+ All Categories
Home > Documents > Genetic dissection of neurodegenerative disease

Genetic dissection of neurodegenerative disease

Date post: 19-Sep-2016
Category:
Upload: john-hardy
View: 215 times
Download: 0 times
Share this document with a friend
8

Click here to load reader

Transcript
Page 1: Genetic dissection of neurodegenerative disease

Genetic dissection of neurodegenerative disease

John Hardy*

Birdsall Building, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA

Abstract

The clinical and molecular of Alzheimer's disease, Parkinson's disease, Pick's disease, progressive supranuclear palsy, corticobasal

degeneration, multiple system atrophy and prion disease are reviewed. The hypothesis that these diseases share pathogenic pathways to

cell death which involve either the tau or the a-synuclein proteins is propounded. The production and use of transgenic animal models of

these disease, based on the genetic ®ndings are brie¯y reviewed in the context of the development of treatments for these prevalent and

distressing disorders. q 2001 Association for Research in Nervous and Mental Disease. All rights reserved.

Keywords: Alzheimer's disease; Parkinson's disease; Pick's disease; Progressive supranuclear palsy; Corticobasal degeneration; Multiple system atrophy;

Prion disease; Genetics; Transgenics

1. Introduction

Alzheimer's disease (AD), Parkinson's disease (PD),

Lewy body disease (LBD), `Pick's' disease and frontal

temporal dementia, progressive supranuclear palsy (PSP),

prion diseases (PrD) and multiple system atrophy (MSA)

are major neurodegenerative diseases which probably

af¯ict, in total, about 3% of the population directly and

many more than that through family relationships. Over

the last 10 years, during the decade of the brain, a combina-

tion of genetic and molecular pathologic investigations has

revealed many of the etiologies of these diseases and is now

beginning to reveal the pathogenic biochemical pathways

that lead to their clinical features. Developing an under-

standing of the etiology should help in developing better

and earlier diagnoses of these diseases, and may possibly

help in developing strategies to avoid them: developing an

understanding of the pathogeneses of these diseases is a

necessary ®rst step in devising rational treatments to replace

the palliative therapies which are used at present.

Surprisingly, these genetic and pathologic investigations

have revealed that these diseases share common pathogenic

mechanisms (Table 1): this commonality may explain the

clinical and pathologic overlaps between them and may also

have implications for the development of treatments for

them.

In this review, I have the following aims.

1. To outline the clinical genetics of these disorders.

2. To discuss the pathogenic relationships between them.

3. To discuss how the genetic ®ndings are helping in devis-

ing animal models of these diseases.

This review does not cover either the triplet repeat

diseases or amyotropic lateral sclerosis.

2. Clinical genetics of neurologic disease

2.1. Early onset autosomal dominant Alzheimer's disease

Three genetic loci have been shown to be important in

causing early onset, autosomal dominant AD. The amyloid

precursor protein (APP) gene on chromosome 21 [1], the

presenilin 1 (PS1) gene on chromosome 14 [2] and the

presenilin 2 (PS2) gene on chromosome 1 [3]. Mutations

at all three loci lead to disease in a generally predictable way

and, in families with these mutations, the age of onset is

relatively constant (see below for exceptions). Nearly all the

mutations are missense mutations (replacement of one

amino acid for another). A few involve the insertion or

deletion of a single amino acid, and one involves the dele-

tion of one exon of the protein, exon 9: even this mutation,

however, does not destroy the protein's structure [4]. Two

websites cover different aspects of these gene mutations:

http://molgen-www.uia.ac.be/ADMutations/ and http://

www.alzforum.org/members/index.html. All the pathogenic

mutations in APP are in and around the amyloid (Ab) part of

the molecule which is encoded by exons 16 and 17 of the

gene [5] (Fig. 1). Typically, the age of disease onset is in the

mid-50s, the disease is fully penetrant but the age of onset is

Clinical Neuroscience Research 1 (2001) 134±141

www.elsevier.nl/locate/clires

1566-2772/01/$ - see front matter q 2001 Association for Research in Nervous and Mental Disease. All rights reserved.

PII: S1566-2772(00)00013-X

* Tel.: 11-904-953-7356; fax: 11-904-953-7356.

E-mail address: [email protected] (J. Hardy).

Page 2: Genetic dissection of neurodegenerative disease

modulated by the apolipoprotein E genotype (see below).

For example, in one well studied family, the age of onset of

individuals with a mutation who are E3 homozygotes is 55,

whereas E4 have an onset of 45, and E2E3 heterozygotes

have an onset of 60 [6]. The phenotype of most families with

pathogenic APP mutations is that of typical AD: there are

two exceptions. First, hereditary cerebral hemorrhage with

amyloidosis, Dutch type, is caused by E693Q [7]: in this

stroke syndrome, individuals from one kindred of Dutch

descent have stroke caused by Ab deposition in the brain

blood vessels, and second the phenotype of A692G is rather

variable: some individuals in this Dutch family have a

stroke syndrome, whereas others have a more typical

Alzheimer phenotype: the pathology in this disease variant

is also unusual [8]. As far as this author is aware, no

commercial testing for APP mutations is available. The

author is reluctantly prepared to consider requests for help

in this regard.

Most of the mutations in PS1 are missense mutations to

residues conserved between PS1 and PS2 (Fig. 2). The

major exception to this rule is the PS1 D9 mutation which

deletes exon 9 and changes a residue at the splice site. For

updated mutation information visit the website referred to

above. Most PS1 mutations are fully penetrant (people with

mutation inevitably develop disease), but there are excep-

tions [9]. In addition the variant E318G is probably a poly-

morphism although it might be a risk factor allele. The

phenotype of PS1 mutations is usually with onset in the

40s, but with typical clinical features. However, a variant

with spastic paraparesis as the presenting feature, which is

associated with unusual neuropathology is well described

[10]. This is a frequent feature of the D9 mutation, but

also occurs with other mutations ([11], and unpublished

data). The age of onset in PS1 mutation families is not

affected by ApoE genotype [12]. Athena Diagnostics offers

diagnostic tests.

Few PS2 mutations have been described (Fig. 3). So far,

they are all missense mutations to residues conserved

between PS1 and PS2. The phenotype of PS2 mutations

seems to be variable: in some individuals, the age of onset

is in the 40s whereas in others, in the same family, it is in the

70s [3,13]. It may well be that there are many other PS2

mutations, but these have not been identi®ed because they

occur in cases without an obvious family history: relatively

few such cases have been sequenced. Two apparently

benign polymorphisms have been identi®ed: R62G and

R62H. No diagnostic tests are available commercially and,

given the variable phenotype, it is dif®cult to thinks of an

effective way that this could be achieved with present tech-

nology.

Mutations in these three genes explain all known cases of

J. Hardy / Clinical Neuroscience Research 1 (2001) 134±141 135

Fig. 1. Amyloid precursor protein. Pathogenic mutations in the APP gene

(in red) in relation to the sites of cleavage of this molecule. Derived from

http://www.alzforum.org with permission.

Table 1

The pathology of human neurodegenerative disease

Alzheimer's disease Plaques (Ab), tangles (tau)

and often, Lewy bodies

(a-synuclein)

Prion disease Often PrP plaques;

sometimes tangles;

sometimes Lewy bodies

FTDP-17/Pick's disease Tangles or Pick bodies

(3-repeat tau)

Parkinson's disease/Lewy

body dementia

Lewy bodies

Progressive supranuclear

palsy and corticobasal

degeneration

Tangles; mainly 4-repeat tau

Multiple system atrophy Oligodendroglial a-synuclein

inclusions

Page 3: Genetic dissection of neurodegenerative disease

AD with clear autosomal dominant, early onset disease.

There are no pedigrees with multiple generations and

cousins affected: there are many nuclear families and

many single cases of early onset disease in which mutations

J. Hardy / Clinical Neuroscience Research 1 (2001) 134±141136

Fig. 2. Presenilin 1. Pathogenic missense mutations in the presenilin 1 protein (in red) drawn on the likely topographic representation of this molecule. Derived

from http://www.alzforum.org with permission.

Fig. 3. Presenilin 2. Pathogenic missense mutations in the presenilin 2 protein (in red) drawn on the likely topographic representation of this molecule. Derived

from http://www.alzforum.org with permission.

Page 4: Genetic dissection of neurodegenerative disease

have not been found but these presumably re¯ect oligogenic

or gene/environment etiologies.

2.2. Late Onset Alzheimer's disease

It is clear that there is much familial clustering in late

onset AD [14]. So far, genetic analysis has only unequivo-

cally demonstrated that the Apolipoprotein E (ApoE) is a

risk factor locus for this form of the disease [15]. The

ApoE4 allele is associated with increased risk of disease

[15], and the ApoE2 allele associated, generally, with

decreased risk [16]. ApoE4 homozygotes have a greatly

increased risk of developing AD. It is likely that there are

several other genetic risk factors for developing disease, but

these have not yet been clearly identi®ed. All authorities are

agreed that ApoE testing is not accurate or speci®c enough

to be used for predictive testing: there is discussion as to

whether it may have a role as a diagnostic aid [17±19].

Apolipoprotein E testing is offered commercially by Athena

Diagnostics.

2.3. Prion diseases

Familial cases of prion disease are caused by mutations in

the prion gene [20]. Most of these are missense mutations

but some are caused by an increase in repeat length of an

octapeptide repeat [21], and there are some premature termi-

nation mutations which occur at the C-terminal of the

protein [22]. There is common polymorphism (M129V).

The occurrence of this variant in cis or trans with pathogenic

mutations can considerably alter both the phenotype and the

age at onset of disease. The phenotype can encompass

Creuzfeld±Jakob disease, Gerstmann±Straussler syndrome

and fatal familial insomnia. The website http://www.mad-

cow.org/ is a useful reference source for of this information.

No commercial company offers prion mutation screening.

Interestingly, sporadic and iatrogenic and `new variant' CJD

cases are nearly always homozygotes for the 129 poly-

morphism [23±25].

2.4. Frontal temporal dementia

Most hereditary cases of frontal temporal dementia are

caused by mutations in the tau gene [26,27] (Fig. 4), although

there is another locus on chromosome 3 [28]. Now, a large

number of tau mutations have been described. For details, see

the website http://www.alzforum.org/members/research/

tau/tau_references.html. Most of the tau mutations are fully

penetrant and fully penetrant, although there is a wide range

of phenotypes [29]. Nearly all the mutations are within the

microtubule binding domains encoded by exons 9±13. A

cluster of mutations occurs just outside exon 10 where it

affects the alternative splicing of that exon. No commercial

company offers sequencing of tau in hereditary frontal

temporal dementia. Clinicians are welcome to contact the

author if there is the suspicion of a tau mutation in family.

The term `Pick's disease' has fallen into confused use:

many cases with tau mutations would have been previously

diagnosed as Pick's disease.

J. Hardy / Clinical Neuroscience Research 1 (2001) 134±141 137

Fig. 4. Mutations in the tau gene, both in the open reading frame (A) and showing the positions which affect splicing alone at the stem loop structure (B) or

affect both splicing and the open reading frame (C).

Page 5: Genetic dissection of neurodegenerative disease

2.5. Progressive supranuclear palsy and corticobasal

degeneration

PSP occasionally occurs in families, but no genetic

linkages have been reported. However, it is now clear that

there are two alleles of tau occurring in the general popula-

tion: H1 and H2, and that PSP is robustly associated with

homozygosity for the H1 allele [30,31]. This is not diagnos-

tically useful: approximately 50% of the general Caucasian

population are H1 homozygotes and 95% of PSP cases.

CBD shows the same genetic association (Houlden, Hutton,

Hardy, unpublished data).

2.6. Parkinson's disease and Lewy body dementia

These diseases are discussed together because they

appear to occur in the same families. Two mutations in

the a-synuclein gene have been described, A53T and

A30P (Fig. 5): the former occurs in many families of

Greek origin [32], and the latter in a single family of

German origin [33]. Both mutations appear to be reasonably

highly penetrant, but more data is needed to establish this

with certainty. The usual phenotype is PD, but in some

cases, dementia, not parkinsonism, is the predominant

feature. Alpha-synuclein sequencing has not been offered

commercially: individuals are welcome to contact the

author if there is a family in which an a-synuclein mutation

may be suspected.

Two other genetic loci have been identi®ed: one on chro-

mosome 2p [34] and the other on chromosome 4p [35].

Neither of these loci are fully penetrant and both are asso-

ciated with variable phenotypes. It is clear that there are

other genetic loci which have not yet been identi®ed

(unpublished data).

Mutations in the parkin gene cause a parkinsonism of

young onset which can be clinically dif®cult to distinguish

from Parkinson's disease [36]. These mutations appear to

always be recessive in their mode of inheritance.

3. The relationship between the etiology andpathogenesis of neurodegenerative disease

The genetic ®ndings, outlined above, ®t with the patho-

logical features summarized in Table 1 and predict several

`rules' pertaining to these diseases [37].

1. If there is an extracellular protein pathology, the primary

genetic lesion relates to that protein. In prion disease, for

example, mutations in the prion gene are pathogenic; in

AD, mutations in the APP gene are one cause of patho-

genesis and mutations in the presenilin genes appear to

act via an effect on APP processing [38]. Another exam-

ple of this `rule' is British dementia, a plaque and tangle

disease, in which the primary lesion is also in the gene

encoding the plaque protein [39].

2. In diseases with extracellular pathology, the intracellular

pathology is secondary. The occurrence of tangles and

Lewy bodies in Alzheimer and prion diseases are thus

secondary, and alternate pathologies [40,41]. It will be of

interest to determine whether British dementia cases

have Lewy bodies as well as tangles.

3. In disease with only intracellular pathology, that pathol-

ogy is primary (mutations in the tau and a-synuclein): the

J. Hardy / Clinical Neuroscience Research 1 (2001) 134±141138

Fig. 5. The sequences of the human synucleins and of mouse a-synuclein: note that the better characterized human mutations (A53T) changes the human

sequence to the rodent sequence at that residue.

Page 6: Genetic dissection of neurodegenerative disease

presumption must be that in the cases of these diseases

without tau or a-synuclein pathology, then the primary

lesion must relate closely to these proteins, as the prese-

nilins relate to APP in AD.

4. When the primary lesion relates to one intracellular

lesion (tau/tangles or a-synuclein/Lewy bodies), then

only that lesion is formed.

These `rules' can be used to construct a diagram of

proposed biochemical relationships based on the genetics

and pathology of these diseases (Fig. 6) [37]. If this diagram

offers an approximation to the truth, it has two implications.

1. The tau/tangle and a-synuclein/Lewy body pathologies

mark alternate routes to cell death [41].

2. Since they both can be activated by both Ab and prion, it is

likely that they share common pathogenic mechanisms.

4. Modeling neurodegenerative disease in animals

A major purpose in de®ning the genetic bases of neuro-

degenerative disease is to develop the resources to enable

transgenic modeling of them, both to test theories of patho-

genesis and to test therapies upon. For reasons of cost,

transgenic mice are the usual model chosen, although the

recent successes in developing partial animal models of both

triplet repeat diseases [42,43] and of Lewy body diseases in

Drosophila offer the prospect that for some applications this

system may be surprisingly valuable.

Use of transgenic mice to model human neurodegenera-

tive disease dates from successful modeling of prion disease

using pathogenic prion alleles as transgenes [44]. This

approach has now been used successfully to model the

amyloid formation of AD using pathogenic APP alleles as

transgenes [45], and even more effectively, using both

mutant PS1 and APP transgenes [46,47] and the tangle

formation in FTDP-17 using pathogenic tau alleles [48]:

reports of successful modeling of Lewy body disease have

not yet been replicated ([49], Duff, Farrer, Hardy, unpub-

lished data). With respect to AD, none of the transgenic

mice yet examined have developed both plaque and tangle

pathology: clearly the separate development of plaque form-

ing mice and tangle forming mice illustrates that this can

now be achieved [45,48]. Determination of whether the two

pathologies interact in such mice will be an important test of

theories of pathogenesis.

From a treatment perspective, the use of these mice is

now guiding the pharmaceutical industry in their drug

development programs, and the potential development of

an Alzheimer `amyloid vaccine' represents the ®rst example

of this approach [50]. Whether this approach to the treat-

ment of neurodegenerative disease will be successful should

become clear in the next 5 years.

J. Hardy / Clinical Neuroscience Research 1 (2001) 134±141 139

Fig. 6. Summary diagram linking all the tau and synucleinopathies is one broad framework of pathogenesis: note that (for example) this scheme is concordant

both with the amyloid cascade hypothesis and with the notion that Alzheimer's disease is a tauopathy (adapted from Ref. [37]).

Page 7: Genetic dissection of neurodegenerative disease

5. Conclusions

Though the application of molecular genetics, we have an

increasingly clear understanding of the etiology of the major

neurodegenerative diseases. Surprisingly, many of these

diseases appear to be pathogenically related to each other.

Furthermore, these genetic ®ndings allow theories of patho-

genesis to be tested in relevant transgenic animals. In addi-

tion, treatment strategies can be assessed in these animals. It

is to be hoped and expected that therapies designed on the

basis of this genetic knowledge will reach clinical practice

within the next decade.

Acknowledgements

Work in the authors laboratory was supported by the NIH

and by the Alzheimer's Association. Updated and modi®ed

from course material prepared for the American Academy

of Neurology.

References

[1] Goate A, Chartier-Harlin MC, Mullan M, et al. Segregation of a

missense mutation in the amyloid precursor protein gene with familial

Alzheimer's disease. Nature 1991;349:704±706.

[2] Sherrington R, Rogaev EI, Liang Y, et al. Cloning of a gene bearing

missense mutations in early-onset familial Alzheimer's disease.

Nature 1995;375:754±760.

[3] Levy-Lahad E, Wasco W, Poorkaj P, et al. Candidate gene for the

chromosome 1 familial Alzheimer's disease locus. Science

1995;269:973±977.

[4] Perez-Tur J, Froelich S, Prihar G, et al. A mutation in Alzheimer's

disease destroying a splice acceptor site in the presenilin-1 gene.

NeuroReport 1995;7:297±301.

[5] Houlden H, Collinge J, Kennedy A, et al. APOE genotype and Alzhei-

mer's disease. Lancet 1993;342:737±738.

[6] Levy E, Carman MD, Fernandez-Madrid IJ, et al. Mutation of the

Alzheimer's disease amyloid gene in hereditary cerebral hemorrhage,

Dutch type. Science 1990;248:1124±1126.

[7] Cras P, van Harskamp F, Hendriks L, et al. Presenile Alzheimer

dementia characterized by amyloid angiopathy and large amyloid

core type senile plaques in the APP 692Ala! Gly mutation. Acta

Neuropathol (Berl) 1998;96:253±260.

[8] Rossor MN, Fox NC, Beck J, Campbell TC, Collinge J. Incomplete

penetrance of familial Alzheimer's disease in a pedigree with a novel

presenilin-1 gene mutation. Lancet 1996;347:1560.

[9] Crook R, Verkkoniemi A, Perez-Tur J, et al. A variant of Alzheimer's

disease with spastic paraparesis and unusual plaques due to deletion

of exon 9 of presenilin 1. Nat Med 1998;4:452±455.

[10] Kwok JB, Taddei K, Hallupp M, et al. Two novel (M233T and

R278T) presenilin-1 mutations in early-onset Alzheimer's disease

pedigrees and preliminary evidence for association of presenilin-1

mutations with a novel phenotype. NeuroReport 1997;8:1537±1542.

[11] Van Broeckhoven C, Backhovens H, Cruts M, et al. APOE genotype

does not modulate age of onset in families with chromosome 14

encoded Alzheimer's disease. Neurosci Lett 1994;169:179±180.

[12] Hardy J. Framing beta-amyloid. Nat Genet 1992;1:233±234.

[13] Rogaev EI, Sherrington R, Rogaeva EA, et al. Familial Alzheimer's

disease in kindreds with missense mutations in a gene on chromosome

1 related to the Alzheimer's disease type 3 gene. Nature

1995;376:775±778.

[14] Heston LL, Mastri AR, Anderson VE, White J. Dementia of the

Alzheimer type. Clinical genetics, natural history, and associated

conditions. Arch Gen Psychiatry 1981;38:1085±1090.

[15] Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apoli-

poprotein E type 4 allele and the risk of Alzheimer's disease in late

onset families. Science 1993;261:921±923.

[16] Chartier-Harlin MC, Par®tt M, Legrain S, et al. Apolipoprotein E

epsilon 4 allele as a major risk factor for sporadic early and late-

onset forms of Alzheimer's disease: analysis of the 19q13.2 chromo-

somal region. Hum Mol Genet 1994;3:569±574.

[17] Post SG, Whitehouse PJ, Binstock RH, et al. The clinical introduction

of genetic testing for Alzheimer disease. An ethical perspective. J Am

Med Assoc 1997;277:832±836.

[18] Relkin NR, Kwon YJ, Tsai J, Gandy S. The National Institute on

Aging/Alzheimer's Association recommendations on the application

of apolipoprotein E genotyping to Alzheimer's disease. Ann N Y

Acad Sci USA 1996;802:149±176.

[19] McConnell LM, Koenig BA, Greely HT, Raf®n TA. Genetic testing

and Alzheimer disease: recommendations of the Stanford Program in

genomics, ethics, and society. Genet Test 1999;3:3±12.

[20] Hsiao K, Baker HF, Crow TJ, et al. Linkage of a prion protein

missense variant to Gerstmann±Straussler syndrome. Nature

1989;338:342±345.

[21] Owen F, Poulter M, Lofthouse R, et al. Insertion in prion protein gene

in familial Creutzfeldt±Jakob disease. Lancet 1989;1:51±52.

[22] Ghetti B, Piccardo P, Spillantini MG, et al. Vascular variant of prion

protein cerebral amyloidosis with tau-positive neuro®brillary tangles:

the phenotype of the stop codon 145 mutation in PRNP. Proc Natl

Acad Sci USA 1996;93:744±748.

[23] Collinge J, Palmer MS, Dryden AJ. Genetic predisposition to iatro-

genic Creutzfeldt±Jakob disease. Lancet 1991;337:1441±1442.

[24] Palmer MS, Dryden AJ, Hughes JT, Collinge J. Homozygous prion

protein genotype predisposes to sporadic Creutzfeldt±Jakob disease.

Nature 1991;352:340±342.

[25] Collinge J, Beck J, Campbell T, Estibeiro K, Will RG. Prion protein

gene analysis in new variant cases of Creutzfeldt±Jakob disease.

Lancet 1996;348:56.

[26] Poorkaj P, Bird TD, Wijsman E, et al. Tau is a candidate gene for

chromosome 17 frontotemporal dementia. Ann Neurol 1998;43:815±

825.

[27] Hutton M, Lendon CL, Rizzu P, et al. Association of missense and 5 0-splice-site mutations in tau with the inherited dementia FTDP-17.

Nature 1998;393:702±705.

[28] Brown J, Ashworth A, Gydesen S, et al. Familial non-speci®c demen-

tia maps to chromosome 3. Hum Mol Genet 1995;4:1625±1628.

[29] Foster NL, Wilhelmsen K, Sima AA, Jones MZ, D'Amato CJ, Gilman

S. Frontotemporal dementia and parkinsonism linked to chromosome

17: a consensus conference. Conference participants. Ann Neurol

1997;41:706±715.

[30] Conrad C, Andreadis A, Trojanowski JQ, et al. Genetic evidence for

the involvement of tau in progressive supranuclear palsy. Ann Neurol

1997;41:277±281.

[31] Baker M, Litvan I, Houlden H, et al. Association of an extended

haplotype in the tau gene with progressive supranuclear palsy. Hum

Mol Genet 1999;8:711±715.

[32] Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the

alpha-synuclein gene identi®ed in families with Parkinson's disease.

Science 1997;276:2045±2047.

[33] Kruger R, Kuhn W, Muller T, et al. Ala30Pro mutation in the gene

encoding alpha-synuclein in Parkinson's disease. Nat Genet

1998;18:106±108.

[34] Gasser T, Muller-Myhsok B, Wszolek ZK, et al. A susceptibility locus

for Parkinson's disease maps to chromosome 2p13. Nat Genet

1998;18:262±265.

[35] Farrer M, Gwinn-Hardy K, Muenter M, et al. A chromosome 4p

haplotype segregating with Parkinson's disease and postural tremor.

Hum Mol Genet 1999;8:81±85.

J. Hardy / Clinical Neuroscience Research 1 (2001) 134±141140

Page 8: Genetic dissection of neurodegenerative disease

[36] Kitada T, Asakawa S, Hattori N, et al. Mutations in the parkin gene

cause autosomal recessive juvenile parkinsonism. Nature

1998;392:605±608.

[37] Hardy J. Pathways to primary neurodegenerative disease. Mayo Clin

Proc 1999;74:835±837.

[38] Scheuner D, Eckman C, Jensen M, et al. Secreted amyloid beta-

protein similar to that in the senile plaques of Alzheimer's disease

is increased in vivo by the presenilin 1 and 2 and APP mutations

linked to familial Alzheimer's disease. Nat Med 1996;2:864±870.

[39] Vidal R, Frangione B, Rostagno A, et al. A stop-codon mutation in the

BRI gene associated with familial British dementia. Nature

1999;399:776±781.

[40] Hardy J. Lewy bodies in Alzheimer's disease in which the primary

lesion is a mutation in the amyloid precursor protein. Neurosci Lett

1994;180:290±291.

[41] Hansen LA, Masliah E, Galasko D, Terry RD. Plaque-only Alzheimer

disease is usually the Lewy body variant, and vice versa. J Neuro-

pathol Exp Neurol 1993;52:648±654.

[42] Warrick JM, Paulson HL, Gray-Board GL, et al. Expanded polyglu-

tamine protein forms nuclear inclusions and causes neural degenera-

tion in Drosophila. Cell 1998;93:939±949.

[43] Feany MB, Bender WW. A Drosophila model of Parkinson's disease.

Nature 2000;404:394±398.

[44] Hsiao KK, Scott M, Foster D, Groth DF, DeArmond SJ, Prusiner SB.

Spontaneous neurodegeneration in transgenic mice with mutant prion

protein. Science 1990;250:1587±1590.

[45] Games D, Adams D, Alessandrini R, et al. Alzheimer-type neuro-

pathology in transgenic mice overexpressing V717F beta-amyloid

precursor protein. Nature 1995;373:523±527.

[46] Borchelt DR, Ratovitski T, van Lare J, et al. Accelerated amyloid

deposition in the brains of transgenic mice coexpressing mutant prese-

nilin 1 and amyloid precursor proteins. Neuron 1997;19:939±945.

[47] Holcomb L, Gordon MN, McGowan E, et al. Accelerated Alzheimer-

type phenotype in transgenic mice carrying both mutant amyloid

precursor protein and presenilin 1 transgenes. Nat Med 1998;4:97±

100.

[48] Lewis J, McGowan E, Rockwood J, Melrose H, Nacharaju P, Van

Slegtenhorst M, Gwinn-Hardy K, Murphy PM, Baker M, Yu X, Duff

K, Hardy J, Corral A, Lin WL, Yen SH, Dickson DW, Davies DW,

Hutton M. Neuro®brillary tangles, amyotrophy and progressive motor

disturbance in mice expressing mutant (P301L) tau protein. Nat Genet

2000;25:402±405.

[49] Masliah E, Rockenstein E, Veinbergs I, et al. Dopaminergic loss and

inclusion body formation in alpha-synuclein mice: implications for

neurodegenerative disorders. Science 2000;287:1265±1269.

[50] Schenk D, Barbour R, Dunn W, et al. Immunization with amyloid-

beta attenuates Alzheimer-disease-like pathology in the PDAPP

mouse. Nature 1999;400:173±177.

J. Hardy / Clinical Neuroscience Research 1 (2001) 134±141 141


Recommended