41
The Prion’s Elusive Reason for Being Adriano Aguzzi, Frank Baumann, and Juliane Bremer Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland, email: [email protected] Annu. Rev. Neurosci. 2008. 31:439–77 First published online as a Review in Advance on April 2, 2008 The Annual Review of Neuroscience is online at neuro.annualreviews.org This article’s doi: 10.1146/annurev.neuro.31.060407.125620 Copyright c 2008 by Annual Reviews. All rights reserved 0147-006X/08/0721-0439$20.00 Key Words transmissible spongiform encephalopathy, prion diseases, PrP C , PrP Sc Abstract The protein-only hypothesis posits that the infectious agent causing transmissible spongiform encephalopathies consists of protein and lacks any informational nucleic acids. This agent, termed prion by Stanley Prusiner, is thought to consist partly of PrP Sc , a conformational isoform of a normal cellular protein termed PrP C . Scientists and lay persons have been fascinated by the prion concept, and it has been subjected to passionate critique and intense experimental scrutiny. As a result, PrP C and its isoforms rank among the most intensively studied proteins encoded by the mammalian genome. Despite all this research, both the physiological function of PrP C 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 PrP C may help explain the toxic phenotypes observed in prion disease. 439 Click here for quick links to Annual Reviews content online, including: • Other articles in this volume • Top cited articles • Top downloaded articles • Our comprehensive search Further ANNUAL REVIEWS Annu. Rev. Neurosci. 2008.31:439-477. Downloaded from www.annualreviews.org by Scuola Internazionale Superiore di Studi Avanzati (SISSA) on 03/21/11. For personal use only.

Artigo 1- The Prion´s Elusive Reason for Being

Embed Size (px)

Citation preview

Page 1: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

The Prion’s Elusive Reasonfor BeingAdriano Aguzzi, Frank Baumann, and Juliane BremerInstitute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland,email: [email protected]

Annu. Rev. Neurosci. 2008. 31:439–77

First published online as a Review in Advance onApril 2, 2008

The Annual Review of Neuroscience is online atneuro.annualreviews.org

This article’s doi:10.1146/annurev.neuro.31.060407.125620

Copyright c! 2008 by Annual Reviews.All rights reserved

0147-006X/08/0721-0439$20.00

Key Wordstransmissible spongiform encephalopathy, prion diseases, PrPC, PrPSc

AbstractThe protein-only hypothesis posits that the infectious agent causingtransmissible spongiform encephalopathies consists of protein and lacksany informational nucleic acids. This agent, termed prion by StanleyPrusiner, is thought to consist partly of PrPSc, a conformational isoformof a normal cellular protein termed PrPC. Scientists and lay personshave been fascinated by the prion concept, and it has been subjectedto passionate critique and intense experimental scrutiny. As a result,PrPC and its isoforms rank among the most intensively studied proteinsencoded by the mammalian genome. Despite all this research, both thephysiological function of PrPC and the molecular pathways leading toneurodegeneration in prion disease remain unknown. Here we reviewthe salient traits of those diseases ascribed to improper behavior of theprion protein and highlight how the physiological functions of PrPC

may help explain the toxic phenotypes observed in prion disease.

439

Click here for quick links to Annual Reviews content online, including:

• Other articles in this volume• Top cited articles• Top downloaded articles• Our comprehensive search

FurtherANNUALREVIEWS

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 2: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

GPI: glycosylphos-phatidylinositol

ContentsTRANSMISSIBLE PRION

DISEASES ANDNONTRANSMISSIBLEPRION-RELATED DISEASES. . . . 440Prion Disease in Humans

and Animals . . . . . . . . . . . . . . . . . . . . 441The Nature of the Prion . . . . . . . . . . . 445Neurotoxicity . . . . . . . . . . . . . . . . . . . . . . 447PHYSIOLOGICAL FUNCTION

OF THE CELLULAR PRIONPROTEIN . . . . . . . . . . . . . . . . . . . . . 447

Prion Protein–Deficient Mice . . . . . . 447Functional Domains

of the Prion Protein . . . . . . . . . . . . . 449Point Mutations within

the Prion Protein . . . . . . . . . . . . . . . 451Evolution of the Prion Protein. . . . . . 452Cellular Processes Influenced

by PrPC Expression . . . . . . . . . . . . . 452PrP and the Immune System . . . . . . . 457Molecular Mechanisms Mediating

the Function of PrPC . . . . . . . . . . . 457Interaction Partners of PrPC . . . . . . . . 461

CONCLUSIONS . . . . . . . . . . . . . . . . . . . . 465

TRANSMISSIBLE PRIONDISEASES ANDNONTRANSMISSIBLEPRION-RELATED DISEASESThe origin of the word “prion” stems from theanagram of “proteinaceous infectious particle.”Naturally, the latter is by no means a qualify-ing attribute because all conventional infectiousagents—including all viruses and bacteria—areproteinaceous to some degree. What sets prionsapart, as proposed by Prusiner, is that the actualinfectious principle consists merely of proteinand is capable of replicating and transmittinginfections without the need for informationalnucleic acids. This postulate counters much ofthe established molecular biological evidence,which predicates that nucleic acids are the basisfor self-replicating biological information in all

living beings, including even the most elemen-tary infectious particles.

Prion diseases are generally characterizedby widespread neurodegeneration and there-fore exhibit clinical signs and symptoms of cog-nitive and motor dysfunction, in addition topropagating infectious prions and, in many in-stances, forming striking amyloid plaques. Thelatter plaques contain aggregates of PrPSc, amisfolded and beta-sheet-rich isoform of theprotein PrPC encoded by the PRNP gene. Fur-ther neuropathological features are neuronalloss, astrocytic activation (gliosis), and spongi-form change. All prion diseases are progressive,fatal, and presently incurable.

Although the normal cellular prion proteinPrPC can easily be digested with proteinase K(PK), the beta-sheet-rich, misfolded form PrPSc

is partially proteinase K (PK) resistant. A cru-cial piece of evidence demonstrating that PrPC

is a key player in prion disease came from ex-periments showing that mice lacking the prionprotein gene are resistant to prions (Bueler et al.1993).

Although the formation of PrPSc accompa-nies neurodegeneration in prion disease, manylines of evidence indicate that PrPSc is not in-trinsically neurotoxic. PrPC needs to be pre-sented by host neurons for neurodegenerationto occur. Thus, when neurografts propagat-ing PrPSc were implanted into Prnpo/o mice,the host mice did not develop prion disease(Brandner et al. 1996). Additionally, trans-genic mice expressing only a secreted form ofPrPC, lacking its membrane attachment via gly-cosylphosphatidylinositol (GPI) anchor, havebeen reported to be refractory to develop clini-cal signs of prion diseases, although prion inoc-ulation induces PrPSc formation and aggrega-tion of amyloid plaques (Chesebro et al. 2005).This finding indicates that membrane attach-ment of PrPC is a prerequisite for neurode-generation to occur and that the presence ofPrPSc alone does not cause disease. The im-portance of neuronal expression of PrPC forprion disease development has been corrobo-rated by the phenotype of mice with neuron-specific ablation of PrPC eight weeks after prion

440 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 3: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

inoculation. Early spongiform changes were re-versed, and clinical disease was prevented. Thisreversal occurred despite the accumulation ofextraneuronal PrPSc (Mallucci et al. 2003).

Bona fide prion diseases are character-ized by their transmissibility and are there-fore also termed transmissible spongiform en-cephalopathies (TSE). Transmissibility is adefining, and hence indispensable, trait of allprion diseases. However, transmissibility hasnot been formally proven for all kinds of dis-eases thought to be caused by prions. In ad-dition, some diseases are genetically associatedwith the prion protein, yet they are nontrans-missible. These diseases are sometimes calledprionopathies. Among these are rare geneticsyndromes that cosegregate with point muta-tions in the open reading frame of the PRNPgene. In addition to these naturally occurringprionopathies, several transgenic mice havebeen used to gain insight into functional do-mains of PrPC (Weissmann & Flechsig 2003).In these mice, deletion of parts of PrPC causedprionopathies, characterized by a shortened lifespan and the development of white matterdisease in the central nervous system (CNS)as well as neuronal cell death in the cere-bellum (Baumann et al. 2007, Li et al. 2007,Shmerling et al. 1998). Overexpression of wild-type PrPC also caused disease in transgenic mice(Westaway et al. 1994).

Prion Disease in Humans and AnimalsPrion diseases have occurred in humans and an-imals for many years. A disease similar to scrapiewas recorded in the mid eighteenth century,and scholars heavily debated its origin. A crucialexperiment showing incontrovertible trans-missibility of scrapie to goats was performed byCuille & Chelle in the 1930s (Cuille & Chelle1939). The first cases of human prion disease,Creutzfeldt-Jakob disease (CJD), were reportedin the 1920s (Creutzfeldt 1920, Jakob 1921).The number of human and animal diseasesrecognized as TSEs has increased steadily andnow includes Gerstmann-Straussler-Scheinkersyndrome (GSS), fatal familial insomnia (FFI),

TSE: transmissiblespongiformencephalopathies

CJD: Creutzfeldt-Jakob disease

BSE: bovinespongiformencephalopathy

CWD: chronicwasting disease

FSE: felinespongiformencephalopathy

sCJD: sporadic CJD

and Kuru in humans; bovine spongiform en-cephalopathy (BSE) in cattle; chronic wastingdisease (CWD) in deer and elk; and trans-missible mink encephalopathy. BSE has beeninadvertently transmitted to a variety of captiveanimals, causing feline spongiform en-cephalopathy (FSE) and a plethora of diseasesin zoo animals including kudus, nyalas, andgreater cats, for example.

Creutzfeldt-Jakob disease. CJD was initiallydescribed as a sporadic disease occurring forno known cause (sCJD). The incidence ofCJD is low in all ethnicities and typically af-fects !1 person in one million each year. Veryrapid cognitive decline, causing dementia, is themain symptom. Cerebellar symptoms includ-ing ataxia and myoclonus are also frequent pre-senting symptoms. Death often occurs withinfew weeks of the first signs of disease, anda fulminant, “apoplectiform” course of dis-ease has been documented in the past. So-matic mutations in the PRNP gene analogousto those in the germline of genetic CJD pa-tients (see below) have been hypothesized tounderlie sporadic CJD. Alternatively, Aguzzi& Glatzel (2006) suggested that some casesof alleged sCJD derive from heretofore un-recognized infections. Finally, PrPC may pos-sess a finite, albeit extremely low, propensity toself-assemble into ordered aggregates of PrPSc,thereby stochastically initiating prion replica-tion and, ultimately, a sporadic form of disease.The latter scenario could be regarded as thebad-luck hypothesis. However, none of this hasbeen proven, and therefore the cause of sCJDis still unknown.

Variant Creutzfeldt-Jakob disease andBovine Spongiform Encephalopathy. Pub-lic understanding of prion disease remainedlimited for a long time: For example, wehave heard neurologists saying that CJD isan essentially nonexistent disease. However,this mindset changed completely when BSEwas first reported in the early 1980s (Wellset al. 1987). In the following years and untilmid 2007, BSE affected !190,000 cows

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 4: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

vCJD: variant CJD

(http://www.oie.int/). Some investigatorssuggested that BSE could cause a new variantform of CJD (vCJD) in humans. A directexperimental proof that vCJD representstransmission of BSE prions to humans cannotbe produced. However, epidemiological,biochemical, neuropathological evidence andtransmission studies strongly suggest that BSEhas transmitted to humans in the form of vCJD(Aguzzi 1996, Aguzzi & Weissmann 1996,Bruce et al. 1997, Hill et al. 1997). The inci-dence of vCJD has been rising between 1994,when the first patients suffering from vCJDpresented with their initial symptoms, and2001, raising fears that a very large epidemicmay be looming. At the time of this writing,vCJD has killed !200 individual victims world-wide (http://www.cjd.ed.ac.uk/). Most of theaffected individuals lived in United Kingdomand France. Fortunately, in the United King-dom the incidence appears to be decreasingfrom the year 2001 to 6 diagnosed cases yearlyin 2005 and 2006. In contrast, in France thenumber of probable and definite cases of vCJDincreased from 0 to 3 diagnosed cases per yearin 1996–2004 to 6 per year in 2005 and 2006. In2007, the number of cases was back to 3 again.(http://www.invs.sante.fr/publications/mcj/donnees mcj.html). A 30+-year mean in-cubation time of BSE/vCJD in humans isnot entirely implausible, and therefore someauthors have predicted a multiphasic humanBSE endemic with a second increase in the in-cidence of vCJD affecting people heterozygousat codon 129 (Collinge et al. 2006). Others,these authors included, regard the incidenceof vCJD as subsiding (Andrews et al. 2003)(Figure 1).

It is important to note, however, that theabove considerations apply primarily to the epi-demiology of primary transmission from cowsto humans. Although, by now a pool of pre-clinically infected humans may have been built.Human-to-human transmission may presentwith characteristics very different from thoseof primary cow-to-human transmission, in-cluding enhanced virulence, shortened incu-bation times, disrespect of allelic PRNP poly-

morphisms (129MM, MV or VV), and hetero-dox modes of infection including blood-bornetransmission. If we account for the time it willtake to eradicate these secondary transmissionsin the population, vCJD is not likely to disap-pear entirely in the coming four decades.

Iatrogenic CJD. Iatrogenic CJD is acciden-tally transmitted during the course of med-ical or surgical procedures. The first docu-mented case of iatrogenic prion transmissionoccurred in 1974 and was caused by cornealtransplantation of a graft derived from a pa-tient suffering from sCJD (Duffy et al. 1974).Iatrogenic CJD is also rare, most often ob-served in individuals that have received cadav-eric dura mater implants and human growthhormone; some of these individuals receivedgonadotrophin extracted from human pitu-itary glands or had stereotactically placed elec-trodes in their brains (Will 2003). Four casesof vCJD transmission by blood transfusionshave been reported recently in the UnitedKingdom (Llewelyn et al. 2004, Peden et al.2004, Wroe et al. 2006) (see also http://www.cjd.ed.ac.uk/TMER/TMER.htm). The factthat preclinically infected individuals can trans-mit vCJD underscores the important medicalneed for sensitive diagnostic tools, which couldbe used for screening blood units prior to trans-fusion, for example.

Kuru. In the mid 1950s, when the remoteparts of Papua New Guinea were first exploredby Australians and Westerners, Kuru was firstdescribed in research (Gajdusek & Zigas 1957).Kuru was, at that time and at least since 1941,an endemic disease among some tribes ofNew Guinea aborigines, especially among theFore linguistic group and neighboring tribes(Gajdusek & Reid 1961). Kuru in the Fore lan-guage means “to shiver,” and along with othersigns of cerebellar ataxia, shivering is a hallmarkof the disease. The ritual consumption of deadrelatives as a symbol of respect and mourningis the attributed route of transmission. As aconsequence, the incidence has steadily fallenafter cessation of cannibalism in Papua New

442 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 5: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

0

5000

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

10,000

15,000

20,000

25,000

30,000

35,000

40,000

Nu

mb

er o

f ye

arly

iden

tifi

ed in

fect

ed c

ow

s

BSE UK

BSE non-UK

a

b

vCJD cases(dead)

SecondaryvCJD (bloodtransfusion)

UK 160 43France 21 –2Republic of Ireland 4 –0Italy 1 ––USA 3 ––Canada 1 ––Saudi Arabia – –1Japan 1 ––Netherlands 2 ––Portugal 1 –1Spain 2 ––

vCJD cases(still alive)

1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 20070

5

10

15

20

25

30

Nu

mb

er o

f ye

arly

dia

gn

ose

d c

ases

Cases UK

Cases France

Figure 1BSE 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 theEuropean 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 ofIreland, Italy, United States, Canada, Saudi Arabia, Japan, the Netherlands, Portugal, and Spain. Data are as of February 2008 andinclude cases of vCJD in patients who resided in the UK in the 1980s or 1990s [see the National Creutzfeldt-Jakob Disease SurveillanceUnit 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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 6: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

gCJD: genetic CJD

OR: octarepeat region

Guinea (Collinge et al. 2006). In a concise andextremely clairvoyant observation published in1959, Bill Hadlow noted the epidemiological,clinical, and neuropathological similaritiesbetween Kuru and scrapie (Hadlow 1959).These were taken up by Carleton Gajdusekwho, in 1966, succeeded in transmitting Kuruto three chimpanzees (Gajdusek et al. 1966).Soon thereafter, serial passage of Kuru and ofseveral other prion diseases was demonstratedin chimpanzees and other primates (Gajduseket al. 1967, 1968). Investigators have sincetransmitted human prion disease to variousspecies including laboratory rodents.

Genetic CJD and Gerstmann-Straussler-Scheinker syndrome. Several mutations inthe prion protein gene (PRNP) have been

found in families with hereditary or geneticCJD (gCJD). Figure 2 summarizes known mu-tations causing human TSEs. gCJD occurswith point mutations mostly affecting the re-gion between the second and the third he-lix of the carboxy-terminus. However, inser-tions in the octarepeat region (OR) in theamino-terminus, and even one instance of apremature termination codon at position 145,have also been associated with human priondisease. The inheritance was, in all cases,autosomal dominant, often with very high pen-etrance. The clinicopathological disease phe-notype varies depending on the actual muta-tion, as well as on polymorphisms at codon 129,and most likely on a plethora of yet uniden-tified modifiers and cofactors (Kovacs et al.2002).

CC1 CC2 HC H1 H2 H3 GPI OR

Mutations in PRNP associated with familial dementiaand/or neuropsychiatric symptoms(not further classified)

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

Mutations causing FFI D178N-129M

V180I E200K

T188K V210I T188R-129V

E196K E211Q

V203I

Mutations causing gCJD Octarepeat insertion D178N-129V E208H M232R

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

Y145*-129M D202N-129V

Q212P

G114V Q160*-129M N171S T183A

H187R

S S

Figure 2The human PrPC protein and its mutants. The mature human PrPC protein contains 208 amino acid residues. It features two positivelycharged amino acid clusters denoted CC1 and CC2 (blue boxes), an octapeptide repeat region (OR) ( green boxes), a hydrophobic core(HC) ( gray box), three !-helixes (H1-H3) (red boxes), one disulphide bond (S–S) between cysteine residues 179 and 214, and twopotential sites for N-linked glycosylation (red forks) at residues 181 and 197. A glycosylphosphatidylinositol anchor (GPI) ( yellow box) isattached to the C-terminus of PrP. This figure indicates in black framed boxes point mutations and insertions found in the humanPRNP 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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 7: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

The first descriptions of Gerstmann-Straussler-Scheinker syndrome (GSS) origi-nate from 1928 and 1936 in an Austrian family(Gerstmann 1928, Gerstmann et al. 1936). Inthe following years, analogous disorders havebeen described, but its classification as a TSElagged until 1981, when Masters and colleagues(1981) reported that inoculation of brain tis-sue from three patients with GSS resulted inspongiform encephalopathy in nonhuman pri-mates. The authors also defined clinical hall-marks of GSS (earlier age at onset, longerdisease duration, and prominent cerebellarataxia) differentiating the disease from CJD.Nowadays GSS is considered an autosomal-dominantly inherited TSE caused by muta-tions in the prion protein open reading frame,manifesting typically with progressive cerebel-lar ataxia or spastic paraparesis and cognitivedecline. The known GSS-causing mutations aresummarized in Figure 2. In addition to the re-gions affected in gCJD, mutations altering thesequence of the central domain can cause GSS.Its distinctive neuropathological feature is thepresence of widespread large and multicentricamyloid plaques (Collins et al. 2001).

GSS is generally transmissible (Hsiao et al.1989, Masters et al. 1981, Tateishi et al. 1988);therefore, its classification as a TSE is widelyaccepted. However, the overall experimentaltransmissibility of GSS to nonhuman primatesand rodents is low. Only for the most com-mon GSS-associated mutations (P102L), andonly in approximately one third of the cases,were brain homogenates derived from patientsreproducibly capable of inducing disease upontransmission (Tateishi et al. 1996a). The lessfrequent mutations causing GSS often failedto induce disease after experimental transmis-sion to nonhuman primates and rodents, and inmany cases transmissibility was never assessed(Brown et al. 1994, Tateishi et al. 1996a).

Fatal familial insomnia. Fatal familial insom-nia (FFI) is the descriptive name given to a dis-ease identified in 1986. Five members of an Ital-ian family presented with insomnia and dysau-tonomia (Lugaresi et al. 1986). In 1992, the

GSS: Gerstmann-Straussler-Scheinkersyndrome

FFI: fatal familialinsomnia

disease-causing mutation in the prion proteingene (D178N) was identified, thereby allow-ing the classification of FFI as a genetically de-termined prion disease (Medori et al. 1992).The final proof that FFI is a TSE was achievedwhen FFI was successfully transmitted to mice(Tateishi et al. 1995). FFI typically affects thethalamus, and accordingly, the core clinical fea-tures are disruption of the normal sleep-wakecycle, sympathetic overactivity, endocrine ab-normalities, and impaired attention (Collinset al. 2001). In addition to the pathogenic pointmutation D178N, the methionine-valine poly-morphism at codon 129 of the PRNP gene con-trols the disease phenotype. Whereas D178N-129MM (homozygosity for methionine atcodon 129) was associated with FFI, heterozy-gosity at codon 129 (D178N-129MV) segre-gated with the familial CJD subtype (Goldfarbet al. 1992). However, Zarranz et al. (2005)reported more recently that this genotype-phenotype association is not absolute. In onestudy, several patients have been identified witha CJD phenotype and a D178N-129MM geno-type. The authors concluded that rather thanbeing separate disease entities, prion diseasephenotypes such as FFI and CJD represent twoextreme manifestations of a continuous diseasespectrum (Zarranz et al. 2005).

In addition to the familial form of fatal in-somnia, a sporadic form of the disease, termedsporadic fatal insomnia, was described. Spo-radic FFI is not associated with mutations inthe PRNP gene (Mastrianni et al. 1999, Parchiet al. 1999).

The Nature of the PrionAlthough formulated a century ago, Koch’s pos-tulates remain the bedrock of microbiology. Ac-cording to Koch, three conditions must be metto identify a microbe as the causative agentof any given infection: (a) The microorganismmust be detectable in all diseased tissues, (b) itsisolation and growth must be achieved in pureculture, and (c) the culture-derived microorgan-isms must be able to induce disease after ex-perimental infection of a subject, from which

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 8: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

PMCA: proteinmisfolding cyclicamplification

a further round of reisolation of the microor-ganism should be possible. Although Koch’swork was performed long before contemporarymolecular biology, his postulates continue toserve remarkably well in defining conventionalviral and bacterial agents.

However, as prions are thought to be in-fectious proteins that amplify in a self-catalyticmisfolding process, their microbiological cul-ture sensu strictiori is not possible. Thereforewhether Koch’s postulates can be meaningfullyapplied to prion disease is questionable. Fur-thermore, Koch’s postulates account for the in-fluence of host susceptibility, which is of utmostimportance in prion disease. Prion disease de-velopment depends on the presence of PrPC

on host cells, and the species-specific aminoacid sequence and polymorphism of codon 129are important. Alternate postulates for infec-tious proteinaceous agents have recently beensuggested (Walker et al. 2006), but it remainsto be seen whether they will garner universalacceptance.

In the prion field, researchers generally ac-cept that a reasonable surrogate for Koch’s sec-ond postulate be fulfilled by the generation ofsynthetic prions in vitro, i.e., the recovery ofperpetually transmissible infectivity from prionprotein produced recombinantly or chemicallyfrom defined constituents. Major progress to-ward this end has been made in recent days. Pu-rified PrPSc was used to generate PK-resistantPrP (PrPres) in a cell-free system that could evenreflect two typical features of prions: speciesbarrier and strain specificity (Bessen et al. 1995,Kocisko et al. 1995).

Another approach used a method calledPMCA (protein misfolding cyclic amplifica-tion), in which PrPres can be amplified byincubating and sonicating PrPres-containingbrain homogenate diluted in normal brain ho-mogenate. Soto and coworkers amplified PrPres

derived from scrapie-infected hamsters indefi-nitely by using PMCA in serial dilutions. Am-plification of PrPres was accompanied by am-plification of infectivity (Castilla et al. 2005a).Certainly PMCA is a very sensitive method todetect PrPSc even in complex samples such as

blood and already in a presymptomatic diseasestate (Castilla et al. 2005b, Saa et al. 2006).The use of purified PrPC instead of brain ho-mogenate as a substrate decreased the efficiencyof amplification, suggesting that additional co-factors may facilitate misfolding (Deleault et al.2005). For a long time, all attempts to use re-combinant PrP as a substrate for PMCA failed.However, Caughey and coworkers have nowsucceeded in carrying out PMCA using bacteri-ally expressed hamster PrP as a substrate. Whilethis represents a major advance in many ways,the sensitivity was not quite as high as that ofthe original PMCA (Aguzzi 2007, Atarashi et al.2007).

Infectivity may not have been generatedde novo in PMCA in these studies. Instead,prion-infected brain could have been inad-vertently added in the beginning. In an fas-cinating study, Supattapone and coworkersidentified the minimal components (PrPC, cop-urified lipids, and single-stranded polyanionicmolecules) required for amplification of PK-resistant PrP, and they convincingly showedthat prion infectivity can be generated de novoin brain homogenates derived from healthyhamsters using PMCA. Inoculation of furtherhealthy hamsters with the de novo–formedprions caused a transmissible prion disease(Deleault et al. 2007). This study might be re-garded as the final proof of the prion hypoth-esis. However, it also acknowledges PMCA’slimitation for diagnostic purposes because PK-resistant material and infectivity can be formedin the absence of prions, thereby risking the re-porting of false positive results.

A second approach comprises de novogeneration of infectivity by misfolding re-combinant PrPC and subsequently inoculatingwild-type animals. In one attempt, a 55-residuepeptide encompassing the GSS mutationP101L was refolded in vitro to a beta-sheetrich peptide and could induce disease similarto GSS in transgenic mice expressing PrP(P101L). Transmission to wild-type mice wasnot successful, and PrP (P101L) was not resis-tant to PK. Because transgenic mice expressingPrP (P101L) develop disease spontaneously,

446 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 9: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

although later in life than those exposed to thepeptide, Nazor et al. (2005) remarked that themisfolded peptide may have simply accelerateda spontaneously occurring disease.

Transmission to wild-type mice of an invitro–generated misfolded part of the prionprotein (amino acid residues 89–231) wasachieved a few years later. Legname andcoworkers produced PrP (89–231) recombi-nantly and generated amyloid fibrils in vitro.These fibrils induced prion disease in trans-genic mice overexpressing PrP (89–231), whichwas subsequently transmissible to wild-typemice (Legname et al. 2004, 2005).

NeurotoxicityCurrent knowledge about the mechanisms be-hind neurodegeneration in prion disease andprionopathies is limited. Apoptosis and oxida-tive stress certainly contribute to some stages ofTSE pathology (Milhavet & Lehmann 2002),but little is known about damage causing pri-mary events. Early pathologic changes that oc-cur during prion disease involve synapses, yetthe molecular underpinnings of these findingsremain unknown.

It is still unclear whether the toxicity ofPrPSc represents a gain of function or whetherloss of function of PrPC is responsible for neu-ropathological changes induced by prions. Al-though some authors belief that the toxicity inprion disease is explainable simply by a loss-of function of PrPC (Nazor et al. 2007), weand others (Westergard et al. 2007) believe again of function is more likely, particularly be-cause the phenotypes of PrPC-deficient miceare very mild. However, a neuroprotective func-tion that may be physiologically provided byPrPC, which would protect neurons duringprion infection, could be reduced following itsconversion to PrPSc.

Our laboratory, and many others, has pur-sued the hypothesis that elucidating the phys-iological function of PrPC might help re-searchers understand the mechanism involvedin prion-induced neurodegeneration. The fol-lowing discussion centers on the discovery that

HC: hydrophobiccore

mice expressing deletion mutants of PrPC de-velop severe neurotoxic syndromes and identi-fies the reasons why we believe that study ofthese syndromes may reveal the mechanismsoperative in prion diseases.

PHYSIOLOGICAL FUNCTION OFTHE CELLULAR PRION PROTEINThe cellular prion protein PrPC is a GPI-linked extracellular membrane protein with twoN-linked complex glycosylation sites. PrPC ishighly abundant in the developing and ma-ture nervous system, where it is expressed byneuronal and glial cells. This mature versionoriginates from a precursor protein proteolyt-ically processed in the endoplasmic reticulumand Golgi (Stahl et al. 1987). As revealed by itsatomic structure, the mature PrPC protein con-tains a well-defined carboxy-terminal globulardomain comprising residues 127–231 (murinenumbering), consisting of three alpha helicesand two beta sheets (Hornemann et al. 1997;Riek et al. 1996) and a structurally less-definedamino proximal region containing a stretch ofseveral octapeptide repeats, termed the OR,and framed by two positively charged chargeclusters, CC1 (aa 23–27) and CC2 (aa 95–110).These domains are linked by a hydrophobicstretch of amino acids [aa 111–134, also termedhydrophobic core (HC)] (Figure 3).

Prion Protein–Deficient MiceAn astonishing number of independent linesof mice lacking PrPC have been generated byhomologous recombination in embryonic stemcells in many laboratories. Mice with disrup-tive modifications restricted to the open readingframe are known as Prnpo/o [Zurich I] (Bueleret al. 1992) or Prnp"/" [Edinburgh] (Mansonet al. 1994). They developed normally, and nosevere pathologies were observed later in life. Aspredicted by the protein-only hypothesis, thesemice were entirely resistant to prion infections(Bueler et al. 1993).

In contrast with these earliest lines, threelines generated afterwards: Prnp"/" [Nagasaki],

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 10: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Rcm0, and Prnp"/" [Zurich II] (Moore et al.1999, Rossi et al. 2001, Sakaguchi et al.1996) developed ataxia and Purkinje cell losslater in life. Because the phenotype wasabolished by reintroduction of Prnp as a

transgene, the originators of the Nagasakimice concluded that it occurred because ofthe lack of PrPC. This, however, wouldrun counter to the lack of pathology inPrnpo/o Zurich-I mice.

S S

S

CC1SP CC2 HC H1 H2 H3 GPIORPheno-

type Trans-

mission Prion

propagation Rescue by PrP Refs.

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

PrPwt

no – yes – a PrP!32–80

no – yes – b;c PrP!32–93

no – no – b;d PrP!32–106

no – yes* – e PrP!23–88

no – no – e PrP!23–88!95–107

no – no – e PrP!23–88!108–121

no – yes – e PrP!23–88!141–176

no – no** – f;g PrP!114–121

no – yes – h PrP!104–114

cerebellar disorder no no yes b PrP!32–121

cerebellar disorder no no yes b;d PrP!32–134

cerebellar disorder no nd yes f PrP!94–134

cerebellar disorder no nd yes k PrP!105–125

cerebellar disorder no yes no l PrP PG 14

nd – – – e PrP 144 #

storage disease no no no e PrP!23–88

!177–200

storage disease no no no e PrP!23–88

!201–217

cerebellar disorder no no no e PrP!23–88

!141–221

nd – – – e PrP!23–88!122–140

nd – – – e PrP!23–88144 #

cerebellar disorder no no no j PrP!1–22

231 #

no – yes – i PrP231 #

no – no – e PrP!23–88C178A A

448 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 11: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

The discrepancy between the differentlines of PrP knockout mice was not resolveduntil a novel gene (Prnd ), encoding a proteincalled Doppel (Dpl), was discovered. Prnd islocalized 16 kb downstream of Prnp. In allthree lines of PrPC-deficient mice developingataxia and Purkinje cell loss, a splice acceptorsite to the third exon of Prnp was deleted. Thisplaced Prnd under transcriptional control ofthe Prnp promoter, resulting in the formationof chimeric transcripts and in overexpressionof Dpl in the brain (Moore et al. 1999, Rossiet al. 2001, Sakaguchi et al. 1996). Preciselywhy the overexpression of Dpl is deleteriousis still unclear. On the basis of the observationthat Dpl expression induced heme oxygenase1 (HO-1) and neuronal and inducible nitricoxide synthases (nNOS and iNOS), suggestingan increased oxidative stress in the brainsof the Dpl-expressing Prnpo/o mice, Wonget al. (2001c) proposed that Dpl expressionexacerbates oxidative damage by antagonizingwild-type PrPC’s antioxidative function.

The latency period before the various trans-genic mice overexpressing Dpl develop patho-logical phenotypes is inversely correlated to theDpl expression level in the brain, indicatinga rather strict gene-dosage effect (Rossi et al.2001). The Dpl-induced disease can be rescuedby coexpression of wild-type PrPC (Nishidaet al. 1999, Rossi et al. 2001), indicating thattoxicity of Dpl and the physiological functionof wild-type PrPC are not independent of eachother, but rather are involved in a commonpathway. Dpl-deficient mice suffer from steril-

ity (Behrens et al. 2002), suggesting that the pri-mary physiological function of Dpl is related tosperm maturation.

Functional Domainsof the Prion ProteinFor all the uncertainties surrounding the physi-ological and molecular functions of PrPC, someknowledge was generated by expressing a seriesof partially deleted Prnp variants in culturedcells and transgenic mice. Some of these mu-tants were made to identify the essential do-mains necessary for restoring prion susceptibil-ity. However, investigators found that domainexpression provoked spontaneous neurodegen-erative disease (Figure 3). In many instances,these syndromes were partially or fully coun-teracted by coexpression of wild-type PrPC. Be-cause the lack of PrPC itself did not induce anobvious phenotype, the latter pathologies indi-cate pathways in which PrPC is functionally ac-tive. Hence mice expressing PrP, which lacksdefined domains, may allow for the identifi-cation of functionally relevant domains withinPrPC.

N-terminal deletion mutants of PrP. TheOR has long been suspected to represent a ma-jor mediator of PrPC’s function, and insertionmutations affecting the OR are associated withhereditary human prion disease. However,transgenic studies indicate that the OR isnot required for PrPC to function or for itsconvertibility into PrPSc (Flechsig et al. 2000).

#"""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""

Figure 3Murine PrPC protein and transgenic mutant PrP. Schematic drawing of full-length murine PrPC, includingthe signal peptide of the precursor protein (SP; brown box). Although amino acid numbering differs betweenhuman and mouse PrP, the organization of domains (including CC1 and CC2, OR, HC, and H1–H3) issimilar to that of human PrPC (see Figure 2). Mouse PrP also contains a disulphide bond (S–S) and aGPI-anchor. The left column denotes the individual mutants described in the text. The right columnsindicate presence or absence of phenotypic abnormalities (Phenotype) in transgenic mice when expressed ona PrP-deficient genetic background, transmissibility of this phenotype to recipient mice (Transmission), andsusceptibility of transgenic mice to prions after intracerebral inoculation with a mouse-adapted strain ofscrapie 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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 12: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

The OR appears to have, at best, a modulatinginfluence on PrP conversion. Mice expressingOR-deficient PrPC mutants do not developpathologies (Fischer et al. 1996, Muramotoet al. 1997, Shmerling et al. 1998). This wasunexpected because a variety of in vitro datahad identified the OR as being responsible forcopper binding (Aronoff-Spencer et al. 2000,Chattopadhyay et al. 2005, Furlan et al. 2007,Leclerc et al. 2006, Qin et al. 2002, Stockelet al. 1998; reviewed in Vassallo & Herms 2003)and for conferring protection against oxidativestress (Brown et al. 1999, Fukuuchi et al. 2006,White et al. 1999, Wong et al. 2001a). On theother hand, transgenic mice expressing ninesupernumerary octapeptide repeats, for a totalof 14 proline and glycine-rich repeats (Chiesaet al. 1998)—which models a human familialCJD-linked mutation—develop ataxia andcerebellar atrophy, granule cell loss, gliosis,progressive myopathy, and PrP deposition.The latter phenotype resembles its humancounterpart in some ways (Chiesa et al. 2000),yet transmission to wild-type mice failed(Chiesa et al. 2003).

In vitro studies indicate that the CC1 regionis involved in recycling and internalizing PrPC

from the cell surface (Sunyach et al. 2003,Taylor et al. 2005). Unfortunately, in vivolittle evidence supports the latter contention.Lack of CC1 in (PrP!23"88) (Muramoto et al.1997) did not induce pathologies in transgenicmice, and convertibility to PrPSc was retained.In PrP!23"88 mice, a second charge cluster(CC2) with several lysine residues aroundposition 100 may replace the function of CC1.However, mice bearing partial deletions ofCC2 (PrP!23"88 !95"107 and PrP!23"88 !108"121)

are also healthy (Muramoto et al. 1997).The combination of amino-terminal deletionwith the elimination of amino acids 141–176(PrP!23"88 !141"176) was also innocuous andrestored susceptibility to prion infection(Muramoto et al. 1996) despite a large deletionwithin the globular domain of PrPC.

The function of PrPC may depend on theHC region in concert with CC2. With the ex-ception of a small deletion between CC2 and

HC (PrP!104"114) (Hegde et al. 1998), abla-tion of CC2 in combination with a partial orcomplete deletion of HC elicits severe patholo-gies in mice. PrP!32"121 and PrP!32"134 trans-genic mice suffer from ataxia and cerebellargranule cell loss in addition to widespreadwhite matter disease (Radovanovic et al. 2005,Shmerling et al. 1998). The latter is also seenin mice expressing deletions encompassing all(PrP!94"134) or part (PrP!105"125) of the centraldomain (CD) (Baumann et al. 2007, Li et al.2007). These pathologies are radically differentfrom those seen in prion infections, and none ofthem goes along with pathological aggregationof PrP.

Each of these pathologies can be coun-teracted by coexpression of wild-type PrPC

(Baumann et al. 2007, Li et al. 2007, Shmer-ling et al. 1998), suggesting a competition ofsorts between PrPC and the toxic mutants. Inone conceivable scenario, PrPC and its variantsmay compete for a common ligand. Binding orcomplex assembly may represent the first stepin a series of events that also involve the inter-action of an effector domain located in or con-trolled by the central domain (CC2 and HC),eventually resulting in signal transduction.

A partial deletion of HC (PrP!114"121)(Baumann et al. 2007) is nontoxic, but its po-tential to counteract the toxicity of PrP!32"134

is lower than that of wild-type PrPC. Micewith deletion of CC2 and HC (PrP!32"121 andPrP!32"134 as well as PrP!104"114, PrP!114"121)did not support prion propagation (Flechsig &Weissmann 2004; Hegde et al. 1998, Holscheret al. 1998), indicating an involvement of theseregions in conversion.

Carboxy-proximal deletion mutants of PrP.Mice expressing PrP mutants with deletionsaffecting Helix 2 (PrP!23"88 !177"200), Helix3 (PrP!23"88 !201"217), or both helices 2 and3 (PrP!23"88 !141"221) suffer from ataxia andpresent with features of neuronal storage dis-ease (Muramoto et al. 1997, Supattapone et al.2001) but fail to replicate prions (Muramotoet al. 1996). Obviously at least Helix 2 and Helix3 are indispensable for stabilizing the structure

450 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 13: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

of PrPC. None of these diseases proved to betransmissible to normal wild-type mice, andthey all manifested themselves independentlyof the presence or absence of wild-type PrP.This stands in sharp contrast to the group ofdeletion mutants affecting CC2 and HC.

Several attempts to generate mice express-ing truncated carboxy-terminal mutants lack-ing membrane anchoring (PrP!23"88 144# andPrP144#) have failed (Fischer et al. 1996,Muramoto et al. 1997). As in the case ofPrP!23"88 !122"140 (Muramoto et al. 1997), linesexpressing high levels of mRNA were gener-ated but protein was never detected. Essentialsignals may have been lost, thereby preventingcorrect sorting, processing, or folding of PrPand resulting in a short-lived polypeptide.

Mutations affecting the localization ofPrPC. The affinity of GPI-linked PrPC forartificial membranes, as measured by surfaceplasmon resonance (Elfrink et al. 2007), sug-gests extremely strong interactions. One mighttherefore expect that most PrPC is attached tocell membranes, with perhaps traces of PrPC

floating in body fluids. Thus it may come asa surprise that plasma contains conspicuousamounts of PrPC (Volkel et al. 2001), and theconcentration of PrPC in cerebrospinal fluidis even higher (Castagna et al. 2002). How-ever, it is unclear whether this soluble PrPC

is chemically identical to its membrane-boundisoform. Treatment of cultured cells with phos-phatidylinositol phospholipase C efficiently re-leases PrPC from cultured cell membranes(Stahl et al. 1987), and a similar mechanism mayunderlie the physiological shedding of PrPC

into body fluids.Release of full-length secreted PrPC was

forced by deletion of its carboxy terminal hy-drophobic domain (PrP231#), which is normallyreplaced by a GPI-anchor. This manipulationdid not induce any pathological phenotype(Chesebro et al. 2005). In contrast, targetingPrPC to the cytosol (cyPrP = PrP!1"22 231#)by deleting its amino-terminal leader peptide(which targets PrPC to the endoplasmic retic-ulum and to the secretory pathway) provoked

ataxia with cerebellar degeneration and glio-sis (Ma et al. 2002). Coexpression of wild-typePrPC did not influence the phenotypes of thesemice. Whether cytoplasmic expression of PrPand its cytotoxicity represent realistic modelsof the events occurring during prion disease re-mains very hotly debated (Fioriti et al. 2005,Roucou et al. 2003).

Point Mutations withinthe Prion ProteinAs previously described, a considerable set ofpoint mutations within PRNP has been linkedto various forms of human prion diseases.Some of these mutations have been expressedin mice. With the possible exception of somestrains of mice expressing the P101L vari-ant of PrPC (Hsiao et al. 1994, Telling et al.1996), none of these attempts succeeded inreproducing the infectiousness of bona fideprions. Point mutations affecting the two N-linked glycosylation sites of PrPC proved, as ex-pected, to alter its glycosylation (Kiachopouloset al. 2005). Point mutations N182T, A198T,or N182T/A198T prevented glycosylation intransgenic mice without grossly affecting cellu-lar sorting in cell culture. Mice developed nor-mally and were readily susceptible to scrapieor BSE (Neuendorf et al. 2004). Knock-in mu-tants carrying either N180T or N196T, orboth mutations, (Cancellotti et al. 2005) didnot suffer from any constitutive phenotype,even if the complete blockade of glycosylationby the N180T/N196T double mutation ledto a mainly intracellular localization of PrPC.This finding is somewhat surprising because re-sults from cultured cells had predicted that un-glycosylated PrPC would be prone to sponta-neous aggregation (Korth et al. 2000, Priola &Lawson 2001).

Two sets of point mutations, PrP3AV (ex-change of alanine to valine at positions 113,115, and 118) (Prusiner & Scott 1997) andPrPKHII (exchange of lysine 109 and histidine110 for isoleucine) (Hegde et al. 1998), gener-ated PrP with altered topology, termed CtmPrPin a cell free assay. CtmPrP supposedly spans

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 14: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

the membrane via the HC domain (Hegdeet al. 1998). Transgenic mice expressing theseproteins developed a fatal neurological disor-der (Hegde et al. 1998). A similar phenotypewas observed in transgenic mice with substi-tution of leucine 9 into arginine in additionto this 3AV mutation (Stewart et al. 2005,Stewart & Harris 2005). However, researchnever formally proved that CtmPrP exists in vivo.It is still noteworthy that coexpression of wild-type PrPC with mutants promoting the CtmPrPtopology aggravated their phenotype. Subtlechanges, such as the removal of disulfide bridges(PrP!23"88 C178A), are tolerated without induc-ing a spontaneous phenotype though reduc-ing the susceptibility for conversion into PrPSc

(Muramoto et al. 1997).

Evolution of the Prion ProteinPrP is present in a broad variety of species(Figure 4). Genes with similarities to Prnp existin birds (Gabriel et al. 1992), reptiles (Simonicet al. 2000), amphibians (Strumbo et al. 2001),and possibly in fish (Favre-Krey et al. 2007,Oidtmann et al. 2003, Rivera-Milla et al. 2003,Suzuki et al. 2002) in addition to all mammals.However, more primitive organisms such as in-sects, cephalopods, and protozoa have not beenreported to contain PrP homologs. All PrPsare glycosylated and membrane attached by aGPI anchor. The sequence identity among theknown PrP homologs is limited, and proteinlength can vary between !250 amino acids intetrapods to !600 amino acids in fish. Fishmay have developed additional Prnp-like genes(Rivera-Milla et al. 2006). The putative fish PrPgenes are thus far identified only on the basis ofrather tenuous sequence similarities. The con-tention that these molecules indeed representparalogs of PrPC would be greatly strengthenedif knockdown-induced phenotypes of zebrafishwould be functionally corrected by mammalianPrPC expression. Such experiments have notbeen reported.

Comparisons between the available struc-tures and molecular models suggest that allPrPs share a common blueprint. A flexible

amino-terminal tail, with a positively chargedCC1 at its far end and repetitive domainsof variable numbers, is hooked to a globu-lar carboxy-terminal domain. The fold of thisdomain is strongly conserved and stabilizedby a disulfide bridge, although the primarysequence shows considerable diversity. Thesetwo domains are linked by a highly conservedhydrophobic linker having a second positive-charge cluster CC2 at its amino-terminus. Thislinker region is by far the most conserved se-quence motive of PrP in all species.

Cellular Processes Influencedby PrPC ExpressionSeveral cellular processes in the nervous systemhave been influenced by the Prnp-genotype, in-cluding neuronal survival; neurite outgrowth;synapse formation, maintenance, and func-tion; and maintenance of myelinated fibers(Figure 5).

One of the most frequently suggested cel-lular functions of PrPC is a survival-promotingeffect on neuronal and nonneuronal cells, whichhas been observed in vitro as well as in in vivostudies.

This neuroprotective function, or cytopro-tective function in general reviewed in Roucou& LeBlanc (2005), has been mediated by anti-apoptotic or antioxidative mechanisms.

Antiapoptotic function. Neurons derivedfrom Prnp"/" mice were originally reported tobe more susceptible to the induction of apop-tosis by serum-deprivation than were cells ex-pressing PrPC (Kuwahara et al. 1999), but thiseffect may have been brought about by Dploverexpression rather than by PrPC ablation.However, several studies indicate that PrPC

has a cytoprotective function by decreasingthe rate of apoptosis after particular apoptoticstimuli such as Bax overexpression or TNF-!.Bax overexpression induces apoptosis in hu-man neuronal cells. Coexpression of wild-typePrPC, but not of PrP lacking the octarepeats,reversed the Bax-mediated induction of apop-tosis (Bounhar et al. 2001).

452 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 15: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

The presence of PrP in the cytosol, be itdue to reverse translocation from the endoplas-mic reticulum or through direct cytosolic ex-pression, was virulently neurotoxic (Ma et al.2002). However, other studies failed to con-firm the toxicity of cytosolic PrP and claimedthat it can instead protect against Bax-mediatedapoptosis in human primary neurons (Roucouet al. 2003). In this context, PrPC inhibitedthe proapoptotic conformational change of Baxand cytochrome c release from mitochondria(Roucou et al. 2005).

In a screening approach for proteins pro-tecting cancer cells from apoptosis, researchersinvestigated the gene-expression profile in anestablished cell clone of MCF-7 breast cancercell line resistant to TNF!-induced apoptosis.PrPC was overexpressed 17-fold. Conversely,overexpression of PrPC converted MCF-7 cellssensitive to TNF!-induced apoptosis into re-sistant cells (Diarra-Mehrpour et al. 2004).

The neuroprotective function of PrPC inthe postischemic rodent brain has been inten-sively studied. Levels of PrPC after ischemiawere increased compared with controls (Shyuet al. 2005, Weise et al. 2004). Moreover,adenovirus-mediated overexpression of PrPC

reduced infarct size in rat brain and improvedneurological behavior after cerebral ischemia(Shyu et al. 2005). Conversely, in a mousemodel of ischemic brain injury Prnpo/o micedisplayed significantly increased infarct vol-umes when compared with wild-type mice(McLennan et al. 2004, Weise et al. 2006). Twogroups of researchers showed that mice lackingPrPC had enhanced postischemic caspase-3 ac-tivation (Spudich et al. 2005, Weise et al. 2006).An increase in Erk-1/-2, STAT-1, and JNK-1/-2 phosphorylation and activation was iden-tified, suggesting PrPC’s possible involvementin cellular signaling (Spudich et al. 2005). Also,a reduced amount of phospho-Akt in the graymatter suggested that PrPC deficiency bringsabout an impairment of the antiapoptotic phos-phatidylinositol 3-kinase/Akt pathway (Weiseet al. 2006). Finally, Mitteregger et al. (2007)claimed that the OR is required within PrPC

for the neuroprotection in the ischemic mouse

brain, although the genetic homogeneity of themice tested in the latter experiment was notcontrolled for.

Protection against oxidative stress. Besidesits possible antiapoptotic function, there aremany reports about an antioxidative effect ofPrPC. These two effects are not necessarily mu-tually exclusive. Oxidative stress may be in-volved in TSE pathogenesis. However, onemust remember that oxidative stress is very un-specific and is seen in different kinds of damageto the nervous system with impaired mitochon-drial function such as defects in the ubiquitin-proteasome system, protein aggregation, andinflammation.

Many investigators believe that the mainfunction of PrPC consists of protecting againstoxidative stress (see Milhavet & Lehmann 2002for a review). First hints came from in vitrostudies of rat pheochromocytoma cells. Thoseselected for resistance to copper toxicity oroxidative stress showed higher levels of PrPC

(Brown et al. 1997a). Primary neuronal cellslacking PrPC were more susceptible to hydro-gen peroxide (H2O2) than were wild-type cells.The increased peroxide toxicity went along witha significant decrease in glutathione reductaseactivity measured in PrPC-deficient neurons(White et al. 1999). Also, PrPC-deficient pri-mary neurons were more susceptible to treat-ment with agents inducing oxidative stress com-pared with wild-type cells, a phenomenon thatwas explained by a reduced Cu/Zn superox-ide dismutase (SOD) activity observed in vivo(Brown et al. 1997b, 2002). Higher levels of ox-idative damage to proteins and lipids were iden-tified in the brain lysates derived from Prnp"/"

compared with wild-type mice (Klamt et al.2001, Wong et al. 2001b).

PrPC itself could have SOD activity andthereby mediate the antioxidative function(Brown et al. 1999). However, there is sig-nificant controversy about this alleged SODactivity. Others, ourselves included, failed toconfirm this proposed SOD activity in vitro(Jones et al. 2005) and in vivo (Hutter et al.2003). Furthermore, PrPC expression level did

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 16: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

–4.01 165 330 494 658

–3.2

–2.4

–1.6

–0.8

0

0.8

1.6

2.4

3.2

4.0 DeerSheepCowCamelRabbitBatChimpanzeeHumanMouseSigmodonPigeonQuailChickenDuck

Zebrafish_prp1Trout_prp1Carp_prp1Zebrafish_prp2Frog

Position

Multiple alignment

a

b

Tetrapod PrP Fish PrP

Hum. Prp253 aa

S R H T N N

S--S

Fug. Prp1461 aa

S R H T N

S--S

S R T N N Zeb. Prp1

606 aaS--S

H

S R H T N N

S--S

Chi. Prp273 aa

S R H T N N

S--S

Xen. Prp216 aa

S R H T N N

S--S

Tur. Prp270 aa

Sal. Prp605 aa

S R

S--S

H N T

Zeb Prp2567 aa

S R

S--S

H N N T

Fug. Prp2435 aa

Hyd

rop

ho

bic

ity

S R

S--S

H H N T

Turtle

Figure 4PrP structural diversity in vertebrates. (a) Schematic drawing of tetrapod PrPs and long (PrP1 and PrP2) fish PrPs. The speciesabbreviations refer to sequences from human (Hum), chicken (Chi), turtle (Tur), Xenopus (Xen), zebrafish (Zeb), salmon (Sal), andFugu (Fug). The location and relative size of conserved structural features are indicated. However, these features were physicallydetermined for the structure of human PrPC and represent mere conjectures in the case of fish. Domains are indicated by differentboxes and/or letters: S, signal peptide sequence; R, repetitive region; H, hydrophobic region; S—S, disulfide bridge; N, glycosylationsite; arrow, GPI anchor residue; and T, hydrophobic tail. (b) Comparison of hydrophobicity plots. Sequences of indicated species werealigned using DNAMAN software (Lynnon BioSoft, Canada), and a hydrophobicity plot was generated using a window of nineamino-acid residues. Numbering of residues is according to alignment matrix. (c) 3-dimensional structures of human (hum based on1QM2.pdb model) chicken (chi based on 1U3M.pdb) turtle (tur based on 1U5L.pdb), and frog (fro based on 1XOU.pdb); pdb files arefrom the protein database (Berman et al. 2000). Note the similarity of the carboxy-terminal globular domain. (d ) Evolutionaryrelationships among vertebrate PrP sequences are based on distance methods (neighbor-joining). Bootstrap values are shown at relevantnodes using DNAMAN software (Lynnon BioSoft, Canada).

454 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 17: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

not significantly influence SOD activity invivo (Hutter et al. 2003, Waggoner et al.2000).

Mitochondria play an important role notonly in oxidative stress but also in the inductionof apoptosis. Morphological alterations in mi-tochondria have been described in scrapie-infected hamsters (Choi et al. 1998) and mice(Lee et al. 1999) as well as in mice lacking PrP,in which the number of mitochondria was re-duced (Miele et al. 2002).

Role of PrPC in synapses. Synapses have de-veloped into a sort of hot spot in prion research.Several immuno-electron microscopy studiescould show that PrPC is localized in synap-tic boutons, whereas it is mainly presynaptic(Fournier et al. 1995, Moya et al. 2000, Saleset al. 1998, Tateishi et al. 1996b). However,others described a much broader distribution ofneuronal PrPC (Laine et al. 2001, Mironov et al.2003). Because PrPC is processed and brokendown into various fragments, not all of which

PrPhum PrPchi PrPtur PrPfro

PigeonQuail

98

Chicken

100

Duck

100

Turtle

92

Frog

DeerCowSheep

92

CamelBatChimpanzeeHuman

100

MouseSigmodon100

95

Rabbit100

Zebrafish_prp1Trout_prp1

100

Carp_prp1Zebrafish_prp2100

100

0.05

c

d

Tetrap

od

Fish

Figure 4(Continued )

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 18: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Neuronal survival• Protection against apoptosis• Protection against oxidative stress

Neurite outgrowth• Dendrites• Axons

Synapse• Formation• Function

Maintenance ofmyelinated axons

Figure 5Physiological 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 topromote neuronal survival, in particular following apoptotic or oxidative stress. Cerebellar granule cell apoptosis was observed in miceexpressing toxic N-terminal deletion mutants of PrP. In addition, the latter transgenic mice show an impaired maintenance ofmyelinated axons in the white matter. Another site of PrPC action might be the synapse, which is often affected in the first stage ofprion diseases and whose formation was found to be reduced in neuronal cultures devoid of PrPC. Furthermore, electrophysiologicalstudies indicate a role of PrPC in synapse function, especially in neurotransmitter release.

are recognized by the antibodies used in thesestudies, one might speculate that some PrPC

degradation products acquire distinct subcellu-lar topologies.

Early pathologic changes occurring in priondiseases involve synapse loss and PrPSc depo-sition in synaptic terminals (Grigoriev et al.1999, Jeffrey et al. 2000, Kitamoto et al. 1992,Matsuda et al. 1999, Roikhel et al. 1983). Synap-tic vesicle proteins associated with exosomesand neurotransmission are reduced in brainsof patients with spongiform encephalopathy(Ferrer et al. 1999). Synaptic disorganizationand loss are fundamental and constant fea-tures of prion disease, irrespective of the pres-ence or absence of spongiform change, neu-ronal loss, and severe gliosis (Clinton et al.1993). Abnormal electrophysiological record-ings in scrapie-infected mouse and hamster hip-pocampal and cortical slices further supportthe synaptic dysfunction during the course ofprion disease (Barrow et al. 1999, Johnston et al.1998). In a terminal disease state, PrPSc accu-mulation in synaptosomes correlated with alter-ations in the GABAergic system (Bouzamondo-Bernstein et al. 2004). Despite the wealth of

the above evidence, however, it should not goundiscussed that synaptic changes can representnonspecific phenomena that are seen in essen-tially all brain diseases at one stage or another.

The generally held view that PrPC is animportant protein in synapses is supportedby electrophysiological studies of CA1 hip-pocampal neurons derived from PrPC-deficientmice. Excitatory glutamatergic synaptic trans-mission, GABAA receptor–mediated fast inhibi-tion, long-term potentiation, and late afterhy-perpolarization were reduced or absent in micelacking PrPC (Carleton et al. 2001; Collinget al. 1994, 1996; Mallucci et al. 2002). Some ofthe findings could be explained by alterationsin Ca-activated K+ currents (Colling et al.1996, Herms et al. 2001). However, the readershould note that alterations in synaptic trans-mission were not confirmed by others (Lledoet al. 1996), and glutamatergic synaptic trans-mission was even observed to be increased inPrPC-deficient mice by yet another laboratory(Maglio et al. 2004, 2006). Another report in-dicates the impact of aging on these alterationsdescribing a reduction in the level of postte-tanic potentiation and long-term potentiation

456 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 19: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

only in old PrPC-deficient mice (Curtis et al.2003). In summary, the impact of the loss ofPrPC on hippocampal electrophysiological pa-rameters is still being hotly debated despite afull decade of research efforts. Some of the dis-crepancies may depend on additional geneticmodifiers for which investigators have not rig-orously controlled.

Other alterations in PrPC-deficient micemight be related to synaptic dysfunctionsuch as altered circadian rhythms and sleep(Tobler et al. 1996) and impaired hippocampal-dependent spatial learning (Criado et al. 2005).The neuromuscular junction is another sitewhere PrPC was concentrated, namely en-riched in subsynaptic endosomes (Gohel et al.1999). A potentiation of acetylcholine releasefrom presynaptic axon terminals was observedafter administration of recombinant PrP atnanomolar concentrations to mouse phrenic-diaphragm preparations (Re et al. 2006). Thesuggestion of an involvement of PrPC insynapse formation originated from in vitro ob-servations in hippocampal neurons, in whichsynaptic-like contacts were increased after addi-tion of recombinant PrP (Kanaani et al. 2005).

It is unknown whether the role of PrPC insynapses is related to its above-mentioned an-tiapoptotic or antioxidative effects or whetherit is mainly the involvement of PrPC in neu-rotransmitter release (e.g., via direct interac-tion with synapsin1 and synaptophysin). How-ever, an as-yet-unidentified process could alsoplay a key role, or several processes could worktogether.

Neurite outgrowth. Several lines of evidenceindicate PrPC’s involvement in neuronal de-velopment, differentiation, and neurite out-growth. Axon or dendrite outgrowth was asso-ciated with PrPC-dependent activation of sig-nal transduction pathways including p59Fyn ki-nase, cAMP/protein kinase A (PKA), proteinkinase C (PKC), and MAP kinase activation(Chen et al. 2003, Kanaani et al. 2005, Lopeset al. 2005, Santuccione et al. 2005). P59Fynkinase activation in this context was depen-

dent on the recruitment of neural cell adhesionmolecule (N-CAM) to lipid rafts (Santuccioneet al. 2005). Recent studies show that PrPC pos-itively regulates neural precursor proliferationduring development and adult mammalian neu-rogenesis (Steele et al. 2006).

Maintenance of the white matter. Centralnervous system white matter, composed mainlyof myelinated axons, might be disrupted inprion diseases and prionopathies. In some casesof GSS, cerebellar and frontal white matter areaffected (Itoh et al. 1994). In an experimentalmodel of human TSEs in rodents, vacuolationin myelinated fibers with splitting of myelinlamellae was observed (Walis et al. 2003). PrPC

is present in purified myelin fractions derivedfrom brain homogenates (Radovanovic et al.2005). Several transgenic mice expressing dele-tion mutants of PrPC (Baumann et al. 2007, Liet al. 2007, Shmerling et al. 1998) as well asPrnp"/" mice accidentally overexpressing Dpl(Nishida et al. 1999) show vacuolation and de-generation of myelinated fibers in the centralnervous system.

PrP and the Immune SystemThe immune system plays a fundamental role inprion disease and PrPC is expressed on cells ofthe immune and hematopoietic system, where itmight have a physiological function. This topicis reviewed in depth in Isaacs et al. (2006). Also,Zhang et al. (2006) reported that PrPC is in-volved in self-renewal of hematopoietic stemcells.

Molecular Mechanisms Mediatingthe Function of PrPC

Despite the overwhelming number of re-ports about alterations in mice and cells lack-ing PrPC summarized above, little is knownabout the molecular mechanisms involved inthese cellular processes. Figure 6 depicts sometheoretical models of how PrPC might influ-ence cell signaling, endocytosis, and cell ad-hesion. Whether these events are mutually

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 20: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

exclusive, or whether they occur only underspecific circumstances in a diversity of tissues,or whether they can act in a combined way,remains speculative. In all cases, PrPC is likely

to mediate its function via one or more interac-tion partners.

One explanation for the diversity of the sug-gested physiological functions of this PrPC is

Interaction with TM protein in trans

• Modulation of signaling pathways

• Modulation of cellular adhesion

Endocytosis of PrPc via Clathrin-coated pits or caveolae

• Cointernalization of anothercell component

• Modulation of signaling pathways

• Degradation of PrPc

cointernalized TM proteins

Interaction with TM protein in cis

c

a

b

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

Lipid raft Non-raft region

Lipid raft Non-raft region

458 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 21: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

that it may exert pleiotropic effects, therebymodulating the function of several cellularpathways. Examples for such a general cellu-lar process would be stabilization of proteincomplexes and the targeting of cell compo-nents to certain cellular sites, such as rafts orendosomes.

Signaling. The attachment of PrPC to themembrane by a GPI anchor, its localization indetergent-resistant membranes, also known aslipid rafts, in many cell types may suggest an in-volvement in cellular signaling (Shmerling et al.1998) as is the case for other raft-associatedproteins. Moreover, as we describe below, PrPC

could also influence cellular signaling events byits involvement in endocytotic pathways.

Several signaling pathways or signalingcomponents, such as Akt, Fyn, cAMP, andErk1/2, are modulated by PrPC expression, itscross-linking, or its interaction with anotherprotein. Antibody-mediated cross-linking ofPrPC induced activation of the p59Fyn ki-nase, a family member of nonreceptor Src-related kinases, in neuronal differentiated cellsin a caveolin-1-dependent manner (Mouillet-Richard et al. 2000). As a downstream event,the same group claims to have identified Erk1/2phosphorylation (Schneider et al. 2003). PrPC

cross-linking additionally modulates seroton-ergic receptor activity in differentiated neu-ronal cells (Mouillet-Richard et al. 2005). Thefinding that PrPC cross-linking modulates ac-tivity of serotonergic receptors in differenti-ated neuronal cells await replication and invivo confirmation. However, p59Fyn activationand downstream activation of Erk1/2 were also

seen in a hypothalamic cell line (Toni et al.2006).

Several studies indicated PrPC involvementin neurite outgrowth and neuronal survival.Chen et al. reported increased neuronal survivaland neurite outgrowth from neurons when cul-tured on Chinese hamster ovary (CHO) cellstransfected to express mouse PrP. Althoughp59Fyn kinase activity in this context was in-volved mainly in neurite outgrowth, the PI3 ki-nase/Akt pathway as well as regulation of Bcl-2and Bax expression contributed to the survivaleffect elicited by PrP. Cyclic AMP/protein ki-nase A (PKA) and Erk signaling pathways con-tributed to both neurite outgrowth and neu-ronal survival (Chen et al. 2003, Santuccioneet al. 2005).

Some investigators suggested that engage-ment of PrPC with stress-inducible proteinI (STI1) induces neuroprotective signals thatrescue cells from apoptosis via cAMP/proteinkinase A and the Erk signaling pathways(Chiarini et al. 2002, Zanata et al. 2002). In-teraction with STI1 induced different signalingpathways, promoting neuroprotection by PKAactivation and neuritogenesis by activation ofthe MAPK pathway (Lopes et al. 2005).

Endocytosis and internalization of PrPC.PrPC is rapidly internalized from the cellmembrane. This internalization of PrPC couldbe crucial for its function, e.g., by influenc-ing signal transduction pathways. Endocyto-sis of membrane receptors does not neces-sarily downregulate receptor activity. Whilebeing internalized, both tyrosine kinase andG protein–coupled receptors may remain

#"""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""""

Figure 6Models 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. Thisregulation could positively or negatively modulate the activity of signal transduction pathways, e.g., viainducing degradation of the cointernalized partner. (b) An interaction with a transmembrane (TM) protein incis independent of an internalization process may lead to modulation of signal transduction pathways in thecell carrying PrPC on its cell surface. (c) Similarly, an interaction with another protein in trans may lead tomodulation of signal-transduction pathways or adhesion to adjacent cells.

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 22: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

activated and produce intracellular responsesalong the endosome-lysosome pathway (Pradoet al. 2004). Internalization of tyrosine kinasereceptors was functionally important in stud-ies: TrkA receptors, for example, mediate nervegrowth factor (NGF)-dependent cell survivalwhile they are located at the cell membrane,whereas internalization is required for induc-tion of neurite outgrowth (York et al. 2000,Zhang et al. 2000).

The mechanism of PrPC internalization isstill controversial because both raft/caveolae orcaveolae-like (Kaneko et al. 1997, Marella et al.2002, Peters et al. 2003, Vey et al. 1996) as wellas clathrin-dependent endocytosis may be op-erative (Shyng et al. 1994, Taylor et al. 2005).These mechanisms might as well be equallyimportant. In addition, internalization of thesame ligand/receptor complex by distinct endo-cytotic pathways can result in different signal-ing outcomes. TGF-" receptor, for example, isdegraded after endocytosis via caveolae, but in-ternalization via clathrin-coated pits promotesits signaling (Di Guglielmo et al. 2003). How-ever, in lymphocytes and neuronal cells that donot express caveolin, internalization can occurin a lipid raft–associated noncaveolar, clathrin-independent process (Kirkham & Parton 2005,Parton & Richards 2003). Therefore addi-tional, less well-characterized endocytosis path-ways including caveolae-like endocytosis mightbe involved in internalization of PrPC.

For endocytosis by clathrin-coated pits,PrPC would need to leave the lipid rafts priorto internalization because the rigid structure ofraft lipids is unlikely to accommodate the tightcurvature of coated pits. This phenomenonoccurred after binding of copper to the OR(Sunyach et al. 2003, Taylor et al. 2005), butits physiological significance is unknown. Low-density lipoprotein receptor-related protein 1(LRP1) was later shown to mediate PrPC en-docytosis (Taylor & Hooper 2007), and CC1region was essential for its internalization inneuroblastoma cells (Sunyach et al. 2003, Tay-lor et al. 2005). Sunyach et al. (2003) suggestedthat heparin sulfate proteoglycans are partof the endocytotic complex involving PrPC.

Glypican-1, a GPI-anchored heparan sulfate-containing cell-associated proteoglycan, inter-acts and cointernalizes with PrPC in N2a afterinduction with copper ions. In cells express-ing PrP, which lacks the OR, internalization ofglypican-1 is reduced, suggesting a possible rolefor PrPC in the cointernalization of other cel-lular components (Cheng et al. 2006).

PrPC might participate in the correct local-ization of some other proteins in lipid rafts.Neuronal nitric oxide synthase (nNOS), for ex-ample, involved in various nervous system pro-cesses such as development, synaptic plasticity,regeneration, and regulation of transmitter re-lease was associated with lipid rafts in wild-typemice. In contrast, in brains of PrPC-deficient aswell as scrapie-infected mice, nNOS was not as-sociated with rafts, and activity of nNOS was re-duced. Therefore PrPC could be important forthe proper cellular localization of other proteins(Keshet et al. 1999). Similarly N-CAM was re-cruited to lipid rafts by PrPC (Santuccione et al.2005).

However, PrPC is involved in a number ofcellular functions and how endocytosis influ-ences them in vivo remains widely unknown; in-ternalization of PrPC could contribute to down-regulation of a signaling event but could also benecessary for signaling. A general involvementof PrPC in vesicle formation could be a possibleexplanation for most of the suggested molecu-lar functions of PrPC because it could regulatesignaling and influence synaptic transmission.

PrP and cell adhesion. Several reports areconsistent with a possible function of PrPC

as a cell adhesion or recognition molecule.Some interaction partners of PrPC identifiedso far have a role in adhesion, including laminin(Graner et al. 2000a,b), a structural componentof basement membrane, laminin-receptor pre-cursor (Gauczynski et al. 2001, Rieger et al.1997), and N-CAM (Schmitt-Ulms et al. 2001).These three molecules are involved in adhe-sion in a diversity of signal transduction path-ways, in differentiation, and in neurite out-growth (Colognato & Yurchenco 2000, Maness& Schachner 2007). Interaction of laminin

460 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 23: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

or N-CAM with PrPC has been associatedmainly with its suggested role in neuritogenesisand neurite outgrowth (Graner et al. 2000a,b;Santuccione et al. 2005). Sales et al. (1998)also proposed that PrPC in its synaptic locationmight stabilize apposing synaptic membranesthrough adhesive mechanisms.

The evidence reported so far could indicatethat PrPC is involved in adhesive mechanisms,but this is likely not its sole function. Adhe-sion molecules that interact biochemically withPrPC could also transduce a PrPC-dependentsignal. The neurite-outgrowth-promoting in-teraction between PrPC and N-CAM, for ex-ample, was associated with activation of thep59Fyn kinase (Santuccione et al. 2005).

Interaction Partners of PrPC

Investigators often hypothesized that PrPC ex-erts its function via interaction with othercell-surface components. GPI-anchored pro-teins would need to interact with a transmem-brane adaptor to influence intracellular signal-transduction pathways, thereby enabling thetransduction of an extracellular signal. An ex-ample of such a protein is the urokinase-typeplasminogen activator receptor uPAR, whichis involved in cellular adhesion, differentia-tion, proliferation, and migration mediated bythe interaction with transmembrane adaptorssuch as integrins, G protein–coupled receptors,and caveolins (Blasi & Carmeliet 2002). Analo-gously, PrPC may bind to a transmembrane pro-tein or to a protein complex that mediates func-tional association with intracellular pathways.

The toxic deletion mutants of PrPC maydestroy such a complex by competing for thebinding of some complex components yet fail-ing to interact with the signal-transducing com-ponents. Indeed, several models for the toxi-city of PrPC deletion mutants have proposedthat PrPC binds to a transmembrane receptorand that deletion mutants induce a toxic sig-nal (Li et al. 2007) or prevent a survival signal(Baumann et al. 2007, Shmerling et al. 1998).

The commonly shared opinion that PrPC

binds to a receptor might explain the tremen-

dous effort put into the identification of its in-teraction partners by different methods such asyeast-two hybrid, coimmunoprecipitations, andcross-linking experiments.

All PrPC interaction partners identified thusfar are summarized in Table 1. They in-clude membrane proteins (receptors, enzymes,Caveolin-1, Na-K-ATPase, and a potassiumchannel), cytoplasmic proteins (componentsof the cytoskeleton, heat-shock proteins, andadaptor proteins involved in signaling), andeven the nuclear protein CBP70. Tantalizingly,several of these interactions partners are knownto play a role in synaptic vesicle function. Un-fortunately, the physiological relevance of mostof the proposed interaction partners remainsunconfirmed. Proteins that are not even lo-calized to the outer leaflet of the cell mem-brane, where PrPC is located and believed toexert its function, would at least require an addi-tional interacting cell component, meaning thattheir interaction with PrPC must be indirectat best.

Another possible explanation for cytosolicinteraction partners is the suggested presenceof transmembranous variants of PrPC, termedNtmPrP and CtmPrP. However, under normalconditions they have been described at best inminute amounts only (Hegde et al. 1998, 1999;Stewart & Harris 2001). Cytosolic PrP was laterdetected (Ma & Lindquist 2002, Ma et al. 2002).The significance of these findings still remainsobscure.

One must consider methodical bottlenecks.Because PrPC is exposed to the extracellularspace, it is questionable whether a yeast two-hybrid screen that artificially exposes PrPC tothe cytosolic compartment with its differentbiochemical composition is the most appropri-ate method by which to study PrPC interactionpartners. A high number of false-positiveresults is to be expected, and therefore it isparticularly important that additional methodsare used to confirm interactions identified byyeast-two hybrid.

One item of paramount importance in im-munoprecipitation experiments is the choiceof the detergent conditions to allow for weak

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 24: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Table 1 Molecules identified as interaction partners to PrPC or PrPSc a

Interactionpartner Subcellular localization Method Binding site

Function of interaction partner andreferences

Hsp60 and GoEL Mitochondria Y2H 180–210 Chaperone (Edenhofer et al. 1996)Laminin receptorprecursor

Plasma membrane Y2H, Co-IP 144–179 Cell adhesion (Hundt et al. 2001,Rieger et al. 1997)

Laminin Extracellular, basementmembrane

Binding assay Unknown Signal transduction, cell adhesion,neuritogenesis (Graner et al. 2000a,b)

Synapsin Ib Synaptic vesicles Y2H, Co-IP 23–100 and90–231

Synaptic vesicle formation, modulation ofneurotransmitter release and in synap-togenesis (Spielhaupter & Schatzl 2001)

Grb2 Cytoplasm Y2H, Co-IP 23–100,90–231

Adaptor protein mediating signal transduc-tion (Spielhaupter & Schatzl 2001)

Pint1 Unknown Y2H, Co-IP 90–231 Unknown (Spielhaupter & Schatzl 2001)N-CAM Plasma membrane

(transmembrane/GPI-anchored)

Formaldehydecross-linking

141–176 Cell adhesion, signaling, etc. (Maness &Schachner 2007, Schmitt-Ulms et al.2001)

Stress-inducibleprotein 1 (STI 1)

Cytoplasm, plasmamembrane?

ComplementaryhydropathyCo-IP

113–128 Heat shock protein (Chiarini et al. 2002,Zanata et al. 2002)

Caveolin-1 Caveolae, plasma membrane(transmembrane)

Co-IP unknown Caveolae formation and endocytosis;cross-linking of PrP induces Fynactivation (Mouillet-Richard et al. 2000)

Fyn kinase Cytoplasm, associated withcytosolic side of plasmamembrane

Co-IP unknown Signal transducer molecule (Mattei et al.2004, Mouillet-Richard et al. 2000)

ZAP70 Cytoplasm Co-IP Unknown Signal transduction during T cellactivation (Mattei et al. 2004)

Synaptophysin Synaptic vesicles(transmembrane)

Co-IP Unknown Presynaptic vesicle protein (Keshet et al.2000)

Neuronal nitricoxide synthase(nNOS)

Intracellular, partlymembrane bound

Co-IP Unknown Production of NO in neuronal tissue,involved in signaling, neurotransmission,etc. (Keshet et al. 2000)

"-dystroglycan Plasma membrane(transmembrane, part of thedystroglycan complex)

Co-IP Unknown Transmembrane protein, bindsextracellularly to !-dystroglycan (boundto laminin) and, intracellularly, todystrophin (Keshet et al. 2000)

Dp71 (dystrophin) Cytoplasmic face of plasmamembrane (part of thedystroglycan complex)

Co-IP Unknown Cytoskeletal protein (Keshet et al. 2000)

!-syntrophin Cytoplasmic face ofmembrane (part of thedystroglycan complex)

Co-IP Unknown Adaptor protein, in sarcolemma and theneuromuscular junction, binds dystro-phins and nNOS (Keshet et al. 2000)

!-tubulin Microtubules, cytoplasm Cross-linking,Co-IP

Unknown Microtubule subunit (Keshet et al. 2000,Nieznanski et al. 2005)

Glutamic aciddecarboxylase(GAD)

Intracellular, vesiclemembrane

Co-IP Unknown Enzyme that synthesizes theneurotransmitter GABA from glutamate(Keshet et al. 2000)

(Continued )

462 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 25: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Table 1 (Continued )

Interactionpartner Subcellular localization Method Binding site

Function of interaction partner andreferences

"-actin Intracellular cytoskeletonprotein

Co-IP, affinitychromatography

Unknown Subunit of microfilaments of thecytoskeleton (Keshet et al. 2000)

BACE-1 Plasma membrane(transmembrane)

Co-IP Unknown APP processing (Parkin et al. 2007)

TREK-1 Plasma membrane(transmembrane)

Bacterial two-hybrid, Co-IP

128–230 Two-pore potassium channel protein(Azzalin et al. 2006)

Bcl-2 Cytoplasmic face of mito-chondrial outer membrane,nuclear envelop, and ER

Yeast two hybrid 72–254 Inhibition of apoptosis (Kurschner &Morgan 1995)

NRAGE(neurotrophinreceptor–interactingMAGE homolog)

Cytoplasm, plasmamembrane associationwhen NGF is bound top75NTR

Y2H, Co-IP 122–231 Binding to p75NTR, permitsNGF/p75NTR-dependent apoptosis(Bragason & Palsdottir 2005, Salehiet al. 2000)

Mouse p45 NF-E2related factor 2(Nrf2)

Cytoplasmic or nuclear(regulated shuttling)

cDNA libraryscreen, interac-tion with AP-tagged soluble PrP

Unknown Transcription factor regulatingexpression of a set of detoxifying andantioxidant enzyme genes

Mouse amyloidprecursor-likeprotein 1 (APLP1)

Plasma membrane(transmembrane)

cDNA libraryscreen, interac-tion with APtagged soluble PrP

Unknown Neuronal survival and neuriteoutgrowth (Sakai & Hohjoh 2006,Yehiely et al. 1997)

!B-crystalline Cytoplasm Y2H Unknown Small heat-shock protein, chaperoneRibosomal proteinP0

Ribosomes Affinitychromatography

Unknown Important for ribosomal activity(Sun et al. 2005)

CNPase Myelin, cytoplasmic face,microtubule-associated

Affinitychromatography

Unknown Membrane anchor for microtubules,required for maintaining the integrityof paranodes and axons (Petrakis &Sklaviadis 2006)

Creatine kinase-B Cytoplasm, intracellularenzyme

Affinitychromatography

Unknown Catalyzes transfer of phosphatebetween ATP and variousphosphogens like creatine phosphate(Petrakis & Sklaviadis 2006)

NSE (neuron-specific enolase)

Cytoplasm, intracellularenzyme

Affinitychromatography

Unknown Enolase is a glycolytic enzyme; NSEcontains #-subunit, expressedprimarily in neurons and inneuroendocrine cells (Petrakis &Sklaviadis 2006)

Clathrin heavychain 1

Cytosolic face of coatedvesicles and coated pits

Affinitychromatography

Unknown Clathrin: major protein component ofthe cytoplasmic face of coated vesiclesand coated pits, involved in intracellu-lar trafficking of receptors and endo-cytosis (Petrakis & Sklaviadis 2006)

!-spectrin Cytoskeletal protein, linesintracellular side of theplasma membrane

Affinitychromatography

Unknown Maintenance of plasma membraneintegrity and cytoskeletal structure(Petrakis & Sklaviadis 2006)

(Continued )

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 26: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Table 1 (Continued )

Interactionpartner Subcellular localization Method Binding site

Function of interaction partner andreferences

Glial fibrillaryacidic protein(GFAP)

Intracellular, intermediatefilament in glial cells such asastrocytes

Affinitychromatography

Unknown Different cellular processes, like cellstructure and movement, cellcommunication (Petrakis & Sklaviadis2006)

Na+/K+ ATPase!3

Plasma membrane Affinitychromatography

Unknown Catalytic subunit of P-type ATPase,active transport of cations across cellmembranes, maintain ionic gradients(Petrakis & Sklaviadis 2006)

PLP (proteolipidprotein)

Myelin (transmembrane) Cross-linking Unknown Major component of central nervoussystem myelin (Petrakis & Sklaviadis2006)

STXBP1 (Syntaxinbinding protein 1)

Membrane-associated inpresence of syntaxin

Cross-linking Unknown Participance in regulation of synapticvesicle docking and fusion (Petrakis &Sklaviadis 2006)

$14–3–3 Intracellular Cross-linking Unknown Phosphoserine or phosphothreoninebinding protein binds diverse signalingproteins, including kinases,phosphatases, and transmembranereceptors, thereby regulating mostcellular processes (Petrakis &Sklaviadis 2006)

BASP1 Bound to plasma membrane,calmodulin, and actin

Cross-linking Unknown Accumulation mainly in axon endings(growth cones and presynaptic area ofsynapses) (Petrakis & Sklaviadis 2006)

Vitronectin Extracellular matrixglycoprotein

Binding andcompetitionassay

105–119 Axonal growth in the peripheralnervous system (Hajj et al. 2007)

CBP70 Nuclear [and cytoplasmatic(Rousseau et al. 1997)]

Co-IP Lectin (Rybner et al. 2002)

Glycosaminoglycans(GAG)

Connective tissues,covalently linked to proteinsto form proteoglycans

ELISA, SPR,heparin-agarosebinding assay

23–35,23–52,53–93,

110–128

Various functions (Caughey et al. 1994,Pan et al. 2002, Warner et al. 2002)

ApoE Secreted Co-IP,cross-linking

23–90109–141

Main apolipoprotein of chylomicrons,involved in neurodegenerative diseasessuch as Alzheimer disease (Baumannet al. 2000, Gao et al. 2006,Schmitt-Ulms et al. 2004)

Plasminogen Secreted Immobilizedserum proteinsprobed withPrPC or PrPres

Unknown Inactive zymogen form of plasmin,which participates in thrombolysis orextracellular matrix degradation (Elliset al. 2002, Fischer et al. 2000)

Rdj2 Cytoplasmic face ofmembranes

Co-IP with GSTfusion proteins

Unknown Chaperone (Beck et al. 2006)

Casein kinase 2!/!’

Cytoplasmic Far-Western, SPR 105–242 Protein kinase (Meggio et al. 2000)

aCo-IP, coimmunoprecipitation; Y2H, yeast-two hybrid; SPR, surface plasmon resonance.

464 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 27: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

and transient protein-protein interactions butdestroy artificial or unspecific interactions. Theinfluence of the detergent used was exemplifiedin one study in which the interactions betweenPrPC and the dystroglycan complex were stud-ied as a function of the detergent used and onthe integrity of lipid rafts (Keshet et al. 2000).Some of the interactions identified by immuno-precipitation may be artifactual—a result thatcould be avoided by using stringent controls in-cluding knockout tissues and specific antibodycompetition experiments.

CONCLUSIONSDespite the progress discussed above, severalimportant issues in the prion field remain un-resolved. Most conspicuously, both the phys-iological function of PrPC and the molecularpathways leading to the fatal neurodegenera-tion in prion diseases remain unknown. Thesetwo issues may be linked, and elucidation of thephysiological function of PrPC has the potentialto help researchers understand the mechanismsinvolved in prion-induced neurodegeneration.

Studies of mice carrying targeted disrup-tions of any given gene have often providedresearchers with useful tools to identify therespective gene product’s function. However,the many lines of mice lacking PrPC that havebeen generated independently by homologousrecombination have failed to uncover a clearmolecular physiological function of PrPC. Themost obvious phenotype was their resistance toprion infection. Nevertheless, an overwhelm-ing number of molecular, structural, or func-tional alterations have been reported in Prnp"/"

mice.Cellular processes in the nervous system that

have been influenced by the Prnp genotypeinclude neuronal survival, neurite outgrowth,synapse formation, maintenance, and function,as well as maintenance of myelinated fibers.

One of the most frequently cited cellular func-tions of PrPC is a survival-promoting effect onneuronal and nonneuronal cells. This cytopro-tective function has been mediated by antiapop-totic or antioxidative mechanisms, but nothingis known about the proximal mechanisms me-diating such effects.

The frequently voiced opinion that PrPC

binds to a receptor has driven a large ef-fort toward the identification of its interac-tion partners by different methods such asyeast-two hybrid, coimmunoprecipitations, andcross-linking experiments. A rather large num-ber of interaction partners have been identified,yet a functional interaction was unambiguouslydiscovered for none of them.

Some of the latter results may need to bereconsidered critically. Several reports sufferfrom intrinsic methodological shortcomings,some of which were unavoidable at the time ofpublication. Furthermore, the likelihood of in-teractions between membrane-attached extra-cellular PrPC with cytosolic or mitochondrialmolecule is counterintuitive and may point toartifactual effects.

Although it is highly plausible in our opin-ion, the connection between the normal func-tion of PrPC and the neurotoxicity of prionsremains admittedly hypothetical and lacks ex-perimental confirmation. Alternative scenariosare thinkable, and it is not impossible that thecascade of events outlined above will prove in-correct. However, the depth of the knowledgegaps in prion biology, along with the medi-cal importance of the neurodegeneration prob-lem, indicates that many exciting discoveriesstill lie ahead. Therefore, despite the disap-pearance of “mad cow disease” from the me-dia and from public awareness, the field ofprion science remains an excellent choice forambitious PhD students and postdocs willingto make an impact in current experimentalbiology.

DISCLOSURE STATEMENTThe authors are not aware of any biases that might be perceived as affecting the objectivity of thisreview.

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 28: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

LITERATURE CITEDAguzzi A. 1996. Between cows and monkeys. Nature 381:734–55Aguzzi A. 2007. Prion biology: the quest for the test. Nat. Methods 4:614–16Aguzzi A, Glatzel M. 2006. Prion infections, blood and transfusions. Nat. Clin. Pract. Neurol.

2:321–29Aguzzi A, Weissmann C. 1996. Spongiform encephalopathies: a suspicious signature. Nature

383:666–67Andrews NJ, Farrington CP, Ward HJ, Cousens SN, Smith PG, et al. 2003. Deaths from variant

Creutzfeldt-Jakob disease in the UK. Lancet 361:751–52Aronoff-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 CDspectroscopy. Biochemistry 39:13760–71

Atarashi R, Moore RA, Sim VL, Hughson AG, Dorward DW, et al. 2007. Ultrasensitive detectionof scrapie prion protein using seeded conversion of recombinant prion protein. Nat. Methods4:645–50

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 withscrapie. Neurobiol. Dis. 6:406–23

Baumann F, Tolnay M, Brabeck C, Pahnke J, Kloz U, et al. 2007. Lethal recessive myelin toxicityof 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 abinding 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 prionPrP(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 proteinhomologue 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-geneticpropagation 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 neuronsagainst Bax-mediated apoptosis. J. Biol. Chem. 276:39145–49

Bouzamondo-Bernstein E, Hopkins SD, Spilman P, Uyehara-Lock J, Deering C, et al. 2004. Theneurodegeneration sequence in prion diseases: evidence from functional, morphological andultrastructural 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 neuronalapoptosis. Mol. Cell Neurosci. 29:232–44

Brandner S, Isenmann S, Raeber A, Fischer M, Sailer A, et al. 1996. Normal host prion proteinnecessary for scrapie-induced neurotoxicity. Nature 379:339–43

Brown DR, Nicholas RS, Canevari L. 2002. Lack of prion protein expression results in a neuronalphenotype sensitive to stress. J. Neurosci. Res. 67:211–24

Brown DR, Schmidt B, Kretzschmar HA. 1997a. Effects of oxidative stress on prion proteinexpression in PC12 cells. Int. J. Dev. Neurosci. 15:961–72

466 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 29: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Brown DR, Schulz-Schaeffer WJ, Schmidt B, Kretzschmar HA. 1997b. Prion protein-deficientcells 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 hasan 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-form encephalopathy: the National Institutes of Health series of 300 cases of experimentallytransmitted disease. Ann. Neurol. 35:513–29

Bruce ME, Will RG, Ironside JW, McConnell I, Drummond D, et al. 1997. Transmissions tomice 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 resistantto scrapie. Cell 73:1339–47

Bueler H, Fischer M, Lang Y, Bluethmann H, Lipp HP, et al. 1992. Normal development andbehaviour 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 glycosylatedPrP proteins can have different neuronal trafficking in brain but do not acquire scrapie-likeproperties. 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. PflugersArch. 442:223–29

Castagna A, Campostrini N, Farinazzo A, Zanusso G, Monaco S, Righetti PG. 2002. Comparativetwo-dimensional mapping of prion protein isoforms in human cerebrospinal fluid and centralnervous system. Electrophoresis 23:339–46

Castilla J, Saa P, Hetz C, Soto C. 2005a. In vitro generation of infectious scrapie prions. Cell121:195–206

Castilla J, Saa P, Soto C. 2005b. Detection of prions in blood. Nat. Med. 11:982–85Caughey 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. Theoctarepeat domain of the prion protein binds Cu(II) with three distinct coordination modesat pH 7.4. J. Am. Chem. Soc. 127:12647–56

Chen S, Mange A, Dong L, Lehmann S, Schachner M. 2003. Prion protein as trans-interactingpartner 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 cointernalizationof 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 proteinresults 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 prionprotein 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 miceexpressing 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 betweenpathogenic and infectious properties of the prion protein. J. Virol. 77:7611–22

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 30: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Choi SI, Ju WK, Choi EK, Kim J, Lea HZ, et al. 1998. Mitochondrial dysfunction inducedby oxidative stress in the brains of hamsters infected with the 263 K scrapie agent. ActaNeuropathol. 96:279–86

Clinton J, Forsyth C, Royston MC, Roberts GW. 1993. Synaptic degeneration is the primaryneuropathological 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: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—anacquired 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 isnecessary for normal synaptic function. Nature 370:295–97

Collins S, McLean CA, Masters CL. 2001. Gerstmann-Straussler-Scheinker syndrome, fatal fa-milial insomnia, and kuru: a review of these less common human transmissible spongiformencephalopathies. 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. Uber eine eigenartige herdformige erkrankung des zentralnervensystems.Z. Ges. Neurol. Psychiatr. 57:1–19

Criado JR, Sanchez-Alavez M, Conti B, Giacchino JL, Wills DN, et al. 2005. Mice devoid ofprion protein have cognitive deficits that are rescued by reconstitution of PrP in neurons.Neurobiol. Dis. 19:255–65

Cuille J, Chelle 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 LTPin 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 substratesand synthetic polyanions. J. Biol. Chem. 280:26873–79

Deleault NR, Harris BT, Rees JR, Supattapone S. 2007. Formation of native prions from minimalcomponents 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 preventshuman breast carcinoma cell line from tumor necrosis factor !-induced cell death. CancerRes. 64:719–27

Di Guglielmo GM, Le Roy C, Goodfellow AF, Wrana JL. 2003. Distinct endocytic pathwaysregulate TGF-" 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-persontransmission 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 proteinPrPc 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-likelipid membranes. Biol. Chem. 388:79–89

Ellis V, Daniels M, Misra R, Brown DR. 2002. Plasminogen activation is stimulated by prionprotein 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) encodinga fish prion protein. Comp. Biochem. Physiol. B 147:566–73

Ferrer I, Rivera R, Blanco R, Martı E. 1999. Expression of proteins linked to exocytosis andneurotransmission in patients with Creutzfeldt-Jakob disease. Neurobiol. Dis. 6:92–100

468 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 31: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

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, Rulicke T, Raeber A, Sailer A, Moser M, et al. 1996. Prion protein (PrP) withamino-proximal deletions restoring susceptibility of PrP knockout mice to scrapie. EMBO J.15:1255–64

Fischer MB, Roeckl C, Parizek P, Schwarz HP, Aguzzi A. 2000. Binding of disease-associatedprion protein to plasminogen. Nature 408:479–83

Flechsig E, Shmerling D, Hegyi I, Raeber AJ, Fischer M, et al. 2000. Prion protein devoid ofthe octapeptide repeat region restores susceptibility to scrapie in PrP knockout mice. Neuron27:399–408

Flechsig E, Weissmann C. 2004. The role of PrP in health and disease. Curr. Mol. Med. 4:337–53Fournier 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 Cubinding 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 syndrometo chimpanzees. Nature 209:794–96

Gajdusek DC, Gibbs CJ Jr, Alpers M. 1967. Transmission and passage of experimenal “kuru” tochimpanzees. Science 155:212–14

Gajdusek DC, Gibbs CJ Jr, Asher DM, David E. 1968. Transmission of experimental kuru to thespider monkey (Ateles geoffreyi). Science 162:693–94

Gajdusek DC, Reid LH. 1961. Studies on kuru. IV. The kuru pattern in Moke, a representativeFore 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 withboth 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 lamininreceptor acts as the cell-surface receptor for the cellular prion protein. EMBO J. 20:5863–75

Gerstmann J. 1928. Uber ein noch nicht beschriebenes Reflexphanomen bei einer Erkrankungdes zerebellaren Systems. Wien. Medizin Wochenschr. 78:906–8

Gerstmann J, Straussler E, Scheinker I. 1936. Uber eine eigenartige hereditar-familiareErkrankung des Zentralnervensystems. Zugleich ein Beitrag zur Frage des vorzeitigen lokalenAlterns. 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 insomniaand 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 prionprotein binds laminin and mediates neuritogenesis. Brain Res. Mol. Brain Res. 76:85–92

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 32: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

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 prionprotein. FEBS Lett. 482:257–60

Grigoriev V, Escaig-Haye F, Streichenberger N, Kopp N, Langeveld J, et al. 1999. Submicroscopicimmunodetection of PrP in the brain of a patient with a new-variant of Creutzfeldt-Jakobdisease. Neurosci. Lett. 264:57–60

Hadlow WJ. 1959. Scrapie and kuru. Lancet 2:289–90Hajj GN, Lopes MH, Mercadante AF, Veiga SS, da Silveira RB, et al. 2007. Cellular prion protein

interaction with vitronectin supports axonal growth and is compensated by integrins. J. CellSci. 120:1915–26

Hegde RS, Mastrianni JA, Scott MR, DeFea KA, Tremblay P, et al. 1998. A transmembrane formof the prion protein in neurodegenerative disease. Science 279:827–34

Hegde RS, Tremblay P, Groth D, DeArmond SJ, Prusiner SB, Lingappa VR. 1999. Transmissibleand 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. 8:324–30Hill AF, Desbruslais M, Joiner S, Sidle KC, Gowland I, et al. 1997. The same prion strain causes

vCJD and BSE. Nature 389:448–50, 526Holscher 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-typePrP(Sc) accumulation. J. Virol. 72:1153–59

Hornemann S, Korth C, Oesch B, Riek R, Wider G, et al. 1997. Recombinant full-length murineprion 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 missensevariant to Gerstmann-Straussler syndrome. Nature 338:342–45

Hsiao KK, Groth D, Scott M, Yang SL, Serban H, et al. 1994. Serial transmission in rodents ofneurodegeneration 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 interactiondomains 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 proteinin vivo. Biol. Chem. 384:1279–85

Isaacs JD, Jackson GS, Altmann DM. 2006. The role of the cellular prion protein in the immunesystem. Clin. Exp. Immunol. 146:1–8

Itoh Y, Yamada M, Hayakawa M, Shozawa T, Tanaka J, et al. 1994. A variant of Gerstmann-Straussler-Scheinker disease carrying codon 105 mutation with codon 129 polymorphism ofthe prion protein gene: a clinicopathological study. J. Neurol. Sci. 127:77–86

Jakob A. 1921. Uber eigenartige erkrankungen des zentralnervensystems mit bemerkenswertemanatomischem befunde (spastische pseudosklerose-encephalomyelopathie mit disseminiertendegenerationsherden). 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 murinehippocampus. Neuropathol. Appl. Neurobiol. 26:41–54

Johnston AR, Fraser JR, Jeffrey M, MacLeod N. 1998. Synaptic plasticity in the CA1 area of thehippocampus of scrapie-infected mice. Neurobiol. Dis. 5:188–95

Jones S, Batchelor M, Bhelt D, Clarke AR, Collinge J, Jackson GS. 2005. Recombinant prionprotein does not possess SOD-1 activity. Biochem. J. 392:309–12

470 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 33: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

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 hippocampalneurons in vitro. J. Neurochem. 95:1373–86

Kaneko K, Vey M, Scott M, Pilkuhn S, Cohen FE, Prusiner SB. 1997. COOH-terminal sequenceof the cellular prion protein directs subcellular trafficking and controls conversion into thescrapie isoform. Proc. Natl. Acad. Sci. USA 94:2333–38

Keshet GI, Bar-Peled O, Yaffe D, Nudel U, Gabizon R. 2000. The cellular prion protein colocalizeswith the dystroglycan complex in the brain. J. Neurochem. 75:1889–97

Keshet GI, Ovadia H, Taraboulos A, Gabizon R. 1999. Scrapie-infected mice and PrP knockoutmice 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 inthe hydrophobic core of the prion protein interfere with folding and attachment of theglycosylphosphatidylinositol anchor. J. Biol. Chem. 280:9320–29

Kirkham M, Parton RG. 2005. Clathrin-independent endocytosis: new insights into caveolae andnoncaveolar 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 ofprion proteins accumulates in the synaptic structures of the central nervous system in patientswith 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 ofantioxidant 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. Speciesspecificity in the cell-free conversion of prion protein to protease-resistant forms: a modelfor the scrapie species barrier. Proc. Natl. Acad. Sci. USA 92:3923–27

Korth C, Kaneko K, Prusiner SB. 2000. Expression of unglycosylated mutated prion proteinfacilitates 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. Mutationsof 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 theyeast 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 preventneuronal cell-line death. Nature 400:225–26

Laine J, Marc ME, Sy MS, Axelrad H. 2001. Cellular and subcellular morphological localizationof 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 metabolismin 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 mammalianprions. 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 lethalityin 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 prionprotein 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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 34: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Llewelyn CA, Hewitt PE, Knight RS, Amar K, Cousens S, et al. 2004. Possible transmission ofvariant 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 cellularprion and stress-inducible protein 1 promotes neuritogenesis and neuroprotection by distinctsignaling pathways. J. Neurosci. 25:11330–39

Lugaresi E, Medori R, Montagna P, Baruzzi A, Cortelli P, et al. 1986. Fatal familial insomnia anddysautonomia 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 thecytosol. 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 LTPexpression in aged mice. Brain Res. 1097:11–18

Maglio LE, Perez MF, Martins VR, Brentani RR, Ramirez OA. 2004. Hippocampal synapticplasticity in mice devoid of cellular prion protein. Brain Res. Mol. Brain Res. 131:58–64

Mallucci G, Dickinson A, Linehan J, Klohn PC, Brandner S, Collinge J. 2003. Depleting neuronalPrP 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 prionprotein alters hippocampal CA1 properties, but does not result in neurodegeneration. EMBOJ. 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 micecarrying 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 fromthe Gerstmann-Straussler syndrome with an analysis of the various forms of amyloid plaquedeposition 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 conformationin 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 componentof 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 proteinaccumulation 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 amodulator 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 proteinexpression affects mitochondrial numbers and morphology. Biochem. Biophys. Res. Commun.291:372–77

472 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 35: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Milhavet O, Lehmann S. 2002. Oxidative stress and the prion protein in transmissible spongiformencephalopathies. 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 theoctarepeat region in neuroprotective function of the cellular prion protein. Brain Pathol.17:174–83

Moore RC, Lee IY, Silverman GL, Harrison PM, Strome R, et al. 1999. Ataxia in prion protein(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. Signaltransduction 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. Heritabledisorder resembling neuronal storage disease in mice expressing prion protein with deletionof an !-helix. Nat. Med. 3:750–55

Muramoto T, Scott M, Cohen FE, Prusiner SB. 1996. Recombinant scrapie-like prion protein of106 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 micedeficient for normal prion protein expression. Biochim. Biophys. Acta 1772:645–53

Neuendorf E, Weber A, Saalmueller A, Schatzl H, Reifenberg K, et al. 2004. Glycosylationdeficiency at either one of the two glycan attachment sites of cellular prion protein preservessusceptibility 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 interactionbetween 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 proteintransgene rescues mice deficient for the prion protein gene from Purkinje cell degenerationand demyelination. Lab. Invest. 79:689–97

Oidtmann B, Simon D, Holtkamp N, Hoffmann R, Baier M. 2003. Identification of cDNAs fromJapanese pufferfish (Fugu rubripes) and Atlantic salmon (Salmo salar) coding for homologuesto 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 interactswith glycosaminoglycans. Biochem. J. 368:81–90

Parchi P, Capellari S, Chin S, Schwarz HB, Schecter NP, et al. 1999. A subtype of sporadic priondisease mimicking fatal familial insomnia. Neurology 52:1757–63

Parkin ET, Watt NT, Hussain I, Eckman EA, Eckman CB, et al. 2007. Cellular prion proteinregulates 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 insightsand common mechanisms. Traffic 4:724–38

Peden AH, Head MW, Ritchie DL, Bell JE, Ironside JW. 2004. Preclinical vCJD after bloodtransfusion in a PRNP codon 129 heterozygous patient. Lancet 364:527–29

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 36: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Peters PJ, Mironov A Jr, Peretz D, van Donselaar E, Leclerc E, et al. 2003. Trafficking of prionproteins 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 nativePrPC. 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

Priola SA, Lawson VA. 2001. Glycosylation influences cross-species formation of protease-resistant prion protein. EMBO J. 20:6692–99

Prusiner SB, Scott MR. 1997. Genetics of prions. Annu. Rev. Genet. 31:139–75Qin 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-timeof 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 andDoppel 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 structureof 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. Disparateevolution of prion protein domains and the distinct origin of Doppel- and prion-related locirevealed 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 earlystages 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 Purkinjecell 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 prionprotein inhibits proapoptotic Bax conformational change in human neurons and in breastcarcinoma MCF-7 cells. Cell Death Differ. 12:783–95

Roucou X, Guo Q, Zhang Y, Goodyer CG, LeBlanc AC. 2003. Cytosolic prion protein is nottoxic 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 inprion diseases. J. Mol. Med. 83:3–11

Rousseau C, Felin M, Doyennette-Moyne MA, Seve AP. 1997. CBP70, a glycosylated nuclearlectin. J. Cell. Biochem. 66:370–85

Rybner C, Finel-Szermanski S, Felin M, Sahraoui T, Rousseau C, et al. 2002. The cellular prionprotein: a new partner of the lectin CBP70 in the nucleus of NB4 human promyelocyticleukemia cells. J. Cell. Biochem. 84:408–19

Saa P, Castilla J, Soto C. 2006. Presymptomatic detection of prions in blood. Science 313:92–94Sakaguchi 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 380:528–31Sakai 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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 37: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

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-dependentapoptosis. Neuron 27:279–88

Sales N, Rodolfo K, Hassig R, Faucheux B, Di Giamberardino L, Moya KL. 1998. Cellular prionprotein 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 neuronalreceptor NCAM to lipid rafts to activate p59fyn and to enhance neurite outgrowth. J. CellBiol. 169:341–54

Schmitt-Ulms G, Hansen K, Liu J, Cowdrey C, Yang J, et al. 2004. Time-controlled transcardiacperfusion 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 neuralcell 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. NADPHoxidase 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. Cell93:203–14

Shyng SL, Heuser JE, Harris DA. 1994. A glycolipid-anchored prion protein is endocytosed viaclathrin-coated pits. J. Cell. Biol. 125:1239–50

Shyu WC, Lin SZ, Chiang MF, Ding DC, Li KW, et al. 2005. Overexpression of PrPC byadenovirus-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 turtleprion protein. FEBS Lett. 469:33–38

Spielhaupter C, Schatzl HM. 2001. PrPC directly interacts with proteins involved in signalingpathways. J. Biol. Chem. 276:44604–12

Spudich A, Frigg R, Kilic E, Kilic U, Oesch B, et al. 2005. Aggravation of ischemic brain injuryby 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 mammalianneurogenesis. Proc. Natl. Acad. Sci. USA 103:3416–21

Stewart RS, Harris DA. 2001. Most pathogenic mutations do not alter the membrane topologyof 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 Golgiapparatus of neurons. J. Biol. Chem. 280:15855–64

Stewart RS, Piccardo P, Ghetti B, Harris DA. 2005. Neurodegenerative illness in transgenic miceexpressing 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 bindscopper(II) ions. Biochemistry 37:7185–93

Strumbo B, Ronchi S, Bolis LC, Simonic T. 2001. Molecular cloning of the cDNA coding forXenopus 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 !B-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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 38: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

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. CellBiol. 21:2608–16

Suzuki T, Kurokawa T, Hashimoto H, Sugiyama M. 2002. cDNA sequence and tissue expressionof Fugu rubripes prion protein-like: a candidate for the teleost orthologue of tetrapod PrPs.Biochem. Biophys. Res. Commun. 294:912–17

Tateishi J, Brown P, Kitamoto T, Hoque ZM, Roos R, et al. 1995. First experimental transmissionof fatal familial insomnia. Nature 376:434–35

Tateishi J, Kitamoto T, Hashiguchi H, Shii H. 1988. Gerstmann-Straussler-Scheinker disease:immunohistological and experimental studies. Ann. Neurol. 24:35–40

Tateishi J, Kitamoto T, Hoque MZ, Furukawa H. 1996a. Experimental transmission ofCreutzfeldt-Jakob disease and related diseases to rodents. Neurology 46:532–37

Tateishi J, Kitamoto T, Kretzschmar H, Mehraein P. 1996b. Immunhistological evaluation ofCreutzfeldt-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 regionsof 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. Interactionsbetween wild-type and mutant prion proteins modulate neurodegeneration in transgenicmice. Genes Dev. 10:1736–50

Tobler I, Gaus SE, Deboer T, Achermann P, Fischer M, et al. 1996. Altered circadian activityrhythms 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 redoxsignalling at the synapse. J. Neurochem. 86:538–44

Vey M, Pilkuhn S, Wille H, Nixon R, DeArmond SJ, et al. 1996. Subcellular colocalization of thecellular 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 sporadicCJD or other neurologic diseases. Transfusion 41:441–48

Waggoner DJ, Drisaldi B, Bartnikas TB, Casareno RL, Prohaska JR, et al. 2000. Brain coppercontent 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. Ultrastructuralchanges in the optic nerves of rodents with experimental Creutzfeldt-Jakob Disease (CJD),Gerstmann-Straussler-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 bindingsites 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 cellularprion protein (PrPc) after focal cerebral ischemia and influence of lesion severity. Neurosci.Lett. 372:146–50

476 Aguzzi · Baumann · Bremer

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 39: Artigo 1- The Prion´s Elusive Reason for Being

ANRV346-NE31-19 ARI 14 May 2008 14:38

Weise J, Sandau R, Schwarting S, Crome O, Wrede A, et al. 2006. Deletion of cellular prionprotein results in reduced Akt activation, enhanced postischemic caspase-3 activation, andexacerbation 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 progressivespongiform encephalopathy in cattle. Vet. Rec. 121:419–20

Westaway D, DeArmond SJ, Cayetano-Canlas J, Groth D, Foster D, et al. 1994. Degenerationof skeletal muscle, peripheral nerves, and the central nervous system in transgenic miceoverexpressing 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-deficientneurons reveal lower glutathione reductase activity and increased susceptibility to hydrogenperoxide 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 stressmarkers 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 andpremortem 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 ofcandidate 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-inducibleprotein 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 variabilityin 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-termrepopulating 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-induceddifferentiation. J. Neurosci. 20:5671–78

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

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 40: Artigo 1- The Prion´s Elusive Reason for Being

AR346-FM ARI 20 May 2008 15:1

Annual Review ofNeuroscience

Volume 31, 2008Contents

Cerebellum-Like Structures and Their Implications for CerebellarFunctionCurtis C. Bell, Victor Han, and Nathaniel B. Sawtell ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 1

Spike Timing–Dependent Plasticity: A Hebbian Learning RuleNatalia Caporale and Yang Dan ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! !25

Balancing Structure and Function at Hippocampal Dendritic SpinesJennifer N. Bourne and Kristen M. Harris ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! !47

Place Cells, Grid Cells, and the Brain’s Spatial Representation SystemEdvard I. Moser, Emilio Kropff, and May-Britt Moser ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! !69

Mitochondrial Disorders in the Nervous SystemSalvatore DiMauro and Eric A. Schon ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! !91

Vestibular System: The Many Facets of a Multimodal SenseDora E. Angelaki and Kathleen E. Cullen ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 125

Role of Axonal Transport in Neurodegenerative DiseasesKurt J. De Vos, Andrew J. Grierson, Steven Ackerley, and Christopher C.J. Miller ! ! ! 151

Active and Passive Immunotherapy for Neurodegenerative DisordersDavid L. Brody and David M. Holtzman ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 175

Descending Pathways in Motor ControlRoger N. Lemon ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 195

Task Set and Prefrontal CortexKatsuyuki Sakai ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 219

Multiple Sclerosis: An Immune or Neurodegenerative Disorder?Bruce D. Trapp and Klaus-Armin Nave ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 247

Multifunctional Pattern-Generating CircuitsK.L. Briggman and W.B. Kristan, Jr. ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 271

Retinal Axon Growth at the Optic Chiasm: To Cross or Not to CrossTimothy J. Petros, Alexandra Rebsam, and Carol A. Mason ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 295

v

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.

Page 41: Artigo 1- The Prion´s Elusive Reason for Being

AR346-FM ARI 20 May 2008 15:1

Brain Circuits for the Internal Monitoring of MovementsMarc A. Sommer and Robert H. Wurtz ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 317

Wnt Signaling in Neural Circuit AssemblyPatricia C. Salinas and Yimin Zou ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 339

Habits, Rituals, and the Evaluative BrainAnn M. Graybiel ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 359

Mechanisms of Self-Motion PerceptionKenneth H. Britten ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 389

Mechanisms of Face PerceptionDoris Y. Tsao and Margaret S. Livingstone ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 411

The Prion’s Elusive Reason for BeingAdriano Aguzzi, Frank Baumann, and Juliane Bremer ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 439

Mechanisms Underlying Development of Visual Maps andReceptive FieldsAndrew D. Huberman, Marla B. Feller, and Barbara Chapman ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 479

Neural Substrates of Language AcquisitionPatricia K. Kuhl and Maritza Rivera-Gaxiola ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 511

Axon-Glial Signaling and the Glial Support of Axon FunctionKlaus-Armin Nave and Bruce D. Trapp ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! ! 535

Signaling Mechanisms Linking Neuronal Activity to Gene Expressionand Plasticity of the Nervous SystemSteven 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 athttp://neuro.annualreviews.org/

vi Contents

Ann

u. R

ev. N

euro

sci.

2008

.31:

439-

477.

Dow

nloa

ded

from

ww

w.a

nnua

lrevi

ews.o

rgby

Scu

ola

Inte

rnaz

iona

le S

uper

iore

di S

tudi

Ava

nzat

i (SI

SSA

) on

03/2

1/11

. For

per

sona

l use

onl

y.