological and morpholog-
ical properties of principal cells of the hippocampus and neocortex in hamsters infected with
scrapie. Neurobiol. Dis. 6:406–23
Baumann F, Tolnay M, Brabeck C, Pahnke J, Kloz U, et al. 2007. Lethal recessive myelin toxicity
of prion protein lacking its central domain. EMBO J. 26:538–47
Baumann MH, Kallijarvi J, Lankinen H, Soto C, Haltia M. 2000. Apolipoprotein E includes a
binding site which is recognized by several amyloidogenic polypeptides. Biochem. J. 349:77–84
Beck KE, Kay JG, Braun JE. 2006. Rdj2, a J protein family member, interacts with cellular prion
PrP(C). Biochem. Biophys. Res. Commun. 346:866–71
Behrens A, Genoud N, Naumann H, Rulicke T, Janett F, et al. 2002. Absence of the prion protein
homologue Doppel causes male sterility. EMBO J. 21:3652–58
Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, et al. 2000. The Protein Data Bank.
Nucleic Acids Res. 28:235–42
Bessen RA, Kocisko DA, Raymond GJ, Nandan S, Lansbury PT, Caughey B. 1995. Non-genetic
propagation of strain-specific properties of scrapie prion protein. Nature 375:698–700
Blasi F, Carmeliet P. 2002. uPAR: a versatile signalling orchestrator. Nat. Rev. Mol. Cell Biol.
3:932–43
Bounhar Y, Zhang Y, Goodyer CG, LeBlanc A. 2001. Prion protein protects human neurons
against Bax-mediated apoptosis. J. Biol. Chem. 276:39145–49
Bouzamondo-Bernstein E, Hopkins SD, Spilman P, Uyehara-Lock J, Deering C, et al. 2004. The
neurodegeneration sequence in prion diseases: evidence from functional, morphological and
ultrastructural studies of the GABAergic system. J. Neuropathol. Exp. Neurol. 63:882–99
Bragason BT, Palsdottir A. 2005. Interaction of PrP with NRAGE, a protein involved in neuronal
apoptosis. Mol. Cell Neurosci. 29:232–44
Brandner S, Isenmann S, Raeber A, Fischer M, Sailer A, et al. 1996. Normal host prion protein
necessary for scrapie-induced neurotoxicity. Nature 379:339–43
Brown DR, Nicholas RS, Canevari L. 2002. Lack of prion protein expression results in a neuronal
phenotype sensitive to stress. J. Neurosci. Res. 67:211–24
Brown DR, Schmidt B, Kretzschmar HA. 1997a. Effects of oxidative stress on prion protein
expression in PC12 cells. Int. J. Dev. Neurosci. 15:961–72

466 Aguzzi · Baumann · Bremer


Brown DR, Schulz-Schaeffer WJ, Schmidt B, Kretzschmar HA. 1997b. Prion protein-deficient
cells show altered response to oxidative stress due to decreased SOD-1 activity. Exp. Neurol.
146:104–12
Brown DR, Wong BS, Hafiz F, Clive C, Haswell SJ, Jones IM. 1999. Normal prion protein has
an activity like that of superoxide dismutase. Biochem. J. 344(Pt.1):1–5
Brown P, Gibbs CJ Jr, Rodgers-Johnson P, Asher DM, Sulima MP, et al. 1994. Human spongi-
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

form encephalopathy: the National Institutes of Health series of 300 cases of experimentally
transmitted disease. Ann. Neurol. 35:513–29
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Bruce ME, Will RG, Ironside JW, McConnell I, Drummond D, et al. 1997. Transmissions to
mice indicate that ‘new variant’ CJD is caused by the BSE agent. Nature 389:498–501
Bueler H, Aguzzi A, Sailer A, Greiner RA, Autenried P, et al. 1993. Mice devoid of PrP are resistant
to scrapie. Cell 73:1339–47
Bueler H, Fischer M, Lang Y, Bluethmann H, Lipp HP, et al. 1992. Normal development and
behaviour of mice lacking the neuronal cell-surface PrP protein. Nature 356:577–82
Cancellotti E, Wiseman F, Tuzi NL, Baybutt H, Monaghan P, et al. 2005. Altered glycosylated
PrP proteins can have different neuronal trafficking in brain but do not acquire scrapie-like
properties. J. Biol. Chem. 280:42909–18
Carleton A, Tremblay P, Vincent JD, Lledo PM. 2001. Dose-dependent, prion protein (PrP)-
mediated facilitation of excitatory synaptic transmission in the mouse hippocampus. Pflugers
Arch. 442:223–29
Castagna A, Campostrini N, Farinazzo A, Zanusso G, Monaco S, Righetti PG. 2002. Comparative
two-dimensional mapping of prion protein isoforms in human cerebrospinal fluid and central
nervous system. Electrophoresis 23:339–46
Castilla J, Saa P, Hetz C, Soto C. 2005a. In vitro generation of infectious scrapie prions. Cell
121:195–206
Castilla J, Saa P, Soto C. 2005b. Detection of prions in blood. Nat. Med. 11:982–85
Caughey B, Brown K, Raymond GJ, Katzenstein GE, Thresher W. 1994. Binding of the protease-
sensitive form of PrP (prion protein) to sulfated glycosaminoglycan and congo red. J. Virol.
68:2135–41
Chattopadhyay M, Walter ED, Newell DJ, Jackson PJ, Aronoff-Spencer E, et al. 2005. The
octarepeat domain of the prion protein binds Cu(II) with three distinct coordination modes
at pH 7.4. J. Am. Chem. Soc. 127:12647–56
Chen S, Mangé A, Dong L, Lehmann S, Schachner M. 2003. Prion protein as trans-interacting
partner for neurons is involved in neurite outgrowth and neuronal survival. Mol. Cell Neurosci.
22:227–33
Cheng F, Lindqvist J, Haigh CL, Brown DR, Mani K. 2006. Copper-dependent cointernalization
of the prion protein and glypican-1. J. Neurochem. 98:1445–57
Chesebro B, Trifilo M, Race R, Meade-White K, Teng C, et al. 2005. Anchorless prion protein
results in infectious amyloid disease without clinical scrapie. Science 308:1435–39
Chiarini LB, Freitas AR, Zanata SM, Brentani RR, Martins VR, Linden R. 2002. Cellular prion
protein transduces neuroprotective signals. EMBO J. 21:3317–26
Chiesa R, Drisaldi B, Quaglio E, Migheli A, Piccardo P, et al. 2000. Accumulation of protease-
resistant prion protein (PrP) and apoptosis of cerebellar granule cells in transgenic mice
expressing a PrP insertional mutation. Proc. Natl. Acad. Sci. USA 97:5574–79
Chiesa R, Piccardo P, Ghetti B, Harris DA. 1998. Neurological illness in transgenic mice express-
ing a prion protein with an insertional mutation. Neuron 21:1339–51
Chiesa R, Piccardo P, Quaglio E, Drisaldi B, Si-Hoe SL, et al. 2003. Molecular distinction between
pathogenic and infectious properties of the prion protein. J. Virol. 77:7611–22

www.annualreviews.org • The Prion’s Elusive Reason for Being 467


Choi SI, Ju WK, Choi EK, Kim J, Lea HZ, et al. 1998. Mitochondrial dysfunction induced
by oxidative stress in the brains of hamsters infected with the 263 K scrapie agent. Acta
Neuropathol. 96:279–86
Clinton J, Forsyth C, Royston MC, Roberts GW. 1993. Synaptic degeneration is the primary
neuropathological feature in prion disease: a preliminary study. NeuroReport 4:65–68
Colling SB, Collinge J, Jefferys JG. 1996. Hippocampal slices from prion protein null mice:
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

disrupted Ca(2+) -activated K+ currents. Neurosci. Lett. 209:49–52


Collinge J, Whitfield J, McKintosh E, Beck J, Mead S, et al. 2006. Kuru in the 21st century—an
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

acquired human prion disease with very long incubation periods. Lancet 367:2068–74
Collinge J, Whittington MA, Sidle KC, Smith CJ, Palmer MS, et al. 1994. Prion protein is
necessary for normal synaptic function. Nature 370:295–97
Collins S, McLean CA, Masters CL. 2001. Gerstmann-Sträussler-Scheinker syndrome, fatal fa-
milial insomnia, and kuru: a review of these less common human transmissible spongiform
encephalopathies. J. Clin. Neurosci. 8:387–97
Colognato H, Yurchenco PD. 2000. Form and function: the laminin family of heterotrimers. Dev.
Dyn. 218:213–34
Creutzfeldt HG. 1920. Über eine eigenartige herdförmige erkrankung des zentralnervensystems.
Z. Ges. Neurol. Psychiatr. 57:1–19
Criado JR, Sánchez-Alavez M, Conti B, Giacchino JL, Wills DN, et al. 2005. Mice devoid of
prion protein have cognitive deficits that are rescued by reconstitution of PrP in neurons.
Neurobiol. Dis. 19:255–65
Cuille J, Chellè PL. 1939. Experimental transmission of trembling to the goat. C. R. Acad. Sci.
208:1058–160
Curtis J, Errington M, Bliss T, Voss K, MacLeod N. 2003. Age-dependent loss of PTP and LTP
in the hippocampus of PrP-null mice. Neurobiol. Dis. 13:55–62
Deleault NR, Geoghegan JC, Nishina K, Kascsak R, Williamson RA, Supattapone S. 2005.
Protease-resistant prion protein amplification reconstituted with partially purified substrates
and synthetic polyanions. J. Biol. Chem. 280:26873–79
Deleault NR, Harris BT, Rees JR, Supattapone S. 2007. Formation of native prions from minimal
components in vitro. Proc. Natl. Acad. Sci. USA 104:9741–46
Diarra-Mehrpour M, Arrabal S, Jalil A, Pinson X, Gaudin C, et al. 2004. Prion protein prevents
human breast carcinoma cell line from tumor necrosis factor α-induced cell death. Cancer
Res. 64:719–27
Di Guglielmo GM, Le Roy C, Goodfellow AF, Wrana JL. 2003. Distinct endocytic pathways
regulate TGF-β receptor signalling and turnover. Nat. Cell Biol. 5:410–21
Duffy P, Wolf J, Collins G, DeVoe AG, Streeten B, Cowen D. 1974. Possible person-to-person
transmission of Creutzfeldt-Jakob disease. N. Engl. J. Med. 290:692–93
Edenhofer F, Rieger R, Famulok M, Wendler W, Weiss S, Winnacker EL. 1996. Prion protein
PrPc interacts with molecular chaperones of the Hsp60 family. J. Virol. 70:4724–28
Elfrink K, Nagel-Steger L, Riesner D. 2007. Interaction of the cellular prion protein with raft-like
lipid membranes. Biol. Chem. 388:79–89
Ellis V, Daniels M, Misra R, Brown DR. 2002. Plasminogen activation is stimulated by prion
protein and regulated in a copper-dependent manner. Biochemistry 41:6891–96
Favre-Krey L, Theodoridou M, Boukouvala E, Panagiotidis CH, Papadopoulos AI, et al. 2007.
Molecular characterization of a cDNA from the gilthead sea bream (Sparus aurata) encoding
a fish prion protein. Comp. Biochem. Physiol. B 147:566–73
Ferrer I, Rivera R, Blanco R, Martı́ E. 1999. Expression of proteins linked to exocytosis and
neurotransmission in patients with Creutzfeldt-Jakob disease. Neurobiol. Dis. 6:92–100

468 Aguzzi · Baumann · Bremer


Fioriti L, Dossena S, Stewart LR, Stewart RS, Harris DA, et al. 2005. Cytosolic prion protein
(PrP) is not toxic in N2a cells and primary neurons expressing pathogenic PrP mutations. J.
Biol. Chem. 280:11320–28
Fischer M, Rülicke T, Raeber A, Sailer A, Moser M, et al. 1996. Prion protein (PrP) with
amino-proximal deletions restoring susceptibility of PrP knockout mice to scrapie. EMBO J.
15:1255–64
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Fischer MB, Roeckl C, Parizek P, Schwarz HP, Aguzzi A. 2000. Binding of disease-associated
prion protein to plasminogen. Nature 408:479–83
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Flechsig E, Shmerling D, Hegyi I, Raeber AJ, Fischer M, et al. 2000. Prion protein devoid of
the octapeptide repeat region restores susceptibility to scrapie in PrP knockout mice. Neuron
27:399–408
Flechsig E, Weissmann C. 2004. The role of PrP in health and disease. Curr. Mol. Med. 4:337–53
Fournier JG, Escaig-Haye F, Billette de Villemeur T, Robain O. 1995. Ultrastructural localization
of cellular prion protein (PrPc) in synaptic boutons of normal hamster hippocampus. C. R.
Acad. Sci. III 318:339–44
Fukuuchi T, Doh-Ura K, Yoshihara S, Ohta S. 2006. Metal complexes with superoxide dismutase-
like activity as candidates for antiprion drug. Bioorg. Med. Chem. Lett. 16:5982–87
Furlan S, La Penna G, Guerrieri F, Morante S, Rossi GC. 2007. Ab initio simulations of Cu
binding sites on the N-terminal region of prion protein. J. Biol. Inorg. Chem. 12:571–83
Gabriel JM, Oesch B, Kretzschmar H, Scott M, Prusiner SB. 1992. Molecular cloning of a can-
didate chicken prion protein. Proc. Natl. Acad. Sci. USA 89:9097–101
Gajdusek DC, Gibbs CJ Jr, Alpers M. 1966. Experimental transmission of a Kuru-like syndrome
to chimpanzees. Nature 209:794–96
Gajdusek DC, Gibbs CJ Jr, Alpers M. 1967. Transmission and passage of experimenal “kuru” to
chimpanzees. Science 155:212–14
Gajdusek DC, Gibbs CJ Jr, Asher DM, David E. 1968. Transmission of experimental kuru to the
spider monkey (Ateles geoffreyi). Science 162:693–94
Gajdusek DC, Reid LH. 1961. Studies on kuru. IV. The kuru pattern in Moke, a representative
Fore village. Am. J. Trop. Med. Hyg. 10:628–38
Gajdusek DC, Zigas V. 1957. Degenerative disease of the central nervous system in New Guinea;
the endemic occurrence of kuru in the native population. N. Engl. J. Med. 257:974–78
Gao C, Lei YJ, Han J, Shi Q, Chen L, et al. 2006. Recombinant neural protein PrP can bind with
both recombinant and native apolipoprotein E in vitro. Acta Biochim. Biophys. Sin. 38:593–601
Gauczynski S, Peyrin JM, Haik S, Leucht C, Hundt C, et al. 2001. The 37-kDa/67-kDa laminin
receptor acts as the cell-surface receptor for the cellular prion protein. EMBO J. 20:5863–75
Gerstmann J. 1928. Über ein noch nicht beschriebenes Reflexphanomen bei einer Erkrankung
des zerebellaren Systems. Wien. Medizin Wochenschr. 78:906–8
Gerstmann J, Sträussler E, Scheinker I. 1936. Über eine eigenartige hereditar-familiäre
Erkrankung des Zentralnervensystems. Zugleich ein Beitrag zur Frage des vorzeitigen lokalen
Alterns. Z. Neurol. 154:736–62
Gohel C, Grigoriev V, Escaig-Haye F, Lasmezas CI, Deslys JP, et al. 1999. Ultrastructural lo-
calization of cellular prion protein (PrPc) at the neuromuscular junction. J. Neurosci. Res.
55:261–67
Goldfarb LG, Petersen RB, Tabaton M, Brown P, LeBlanc AC, et al. 1992. Fatal familial insomnia
and familial Creutzfeldt-Jakob disease: disease phenotype determined by a DNA polymor-
phism. Science 258:806–8
Graner E, Mercadante AF, Zanata SM, Forlenza OV, Cabral AL, et al. 2000a. Cellular prion
protein binds laminin and mediates neuritogenesis. Brain Res. Mol. Brain Res. 76:85–92

www.annualreviews.org • The Prion’s Elusive Reason for Being 469


Graner E, Mercadante AF, Zanata SM, Martins VR, Jay DG, Brentani RR. 2000b. Laminin-
induced PC-12 cell differentiation is inhibited following laser inactivation of cellular prion
protein. FEBS Lett. 482:257–60
Grigoriev V, Escaig-Haye F, Streichenberger N, Kopp N, Langeveld J, et al. 1999. Submicroscopic
immunodetection of PrP in the brain of a patient with a new-variant of Creutzfeldt-Jakob
disease. Neurosci. Lett. 264:57–60
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Hadlow WJ. 1959. Scrapie and kuru. Lancet 2:289–90


Hajj GN, Lopes MH, Mercadante AF, Veiga SS, da Silveira RB, et al. 2007. Cellular prion protein
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

interaction with vitronectin supports axonal growth and is compensated by integrins. J. Cell
Sci. 120:1915–26
Hegde RS, Mastrianni JA, Scott MR, DeFea KA, Tremblay P, et al. 1998. A transmembrane form
of the prion protein in neurodegenerative disease. Science 279:827–34
Hegde RS, Tremblay P, Groth D, DeArmond SJ, Prusiner SB, Lingappa VR. 1999. Transmissible
and genetic prion diseases share a common pathway of neurodegeneration. Nature 402:822–
26
Herms JW, Tings T, Dunker S, Kretzschmar HA. 2001. Prion protein affects Ca2+ -activated K+
currents in cerebellar purkinje cells. Neurobiol. Dis. 8:324–30
Hill AF, Desbruslais M, Joiner S, Sidle KC, Gowland I, et al. 1997. The same prion strain causes
vCJD and BSE. Nature 389:448–50, 526
Holscher C, Delius H, Burkle A. 1998. Overexpression of nonconvertible PrPc delta114–121 in
scrapie-infected mouse neuroblastoma cells leads to trans-dominant inhibition of wild-type
PrP(Sc) accumulation. J. Virol. 72:1153–59
Hornemann S, Korth C, Oesch B, Riek R, Wider G, et al. 1997. Recombinant full-length murine
prion protein, mPrP(23–231): purification and spectroscopic characterization. FEBS Lett.
413:277–81
Hsiao K, Baker HF, Crow TJ, Poulter M, Owen F, et al. 1989. Linkage of a prion protein missense
variant to Gerstmann-Sträussler syndrome. Nature 338:342–45
Hsiao KK, Groth D, Scott M, Yang SL, Serban H, et al. 1994. Serial transmission in rodents of
neurodegeneration from transgenic mice expressing mutant prion protein. Proc. Natl. Acad.
Sci. USA 91:9126–30
Hundt C, Peyrin JM, Haik S, Gauczynski S, Leucht C, et al. 2001. Identification of interaction
domains of the prion protein with its 37-kDa/67-kDa laminin receptor. EMBO J. 20:5876–86
Hutter G, Heppner FL, Aguzzi A. 2003. No superoxide dismutase activity of cellular prion protein
in vivo. Biol. Chem. 384:1279–85
Isaacs JD, Jackson GS, Altmann DM. 2006. The role of the cellular prion protein in the immune
system. Clin. Exp. Immunol. 146:1–8
Itoh Y, Yamada M, Hayakawa M, Shozawa T, Tanaka J, et al. 1994. A variant of Gerstmann-
Sträussler-Scheinker disease carrying codon 105 mutation with codon 129 polymorphism of
the prion protein gene: a clinicopathological study. J. Neurol. Sci. 127:77–86
Jakob A. 1921. Über eigenartige erkrankungen des zentralnervensystems mit bemerkenswertem
anatomischem befunde (spastische pseudosklerose-encephalomyelopathie mit disseminierten
degenerationsherden). Z. Ges. Neurol. Psychiatr. 64:147–228
Jeffrey M, Halliday WG, Bell J, Johnston AR, MacLeod NK, et al. 2000. Synapse loss asso-
ciated with abnormal PrP precedes neuronal degeneration in the scrapie-infected murine
hippocampus. Neuropathol. Appl. Neurobiol. 26:41–54
Johnston AR, Fraser JR, Jeffrey M, MacLeod N. 1998. Synaptic plasticity in the CA1 area of the
hippocampus of scrapie-infected mice. Neurobiol. Dis. 5:188–95
Jones S, Batchelor M, Bhelt D, Clarke AR, Collinge J, Jackson GS. 2005. Recombinant prion
protein does not possess SOD-1 activity. Biochem. J. 392:309–12

470 Aguzzi · Baumann · Bremer


Kanaani J, Prusiner SB, Diacovo J, Baekkeskov S, Legname G. 2005. Recombinant prion pro-
tein induces rapid polarization and development of synapses in embryonic rat hippocampal
neurons in vitro. J. Neurochem. 95:1373–86
Kaneko K, Vey M, Scott M, Pilkuhn S, Cohen FE, Prusiner SB. 1997. COOH-terminal sequence
of the cellular prion protein directs subcellular trafficking and controls conversion into the
scrapie isoform. Proc. Natl. Acad. Sci. USA 94:2333–38
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Keshet GI, Bar-Peled O, Yaffe D, Nudel U, Gabizon R. 2000. The cellular prion protein colocalizes
with the dystroglycan complex in the brain. J. Neurochem. 75:1889–97
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Keshet GI, Ovadia H, Taraboulos A, Gabizon R. 1999. Scrapie-infected mice and PrP knockout
mice share abnormal localization and activity of neuronal nitric oxide synthase. J. Neurochem.
72:1224–31
Kiachopoulos S, Bracher A, Winklhofer KF, Tatzelt J. 2005. Pathogenic mutations located in
the hydrophobic core of the prion protein interfere with folding and attachment of the
glycosylphosphatidylinositol anchor. J. Biol. Chem. 280:9320–29
Kirkham M, Parton RG. 2005. Clathrin-independent endocytosis: new insights into caveolae and
noncaveolar lipid raft carriers. Biochim. Biophys. Acta 1746:349–63
Kitamoto T, Shin RW, Doh-ura K, Tomokane N, Miyazono M, et al. 1992. Abnormal isoform of
prion proteins accumulates in the synaptic structures of the central nervous system in patients
with Creutzfeldt-Jakob disease. Am. J. Pathol. 140:1285–94
Klamt F, Dal-Pizzol F, Conte da Frota MJ, Walz R, Andrades ME, et al. 2001. Imbalance of
antioxidant defense in mice lacking cellular prion protein. Free Radic. Biol. Med. 30:1137–44
Kocisko DA, Priola SA, Raymond GJ, Chesebro B, Lansbury PT Jr, Caughey B. 1995. Species
specificity in the cell-free conversion of prion protein to protease-resistant forms: a model
for the scrapie species barrier. Proc. Natl. Acad. Sci. USA 92:3923–27
Korth C, Kaneko K, Prusiner SB. 2000. Expression of unglycosylated mutated prion protein
facilitates PrP(Sc) formation in neuroblastoma cells infected with different prion strains. J.
Gen. Virol. 81:2555–63
Kovacs GG, Trabattoni G, Hainfellner JA, Ironside JW, Knight RS, Budka H. 2002. Mutations
of the prion protein gene phenotypic spectrum. J. Neurol. 249:1567–82
Kurschner C, Morgan JI. 1995. The cellular prion protein (PrP) selectively binds to Bcl-2 in the
yeast two-hybrid system. Brain Res. Mol. Brain Res. 30:165–68
Kuwahara C, Takeuchi AM, Nishimura T, Haraguchi K, Kubosaki A, et al. 1999. Prions prevent
neuronal cell-line death. Nature 400:225–26
Laine J, Marc ME, Sy MS, Axelrad H. 2001. Cellular and subcellular morphological localization
of normal prion protein in rodent cerebellum. Eur. J. Neurosci. 14:47–56
Leclerc E, Serban H, Prusiner SB, Burton DR, Williamson RA. 2006. Copper induces conforma-
tional changes in the N-terminal part of cell-surface PrPC. Arch. Virol. 151:2103–9
Lee DW, Sohn HO, Lim HB, Lee YG, Kim YS, et al. 1999. Alteration of free radical metabolism
in the brain of mice infected with scrapie agent. Free Radic. Res. 30:499–507
Legname G, Baskakov IV, Nguyen HO, Riesner D, Cohen FE, et al. 2004. Synthetic mammalian
prions. Science 305:673–76
Legname G, Nguyen HO, Baskakov IV, Cohen FE, Dearmond SJ, Prusiner SB. 2005. Strain-
specified characteristics of mouse synthetic prions. Proc. Natl. Acad. Sci. USA 102:2168–73
Li A, Christensen HM, Stewart LR, Roth KA, Chiesa R, Harris DA. 2007. Neonatal lethality
in transgenic mice expressing prion protein with a deletion of residues 105–125. EMBO J.
26:548–58
Lledo PM, Tremblay P, DeArmond SJ, Prusiner SB, Nicoll RA. 1996. Mice deficient for prion
protein exhibit normal neuronal excitability and synaptic transmission in the hippocampus.
Proc. Natl. Acad. Sci. USA 93:2403–7

www.annualreviews.org • The Prion’s Elusive Reason for Being 471


Llewelyn CA, Hewitt PE, Knight RS, Amar K, Cousens S, et al. 2004. Possible transmission of
variant Creutzfeldt-Jakob disease by blood transfusion. Lancet 363:417–21
Lopes MH, Hajj GN, Muras AG, Mancini GL, Castro RM, et al. 2005. Interaction of cellular
prion and stress-inducible protein 1 promotes neuritogenesis and neuroprotection by distinct
signaling pathways. J. Neurosci. 25:11330–39
Lugaresi E, Medori R, Montagna P, Baruzzi A, Cortelli P, et al. 1986. Fatal familial insomnia and
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

dysautonomia with selective degeneration of thalamic nuclei. N. Engl. J. Med. 315:997–1003


Ma J, Lindquist S. 2002. Conversion of PrP to a self-perpetuating PrPSc-like conformation in the
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

cytosol. Science 298:1785–88


Ma J, Wollmann R, Lindquist S. 2002. Neurotoxicity and neurodegeneration when PrP accumu-
lates in the cytosol. Science 298:1781–85
Maglio LE, Martins VR, Izquierdo I, Ramirez OA. 2006. Role of cellular prion protein on LTP
expression in aged mice. Brain Res. 1097:11–18
Maglio LE, Perez MF, Martins VR, Brentani RR, Ramirez OA. 2004. Hippocampal synaptic
plasticity in mice devoid of cellular prion protein. Brain Res. Mol. Brain Res. 131:58–64
Mallucci G, Dickinson A, Linehan J, Klöhn PC, Brandner S, Collinge J. 2003. Depleting neuronal
PrP in prion infection prevents disease and reverses spongiosis. Science 302:871–74
Mallucci GR, Ratte S, Asante EA, Linehan J, Gowland I, et al. 2002. Post-natal knockout of prion
protein alters hippocampal CA1 properties, but does not result in neurodegeneration. EMBO
J. 21:202–10
Maness PF, Schachner M. 2007. Neural recognition molecules of the immunoglobulin superfam-
ily: signaling transducers of axon guidance and neuronal migration. Nat. Neurosci. 10:19–26
Manson JC, Clarke AR, Hooper ML, Aitchison L, McConnell I, Hope J. 1994. 129/Ola mice
carrying a null mutation in PrP that abolishes mRNA production are developmentally normal.
Mol. Neurobiol. 8:121–27
Marella M, Lehmann S, Grassi J, Chabry J. 2002. Filipin prevents pathological prion protein accu-
mulation by reducing endocytosis and inducing cellular PrP release. J. Biol. Chem. 277:25457–
64
Masters CL, Gajdusek DC, Gibbs CJ Jr. 1981. Creutzfeldt-Jakob disease virus isolations from
the Gerstmann-Straussler syndrome with an analysis of the various forms of amyloid plaque
deposition in the virus-induced spongiform encephalopathies. Brain 104:559–88
Mastrianni JA, Nixon R, Layzer R, Telling GC, Han D, et al. 1999. Prion protein conformation
in a patient with sporadic fatal insomnia. N. Engl. J. Med. 340:1630–38
Matsuda H, Mitsuda H, Nakamura N, Furusawa S, Mohri S, Kitamoto T. 1999. A chicken mono-
clonal antibody with specificity for the N-terminal of human prion protein. FEMS Immunol.
Med. Microbiol. 23:189–94
Mattei V, Garofalo T, Misasi R, Circella A, Manganelli V, et al. 2004. Prion protein is a component
of the multimolecular signaling complex involved in T cell activation. FEBS Lett. 560:14–18
McLennan NF, Brennan PM, McNeill A, Davies I, Fotheringham A, et al. 2004. Prion protein
accumulation and neuroprotection in hypoxic brain damage. Am. J. Pathol. 165:227–35
Medori R, Tritschler HJ, LeBlanc A, Villare F, Manetto V, et al. 1992. Fatal familial insomnia,
a prion disease with a mutation at codon 178 of the prion protein gene. N. Engl. J. Med.
326:444–49
Meggio F, Negro A, Sarno S, Ruzzene M, Bertoli A, et al. 2000. Bovine prion protein as a
modulator of protein kinase CK2. Biochem. J. 352(Pt. 1):191–96
Miele G, Jeffrey M, Turnbull D, Manson J, Clinton M. 2002. Ablation of cellular prion protein
expression affects mitochondrial numbers and morphology. Biochem. Biophys. Res. Commun.
291:372–77

472 Aguzzi · Baumann · Bremer


Milhavet O, Lehmann S. 2002. Oxidative stress and the prion protein in transmissible spongiform
encephalopathies. Brain Res. Brain Res. Rev. 38:328–39
Mironov A Jr, Latawiec D, Wille H, Bouzamondo-Bernstein E, Legname G, et al. 2003.
Cytosolic prion protein in neurons. J. Neurosci. 23:7183–93
Mitteregger G, Vosko M, Krebs B, Xiang W, Kohlmannsperger V, et al. 2007. The role of the
octarepeat region in neuroprotective function of the cellular prion protein. Brain Pathol.
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

17:174–83
Moore RC, Lee IY, Silverman GL, Harrison PM, Strome R, et al. 1999. Ataxia in prion protein
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

(PrP)-deficient mice is associated with upregulation of the novel PrP-like protein Doppel. J.
Mol. Biol. 292:797–817
Mouillet-Richard S, Ermonval M, Chebassier C, Laplanche JL, Lehmann S, et al. 2000. Signal
transduction through prion protein. Science 289:1925–28
Mouillet-Richard S, Pietri M, Schneider B, Vidal C, Mutel V, et al. 2005. Modulation of seroton-
ergic receptor signaling and cross-talk by prion protein. J. Biol. Chem. 280:4592–601
Moya KL, Sales N, Hassig R, Creminon C, Grassi J, Di Giamberardino L. 2000. Immunolocal-
ization of the cellular prion protein in normal brain. Microsc. Res. Tech. 50:58–65
Muramoto T, DeArmond SJ, Scott M, Telling GC, Cohen FE, Prusiner SB. 1997. Heritable
disorder resembling neuronal storage disease in mice expressing prion protein with deletion
of an α-helix. Nat. Med. 3:750–55
Muramoto T, Scott M, Cohen FE, Prusiner SB. 1996. Recombinant scrapie-like prion protein of
106 amino acids is soluble. Proc. Natl. Acad. Sci. USA 93:15457–62
Nazor KE, Kuhn F, Seward T, Green M, Zwald D, et al. 2005. Immunodetection of disease-
associated mutant PrP, which accelerates disease in GSS transgenic mice. EMBO J. 24:2472–
80
Nazor KE, Seward T, Telling GC. 2007. Motor behavioral and neuropathological deficits in mice
deficient for normal prion protein expression. Biochim. Biophys. Acta 1772:645–53
Neuendorf E, Weber A, Saalmueller A, Schatzl H, Reifenberg K, et al. 2004. Glycosylation
deficiency at either one of the two glycan attachment sites of cellular prion protein preserves
susceptibility to bovine spongiform encephalopathy and scrapie infections. J. Biol. Chem.
279:53306–16
Nieznanski K, Nieznanska H, Skowronek KJ, Osiecka KM, Stepkowski D. 2005. Direct interaction
between prion protein and tubulin. Biochem. Biophys. Res. Commun. 334:403–11
Nishida N, Tremblay P, Sugimoto T, Shigematsu K, Shirabe S, et al. 1999. A mouse prion protein
transgene rescues mice deficient for the prion protein gene from Purkinje cell degeneration
and demyelination. Lab. Invest. 79:689–97
Oidtmann B, Simon D, Holtkamp N, Hoffmann R, Baier M. 2003. Identification of cDNAs from
Japanese pufferfish (Fugu rubripes) and Atlantic salmon (Salmo salar) coding for homologues
to tetrapod prion proteins. FEBS Lett. 538:96–100
Pan T, Wong BS, Liu T, Li R, Petersen RB, Sy MS. 2002. Cell-surface prion protein interacts
with glycosaminoglycans. Biochem. J. 368:81–90
Parchi P, Capellari S, Chin S, Schwarz HB, Schecter NP, et al. 1999. A subtype of sporadic prion
disease mimicking fatal familial insomnia. Neurology 52:1757–63
Parkin ET, Watt NT, Hussain I, Eckman EA, Eckman CB, et al. 2007. Cellular prion protein
regulates beta-secretase cleavage of the Alzheimer’s amyloid precursor protein. Proc. Natl.
Acad. Sci. USA 104:11062–67
Parton RG, Richards AA. 2003. Lipid rafts and caveolae as portals for endocytosis: new insights
and common mechanisms. Traffic 4:724–38
Peden AH, Head MW, Ritchie DL, Bell JE, Ironside JW. 2004. Preclinical vCJD after blood
transfusion in a PRNP codon 129 heterozygous patient. Lancet 364:527–29

www.annualreviews.org • The Prion’s Elusive Reason for Being 473


Peters PJ, Mironov A Jr, Peretz D, van Donselaar E, Leclerc E, et al. 2003. Trafficking of prion
proteins through a caveolae-mediated endosomal pathway. J. Cell Biol. 162:703–17
Petrakis S, Sklaviadis T. 2006. Identification of proteins with high affinity for refolded and native
PrPC. Proteomics 6:6476–84
Prado MA, Alves-Silva J, Magalhaes AC, Prado VF, Linden R, et al. 2004. PrPc on the road:
trafficking of the cellular prion protein. J. Neurochem. 88:769–81
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Priola SA, Lawson VA. 2001. Glycosylation influences cross-species formation of protease-
resistant prion protein. EMBO J. 20:6692–99
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Prusiner SB, Scott MR. 1997. Genetics of prions. Annu. Rev. Genet. 31:139–75
Qin K, Yang Y, Mastrangelo P, Westaway D. 2002. Mapping Cu(II) binding sites in prion proteins
by diethyl pyrocarbonate modification and matrix-assisted laser desorption ionization-time
of flight (MALDI-TOF) mass spectrometric footprinting. J. Biol. Chem. 277:1981–90
Radovanovic I, Braun N, Giger OT, Mertz K, Miele G, et al. 2005. Truncated prion protein and
Doppel are myelinotoxic in the absence of oligodendrocytic PrPC. J. Neurosci. 25:4879–88
Re L, Rossini F, Re F, Bordicchia M, Mercanti A, et al. 2006. Prion protein potentiates acetyl-
choline release at the neuromuscular junction. Pharmacol. Res. 53:62–68
Rieger R, Edenhofer F, Lasmezas CI, Weiss S. 1997. The human 37-kDa laminin receptor pre-
cursor interacts with the prion protein in eukaryotic cells. Nat. Med. 3:1383–88
Riek R, Hornemann S, Wider G, Billeter M, Glockshuber R, Wuthrich K. 1996. NMR structure
of the mouse prion protein domain PrP(121–321). Nature 382:180–82
Rivera-Milla E, Oidtmann B, Panagiotidis CH, Baier M, Sklaviadis T, et al. 2006. Disparate
evolution of prion protein domains and the distinct origin of Doppel- and prion-related loci
revealed by fish-to-mammal comparisons. FASEB J. 20:317–19
Rivera-Milla E, Stuermer CA, Malaga-Trillo E. 2003. An evolutionary basis for scrapie disease:
identification of a fish prion mRNA. Trends Genet. 19:72–75
Roikhel VM, Fokina GI, Sobolev SG, Korolev MB, Ravkina LI, Pogodina VV. 1983. Study of early
stages of the pathogenesis of scrapie in experimentally infected mice. Acta Virol. 27:147–53
Rossi D, Cozzio A, Flechsig E, Klein MA, Rulicke T, et al. 2001. Onset of ataxia and Purkinje
cell loss in PrP null mice inversely correlated with Dpl level in brain. EMBO J. 20:694–702
Roucou X, Giannopoulos PN, Zhang Y, Jodoin J, Goodyer CG, LeBlanc A. 2005. Cellular prion
protein inhibits proapoptotic Bax conformational change in human neurons and in breast
carcinoma MCF-7 cells. Cell Death Differ. 12:783–95
Roucou X, Guo Q, Zhang Y, Goodyer CG, LeBlanc AC. 2003. Cytosolic prion protein is not
toxic and protects against Bax-mediated cell death in human primary neurons. J. Biol. Chem.
278:40877–81
Roucou X, LeBlanc AC. 2005. Cellular prion protein neuroprotective function: implications in
prion diseases. J. Mol. Med. 83:3–11
Rousseau C, Felin M, Doyennette-Moyne MA, Seve AP. 1997. CBP70, a glycosylated nuclear
lectin. J. Cell. Biochem. 66:370–85
Rybner C, Finel-Szermanski S, Felin M, Sahraoui T, Rousseau C, et al. 2002. The cellular prion
protein: a new partner of the lectin CBP70 in the nucleus of NB4 human promyelocytic
leukemia cells. J. Cell. Biochem. 84:408–19
Saa P, Castilla J, Soto C. 2006. Presymptomatic detection of prions in blood. Science 313:92–94
Sakaguchi S, Katamine S, Nishida N, Moriuchi R, Shigematsu K, et al. 1996. Loss of cerebellar
Purkinje cells in aged mice homozygous for a disrupted PrP gene. Nature 380:528–31
Sakai T, Hohjoh H. 2006. Gene silencing analyses against amyloid precursor protein (APP) gene
family by RNA interference. Cell Biol. Int. 30:952–56

474 Aguzzi · Baumann · Bremer


Salehi AH, Roux PP, Kubu CJ, Zeindler C, Bhakar A, et al. 2000. NRAGE, a novel MAGE protein,
interacts with the p75 neurotrophin receptor and facilitates nerve growth factor-dependent
apoptosis. Neuron 27:279–88
Sales N, Rodolfo K, Hassig R, Faucheux B, Di Giamberardino L, Moya KL. 1998. Cellular prion
protein localization in rodent and primate brain. Eur. J. Neurosci. 10:2464–71
Santuccione A, Sytnyk V, Leshchyns’ka I, Schachner M. 2005. Prion protein recruits its neuronal
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

receptor NCAM to lipid rafts to activate p59fyn and to enhance neurite outgrowth. J. Cell
Biol. 169:341–54
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Schmitt-Ulms G, Hansen K, Liu J, Cowdrey C, Yang J, et al. 2004. Time-controlled transcardiac


perfusion cross-linking for the study of protein interactions in complex tissues. Nat. Biotechnol.
22:724–31
Schmitt-Ulms G, Legname G, Baldwin MA, Ball HL, Bradon N, et al. 2001. Binding of neural
cell adhesion molecules (N-CAMs) to the cellular prion protein. J. Mol. Biol. 314:1209–25
Schneider B, Mutel V, Pietri M, Ermonval M, Mouillet-Richard S, Kellermann O. 2003. NADPH
oxidase and extracellular regulated kinases 1/2 are targets of prion protein signaling in neu-
ronal and nonneuronal cells. Proc. Natl. Acad. Sci. USA 100:13326–31
Shmerling D, Hegyi I, Fischer M, Blattler T, Brandner S, et al. 1998. Expression of amino-
terminally truncated PrP in the mouse leading to ataxia and specific cerebellar lesions. Cell
93:203–14
Shyng SL, Heuser JE, Harris DA. 1994. A glycolipid-anchored prion protein is endocytosed via
clathrin-coated pits. J. Cell. Biol. 125:1239–50
Shyu WC, Lin SZ, Chiang MF, Ding DC, Li KW, et al. 2005. Overexpression of PrPC by
adenovirus-mediated gene targeting reduces ischemic injury in a stroke rat model. J. Neurosci.
25:8967–77
Simonic T, Duga S, Strumbo B, Asselta R, Ceciliani F, Ronchi S. 2000. cDNA cloning of turtle
prion protein. FEBS Lett. 469:33–38
Spielhaupter C, Schatzl HM. 2001. PrPC directly interacts with proteins involved in signaling
pathways. J. Biol. Chem. 276:44604–12
Spudich A, Frigg R, Kilic E, Kilic U, Oesch B, et al. 2005. Aggravation of ischemic brain injury
by prion protein deficiency: role of ERK-1/-2 and STAT-1. Neurobiol. Dis. 20:442–49
Stahl N, Borchelt DR, Hsiao K, Prusiner SB. 1987. Scrapie prion protein contains a phosphatidyli-
nositol glycolipid. Cell 51:229–40
Steele AD, Emsley JG, Ozdinler PH, Lindquist S, Macklis JD. 2006. Prion protein (PrPc) pos-
itively regulates neural precursor proliferation during developmental and adult mammalian
neurogenesis. Proc. Natl. Acad. Sci. USA 103:3416–21
Stewart RS, Harris DA. 2001. Most pathogenic mutations do not alter the membrane topology
of the prion protein. J. Biol. Chem. 276:2212–20
Stewart RS, Harris DA. 2005. A transmembrane form of the prion protein is localized in the Golgi
apparatus of neurons. J. Biol. Chem. 280:15855–64
Stewart RS, Piccardo P, Ghetti B, Harris DA. 2005. Neurodegenerative illness in transgenic mice
expressing a transmembrane form of the prion protein. J. Neurosci. 25:3469–77
Stockel J, Safar J, Wallace AC, Cohen FE, Prusiner SB. 1998. Prion protein selectively binds
copper(II) ions. Biochemistry 37:7185–93
Strumbo B, Ronchi S, Bolis LC, Simonic T. 2001. Molecular cloning of the cDNA coding for
Xenopus laevis prion protein. FEBS Lett. 508:170–74
Sun G, Guo M, Shen A, Mei F, Peng X, et al. 2005. Bovine PrPC directly interacts with αB-
crystalline. FEBS Lett. 579:5419–24
Sunyach C, Jen A, Deng J, Fitzgerald KT, Frobert Y, et al. 2003. The mechanism of internaliza-
tion of glycosylphosphatidylinositol-anchored prion protein. EMBO J. 22:3591–601

www.annualreviews.org • The Prion’s Elusive Reason for Being 475


Supattapone S, Bouzamondo E, Ball HL, Wille H, Nguyen HO, et al. 2001. A protease-
resistant 61-residue prion peptide causes neurodegeneration in transgenic mice. Mol. Cell
Biol. 21:2608–16
Suzuki T, Kurokawa T, Hashimoto H, Sugiyama M. 2002. cDNA sequence and tissue expression
of Fugu rubripes prion protein-like: a candidate for the teleost orthologue of tetrapod PrPs.
Biochem. Biophys. Res. Commun. 294:912–17
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Tateishi J, Brown P, Kitamoto T, Hoque ZM, Roos R, et al. 1995. First experimental transmission
of fatal familial insomnia. Nature 376:434–35
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Tateishi J, Kitamoto T, Hashiguchi H, Shii H. 1988. Gerstmann-Sträussler-Scheinker disease:


immunohistological and experimental studies. Ann. Neurol. 24:35–40
Tateishi J, Kitamoto T, Hoque MZ, Furukawa H. 1996a. Experimental transmission of
Creutzfeldt-Jakob disease and related diseases to rodents. Neurology 46:532–37
Tateishi J, Kitamoto T, Kretzschmar H, Mehraein P. 1996b. Immunhistological evaluation of
Creutzfeldt-Jakob disease with reference to the type PrPres deposition. Clin. Neuropathol.
15:358–60
Taylor DR, Hooper NM. 2007. The low-density lipoprotein receptor-related protein 1 (LRP1)
mediates the endocytosis of the cellular prion protein. Biochem. J. 402:17–23
Taylor DR, Watt NT, Perera WS, Hooper NM. 2005. Assigning functions to distinct regions
of the N-terminus of the prion protein that are involved in its copper-stimulated, clathrin-
dependent endocytosis. J. Cell Sci. 118:5141–53
Telling GC, Haga T, Torchia M, Tremblay P, DeArmond SJ, Prusiner SB. 1996. Interactions
between wild-type and mutant prion proteins modulate neurodegeneration in transgenic
mice. Genes Dev. 10:1736–50
Tobler I, Gaus SE, Deboer T, Achermann P, Fischer M, et al. 1996. Altered circadian activity
rhythms and sleep in mice devoid of prion protein. Nature 380:639–42
Toni M, Spisni E, Griffoni C, Santi S, Riccio M, et al. 2006. Cellular prion protein and caveolin-
1 interaction in a neuronal cell line precedes fyn/erk 1/2 signal transduction. J. Biomed.
Biotechnol. 2006:69469
Vassallo N, Herms J. 2003. Cellular prion protein function in copper homeostasis and redox
signalling at the synapse. J. Neurochem. 86:538–44
Vey M, Pilkuhn S, Wille H, Nixon R, DeArmond SJ, et al. 1996. Subcellular colocalization of the
cellular and scrapie prion proteins in caveolae-like membranous domains. Proc. Natl. Acad.
Sci. USA 93:14945–49
Volkel D, Zimmermann K, Zerr I, Bodemer M, Lindner T, et al. 2001. Immunochemical deter-
mination of cellular prion protein in plasma from healthy subjects and patients with sporadic
CJD or other neurologic diseases. Transfusion 41:441–48
Waggoner DJ, Drisaldi B, Bartnikas TB, Casareno RL, Prohaska JR, et al. 2000. Brain copper
content and cuproenzyme activity do not vary with prion protein expression level. J. Biol.
Chem. 275:7455–58
Walis A, Bratosiewicz J, Sikorska B, Brown P, Gajdusek DC, Liberski PP. 2003. Ultrastructural
changes in the optic nerves of rodents with experimental Creutzfeldt-Jakob Disease (CJD),
Gerstmann-Sträussler-Scheinker disease (GSS) or scrapie. J. Comp. Pathol. 129:213–25
Walker L, Levine H, Jucker M. 2006. Koch’s postulates and infectious proteins. Acta Neuropathol.
112:1–4
Warner RG, Hundt C, Weiss S, Turnbull JE. 2002. Identification of the heparan sulfate binding
sites in the cellular prion protein. J. Biol. Chem. 277:18421–30
Weise J, Crome O, Sandau R, Schulz-Schaeffer W, Bahr M, Zerr I. 2004. Upregulation of cellular
prion protein (PrPc) after focal cerebral ischemia and influence of lesion severity. Neurosci.
Lett. 372:146–50

476 Aguzzi · Baumann · Bremer


Weise J, Sandau R, Schwarting S, Crome O, Wrede A, et al. 2006. Deletion of cellular prion
protein results in reduced Akt activation, enhanced postischemic caspase-3 activation, and
exacerbation of ischemic brain injury. Stroke 37:1296–300
Weissmann C, Flechsig E. 2003. PrP knock-out and PrP transgenic mice in prion research. Br.
Med. Bull. 66:43–60
Wells GA, Scott AC, Johnson CT, Gunning RF, Hancock RD, et al. 1987. A novel progressive
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

spongiform encephalopathy in cattle. Vet. Rec. 121:419–20


Westaway D, DeArmond SJ, Cayetano-Canlas J, Groth D, Foster D, et al. 1994. Degeneration
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

of skeletal muscle, peripheral nerves, and the central nervous system in transgenic mice
overexpressing wild-type prion proteins. Cell 76:117–29
Westergard L, Christensen HM, Harris DA. 2007. The cellular prion protein (PrP(C)): its phys-
iological function and role in disease. Biochim. Biophys. Acta 1772:629–44
White AR, Collins SJ, Maher F, Jobling MF, Stewart LR, et al. 1999. Prion protein-deficient
neurons reveal lower glutathione reductase activity and increased susceptibility to hydrogen
peroxide toxicity. Am. J. Pathol. 155:1723–30
Will RG. 2003. Acquired prion disease: iatrogenic CJD, variant CJD, kuru. Br. Med. Bull. 66:255–
65
Wong BS, Brown DR, Pan T, Whiteman M, Liu T, et al. 2001a. Oxidative impairment in scrapie-
infected mice is associated with brain metals perturbations and altered antioxidant activities.
J. Neurochem. 79:689–98
Wong BS, Liu T, Li R, Pan T, Petersen RB, et al. 2001b. Increased levels of oxidative stress
markers detected in the brains of mice devoid of prion protein. J. Neurochem. 76:565–72
Wong BS, Liu T, Paisley D, Li R, Pan T, et al. 2001c. Induction of HO-1 and NOS in Doppel-
expressing mice devoid of PrP: implications for Doppel function. Mol. Cell Neurosci. 17:768–75
Wroe SJ, Pal S, Siddique D, Hyare H, Macfarlane R, et al. 2006. Clinical presentation and
premortem diagnosis of variant Creutzfeldt-Jakob disease associated with blood transfusion:
a case report. Lancet 368:2061–67
Yehiely F, Bamborough P, Da Costa M, Perry BJ, Thinakaran G, et al. 1997. Identification of
candidate proteins binding to prion protein. Neurobiol. Dis. 3:339–55
York RD, Molliver DC, Grewal SS, Stenberg PE, McCleskey EW, Stork PJ. 2000. Role of phos-
phoinositide 3-kinase and endocytosis in nerve growth factor-induced extracellular signal-
regulated kinase activation via Ras and Rap1. Mol. Cell Biol. 20:8069–83
Zanata SM, Lopes MH, Mercadante AF, Hajj GN, Chiarini LB, et al. 2002. Stress-inducible
protein 1 is a cell surface ligand for cellular prion that triggers neuroprotection. EMBO J.
21:3307–16
Zarranz JJ, Digon A, Atares B, Rodriguez-Martinez AB, Arce A, et al. 2005. Phenotypic variability
in familial prion diseases due to the D178N mutation. J. Neurol. Neurosurg. Psychiatry 76:1491–
96
Zhang CC, Steele AD, Lindquist S, Lodish HF. 2006. Prion protein is expressed on long-term
repopulating hematopoietic stem cells and is important for their self-renewal. Proc. Natl. Acad.
Sci. USA 103:2184–89
Zhang Y, Moheban DB, Conway BR, Bhattacharyya A, Segal RA. 2000. Cell surface Trk re-
ceptors mediate NGF-induced survival while internalized receptors regulate NGF-induced
differentiation. J. Neurosci. 20:5671–78

www.annualreviews.org • The Prion’s Elusive Reason for Being 477


AR346-FM ARI 20 May 2008 15:1

Annual Review of
Neuroscience

Contents Volume 31, 2008


by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Cerebellum-Like Structures and Their Implications for Cerebellar


Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

Function
Curtis C. Bell, Victor Han, and Nathaniel B. Sawtell ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 1
Spike Timing–Dependent Plasticity: A Hebbian Learning Rule
Natalia Caporale and Yang Dan ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! !25
Balancing Structure and Function at Hippocampal Dendritic Spines
Jennifer N. Bourne and Kristen M. Harris ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! !47
Place Cells, Grid Cells, and the Brain’s Spatial Representation System
Edvard I. Moser, Emilio Kropff, and May-Britt Moser ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! !69
Mitochondrial Disorders in the Nervous System
Salvatore DiMauro and Eric A. Schon ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! !91
Vestibular System: The Many Facets of a Multimodal Sense
Dora E. Angelaki and Kathleen E. Cullen ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 125
Role of Axonal Transport in Neurodegenerative Diseases
Kurt J. De Vos, Andrew J. Grierson, Steven Ackerley, and Christopher C.J. Miller ! ! ! 151
Active and Passive Immunotherapy for Neurodegenerative Disorders
David L. Brody and David M. Holtzman ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 175
Descending Pathways in Motor Control
Roger N. Lemon ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 195
Task Set and Prefrontal Cortex
Katsuyuki Sakai ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 219
Multiple Sclerosis: An Immune or Neurodegenerative Disorder?
Bruce D. Trapp and Klaus-Armin Nave ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 247
Multifunctional Pattern-Generating Circuits
K.L. Briggman and W.B. Kristan, Jr. ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 271
Retinal Axon Growth at the Optic Chiasm: To Cross or Not to Cross
Timothy J. Petros, Alexandra Rebsam, and Carol A. Mason ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 295

v
AR346-FM ARI 20 May 2008 15:1

Brain Circuits for the Internal Monitoring of Movements


Marc A. Sommer and Robert H. Wurtz ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 317
Wnt Signaling in Neural Circuit Assembly
Patricia C. Salinas and Yimin Zou ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 339
Habits, Rituals, and the Evaluative Brain
Ann M. Graybiel ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 359
Mechanisms of Self-Motion Perception
Kenneth H. Britten ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 389
by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Mechanisms of Face Perception


Doris Y. Tsao and Margaret S. Livingstone ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 411
Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org

The Prion’s Elusive Reason for Being


Adriano Aguzzi, Frank Baumann, and Juliane Bremer ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 439
Mechanisms Underlying Development of Visual Maps and
Receptive Fields
Andrew D. Huberman, Marla B. Feller, and Barbara Chapman ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 479
Neural Substrates of Language Acquisition
Patricia K. Kuhl and Maritza Rivera-Gaxiola ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 511
Axon-Glial Signaling and the Glial Support of Axon Function
Klaus-Armin Nave and Bruce D. Trapp ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 535
Signaling Mechanisms Linking Neuronal Activity to Gene Expression
and Plasticity of the Nervous System
Steven W. Flavell and Michael E. Greenberg ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 563

Indexes

Cumulative Index of Contributing Authors, Volumes 22–31 ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 591


Cumulative Index of Chapter Titles, Volumes 22–31 ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 595

Errata

An online log of corrections to Annual Review of Neuroscience articles may be found at


http://neuro.annualreviews.org/

vi Contents

Anda mungkin juga menyukai