articulo 6. priones

Upload: saray-ramos

Post on 21-Feb-2018

230 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/24/2019 Articulo 6. Priones

    1/23

    http://jgp.sagepub.com/Journal of Geriatric Psychiatry and Neurology

    http://jgp.sagepub.com/content/23/4/277Theonline version of this article can be foundat:

    DOI: 10.1177/0891988710383576

    2010 23: 277 originally published online 11 October 2010J Geriatr Psychiatry NeurolKhalilah Brown and James A. Mastrianni

    The Prion Diseases

    Published by:

    http://www.sagepublications.com

    can be found at:Journal of Geriatric Psychiatry and NeurologyAdditional services and information for

    http://jgp.sagepub.com/cgi/alertsEmail Alerts:

    http://jgp.sagepub.com/subscriptionsSubscriptions:

    http://www.sagepub.com/journalsReprints.navReprints:

    http://www.sagepub.com/journalsPermissions.navPermissions:

    http://jgp.sagepub.com/content/23/4/277.refs.htmlCitations:

    What is This?

    - Oct 11, 2010OnlineFirst Version of Record

    - Nov 2, 2010Version of Record>>

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/content/23/4/277http://jgp.sagepub.com/content/23/4/277http://jgp.sagepub.com/content/23/4/277http://www.sagepublications.com/http://jgp.sagepub.com/cgi/alertshttp://jgp.sagepub.com/cgi/alertshttp://jgp.sagepub.com/subscriptionshttp://www.sagepub.com/journalsReprints.navhttp://www.sagepub.com/journalsReprints.navhttp://www.sagepub.com/journalsPermissions.navhttp://jgp.sagepub.com/content/23/4/277.refs.htmlhttp://online.sagepub.com/site/sphelp/vorhelp.xhtmlhttp://online.sagepub.com/site/sphelp/vorhelp.xhtmlhttp://jgp.sagepub.com/content/early/2010/10/06/0891988710383576.full.pdfhttp://jgp.sagepub.com/content/early/2010/10/06/0891988710383576.full.pdfhttp://jgp.sagepub.com/content/23/4/277.full.pdfhttp://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://online.sagepub.com/site/sphelp/vorhelp.xhtmlhttp://jgp.sagepub.com/content/early/2010/10/06/0891988710383576.full.pdfhttp://jgp.sagepub.com/content/23/4/277.full.pdfhttp://jgp.sagepub.com/content/23/4/277.refs.htmlhttp://www.sagepub.com/journalsPermissions.navhttp://www.sagepub.com/journalsReprints.navhttp://jgp.sagepub.com/subscriptionshttp://jgp.sagepub.com/cgi/alertshttp://www.sagepublications.com/http://jgp.sagepub.com/content/23/4/277http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    2/23

    The Prion Diseases

    Khalilah Brown, MD1 and James A. Mastrianni, MD, PhD1

    Abstract

    The prion diseases are a family of rare neurodegenerative disorders that result from the accumulation of a misfolded isoform ofthe prion protein (PrP), a normal constituent of the neuronal membrane. Five subtypes constitute the known human priondiseases; kuru, Creutzfeldt-Jakob disease (CJD), Gerstmann-Straussler-Scheinker syndrome (GSS), fatal insomnia (FI), and variant

    CJD (vCJD). These subtypes are distinguished, in part, by their clinical phenotype, but primarily by their associated brain histo-pathology. Evidence suggests these phenotypes are defined by differences in the pathogenic conformation of misfolded PrP.Although the vast majority of cases are sporadic, 10% to15% result from an autosomal dominant mutation of the PrPgene (PRNP).General phenotype-genotype correlations can be made for the major subtypes of CJD, GSS, and FI. This paper will review some of

    the general background related to prion biology and detail the clinical and pathologic features of the major prion diseases, with a

    particular focus on the genetic aspects that result in prion disease or modification of its risk or phenotype.

    Keywords

    genetics, neurodegeneration, prion diseases, CJD, GSS, FI

    Introduction

    The human prion disorders include kuru, sporadic Creutzfeldt-

    Jakob disease (sCJD), familial CJD (fCJD), iatrogenic CJD

    (iCJD), Gerstmann-Straussler-Scheinker disease (GSS), fatal

    insomnia (FI), and new variant CJD (vCJD). As a group, they

    are unique among neurodegenerative diseases in that they not

    only have a sporadic and genetic occurrence, but they are hor-

    izontally transmissible. Additionally, prion diseases affect ani-

    mals, some of which include scrapie of sheep, chronic wasting

    disease of deer and elk (CWD), and bovine spongiform ence-

    phalopathy (BSE). The transmissible nature of these diseases

    was first demonstrated experimentally in 1936 by Cuille and

    Chelle through intraocular administration of scrapie-infected

    spinal cord to a goat.1 Thirty years later, kuru, a disease of the

    Fore people of New Guinea related to the practice of cannibal-

    ism, was transmitted to chimpanzees by Gajdusek,2 and 2 years

    later, Gibbs followed suit with transmission of CJD.3 Initially

    proposed to be a slow virus,4,5 the etiologic agent of these

    diseases is now recognized as the prion, a misfolded isoform

    of the prion protein (PrP).6-8 PrP exists in 2 major conforma-

    tional isoforms: the nonpathogenic, predominantly a-helical,

    protease-sensitive, cellular isoform (PrPC) and the pathogenic,

    protease-resistant scrapie-inducing isoform (PrPSc) that is high

    in b-pleated sheet structure9 (Figure 1). The initial conversion

    of PrPC to PrPSc may be spontaneous or mutation-induced; how-

    ever, once an infectious unit has been generated, propagation

    of PrPSc occurs via protein-protein interaction, such that PrPSc

    acts as a template to transfer its conformation onto PrPC, thereby

    generating new PrPSc. Thus, PrP knockout mice (Prnp /) donot

    develop prion disease when challenged with scrapie.10-12

    In humans, a single exon on the short arm of chromosome

    20 encodes the 253 amino acid PrP.13 Differential splicing does

    not occur. A 22-amino acid signal peptide is cleaved from theamino terminus during synthesis in the endoplasmic reticulum

    and a 23-residue signal sequence on the carboxy terminus is

    cleaved upon addition of a glycoinositol phospholipid (GPI)

    anchor,14 through which PrP attaches to the outer leaflet of the

    plasma membrane. Two asparagine-linked glycosylation sites

    lie within a loop region of the protein created by a disulfide

    bond (Figure 2). PrP is regulated during development and con-

    stitutively expressed in the adult. An increase in mRNA levels

    has not been detected during disease development in ani-

    mals.8,15 The highest levels of expression are within neurons,16

    but lower levels are detected in other peripheral tissues includ-

    ing lung, heart, kidney, pancreas, testis, white blood cells,17

    and platelets.18 Peripheral and central anterograde neuronal

    transport of PrPC has also been reported.15,19

    1 Center for Comprehensive Care and Research on Memory Disorders,

    Department of Neurology, University of Chicago, Chicago, IL, USA

    Corresponding Author:

    James A. Mastrianni, MD, Center for Comprehensive Care and Research on

    Memory Disorders, Department of Neurology, University of Chicago,

    Chicago IL, 60637, USA

    Email: [email protected]

    Journal of Geriatric Psychiatry

    and Neurology

    23(4) 277-298

    The Author(s) 2010

    Reprints and permission:

    sagepub.com/journalsPermissions.nav

    DOI: 10.1177/0891988710383576

    http://jgpn.sagepub.com

    277

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    3/23

    The normal function of PrPC is not known, although several

    lines of evidence have led to a variety of proposed functions,

    including the formation, function, and maintenance of

    synapses20-23; signaling,24-26 neuritogenesis and neuroprotec-

    tion27,28; copper binding and cellular delivery29-31; antioxidant

    activity32; cellular adhesion33; and a possible role in immune

    modulation (see ref34 for review). Interestingly, in PrP

    knockout mice, no obvious developmental or behavioral

    abnormalities were noted,10 suggesting redundancy of func-

    tion, although an alteration in synaptic function within the hip-

    pocampus of these animals has been reported by some20but not

    others.35 Recent work also suggests that adult mice lacking PrP

    display some hippocampal-dependent spatial memory defi-

    cits36 and have impaired peripheral myelin maintenance.37

    Some PrP knockout mouse lines also displayed disrupted circa-

    dian rhythm,38 and ataxia, the latter found to be related to upre-

    gulation ofPrnd, a downstream gene that encodes a truncated

    homolog of PrP known as Doppel, as a result of gene

    splicing that occurred during the deletion ofPrnp.39 An intri-

    guing line of work has recently suggested a link between PrP

    and Alzheimer disease, although the balance of its effect

    has not been determined. For instance, 1 study suggests PrPC

    normally inhibits b-secretase,40 a key enzyme involved in the

    cleavage of amyloid precursor protein (APP) that generates

    b-amyloid, thereby functioning in a protective role. In contrast,

    other reports suggest PrPC might function as a receptor to

    b-amyloid oligomers41 or to foster the generation of amyloid

    plaques of Alzheimer disease,42 thereby functioning to promote

    Alzheimer disease.

    Molecular Genetics of Prion Disease

    The worldwide incidence of prion disease is roughly 1 per 106

    population per year for sporadic disease and 1 per 107 per year

    Figure 1.Comparison of protease-sensitive, cellular isoform (PrPC) and protease-resistant scrapie-related isoform (PrPSc). Protease-resistantscrapie-related isoform carries greater b-sheet structure (flat ribbons), with loss of the a-helical structure (curly ribbons), and displays theproperties of infectivity, insolubility, and protease-resistance. The western blot demonstrates the protease-sensitive nature of PrPC and theprotease-resistant core of PrPSc that migrates faster in the gel because it is partially cleaved at the amino-terminal. The primary differencebetween PrPSc and PrPC is the proteinase-K resistance.

    278 Journal of Geriatric Psychiatry and Neurology 23(4)

    278

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    4/23

    for familial disease. There is no obvious gender bias.43 The

    peak age for the sporadic form of disease is *67 years,

    although patients aged 17 to 90 years have been reported.

    Sporadic forms of the disease tend to occur in the seventh

    decade and eighth and have a rapidly progressive course of

    under a year (avg. 4 to 6 months), whereas inherited forms usu-

    ally manifest at a younger age and have a more protracted

    course (Table 1). True autosomal dominant forms of prion dis-

    ease occur in about 10% to 15% of reported cases, although this

    estimate may be spuriously low because of the variable and

    often late age of disease onset in some forms of inherited prion

    disease.44 No obvious environmental risk to disease has been

    recognized, with the possible exception of vCJD that is linked

    to BSE exposure and iCJD resulting from prion-contaiminated

    biologicals or surgical instruments.

    A single base pair (bp) change within codon 102 of the

    PRNP gene, resulting in an amino acid change from proline

    to leucine (P102L) was first linked to the typical clinical pre-

    sentation of GSS in 1989.45 Since then, multiple-point muta-

    tions, insertions, and deletions of the gene have been

    identified (Figure 2, Tables 2 and 3). A handful of mutations

    introduce an early stop signal at different positions within the

    coding segment resulting in truncated forms of PrP, in 1 case

    (Y145X) generating a protein roughly half the normal length.

    Of the currently identified mutations, only 4 (P102L, D178N,

    E200K, and a 144-bp insert) have been observed with high

    enough frequency to generate significant logarithm of the odds

    (LOD) scores for linkage. Several polymorphic sites of the

    PRNP gene are known, the most common and best studied is

    codon 129 that acts as a predisposing factor to sporadic,

    Figure 2.General organization of human prion protein (PrP) and related mutations and polymorphisms. The 762 base pair (bp) open-readingframe ofPRNPencodes the 253 amino acid protease-sensitive, cellular isoform (PrPC). Nuclear magnetic resonance (NMR) studies predict3 a-helices (H1, H2, and H3), and 2 b strands (S1 and S2). Asn-linked glycosylation sites (CHO) occur at residues 181 and 197. The

    octapeptide repeat segment extends between residues 51 and 91. Pathogenic mutations and polymorphisms of thePRNPgene are representedbelow and above the schematic, respectively. A single octapeptide repeat deletion and a small number of single bp changes are considerednonpathogenic polymorphisms, some of which act as phenotypic modifiers, most notably, residue 129. Octapeptide repeat insertions (OPRI)of 1 to 9, excluding 3, are pathogenic, as are *30 bp changes. Letters preceding the numbers indicate the normal amino acid residue forthe position and letters following the numbers designate the new residue due to the mutation. Bold mutations are associated with Gerstmann-Straussler-Scheinker syndrome (GSS), the remainder cause Creutzfeldt-Jakob disease (CJD). * D178N is associated with either CJD or familialfatal insomnia (FFI), depending on the allelic codon 129 sequence (Met FFI; Val CJD). H187R displays variable pathology in the limitedcases reported. Amino acid letter designations are as follows: A indicates alanine, D aspartate, E glutamate, F phenylalanine, H histidine, I isoleucine, K lysine, L leucine, P proline, Q glutamine, R arginine, S serine, T threonine, V valine, Y tyrosine,(-) stop signal.

    Brown and Mastrianni 279

    279

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    5/23

    iatrogenic, and vCJD, in addition to a phenotypic modifier of

    sporadic and familial prion disease (discussed later).As mutations of PRNP have been discovered, genotype-

    phenotype correlations have been made, many of which

    generally hold true, especially with regard to the general

    pathology associated with each mutation. However, as more

    patients with similar mutations are described, it has become

    increasingly evident that the clinical features associated with

    specific mutations may vary, sometimes considerably and

    even within the same pedigree. Whether this feature repre-

    sents subtle alterations in the conformational subtype

    of PrPSc (see below), or an influence of other modifying

    genes, is not clear.

    A general classification scheme for the major phenotypes ofprion disease is based primarily on the pathologic features of

    disease, which appear to be more consistently linked to a spe-

    cific set of mutations. Using this parameter, excluding kuru,

    4 major prion disease subtypes in humans are known; CJD,

    GSS, FI, and vCJD. The pathognomonic feature of diffuse

    spongiosis with few or no PrP-amyloid plaques is characteristic

    of CJD, while extensive PrP-amyloid deposits with minimal

    spongiform change denotes GSS (Figure 3). Focal thalamic

    gliosis with minimal or absent spongiosis is seen in FI, and the

    presence of dense core PrP-amyloid plaques surrounded by a

    halo of vacuolar change, designated florid plaques, are seen

    in vCJD. In addition to PrP immunoreactive plaques in GSS,

    neurofibrillary tangles have been detected with at least 4 muta-

    tions (P105L, Y145Stop, F198S, and Q217R).102-105

    Although it is not understood how PrPSc induces such a

    broad range of pathological phenotypes, the distinct association

    of the major subtypes with specific mutations and characteristic

    pattern of protease-resistant PrPSc on Western blot (an indirect

    measure of PrPSc conformation) argues that the conformational

    subtype of PrPSc is ultimately responsible for the observed phe-

    notypes (Figure 4). This was confirmed by the recognition that

    sporadic FI is a phenocopy of familial FI that lacks the muta-

    tion associated with the latter but displays a similar PrPSc by

    Western blot and all other histopathologic and transmissible

    characteristics of familial FI.106

    These findings in human disease have their origin in the study

    of animal strains of prion disease. The first evidence for thiscame from studies of hamsters infected with 2 phenotypically

    different strains of transmissible mink encephalopathy (TME),

    known as HYPER and DROWSY, which were found to be

    linked to 2 distinct PrPSc conformations.107,108 Passage of these

    2 PrPSc strains to new hamster hosts resulted in the stable trans-

    mission of clinical and pathologic phenotypes. This property of

    prions was instrumental in determining that vCJD resulted from

    exposure to BSE, since passage of BSE to a variety of animal

    hosts including mice, macaques, and hamsters resulted in a char-

    acteristic migration pattern and glycosylation profile, by West-

    ern blot, that was similar to that found in PrPSc isolated from

    brains of participants with vCJD.

    109-113

    Evidence suggests thatboth host PrPC and prion strain (ie, conformation of PrPSc) are

    important contributors to the observed phenotype,114 although

    the mechanisms that underlie the targeting of PrPSc to specific

    cell populations, which leads to the phenotype, is still obscure.

    Major Subtypes of Prion Disease

    Creutzfeldt-Jakob disease

    The majority of patients with CJD present with confusion and

    memory loss that progress to severe cortical dementia, generally

    in combination with ataxia and myoclonus. The electroencepha-

    logram (EEG) typically shows bilateral periodic discharges with

    a frequency of 0.5 to 2 per second and, when seen in combina-

    tion with the above presentation, predicts the diagnosis with

    about 90% certainty.115-117 In addition to these defining features,

    a host of neurologic signs and symptoms have been reported,

    including weakness, rigidity, bradykinesia, tremor, chorea, alien

    hand syndrome, and sensory disturbances, among others. Vague

    complaints of fatigue, headache, sleep disturbance, vertigo, and

    behavioral changes may precede the development of frank pro-

    gressive dementia in many cases.116 As many as 25%begin with

    ataxia118 while a smaller percentage begins with cortical blind-

    ness, designated as the Heidenhain variant. Familial CJD has

    been associated with point mutations at codons 178, 180, 183,

    Table 1.Clinical Phenotypes of Prion Disease

    Disease Primary features Age at Onset (Range) Duration Pathology

    Kuru Ataxia, then dementia 40 years (29-60) 3 months1 year Kuru plaquessCJD Dementia, ataxia, myoclonus 61 years (17-83) rare

  • 7/24/2019 Articulo 6. Priones

    6/23

    196, 200, 203, 208, 210, 211, 232, and insertions of 1, 2, 4, 5, 6,

    7, and 9 octarepeat segments.

    The defining pathology for this prion disease is the presence of

    spongiform degeneration, or vacuolation, of cortical grey mat-

    ter (Figure 3). The vacuoles observed by light microscopy repre-

    sent focal swellings of axonal and dendritic neuronal processes,

    associated with the loss of synaptic organelles and accumulation

    of abnormal membranes by electron microscopy.119-121 They

    may be distributed throughout the gray and white matter but are

    generally most prominent in the gray matter neuropil. They

    typically range in size from 5 to 25 mm,122 but in advanced

    cases, they may be as large as 100 mm. When large vacuoles

    are seen in the presence of severe astrogliosis and neuronal

    loss, the constellation is termed status spongiosis. The dis-

    tribution of vacuolation is typically within the cerebral

    neocortex, subiculum of the hippocampus, caudate, putamen,

    thalamus, and the molecular layer of the cerebellar cortex.122

    A reactive gliosis is also consistently present, while an inflam-

    matory response is conspicuously absent. Protease-resistant

    PrP is easily detected in brain tissue from the majority of

    patients with CJD. Transmission of CJD to nonhuman pri-

    mates is relatively efficient (85%)123 and highly efficient

    (approaching 100%) to Tg mice that express human PrP.124,125

    In recent years, the bank vole has also been shown to be an

    efficient host for human prion transmission.126

    Iatrogenic CJD

    Prior to the introduction of recombinant human growth hor-

    mone (hGH) in 1985, more than 80 individuals developed CJD

    Table 2. Single Base Pair Changes ofPRNP

    Codon Sequence Change Amino Acid Changea Codon 129 Pathologic Phenotype Reference

    102 CCG ! CTG Pro (P) ! Leu (L) Met GSS 46102 CTG Leu (L) Val GSS 47105 CCA ! CTA Pro (P) ! Leu (L) Val GSS 48

    105 ACA Thr (T) Val GSS 49105 TCA Ser (S) Val Atypical GSS 50114 GGT ! GTT Gly (G) ! Val (V) Met CJD 51117 GCA ! GTG Ala (A) ! Val (V) Val GSS 52-54129 ATG or GTG Met (M) or Val (V) b 45, 55131 GGA ! GTA Gly (G) ! Val (V) Met Atypical GSS 56133 GCA ! GTG Ala (A) ! Val (V) Met Atypical GSS 57145 TAT ! TAG Tyr (Y) ! Stop (-) Met GSS w/NFTs (PrP-CAA) 58148 CGT ! CAT Arg (R) ! His (H) Met CJD 59160 Gln (Q) ! Stop (-) Met GSS 60163 Tyr (Y) ! Stop (-) GSS 61171 AAC ! AGC Asn (N) ! Ser (S) Val b 62178 GAC ! AAC Asp (D) ! Asn (N) Val CJD 63178 AAC Asn (N) Met FFI 64

    180 GTC ! ATC Val (V) ! Ile (I) Met CJD 65183 ACA ! ACG Thr (T) ! Ala (A) Met CJD 66187 CAC ! CGC His (H) ! Arg (R) Val Atypicalc 67, 68188 ACG ! AAG Thr (T) ! Lys (K) ? CJD 60196 GAG ! AAG Glu (E) ! Lys (K) Met CJD 69198 TTC ! TCC Phe (F) ! Ser (S) Val GSS w/NFTs 70200 GAG ! AAG Glu (E) ! Lys (K) Met/Val CJD 71, 72202 GAC ! AAC Asp (D) ! Asn (N) Val GSS 73203 GTT ! ATT Val (V) ! Ile (I) Met CJD 69208 CGC ! CAC Arg (R) ! His (H) Met CJD 74210 GTT ! ATT Val (V) ! Ile (I) Met CJD 75-77211 GAG ! CAG Glu (E) ! Gln (Q) Met CJD 69212 CAG ! CCG Gln (Q) ! Pro (P) Val GSS 73217 CAG ! CGG Gln (Q) ! Arg (R) Val GSS w/NFTs 78

    219 GAG ! AAG Glu (E) ! Lys (K) Met b

    79226 TAC ! TAA Tyr (Y) ! Stop (-) Val PrP-CAA 80227 CAG ! TAG Gln (Q) ! Stop (-) Val GSS 80232 ATG ! AGG Met (M) ! Arg (R) Met CJD 65, 81238 CCA ! TCA Pro (P) ! Ser (S) Met CJD 82

    Abbreviations: CJD, Creutzfeldt-Jakob disease; CAA, cerebral amyloid angiopathy; GSS, Gerstmann-Stra ussler-Scheinker syndrome; NFTs, neurofibrillary tangles;PrP, prion protein.a Letters in parentheses indicate letter codes for amino acids.b Polymorphismsmay modify disease phenotype.c Either early psychiatric symptoms with or without dementia preceding ataxia with curly PrP deposits.

    Brown and Mastrianni 281

    281

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    7/23

    through exposure to cadaver-derived human growth hormone

    (hGH) from at least 3 separate sources in the United Kingdom,

    France, and the United States.127-131 In contrast to sCJD,

    patients with iCJD more often display cerebellar ataxia rather

    than cognitive problems, and the EEG shows a diffusely slow

    wave pattern instead of periodic sharp waves complexes.132,133

    Table 3.Coding Alterations in Octarepeat Segmenta

    Coding Change Extra inserts Sequence Codon 129 Pathologic Phenotype Reference

    None R1,R2,R2,R3,R4 Met / Val24-bp deletion R3-R4 or R2 or R2-R3 Met 8324-bp insertion 1 R1,R2,R2,R2,R3,R4 Met N/A 84

    48-bp insertion 2 R1,R2,R2, R3,R2a,R2a,R4 Met Spongiosis (CJD-like) 8596-bp insertion 4 R2,R3,R2,R3 Met 8696-bp insertion 4 R1,R2(6),R3,R4 Met CJD (spongiosis) 8796-bp insertion 4 R1,R2,R2,R3,R2,R2,R2,R3,R4 Val N/A 84120-bp insertion 5 R1,R2(2),R3,R2,R3g,R2(2),R3,R4 Met CJD (spongiosis) 86120-bp insertion 5 R1,R2(2),R3,R2,R2,R2,R2,R3,R4 Met CJD (spongiosis) 88144-bp insertion 6 R1,R2(3),R3,R2,R3g,R2(2),R3,R4 Met Variable, usu. spongiosis,

    one pt. with PrP plaques89-91

    144-bp insertion 6 R1,R2(2),R3g,R2(2),R3g,R2(2),R3,R4 Met CJD (spongiosis) 92144-bp insertion 6 R1,R2,R2,R3,R2,R3g,R2,R3g,R2,R3,R4 Met CJD (spongiosis-variable) 93144-bp insertion 6 R1,R2,R2,R2(6),R3,R4 Met CJD (spongiosis- variable) 94168-bp insertion 7 R1,R2,R2c,R3,R2,R3,R2,R3,R2,R3g,R3,R4 Met CJD (spongiosis) 95168-bp insertion 7 Met gliosis, no spongiosis

    +PrP deposits96

    192-bp insertion 8 R1,R2,R2,R3,R2(7),R2a,R4 Val GSS (PrP plaques) 86, 97192-bp insertion 8 R1,R2,R2,R3g,R3,R2(6),R3,R4 Val GSS (PrP plaques) 98216-bp insertion 9 R1,R2,R2,R3,R2,R3g,R2a,R2,R2,R2,R3g,R2,R3,R4 Met GSS (PrP plaques) 99, 100216-bp insertion 9 R1,R2,R2,R3,R2,R3,R3g,R2,R2a,R2,R3,R2,R3,R4 Met N/A 101

    Abbreviations: R, an octarepeat unit (Pro-(His/Gln)-Gly-Gly-Gly-(-/Gly)-Trp-Gly-Gln); N/A, pathology not available.a Small case letters indicate repeat units which contain a single base pair alteration, although the amino acid coding is unchanged.

    Figure 3.Pathologic features of prion disease. A, Hemotoxylin and Eosin staining demonstrates typical spongiform degeneration (vacuolation)of the grey matter neuropil characteristic of Creutzfeldt-Jakob disease (CJD). This feature is less obvious in fatal insomnia (FI) and Gerstmann-Straussler-Scheinker syndrome (GSS). B, PrP-positive multicentric plaques are pathognomonic for GSS. These are mostly present within themolecular layer of the cerebellum but may be diffusely present throughout the cerebrum. C, Glial fibrillary astrocytic protein (GFAP)antibodies demonstrate hypertrophy and proliferation of astrocytes. This feature is present in all prion subtypes. In FI, this is often found focallywithin the anterior nucleus and dorsomedial nucleus of the thalamus and brainstem, in combination with neuronal dropout. In GSS it mayparallel PrP plaque pathology. D, The florid plaques of variant CJD (vCJD) consist of dense core PrP amyloid deposits surrounded by vacuoles.

    282 Journal of Geriatric Psychiatry and Neurology 23(4)

    282

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    8/23

    This disease has also been linked to dura mater obtained

    primarily from a single manufacturer in Germany whose

    preparative procedures were inadequate to decontaminate

    prions.132,134-139 Throughout the world, more than 100 cases

    of iCJD related to contaminated dura exposure between 1979

    and 1996 have been reported.138,140 Other iatrogenic forms of

    prion disease include a single corneal transplant,141 2 patients

    exposed to improperly decontaminated depth electrodes used

    for seizure focus localization,142 and at least 5 cases of CJD

    in women after receiving human pituitary gonadotropin.143-145

    Variant CJD

    To date, since 1995, vCJD has been reported in more than 200

    patients throughout the world, with the greatest number of

    cases in the United Kingdom (170) and France (25), but

    also reported in the Republic of Ireland, Italy, the United States

    (3 emigrants from the United Kingdom and Saudi Arabia),

    Canada, Japan, The Netherlands, Portugal, and Spain.146-151

    Compared with sCJD, vCJD more often presents with psy-

    chiatric features, particularly apathy and depression, in addi-

    tion to painful distal sensations, it occurs in younger

    individuals (ages 17 to 42 years), and has a slightly protracted

    course of greater than 1 year. The pathognomonic feature in

    the brain is the presence of dense core PrP plaques surrounded

    by a halo of spongiform change, also known as florid pla-

    ques (Figure 3). The protease-resistant fraction of PrPSc in

    the brain of these patients has a faster migration rate on West-

    ern blot than typical sCJD, and the diglycosylated PrP (the

    highest of the 3 bands of PrP) is more prominent, in contrast

    to sCJD, in which PrPSc is predominantly monoglycosylated.152

    As mentioned above, this predominance of the diglycosylated

    form was a key feature used in transmission studies to support

    the hypothesis that vCJD resulted from exposure to BSE-

    tainted beef. All but 1 case of vCJD resulting from primary

    infection with BSE have been homozygous for Met at codon

    129 (see below), further supporting this as a risk factor to prion

    disease. Secondary infection was first reported in an asympto-

    matic codon 129 heterozygote (129MV) who died of unrelated

    causes 5 years after receiving a transfusion of blood derived

    from a patient who developed signs of vCJD 18 months follow-

    ing blood donation.153 Since then, 4 additional cases of potential

    transfusion-related vCJD have been reported, one of which was a

    hemophiliac who received blood products rather than whole

    Figure 4.Western blot comparing the major isoforms observed in the 4 principal subtypes of prion disease. To the left of the blot displays theprion protein (PrP) segment that is represented in the adjacent blot. The highest molecular weight of PrP is the diglycosylated fraction of PrP,whereas the monoglycosylated and unglycosylated fractions run faster in the gel, because of their lower molecular weight. In CJD, FI, and vCJD,

    proteinase-K cleaves the first *67 amino acids of protease-resistant scrapie-related isoform (PrPSc

    ), leaving the PK-resistant core, PrP90-231.In most cases of Gerstmann-Straussler-Scheinker syndrome (GSS), a second C-terminal cleavage that removes the glycosylated segmentoccurs endogenously, leaving a nonglycosylated central segment.

    Brown and Mastrianni 283

    283

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    9/23

    blood.154-156 These findings have raised concerns about the

    prevalence of asymptomatic carriers of vCJD in the United

    Kingdom and the safety of the blood supply and has led to

    major changes in the handling of blood products in the United

    Kingdom and the United States.

    Gerstmann-Straussler-Scheinker Syndrome

    This subtype of prion disease is always familial and has been

    found associated with point mutations at codons 102, 105,

    117, 131, 145, 160, 198, 202, 212, 217, and in some cases of

    octapeptide repeat insertions (OPRI), especially those with a

    higher number of inserts. In its classic form, as originally

    described by Gerstmann,157,158 ataxia of gait and/or dysarthria

    are presenting features followed by variable degrees of pyrami-

    dal and extrapyramidal symptoms and often late development

    of dementia. There are, however, several variants of this dis-

    ease in which ataxia is not a prominent feature. Reports of

    impaired memory, spastic paraparesis, movement disorder, orbehavioral features, with a presentation suggestive of fronto-

    temporal dementia or Alzheimer disease have been reported.

    The EEG commonly lacks periodic discharges but may show

    slow waves. Duration of disease typically ranges from 2 to 7,

    but up to 10 or more years. Aspiration pneumonia is a signifi-

    cant risk because of impaired coordination of swallowing.

    The presence of plaque deposits immunoreactive to anti-PrP

    antibodies is the defining hallmark of GSS. Vacuolation may

    be minimal. PrP amyloid is PAS positive and shows birefrin-

    gence under polarized light after Congo Red staining in most

    cases. The most characteristic is the multicentric plaque,

    defined as a dense central core surrounded by smaller satel-lites122 (Figure 3), although other morphologies, from punctate

    to diffuse, have been recognized. Various-sized plaques, often

    termed primitive because they lack the characteristic green

    birefringence with Congo red staining, are seen in many of the

    GSS cases, suggesting there are variations in the maturity of

    amyloid formed among the different mutations. Plaque depos-

    its isolated from at least 4 of the GSS-associated mutations

    (P102L, A117V, F198S, Q217R) appear to be composed of

    peptide fragments of 7 and/or 11-14 kDa, which have been

    amino- and carboxy-terminally clipped and span residues of

    58-150 and 81-150 of the mutant alleles.159

    Fatal insomnia

    Originally reported and defined as a familial form of prion dis-

    ease, this is now known to occur on a familial (FFI) and spora-

    dic (sFI) basis.64,160-162 The characteristic clinical profile

    includes intractable insomnia, which may be observed for sev-

    eral months prior to the obvious onset of disease that may

    include dysautonomia, ataxia, variable pyramidal and extrapyr-

    amidal signs and symptoms, and relatively spared cognitive

    function until late in the course. Diffuse slowing rather than

    periodic discharges is observed on the EEG.161 Magnetic reso-

    nance imaging (MRI) is unhelpful, but functional imaging

    (PET or SPECT) shows a reduction in metabolic activity or

    blood flow to the thalamus early in the disease.163 Age at

    disease onset ranges from 25 to 61 years (avg. 48 years), and

    time to death is generally 1 to 2 years (range of 7 to 33

    months).161 Not all cases are clear-cut. A sleep study may be

    required in less clinically obvious cases to recognize a shorten-

    ing of total sleep time. A family with phenotypic heterogeneity

    from Australia was reported with the FFI haplotype(D178N,129M),164 and while some members were found to have

    clinically apparent insomnia early or late in the presentation, oth-

    ers did not exhibit it, suggesting even this disease may have a

    variable presentation.164

    The neuropathologic features of FI include neuronal loss

    and astrogliosis within the thalamus and inferior olives, and

    to a lesser degree, the cerebellum. Vacuolation is minimal to

    absent in typical cases. Protease-resistant PrP is detectable in

    the brains of affected patients but is usually present only in

    small amounts and is often restricted to specific regions such

    as the thalamus and temporal lobe.165

    Polymorphisms and Mutations ofPRNP

    Several polymorphisms have been identified in thePRNPgene.

    The most common and important ones are highlighted.

    Codon 129

    This codon may be either ATG, which codes for Met, or GTG,

    which codes for Val. The allelic frequency of Val in the general

    Caucasian population is 0.34 while that of Met is 0.66.55 The

    genotype distribution in this population is 37% Met/Met,

    51%

    Met/Val, and 12%

    Val/Val. Homozygosity at this positionpredominates in sCJD. In 1 series of 45 patients with sCJD

    from the United Kingdom, 89% were homozygous (Met/Met

    or Val/Val) at codon 129,166 compared with only 49% of 106

    normal participants.55 Similar findings were reported in France

    and Italy. The frequency of Met homozygosity was found to be

    between 41% and 45% in unaffected control participants and

    between 71% and 81% in 41 definite and/or probable sCJD

    patients.167,168 Homozygosity at codon 129 is also overrepre-

    sented in iCJD, but most dramatically in vCJD, where all but

    1 patient with confirmed vCJD, resulting from primary expo-

    sure to BSE, carried the 129MM genotype.169 The importance

    of homozygosity in prion susceptibility is further supported by

    studies in transgenic (Tg) mice that demonstrate more efficient

    transmission of prions when the recipient Tg mouse expresses

    human PrPC carrying the same amino acid at position 129 of

    the human PrPSc inoculum.170-172 These epidemiologic and

    Tg animal studies support the concept that sequence homology

    between PrPSc and PrPC facilitates their interaction and subse-

    quent generation of more PrPSc, which have also recently been

    demonstrated using FRET to assess the specific interaction of

    homologous and heterologous PrP molecules.171 It should be

    noted that the allelic frequencies in Japan are 0.96 for Met and

    0.04 for Val, masking a relative risk of homozygosity.173

    The codon 129 genotype also modifies the phenotype of

    sCJD. A cognitive onset with rapid progression is associated

    284 Journal of Geriatric Psychiatry and Neurology 23(4)

    284

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    10/23

    with the 129MM genotype, while slower progression and an

    ataxic onset is more common with the 129MV or 129VV gen-

    otype.174 The conformation of PrPSc, which is approximated by

    limited protease digestion and gel electrophoresis, differs

    between 129MM patients (Type 1 PrPSc migrates *21 kDa)

    and 129VV (Type 2 PrPSc migrates *19kDa).174,175 These

    findings imply an important function of the amino acid residueat position 129 in protein-protein interaction and PrPSc confor-

    mation determination.171

    In familial prion disease, the polymorphic codon 129 also

    plays a modifying role. This is most striking with the D178N

    mutation; when D178N is coupled to 129M, the clinicopatho-

    logic phenotype of FFI results in a protease-resistant PrPSc

    fragment of *19 kDa, whereas coupling of D178N with

    129V results in the fCJD phenotype and a PrPSc fragment of

    *21 kDa.176 Coupling of the F198S and 144-bp insert muta-

    tions with homozygosity at 129 also appears to lower the age

    at onset and decrease the duration of disease.

    Other Polymorphisms:

    a. 24-base pair (bp) deletion:This results in the loss of a

    stretch of 8 amino acids within the octapeptide repeat

    segment of PrP. Codons 51 through 91 encode a series of

    5 repeating elements of Pro-(His/Gln)-Gly-Gly-Gly-

    (-/Gly)-Trp-Gly-Gln, the first of which includes 27

    nucleotides encoding 9 amino acid residues, while the

    4 subsequent ones are 24 bp in length encoding 8 residues

    each. A deletion of 1 of the repeat elements was initially

    detected in 3 healthy members of a Moroccan family177

    and then incidentally in a cosmid library construct derivedfrom the HeLa human cell line.178 It is generally consid-

    ered to be a nonpathogenic polymorphism occurring in

    about 3% of the normal population.83,177,179

    b. N171S: This rare polymorphism was incidentally noted in

    a 69-year-old healthy control participant.62 A family with

    psychiatric disease was reported with this polymorphism,

    and although it was suggested that it may be linked to schi-

    zophrenia, 1 healthy member carried the polymorphism180

    and another study found no evidence for the N171S poly-

    morphism in a schizophrenia population.181

    c. E219K polymorphism: Codon 219 normally encodes glu-

    tamate (E) in the Caucasian population, although lysine

    (K) coding is found in about 6% of the Japanese popula-

    tion.79 This polymorphism was also reported on the same

    allele as the P102L mutation in a Japanese family in which

    dementia rather than ataxia was prominent and cerebellar

    plaque pathology was less prominent compared with

    cases of the P102L mutation alone.182 However, variabil-

    ity in presentation of GSS was also observed in an Italian

    family in the absence of the E219K polymorphism.

    Recent transmission studies in transgenic knock-in mice

    suggest a heterozygous state at codon 219 confers

    reduced susceptibility to prion transmission.183

    d. G127V polymorphism: This rare polymorphism was

    recently reported in the population within New Guinea, in

    the area where kuru was endemic. Sampling the population

    suggests that this polymorphism is protective against

    prion infection.184 Studies are in progress to determine this

    experimentally.

    SelectedPRNP

    Mutations That Cause CJDD178N, 129V

    A transition of G to A at the first nucleotide of codon 178 of the

    PRNP gene was initially reported in a Finnish family63 and

    then shortly thereafter in 2 American families, one of Dutch

    and the other of Hungarian origin,185 and in a French family

    from Brittany 186. The clinical phenotype displays a younger

    mean age at onset (46 vs 62 years), a more prolonged disease

    duration (avg 2 years, range 9 to 51 months), and slowing of the

    EEG rather than periodic triphasic waves, compared with

    typical sCJD.185,187,188 Memory disturbance is the presenting

    feature in the vast majority of carriers, followed typically by

    cerebellar ataxia and myoclonus, in addition to varying degreesof visual disturbance, reduced speech output, extrapyramidal,

    and pyramidal tract features. Less than 10% develop seizures.

    Brain pathology shows diffuse spongiform degeneration, glio-

    sis, and neuronal loss within the frontotemporal cerebral cor-

    tex, caudate, and basal ganglia, with relative sparing of deep

    thalamic nuclei and the cerebellum.161,185,189

    V180I

    This point mutation (GTC to ATC) was initially reported in

    2 unrelated individuals with no obvious family history and a

    presentation similar to fCJD associated with the D178N muta-tion.65 Since then, only a handful of cases have been detailed,

    although a recent genetic survey in Japan identified the muta-

    tion in 84 cases of prion disease over the past 10 years.190 Most

    reported cases appear to lack a clear family history. The muta-

    tion has been consistently found allelic with 129M. A presenta-

    tion of subacute dementia, often with aphasia, and subsequent

    myoclonus, but no periodic discharges on EEG, is most com-

    mon. Neuropathological findings include typical CJD changes

    of diffuse vacuolation, neuronal loss, and gliosis of the cortex.

    Western analysis shows protease-resistant PrP that lacks the

    higher molecular weight diglycosylated fraction of PrPSc,

    although expression of PrP

    V180I

    in cell culture appears nor-mally glycosylated, suggesting selective conversion of mono-

    and unglycosylated PrPV180I.50,191

    T183A

    This was first described in a Brazilian family with 9 affected

    members who developed a progressive dementia with clinical

    features suggesting a frontotemporal neurodegenerative pro-

    cess.66 Behavioral features, including aggressiveness, hyperoral-

    ity, and verbal stereotypes, were prominent early in the disease

    course with all patients. Disease begins in the fourth or fifth

    decade (average 45 years, range 37 to 49 years) and has a some-

    what protracted course of 2 to 9 years (average 4.2 years).

    Brown and Mastrianni 285

    285

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    11/23

    Spongiform change was prominent in frontotemporal regions.

    Another report in a 40-year-old with more typical CJD and rel-

    atively prolonged course of 4 years, with similar pathology, was

    also reported.192 This mutation was allelic with 129M.

    E200KThis is the most common mutation of PRNP worldwide. A

    change in coding from GAG to AAG at codon 200 of the gene

    was first detected in an unusual cluster of CJD cases in rural

    Slovakia72 where the annual mortality rate was about 100 per

    million population and almost simultaneously in a Libyan Jew-

    ish family.193 It is linked to CJD with a LOD score of 4.85.194

    Carriers of this mutation have been identified from more than

    10 different countries71,195-197 and were initially thought to

    have a common ancestral origin of a Sephardic Jew whose des-

    cendents emigrated from Spain and Portugal during the Inqui-

    sition. This was further supported by the consistent association

    of the E200K mutation with the 129M. A report of this muta-tion in a Japanese family with no evidence of racial intermix-

    ing198 and the detection of the mutation in association with

    Val coding at codon 12972 indicates at least 2 additional foun-

    ders and supports the more likely possibility that the mutation

    arose spontaneously in several populations from the deamination

    of a methylated CpG in a germline PRNPgene. Clusters of this

    mutation are seen in populations from Israel, Chile, and Eastern

    Europe. Surveillance studies from France and England have

    detected the E200K mutation in patients without a clear family

    history, supporting the variable onset of CJD(E200K).167,199

    The phenotype associated with this mutation is quite vari-

    able but is generally comparable to sCJD.

    200

    Forgetfulness andconfusion are typically early manifestations, followed by ataxia

    and myoclonus and the appearance of periodic discharges on

    EEG.201 The average age at disease onset is *61 and time to

    death is typically under 1 year (*4.5 months), similar charac-

    teristics to that of sCJD. Pathology is also similar to sCJD, with

    widespread spongiform degeneration and no PrP plaque pathol-

    ogy. However, the fCJD(E200k,129V) case included PrP pla-

    que deposits within the cerebellum.72 Chapman et al,202

    compared clinical data from 14 patients with fCJD(E200K) and

    noted a wide variability in age at onset (34 to 65 years) and in

    disease duration (2 to 66 months), along with a host of varied

    clinical features that included cerebral, basal ganglia, brain-

    stem, cerebellar, and spinal cord dysfunction. A large kindred

    of German ancestry was reported in which 5 of 9 members with

    CJD, for whom neuro-ophthalmologic data were available, had

    supranuclear palsy as an early feature, while myoclonus and

    periodic EEG were uncommon.196 Demyelinating peripheral

    neuropathy, which could not be attributed to a coexisting dis-

    ease process, was reported in 2 patients who developed CJD

    related to the E200K mutation,203 while three others have been

    described with motor axonal neuropathy.202 Severe insomnia

    was prominent, but not the presenting feature of disease in a

    single E200K carrier.204

    The variable age at onset with this disease suggests reduced

    penetrance; however, using life-table analyses, the risk to

    disease development for fCJD(E200K) is age-dependent and

    penetrance is nearly complete by the ninth decade of life.44,205

    The situation in which a child develops the disease yet one of

    the parents either is a healthy carrier or died from other causes

    prior to developing disease is therefore a potential feature to

    consider when ruling out familial disease by history alone.

    Transmission of this familial form of prion disease to experi-mental animals was initially demonstrated in nonhuman pri-

    mates,206 and later in Tg transgenic mice.125

    R208H

    A transition at the second nucleotide of codon 208 from G to A

    resulting in a missense coding for histidine rather than arginine

    was reported in a 60-year-old who presented with confusion

    and memory problems and later developed paranoia, ataxia,

    myoclonus, and a positive EEG, over a 7-month course.74

    Neuropathology was typical of CJD (diffuse spongiosis, neu-

    ronal loss, and gliosis) and protease-resistant PrP was presentthroughout the brain. A family history was not evident, possi-

    bly related to the premature deaths of the patients father,

    from an unrelated condition. Additional reports of R208H

    linked to 129M, include a clinicopathologic phenotype char-

    acteristic of sCJD,207 another with tau pathology in the

    entorhinal cortex, ballooned neurons, and an extra 17 kDa

    PK-resistant PrPSc fragment,208 and a unique case, allelic with

    129V, was reported with a rapidly progressive syndrome of

    behavioral changes, cerebellar ataxia, and kuru type cerebel-

    lar plaques.209

    V210I

    This was initially reported in Italy75 and France76,77 without

    evidence of a family history but has now been reported in sev-

    eral countries, including the United States.77,210,211 As with the

    E200K mutation, the presentation is somewhat variable and

    includes a cerebellar syndrome, bilateral myoclonic jerks,

    dysarthria, stroke-like features such as hemisensory loss and

    hemiparesis, sudden onset of visual disturbances, or the onset

    of behavioral changes, all beginning between 50 and 70 years

    of age. The EEG displays periodic discharges. Disease dura-

    tion is typically less than 6 months. Protease-resistant PrP is

    similar to that of typical sCJD. Spongiform degeneration isdiffusely present in the brain. Because healthy carriers of the

    V210I mutation aged 67 to 82 years76 have been detected and

    some patients lack a clear family history, this mutation also

    appears to display variable penetrance compared with other

    PRNP mutations.

    M232R

    Clear evidence for an autosomal dominant effect of this

    mutation is lacking, but it has been reported in several cases,

    primarily in the Japanese population. It too presents as sCJD,

    with a typically rapid disease course and an absent family

    286 Journal of Geriatric Psychiatry and Neurology 23(4)

    286

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    12/23

    history.65,82,212,213 In addition, this sequence alteration was

    reported in a V180I carrier on the normal allele.

    144-bp insertion

    This 6 octapeptide repeat insertion (OPRI) was described in a

    large kindred in the United Kingdom. It is linked to an interest-ing disease phenotype in which behavioral symptoms and per-

    sonality disorders of long-standing duration are observed prior

    to the onset of a slowly progressive dementia.89,90,91,214-216 A

    variation in the degree of cerebellar ataxia, dysarthria, and pyr-

    amidal and extrapyramidal signs are observed among affected

    individuals, as is myoclonus. The pathology is also quite vari-

    able, ranging from severe spongiosis to no obvious signs of

    pathology, generally without plaques, although in 1 patient,

    cerebellar plaques were observed.90,216 Since the initial report

    of a family with this insertion, additional families from the

    United Kingdom,93 Japan,92 the United States,88 and Basque94

    have been described. All have similar variations in disease phe-notype and all have the mutation on the Met 129 allele. Cur-

    iously, there are slight variations in the sequence of the insert

    (Table 3), although their role in phenotype modification is not

    apparent.

    Additional OPRI Mutations

    Although there is significant variability in the presentation

    and pathologic features of prion disease related to the repeat

    expansions, they are discussed here as a group for ease of pre-

    sentation. Between codons 51 and 91 of PRNP, 9 different

    insertions of the gene have been described, none of which dis-rupt the overall sequence of the remainder of the protein

    beyond residue 91. Most are coupled to Met coding at codon

    129. The largest family reported is the British family with

    6-OPRI described above (144-bp insert). Octapeptide repeat

    insertions of 1 to 9 additional repeats have been described

    (Table 3). Genotype-phenotype correlations have been difficult

    to make, based on the small number of affected patients within

    most families and because variability of presentation is com-

    mon with these mutations. There is no obvious anticipation and

    the length of the insert appears stable during meiosis.217 In gen-

    eral, the length of the insert correlates inversely with the age at

    onset of disease. In patients with 7 to 9 extra repeats, disease

    presents in the 30s, while in those with 1 to 4 extra repeats, dis-

    ease may be delayed until the sixth to seventh decade.94,218 The

    duration of illness, however, appears to be proportional to the

    length of insert, ranging from 5 to 120 months, with an increase

    in insert number from 1 to 7.218 A Huntington disease-like phe-

    notype (HDL-1) was reported in a family with an 8-OPRI.219

    Interestingly, another individual was reported with the HDL-

    1 phenotype but later found to be a member of the 6-OPRI

    kindred described above, further supporting the variable phe-

    notype observed with OPRI mutations, in addition to the

    importance of genetic testing in neurodegenerative disease.220

    A minority of patients has the typical clinicopathologic fea-

    tures of sCJD; rather, the majority has a more chronic course

    often including atypical features such as aphasia, apraxia, and

    a personality disorder associated with memory loss. The per-

    sonality disorder may appear as a primary or early feature in

    as many as half of the carriers of these mutations. Dementia

    eventually occurs in all and may be precipitous after a long

    prodrome of mild cognitive problems or behavioral features.

    Cerebellar ataxia and extrapyramidal features are also some-what common in their course. Myoclonus may occur in less

    than one half of carriers, while periodic discharges on EEG are

    even less frequent (

  • 7/24/2019 Articulo 6. Priones

    13/23

    the P102L mutation in a Japanese family that showed variabil-

    ity in presentation and weak Congo Red staining of plaques

    compared with typical GSS plaques associated with the

    P102L mutation alone,182 suggesting that polymorphisms other

    than codon 129 (ie, codon 219) might influence the phenotype

    of dominant mutations.

    The clinical and pathologic features of GSS, including obvi-ous ataxia and PrP plaque deposits, were duplicated in a mouse

    constructed with a transgene harboring the equivalent P102L

    human mutation.230 These mice, which overexpress the mouse

    equivalent (PrP-P101L) of human PrP-P102L, develop sponta-

    neous ataxia at about 150 days. Transmission of GSS(P102L)

    has been reported in nonhuman primates and mice, although

    the rates of transmission (*40%) is much less than sporadic

    and familial CJD (*85%).231

    P105L

    A cytosine (C) to thymine (T) transition at the second nucleo-

    tide of codon 105 (CCA to CTA) results in the substitution of

    leucine for proline in the protein, and in all cases, is coupled

    to 129V. This mutation, recognized primarily in Japan, is

    classically associated with the development of spastic para-

    paresis, manifest as weakness with hyperreflexia and extensor

    plantar responses, prior to, or along with, the development of

    dementia, progressing eventually to tetraparesis with a pseu-

    dobulbar affect over a 7- to 12-year course.48,232,233 Although

    cerebellar symptoms were uncommon and neither periodic dis-

    charges on the EEG nor myoclonus were present in most

    patients reported with this mutation, subsequent reports sug-

    gested some members of this pedigree did develop more typ-

    ical signs of GSS. Age at onset ranged from 38 to 48 years.

    Pathologic findings were confined to the telencephalon where

    diffuse-type PrP immunoreactive plaques were present

    throughout the gray matter of the cortex, particularly within

    frontal motor cortex and temporal lobes and deep gray nuclei

    of the basal ganglia and thalamus. Severe neuronal loss and

    gliosis were also present within those same areas, although

    spongiform change was absent.

    In addition to the leucine mutation at codon 105, a threo-

    nine substitution (ie, P105T) was identified in a Canadian

    family49 and a serine substitution (P105S) in an American

    family.50 P105T appears to have a generally typical GSSclinical phenotype, with the exception of an unusual onset

    of psychiatric symptoms found in a 13-year-old family

    member, whereas the P105S mutation was detected in a

    31-year-old woman who developed a clinical presentation

    characteristic of frontotemporal lobar dementia without

    ataxia or spastic paraplegia, and a combination of intense

    focal vacuolation of the basal ganglia and dramatic plaque

    pathology in the cerebellum and hippocampus. These find-

    ings provide support to the concept that the substitution

    itself, rather than the loss of the normal amino acid, is the

    determinant of phenotype, presumably by inducing different

    PrPSc conformations.

    A117V

    This mutation was initially identified in a French family with

    8 affected members spanning 4 generations52 and later in 2

    American families of German descent.53,54 In all cases, the

    dominant mutation is carried on the 129V allele. In the

    French family, the presentation was consistent with a primary

    dementing syndrome with variable degrees of pyramidal,

    extrapyramidal, and cerebellar features. The affected mem-

    bers of one American family (designated GCSA) presented

    with presenile dementia followed by pyramidal and extrapyr-

    amidal features without obvious cerebellar involvement.53,234

    The disease was originally considered to be familial Alzhei-

    mer disease, based on this clinical presentation and the neu-

    ropathologic finding of multiple amyloid plaques scattered

    throughout the cerebral but not the cerebellar cortex.234 The

    plaques, however, did not immunoreact with anti-Ab anti-

    body but did with anti-PrP antibody,235 confirming it to be

    a prion disease. In contrast to this telencephalic presenta-

    tion, the second US family with the A117V mutation dis-

    played the classic GSS phenotype of ataxia with pyramidal

    and extrapyramidal features and late-developing dementia.54

    The proband of that family had widespread deposition of

    PrP-plaques throughout both the cerebral and cerebellar cor-

    tex. Follow-up reports of some members from subsequent

    generations of the French family also suggested a cerebellar

    onset with progressive gait difficulties, dysarthria, and dys-

    phagia, leading eventually to mental deterioration and

    dementia,236 and plaque deposition in some was wide-

    spread.237 These findings suggest a clear correlation of pla-

    que pathology with clinical phenotype and variability of

    clinical phenotype due to the same point mutation. Commonfeatures include the absence of periodic discharges on EEG,

    an early onset (third to fifth decade), moderately prolonged

    duration (average 3 years), and the presence of GSS plaque

    pathology. In some cases, protease-resistant PrP is difficult

    to detect, whereas in most, a low level of a 14 kDa fragment

    is observed. Several attempts to transmit this disease to

    rodents have been unsuccessful.236 A Tg mouse that

    expresses the mouse-equivalent PrP-A116V mutation allelic

    with 129V and develops severe progressive ataxia beginning

    at *120 days until their death *30 days later, has been gen-

    erated.238 The brains demonstrate PrP amyloid deposits in the

    cerebellum and hippocampus and a *13 to 14 kDa protease-resistant PrP fragment. In humans with GSS(A117V), PrP

    displayed a significantly higher level of a transmembrane

    form of PrP, suggesting that this mutation affects the translo-

    cation of PrP.239 Whether this can fully explain the patho-

    genic properties of the molecule and the reduced

    transmissibility seen with this particular prion disease variant

    remains to be determined.

    F198S

    This mutation, which is coupled to Val at codon 129, was

    identified in a very large kindred in Indiana.240 Of the

    288 Journal of Geriatric Psychiatry and Neurology 23(4)

    288

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    14/23

    1200-member family spanning 6 generations, 67 individuals

    were affected at the time of the report in 1989. Linkage of the

    mutation was demonstrated with a LOD score of 6.37.240 The

    presentation is generally one of progressive gait ataxia, dysar-

    thria, and impaired short-term memory, often associated with

    extrapyramidal symptoms of bradykinesia and rigidity, and less

    often with pyramidal tract signs. The dementia is slowlyprogressive, occurring at a mean age of 52 (range 30s to 60s)

    and lasting on average 6 years (range 2 to 12).241 Disease may

    present 10 years earlier in patients homozygous for Val at codon

    129.240 Ocular signs, including supranuclear gaze palsy, jerky

    pursuits, and gaze-evoked nystagmus in all directions, were

    noted in the early phase of the disease. Myoclonus is present

    in some but not all patients. The EEG may show slowing, but

    it does not exhibit periodic discharges, consistent with other GSS

    diseases. Histopathology includes diffusely distributed PrP pla-

    ques throughout the cerebrum and cerebellum with minimal

    spongiform change.242 In addition, tau-positive neurofibrillary

    tangles are found primarily in frontal, parahippocampal cortex,cingulate gyrus, and insular cortex.243,244

    Q217R

    An A to G transition at the second nucleotide of codon 217

    (CAG to CGG) results in the missense coding of arginine

    instead of glutamine. A small number of individuals in a single

    American family of Swedish origin have been described, each

    of whom developed a progressive dementia at least 4 years

    before they developed progressive gait ataxia, dysphagia, and

    parkinsonism several months prior to their deaths at the ages

    of 67 and 71 years.78 Disease duration was 5 and 6 years. EEG

    results were not reported. The primary neuropathologic feature

    was diffusely deposited PrP plaques throughout the neocortex.

    The plaques were shown to be derived from the mutant PrP and

    composed of amino and carboxy-terminal clipped fragments

    extending from residues 81 or 82 to 145 or 146.245 In addition,

    neurofibrillary tangles were diffusely present throughout the

    neocortex and several subcortical grey structures that denote

    the similarity of pathologic phenotype with that of GSS(F198S)

    and GSS(Y145Stop). Experimental transmission of this prion

    disease has not been demonstrated.

    Truncation Mutations of GSSA stop sequence (TAG) at codon 145 was initially described on

    the 129M allele in a Japanese patient with a clinical diagnosis

    of Alzheimer disease, with plaque deposits of truncated

    PrP.58,104 The patient developed a 20-year course of distur-

    bance in memory, eventually progressing to severe dementia

    and death at 59 years of age. Periodic discharges were not evi-

    dent on EEG. Neuronal loss and gliosis was severe, but spongi-

    form change was absent. PrP deposits were found in and around

    small- and medium-sized blood vessels and neurofibrillary tan-

    gles (NFTs) were evident throughout the cerebral cortex,104

    labeling this as a vascular variant of GSS and a cerebral

    amyloid angiopathy (PrP-CAA) 159.

    Since that case, 4 additional truncation mutations have been

    described; Q160X,60 Y163X,61 Y226X, and Q227X.80 The

    Q160X case does show GSS type pathology, but it has not yet

    been fully described (personal communication). The Y163X

    case also displayed PrP-CAA, whereas the Y226X and

    Q227X mutations, although differing by only 1 residue, were

    found in 2 Dutch patients with 2 distinct disease subtypes. TheY226X mutation was found in a 55-year-old woman who

    developed cognitive problems, aphasia, and hallucinations. In

    contrast to typical GSS, the CSF 14-3-3 was positive and the

    EEG displayed PSWCs. The disease ran a course of 27 months

    and the histopathology revealed PrP-CAA in the absence of

    neurofibrillary tangles. The Western blot was not described.

    The Q227X mutation was found in a 44-year-old woman with

    a 72-month slowly progressive course of hypokinetic rigid gait

    with cognitive and behavioral changes, and parkinsonian fea-

    tures, suggesting frontotemporal dementia (FTD). The histo-

    pathology displayed multicentric amyloid plaques throughout

    the cerebrum and severe neurofibrillary lesions without PrP-CAA, and Western blot revealed a 7 kDa unglycosylated PrPSc

    fragment truncated at both the N- and C-terminal ends.

    The consistent demonstration that PrP amyloid deposits are

    linked to truncation mutations validates the finding in Tg

    mice that express PrP lacking the GPI anchor, and develop

    extensive PrP amyloid plaque deposits.246 Thus, the absence

    of the GPI anchor results in PrP secretion to the extracellular

    space, leading to amyloid formation. Why specific point

    mutations result in this presumed secretion of PrP is currently

    undetermined.

    Diagnostic ConsiderationsA rapidly progressive dementia associated with ataxia, myoclo-

    nus, and periodic discharges on the EEG, in an afebrile 65-

    year-old individual provides a straightforward diagnosis of

    CJD. However, this constellation of features may be observed

    in less than 60% of cases. It should be clear from the above

    descriptions of the presentations of prion disease that the range

    of clinical phenotypes now is quite broad. As such, the diagno-

    sis of prion disease should be considered as part of the differ-

    ential in all cases presenting with dementia, an atypical

    movement disorder, or late onset psychiatric disease, especially

    if the rate of disease progression is rapid or if it is accompanied

    or preceded by other neurological signs or symptoms and/or a

    family history of a similar disease. Other diseases with overlap-

    ping symptoms include Alzheimer disease, cortical basal

    degeneration (CBD), dementia with Lewy bodies (DLB), Hun-

    tington disease (HD), Spinocerebellar ataxias (SCAs), and the

    frontotemporal lobar dementias (FTLD), including the beha-

    vioral variant of FTD, semantic dementia, and progressive non-

    fluent aphasia, among other conditions. Central nervous system

    (CNS) vasculitis may be entertained in some rapidly progres-

    sive forms of prion disease, although a normal MRI usually

    argues against it. Angiogram may be considered in some cases.

    The inherited metabolic disorder of ceroid lipofuscinosis (Kufs

    disease in the adult) can also present with dementia and

    Brown and Mastrianni 289

    289

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    15/23

    myoclonus. Metabolic causes of a prion-like presentation

    include bismuth or lithium toxicity. Hashimotos encephalopa-

    thy is an important treatable condition to rule out. It can pro-

    duce intermittent episodes of confusion, cerebellar ataxia,

    and may even demonstrate periodic discharges. Antithyroper-

    oxidase antibody (anti-TPO) and antithyroglobulin will be con-

    siderably elevated. Corticosteroid therapy may completelyreverse the symptoms.

    Key diagnostic studies include MRI, EEG, cerebrospinal

    fluid (CSF) analysis, and sometimes PET scan. EEG findings

    of periodic sharp wave complexes (PSWCs) consisting of

    triphasic or sharp wave bursts every 0.5 to 2.0 seconds support

    the diagnosis of prion disease. Although PSWCs are observed

    in a small percentage of individuals with genetic prion

    disease, their presence seems to be highly dependent on the

    associated causal mutation and resultant clinical phenotype;

    those mutations that produce a CJD-like clinical phenotype

    and spongiform degeneration pathology seem more likely to

    have a positive EEG.Hyperintensity on diffusion weighted magnetic resonance

    imaging (DWI), especially within the basal ganglia or involv-

    ing the cortical ribbon, appears to be highly predictive of CJD,

    although the studies with familial CJD are limited, as are those

    with GSS. A PET scan is typically uninformative for CJD or

    GSS, but it may show focal hypometabolism of the thalamus

    in sporadic and familial forms of FI. In addition, a clear pattern

    of Alzheimer disease (hypometabolism of temporoparietal

    lobes) or frontal lobar dementia (focal frontal and temporal

    lobe hypometabolism) may be useful to rule out prion disease,

    but this may not be 100% certain.

    Cerebrospinal fluid often shows a mild,*10%

    , elevation oftotal protein without a cytological response. Significant eleva-

    tions in 14-3-3, total tau, or neuron-specific enolase have been

    shown to be highly predictive of prion disease, more specifi-

    cally sCJD.247-250 An elevation in the level of these proteins

    represents rapid neuronal death with leakage of the cell con-

    tents into the spinal fluid. Thus, these appear to be more predic-

    tive of faster-progressing, nonfamilial forms of prion disease.

    False negatives are known in some reports over 40% of the

    time.250 False positives have been reported with herpes ence-

    phalitis, hypoxic brain damage, acute stroke, and other condi-

    tions that induce neuronal damage. Genetic analysis of the

    PRNPgene may be helpful whether a family history of disease

    is present, as the late and often variable age at onset of the prion

    diseases may obscure a positive family history.

    A definitive diagnosis of prion disease can only be made by

    pathologic confirmation following biopsy (no longer a com-

    mon practice because of the preparation and cleanup required

    for the operating room) or autopsy. Transmission of disease

    to a proper host animal is considered the ultimate diagnostic

    test for the presence of prions; however, these studies are

    expensive and time consuming, in some cases requiring more

    than 2 years to complete. Newer technologies, using in vitro

    methods such as the protein misfolding cyclic amplification

    (PMCA) process may prove more useful. In this process, a

    small amount of prion-affected sample is mixed with a large

    excess of normal brain homogenate and subjected to a series

    of incubations and sonications to break up PrP fibrils and cre-

    ate new PrPSc seeds resulting in the amplification of PrPSc

    several fold.251 This technique has been shown to detect

    prions in blood and, as such, might eventually be used in diag-

    nostic testing of peripherally infected individuals, such as in

    vCJD.252

    Therapy

    There are currently no therapeutic agents designed to slow the

    progression or reverse the effects of prion disease. Thus, therapy

    is aimed at controlling symptoms. If present, seizures may be

    treated with general antiepileptic agents such as phenytoin or car-

    bamazepine. Myoclonus often responds to low doses of clonaze-

    pam. Issues related to dysphagia are often difficult to resolve and

    the decision to place a feeding tube should be weighed against the

    confidence of the diagnosis of these terminal diseases. Severe

    psychiatric symptoms that may include hallucinations and/ordelusionsare best managed by small doses of atypicalantipsycho-

    tics, such as quetiapine. Evaluation by a social worker is manda-

    tory to assist the family in management planning.

    The most extensive clinical trials for prion therapy focused

    on quinacrine, an antimalarial that showed promise in curing

    infectious PrPSc from cultured neuroblastoma cells chronically

    infected with scrapie.253 The results of studies from the United

    States and the United Kingdom did not support a clinical ben-

    efit of quinacrine for CJD. Other agents, particularly polysul-

    fated compounds such as pentosan polysulfate, suramin, and

    heparan sulfate,254,255 prolong the incubation period of experi-

    mental scrapie in animals, although they must be administeredprior to, or simultaneous with, the inoculation of the animal

    with prions, making these impractical for the symptomatic

    patient who presented for medical attention.256 Pentosan poly-

    sulfate was administered intracerebrally to a single patient with

    vCJD, with unclear clinical benefit although his course

    appeared to be prolonged.257 The mechanism by which these

    drugs exert their effects is unclear, but some consider that the

    highly charged molecules sequester prions away from other

    interacting proteins. In addition to these agents, anti-PrP anti-

    body therapy, designed to block the interaction of PrPSc with

    PrPC,258-260 is under investigation, as is the attractive approach

    of siRNA therapy, which will act to knock down PrPC expres-

    sion, leading to less substrate for conversion to PrPSc.261

    Although such treatments are in dire need, better methods of

    detection and early diagnosis will need to be developed, to

    afford the best chance for successfully inhibiting disease pro-

    gression. Genetic detection of familial forms paired with devel-

    oping strategies of brain imaging and PMCA of body fluids

    may assist in that goal.

    Declaration of Conflicting Interests

    The author(s) declared a potential conflict of interest (e.g. a financial

    relationship with the commercial organizations or products discussed

    in this article) as follows: Dr. Mastrianni is a consultant to the FDA

    TSE Advisory Committee.

    290 Journal of Geriatric Psychiatry and Neurology 23(4)

    290

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    16/23

    Funding

    The author(s) disclosed receipt of the following financial support for

    the research and/or authorship of this article: Dr. Mastrianni receives

    funding from the NIH for prion disease research.

    References1. Cuille J, Chelle PL. Experimental transmission of trembling to the

    goat.CR Seances Acad Sci. 1939;208:1058-1060.

    2. Gajdusek DC, Gibbs CJ Jr, Alpers M. Experimental transmission

    of a kuru-like syndrome to chimpanzees. Nature. 1966;

    209(5025):794-796.

    3. Gibbs CJ Jr, Gajdusek DC, Asher DM, et al. Creutzfeldt-Jakob

    disease (spongiform encephalopathy): transmission to the chim-

    panzee.Science. 1968;161(839):388-389.

    4. Gibbs CJ Jr, Amyx HL, Bacote A, Masters CL, Gajdusek DC.

    Oral transmission of kuru, Creutzfeldt-Jakob disease and scrapie

    to nonhuman primates. J Infect Dis. 1980;142(2):205-208.

    5. Chandler RL. Encephalopathy in mice produced by inoculationwith scrapie brain material. Lancet. 1961;1(7191):1378-1379.

    6. Prusiner SB, McKinley MP, Groth DF, et al. Scrapie agent con-

    tains a hydrophobic protein. Proc Natl Acad Sci U S A.

    1981;78(11):6675-6679.

    7. Bolton DC, McKinley MP, Prusiner SB. Identification of a

    protein that purifies with the scrapie prion. Science. 1982;

    218(4579):1309-1311.

    8. Oesch B, Westaway D, Walchli M, et al. A cellular gene encodes

    scrapie PrP 27-30 protein. Cell. 1985;40(4):735-746.

    9. Safar J, Roller PP, Gajdusek DC, Gibbs CJ Jr. Conformational

    transitions, dissociation, and unfolding of scrapie amyloid (prion)

    protein.J Biol Chem

    . 1993;268(27):20276-20284.10. Bueler H, Fischer M, Lang Y, et al. Normal development and

    behaviour of mice lacking the neuronal cell-surface PrP protein.

    Nature. 1992;356(6370):577-582.

    11. Bueler H, Aguzzi A, Sailer A, et al. Mice devoid of PrP are

    resistant to scrapie. Cell. 1993;73(7):1339-1347.

    12. Sailer A, Bueler H, Fischer M, Aguzzi A, Weissmann C. No

    propagation of prions in mice devoid of PrP. Cell. 1994;77(7):

    967-968.

    13. Liao YC, Lebo RV, Clawson GA, Smuckler EA. Human

    prion protein cDNA: molecular cloning, chromosomal map-

    ping, and biological implication. Science. 1986;233(4761):

    364-367.

    14. Stahl N, Borchelt DR, Hsiao K, Prusiner SB. Scrapie prion protein

    contains a phosphatidylinositol glycolipid. Cell. 1987;51(2):

    229-240.

    15. Jendroska K, Heinzel FP, Torchia M, et al. Proteinase-resistant

    prion protein accumulation in Syrian hamster brain correlates

    with regional pathology and scrapie infectivity. Neurology.

    1991;41(9):1482-1490.

    16. Kretzschmar HA, Prusiner SB, Stowring LE, DeArmond SJ. Scra-

    pie prion proteins are synthesized in neurons. Am J Pathol.

    1986;122(1):1-5.

    17. Bendheim PE, Brown HR, Rudelli RD, et al. Nearly ubiquitous

    tissue distribution of the scrapie agent precursor protein. Neurol-

    ogy. 1992;42(1):149-156.

    18. Perini F, Vidal R, Ghetti B, Tagliavini F, Frangione B, Prelli F.

    PrP27-30 is a normal soluble prion protein fragment released by

    human platelets. Biochem Biophys Res Commun. 1996;223(3):

    572-577.

    19. Borchelt DR, Koliatsis VE, Guarnieri M, Pardo CA, Sisodia SS,

    Price DL. Rapid anterograde axonal transport of the cellular prion

    glycoprotein in the peripheral and central nervous systems. J BiolChem. 1994;269(20):14711-14714.

    20. Collinge J, Whittington MA, Sidle KC, et al. Prion protein is nec-

    essary for normal synaptic function. Nature. 1994;370(6487):

    295-297.

    21. Whittington MA, Sidle KCL, Gowland I, et al. Rescue of

    neurophysiological phenotype seen in PrP null mice by transgene

    encoding human prion protein. Nat Genet. 1995;9(2):197-201.

    22. Manson JC, Hope J, Clarke AR, Johnston A, Black C,

    MacLeod N. PrP gene dosage and long term potentiation. Neuro-

    degeneration. 1995;4(1):113-114.

    23. Kanaani J, Prusiner SB, Diacovo J, Baekkeskov S, Legname G.

    Recombinant prion protein induces rapid polarization and devel-opment of synapses in embryonic rat hippocampal neurons in

    vitro.J Neurochem. 2005;95(5):1373-1386.

    24. Spielhaupter C, Schatzl HM. PrPC directly interacts with proteins

    involved in signaling pathways. J Biol Chem. 2001;276(48):

    44604-44612.

    25. Solforosi L, Criado JR, McGavern DB, et al. Cross-linking cellu-

    lar prion protein triggers neuronal apoptosis in vivo. Science.

    2004;303(5663):1514-1516.

    26. Mouillet-Richard S, Ermonval M, Chebassier C, et al. Signal

    transduction through prion protein. Science. 2000;289:

    1925-1928.

    27. Lopes MH, Hajj GN, Muras AG, et al. Interaction of cellular prion

    and stress-inducible protein 1 promotes neuritogenesis and neuro-

    protection by distinct signaling pathways. J Neurosci. 2005;

    25(49):11330-11339.

    28. Roucou X, LeBlanc AC. Cellular prion protein neuroprotective

    function: implications in prion diseases. J Mol Med. 2005;83(1):

    3-11.

    29. Brown DR, Qin K, Herms JW, et al. The cellular prion protein

    binds copperin vivo.Nature. 1997;390(6661):684-687.

    30. Stockel J, Safar J, Wallace AC, Cohen FE, Prusiner SB. Prion pro-

    tein selectively binds copper (II) ions.Biochemistry. 1998;37(20):

    7185-7193.

    31. Zanata SM, Lopes MH, Mercadante AF, et al. Stress-inducible

    protein 1 is a cell surface ligand for cellular prion that triggers

    neuroprotection.Embo J. 2002;21(13):3307-3316.

    32. Brown DR, Wong BS, Hafiz F, Clive C, Haswell SJ, Jones IM.

    Normal prion protein has an activity like that of superoxide dis-

    mutase.Biochem J. 1999;344(pt 1):1-5.

    33. Malaga-Trillo E, Solis GP, Schrock Y, et al. Regulation of

    embryonic cell adhesion by the prion protein. PLoS Biol. 2009;

    7(3):e55.

    34. Isaacs JD, Jackson GS, Altmann DM. The role of the cellular

    prion protein in the immune system. Clin Exp Immunol. 2006;

    146(1):1-8.

    35. Lledo PM, Tremblay P, DeArmond SJ, Prusiner SB, Nicoll RA.

    Mice deficient for prion protein exhibit normal neuronal

    Brown and Mastrianni 291

    291

    by guest on August 29, 2012jgp.sagepub.comDownloaded from

    http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/http://jgp.sagepub.com/
  • 7/24/2019 Articulo 6. Priones

    17/23

    excitability and synaptic transmission in the hippocampus. Proc

    Natl Acad Sci U S A. 1996;93(6):2403-2407.

    36. Criado JR, Sanchez-Alavez M, Conti B, et al. Mice devoid of

    prion protein have cognitive deficits that are rescued by reconsti-

    tution of PrP in neurons. Neurobiol Dis. 2005;19(1-2):255-265.

    37. Bremer J, Baumann F, Tiberi C, et al. Axonal prion protein is

    required for peripheral myelin maintenance. Nat Neurosci.2010;13(3):310-318.

    38. Tobler I, Gaus SE, Deboer T, et al. Altered circadian activity

    rhythms and sleep in mice devoid of prion protein. Nature.

    1996;380(6575):639-642.

    39. Moore RC, Lee IY, Silverman GL, et al. Ataxia in prion protein

    (PrP)-deficient mice is associated with upregulation of the novel

    PrP-like protein doppel. J Mol Biol. 1999;292(4):797-817.

    40. Parkin ET, Watt NT, Hussain I, et al. Cellular prion protein

    regulates beta-secretase cleavage of the Alzheimers amyloid

    precursor protein. Proc Natl Acad Sci USA. 2007;104(26):

    11062-11067.

    41. Nygaard HB, Strittmatter SM. Cellular prion protein mediates thetoxicity of beta-amyloid oligomers: implications for Alzheimer

    disease.Arch Neurol. 2009;66(11):1325-1328.

    42. Schwarze-Eicker K, Keyvani K, Gortz N, Westaway D, Sachser N,

    Paulus W. Prion protein (PrPc) promotes beta-amyloid plaque

    formation. Neurobiol Aging. 2005;26(8):1177-1182.

    43. Brown P, Cathala F, Raubertas RF, Gajdusek DC, Castaigne P.

    The epidemiology of Creutzfeldt-Jakob disease: conclusion of a

    15-year investigation in France and review of the world literature.

    Neurology. 1987;37(6):895-904.

    44. Spudich S, Mastrianni JA, Wrensch M, et al. Complete penetrance

    of Creutzfeldt-Jakob disease in Libyan Jews carrying the E200K

    mutation in the prion protein gene.Mol Med. 1995;1(6):607-613.

    45. Hsiao KK, Doh-ura K, Kitamoto T, Tateishi J, Prusiner SB. A

    prion protein amino acid substitution in ataxic Gerstmann-

    Straussler syndrome. Ann Neurol. 1989;26:137.

    46. Hsiao K, Baker HF, Crow TJ, et al. Linkage of a prion protein

    missense variant to Gerstmann-Straussler syndrome. Nature.

    1989;338(6213):342-345.

    47. Young K, Clark HB, Piccardo P, Dlouhy SR, Ghetti B.

    Gerstmann-Straussler-Scheinker disease with the PRNP P102L

    mutation and valine at codon 129. Brain Res Mol Brain Res.

    1997;44(1):147-150.

    48. Kitamoto T, Amano N, Terao Y, et al. A new inherited prion dis-

    ease (PrP-P105L mutation) showing spastic paraparesis.Ann Neu-

    rol. 1993;34(6):808-813.

    49. Rogaeva E, Zadikoff C, Ponesse J, et al. Childhood onset in famil-

    ial prion disease with a novel mutation in the PRNP gene. Arch

    Neurol. 2006;63(7):1016-1021.

    50. Tunnell E, Wollman R, Mallik S, Cortes CJ, Dearmond SJ,

    Mastrianni JA. A novel PRNP-P105S mutation associated with

    atypical prion disease and a rare PrPSc conformation. Neurology.

    2008;71(18):1431-1438.

    51. Rodriguez MM, Peoch K, Haik S, et al. A novel mutation

    (G114V) in the prion protein gene in a family with inherited prion

    disease.Neurology. 2005;64(8):1455-1457.

    52. Doh-ura K, Tateishi J, Sasaki H, Kitamoto T, Sakaki Y. Pro->Leu

    change at position 102 of prion protein is the most common but

    not the sole mutation related to Gerstmann-Straussler syndrome.

    Biochem Biophys Res Commun. 1989;163(2):974-979.

    53. Hsiao KK, Cass C, Schellenberg GD, et al. A prion protein variant

    in a family with the telencephalic form of Gerstmann-Straussler-

    Scheinker syndrome.Neurology. 1991;41(5):681-684.

    54. Mastrianni JA, Curtis MT, Oberholtzer JC, et al. Prion disease

    (PrP-A117V) presenting with ataxia instead of dementia. Neurol-ogy. 1995;45(11):2042-2050.

    55. Owen F, Poulter M, Collinge J, Crow TJ. Codon 129 changes in

    the prion protein gene in Caucasians. Am J Hum Genet.

    1990;46(6):1215-1216.

    56. Panegyres PK, Toufexis K, Kakulas BA, et al. A new PRNP muta-

    tion (G131V) associated with Gerstmann-Straussler-Scheinker

    disease.Arch Neurol. 2001;58(11):1899-1902.

    57. Rowe DB, Lewis V, Needham M, et al. Novel prion protein

    gene mutation presenting with subacute PSP-like syndrome.

    Neurology. 2007;68(11):868-870.

    58. Kitamoto T, Iizuka R, Tateishi J. An amber mutation of prion pro-

    tein in Gerstmann-Straussler syndrome with mutant PrP plaques.Biochem Biophys Res Commun. 1993;192(2):525-531.

    59. Pastore M, Chin SS, Bell KL, et al. Creutzfeldt-Jakob disease

    (CJD) with a mutation at codon 148 of prion protein gene:

    relationship with sporadic CJD. Am J Pathol. 2005;167(6):

    1729-1738.

    60. Finckh U, Muller-Thomsen T, Mann U, et al. High prevalence of

    pathogenic mutations in patients with early-onset dementia

    detected by sequence analyses of four different genes. Am J Hum

    Genet. 2000;66(1):110-117.

    61. Revesz T, Holton JL, Lashley T, et al. Genetics and molecular

    pathogenesis of sporadic and hereditary cerebral amyloid angio-

    pathies.Acta Neuropathol. 2009;118(1):115-130.

    62. Fink JK, Peacock ML, Warren JT, Roses AD, Prusiner SB.

    Detecting prion protein gene mutations by denaturing gradient gel

    electrophoresis.Hum Mutat. 1994;4(1):42-50.

    63. Goldfarb LG, Haltia M, Brown P, et al. New mutation in scrapie

    amyloid precursor gene (at codon 178) in Finnish Creutzfeldt-

    Jakob kindred.Lancet. 1991;337(8738):425.

    64. Medori R, Tritschler HJ, LeBlanc A, et al. Fatal familial insom-

    nia, a prion disease with a mutation at codon 178 of the prion pro-

    tein gene.N Engl J Med. 1992;326(7):444-449.

    65. Kitamoto T, Ohta M, Doh-ura K, Hitoshi S, Terao Y, Tateishi J.

    Novel missense variants of prion protein in Creutzfeldt-Jakob dis-

    ease or Gerstmann-Straussler syndrome. Biochem Biophys Res

    Commun 1993;191:709-14.

    66. Nitrini R, Rosemberg S, Passos-Bueno M, et al. Fa