• Tidak ada hasil yang ditemukan

Directory UMM :Data Elmu:jurnal:B:Brain Research:Vol886.Issue1-2.Nov2000:

N/A
N/A
Protected

Academic year: 2017

Membagikan "Directory UMM :Data Elmu:jurnal:B:Brain Research:Vol886.Issue1-2.Nov2000:"

Copied!
13
0
0

Teks penuh

(1)

www.elsevier.com / locate / bres

Interactive report

1

Alzheimer’s disease: a dysfunction of the amyloid precursor protein

*

Rachael L. Neve , Donna L. McPhie, Yuzhi Chen

Department of Psychiatry, Harvard Medical School, MRC 223 McLean Hospital, 115 Mill St., Belmont, MA 02478, USA

Accepted 25 August 2000

Abstract

In this review, we argue that at least one insult that causes Alzheimer’s disease (AD) is disruption of the normal function of the amyloid precursor protein (APP). Familial Alzheimer’s disease (FAD) mutations in APP cause a disease phenotype that is identical (with the exception that they cause an earlier onset of the disease) to that of ‘sporadic’ AD. This suggests that there are molecular pathways common to FAD and sporadic AD. In addition, all individuals with Down syndrome, who carry an extra copy of chromosome 21 and overexpress APP several-fold in the brain, develop the pathology of AD if they live past the age of 40. These data support the primacy of APP in the disease. Although APP is the source of theb-amyloid (Ab) that is deposited in amyloid plaques in AD brain, the primacy of APP in AD may not lie in the production of Abfrom this molecule. We suggest instead that APP normally functions in the brain as a cell surface signaling molecule, and that a disruption of this normal function of APP is at least one cause of the neurodegeneration and consequent dementia in AD. We hypothesize in addition that disruption of the normal signaling function of APP causes cell cycle abnormalities in the neuron, and that these abnormalities constitute one mechanism of neuronal death in AD. Data supporting these hypotheses have come from investigations of the molecular consequences of neuronal expression of FAD mutants of APP or overexpression of wild type APP, as well as from identification of binding proteins for the carboxyl-terminus (C-terminus) of APP.

 2000 Elsevier Science B.V. All rights reserved.

Theme: Disorders of the nervous system

Topic: Degenerative disease: Alzheimer’s – beta amyloid

Keywords: Alzheimer’s disease; Amyloid precursor protein; Neuronal death

1. Introduction

possession of an extra copy of chromosome 21 (Down

syndrome); or it can be caused by mutations in the amyloid

1.1. APP, A

b

, and Alzheimer disease

precursor protein (APP) gene on chromosome 21 or by

mutations in the presenilin genes on chromsome 1 and 14.

All individuals with Alzheimer disease (AD) experience

Additional genetic complexicity is conferred on it by the

a progressive loss of cognitive function, resulting from a

fact that the

e

4 allele of the APOE gene is a major risk

neurodegenerative process characterized classically by the

factor for the development of AD. Thus, it is not likely that

deposition of

b

-amyloid (A

b

) in plaques and in the

AD is caused by a single molecular event.

cerebrovasculature, and the formation of neurofibrillary

Numerous mechanisms for the neuronal cell death in

tangles in neurons. Additional pathological hallmarks of

AD have been proposed. One of these is the amyloid

AD include granulovacular degeneration, loss of synapses

hypothesis, which suggests that deposition of A

b

is a

and decreases in cell density in distinct regions of the

primary event in the pathological cascade for AD. This

brain. Alzheimer disease does not have a simple etiology.

argument is based on in vitro studies showing that A

b

is

It can occur as a ‘sporadic’ event; it can result from the

toxic to neurons and on the measurement of increased

release of A

b

by cells carrying familial AD (FAD) mutant

genes. There are two major carboxyl-terminal variants of

1

Published on the World Wide Web on 11 September 2000.

A

b

. A

b

1 – 40

is the major species secreted from cultured

*Corresponding author. Tel.: 11-617-855-2413; fax:

11-617-855-cells and found in cerebrospinal fluid, while A

b

is the

3793. 1 – 42

E-mail address: neve@helix.mgh.harvard.edu (R.L. Neve).

major component of amyloid deposits (reviewed in Ref.

(2)

[118]). Cells expressing FAD mutants of APP and the

transmission [28]. There has been some question of

presenilins are reported to secrete increased amounts of

whether C100 exerts its neurotoxic effects from the inside

A

b

1 – 42

, suggesting a link of this variant of A

b

to AD

or the outside of the cell [23,117]. Our data of the past 6

pathogenesis. Consequently, a leading hypothesis for the

years suggest strongly that C100 kills from inside the cell;

etiology of AD is that increased A

b

1 – 42

is a shared

this is supported by the observation that C100 is not

molecular correlate of FAD mutations, and that it repre-

secreted, even when it carries a signal peptide [19,14,66].

sents a gain of deleterious function that can cause FAD

Although at least one group has reported neurotoxicity due

[38] and may be an essential early event in AD [118].

to the addition of C100 to the culture medium [50], we

While this ‘amyloid hypothesis’ is attractive, molecular

believe that that type of neurotoxicity is mechanistically

mechanisms other than those mediated by extracellular A

b

different from the neurodegeneration that we observe upon

could also lead to AD neurodegeneration.

expression of C100 within primary neurons.

These mechanisms are likely to be linked in some way

The findings that APP interacts with the signaling

to the

b

-amyloid protein precursor (APP), the source of

molecule G , that FAD mutants of APP can cause G -

o o

A

b

. One of the most compelling pieces of evidence that

mediated apoptosis in neuronal cells, and that these same

links AD neurodegeneration to APP and / or its A

b

-con-

FAD mutants of APP cause the intracellular accumulation

taining derivatives is the early finding that the APP gene is

of C100, suggested to us the following working

hypoth-on chromosome 21: virtually all individuals trisomic for

esis: In the brain a portion of APP is present as an integral

this chromosome show AD-like neuropathology by the age

plasma membrane protein that mediates the transduction of

of 40. Additionally, it has been discovered that specific

extracellular signals into the cell via its C-terminal tail, and

mutations in APP cause some forms of familial FAD.

abnormal accumulation of its A

b

-containing C-terminal

These data have raised the possibility that AD may result

tail in the neuron causes progressive dysfunction of APP

from an alteration in the normal function of APP [74,76],

signaling in AD, resulting in apoptosis. This hypothesis has

and have refocused attention on the delineation of the

been supported by the finding that the intracellular

C-function that APP subserves in the brain. It has been

terminal tail of APP interacts with the cell cycle protein

shown [47,83] that in the brain a percentage of APP is

APP-BP1 [10,9], and with members of the Fe65 family of

present on the cell surface, and it is proposed [76,83] that

adaptor proteins (reviewed in Ref. [89]). Additional

sup-this cell surface APP mediates the transduction of extracel-

port for this hypothesis emerged with the recent report [60]

lular signals into the cell via its C-terminal tail.

that the C31 peptide of APP, which is derived from C100

Nishimoto and his colleagues [75] showed that APP

and within which are contained the binding sites for the

binds to the brain-specific signal transducing G protein G ;

o

above proteins, is elevated in AD brain and is a potent

independent confirmation of this finding has subsequently

inducer of apoptosis.

been published [4,5]. It was then discovered [113] that

V642 (‘London’) FAD mutants of APP induce neuronal

DNA fragmentation, a feature of apoptosis, in a neuronal

2. Processing of APP

cell line. This fragmentation is independent of A

b

1 – 42

production [114] and is mediated by the G

b g

2 2

complex of

Most of what we know about APP processing has come

G [29]. These data support the notion that APP has an

o

from work with cultured cells. APP matures through the

intrinsic signaling function in the neuron, which becomes

constitutive secretory pathway. Some fraction of the APP

ligand-independent when APP is mutated at V642.

is endoproteolytically cleaved at the cell surface within the

To examine the mechanism by which FAD APP might

A

b

sequence by the

a

-secretase, which generates the

cause apoptosis in neurons, we [66] expressed five differ-

neuroprotective

secreted

amyloid

precursor

protein

ent Alzheimer mutations of APP in primary neurons via

(APP ) and nonamyloidogenic 3 kDa A

sa

b

secreted

prod-recombinant herpes simplex virus (HSV) vectors, and

ucts [81,37,65]. APP

Sa

is readily detected in human plasma

quantified the levels of APP metabolites. The predominant

and cerebrospinal fluid.

(3)

can be detected intracellularly. C100-like amyloidogenic

and colleagues have shown that

b

-secretase cleavage

fragments have been detected only intracellularly.

products of APP are present in fetal and neonatal Down

It is important to note that APP processing is cell

syndrome brain at twice normal levels ([93] and personal

type-specific. LeBlanc and colleagues have reported that

communication with Dr. Russo). We have hypothesized

human neurons secrete more 4 kDa than 3 kDa A

b

, and

that abnormal accumulation of the A

b

-containing

C-termi-metabolize approximately 40% of newly-synthesized APP

nal tail (C100) of APP in neurons also occurs in Alzheimer

through the

a

-secretase pathway [53,55]. Moreover,

disease.

human neurons produce five C-terminal fragments of APP

in a pattern seen uniquely in human brain [21,53]. The two

largest C-terminal derivatives have the entire A

b

sequence

3. APP as a signaling molecule

at or near their amino terminus [21], and most likely

represent endogenous ‘C100’ fragments. Thus, C100 is a

The possibility that APP may act as a signaling receptor

physiologically relevant fragment of APP in the human

was first proposed on the basis of its predicted amino acid

brain. In contrast to human neurons, most APP-transfected

sequence, which suggested that APP was a type 1 intrinsic

human or nonhuman cell lines produce more 3 kDa than 4

membrane protein consistent with the structure of a ‘cell

kDa A

b

and show a relatively nonamyloidogenic pattern

surface receptor.’ [49]. However, subsequent studies of the

of C-terminal fragments [31,36,37,54].

function of APP concentrated largely on the secreted

Analyses of

b

-amyloid (A

b

) in genetically engineered

ectodomain, because of a lack of direct evidence that

cell lines expressing FAD mutations in both APP and the

mature APP exists on the cell surface with intact

intracel-presenilins (PS) have shown that all of the mutations cause

lular, transmembrane, and intracellular domains. Surface

either increased overall secretion of A

b

or secretion of the

APP was inferred to exist on a variety of cultured cells

‘long’ (42–43-amino acid) form of A

b

(A

b

1 – 42

) relative to

[36,95,114], but some laboratories could not detect it

the shorter 40-amino acid form (reviewed in Ref. [38]).

[90,1]. Nevertheless some reports demonstrating

in-Increases in A

b

1 – 42

have also been detected in transgenic

volvement of APP in neuronal development,

synap-mice expressing FAD mutations of both APP and PS

togenesis, and synaptic plasticity [62,68,69,86,70,85] did

(reviewed in Ref. [38]). A

b

1 – 42

is the major component of

not restrict the observed function to secreted APP, raising

brain amyloid deposits in AD. Consequently, a leading

the possibility that some aspects of synaptic plasticity are

hypothesis for the etiology of AD is that increased A

b

1 – 42

mediated by cell-associated APP. Indeed, it has now been

is a shared molecular correlate of FAD mutations which

demonstrated directly that a percentage of APP is found on

may also be operative in ‘sporadic’ AD. Increases in

the cell surface in neurons [47,99,83]. Cell-surface APP

A

b

1 – 42

have not been shown directly in human AD brain

possesses a neurite-promoting activity that is distinct from

homogenates, although it is clear that amyloid plaques

that of the secreted APP [85], co-localizes with adhesion

contain a disproportionate amount of A

b

1 – 42

. Furthermore,

plaque components [99,114], and participates in synaptic

analyses of levels of this peptide in the plasma and

vesicle recycling [63], suggesting that a percentage of APP

cerebrospinal fluid (CSF) of AD patients have revealed no

may function as a cell surface receptor, transducing signals

differences between AD patients and controls in the

from the extracellular matrix to the interior of the cell.

plasma [43], and a reduction of A

b

1 – 42

in the CSF of AD

The growth cone G protein G

o

[75], the presumptive

patients relative to controls [67,43]. However, increased

adaptor proteins Fe65 and X11 (reviewed in Ref. [89]),

release of A

b

( 1 – 42 )

from fibroblasts of AD patients with

and APP-BP1 [10], a protein involved in cell cycle

presenilin mutations, as well as increased levels of

regulation [9], have been reported to interact with the

A

b

( 1 – 42 )

in their plasma, have been demonstrated [94].

C-terminus of APP (Fig. 1), presumably to initiate

intracel-The

b

-secretase cleavage product of APP, C100, is

lular signaling. While the functions of Fe65 and X11 are

increased in cell lines expressing the Swedish FAD

not known, Fe65 has the characteristics of adaptor

pro-mutation of APP [12–14,8], but not in cell lines expressing

teins, which are thought to link signal transduction events

the London V642 mutation of APP [102]. Because neurons

emanating from plasma membrane receptors to

intracellu-process APP differently from cell lines, we expressed all

lar molecules, by forming complexes of these proteins.

known FAD mutants of APP in primary neurons in culture,

Therefore, one could envision APP being part of a G

o

(4)

Fig. 1. Schematic depicting the structural and functional domains of the amyloid precursor protein.

defined in greatest detail is G . Nishimoto and colleagues

o

respective roles of G , APP-BP1, Fe65 and X11, and

o

657 676

have demonstrated that the His

–Lys

domain of APP-

UV-DDB in the normal function of APP, to test the

695 activates the heterotrimeric GTP-binding protein G in

o

hypothesis that progressive dysfunction of these roles

a GTP S-inhibitable manner [75,52]. Their demonstration

g

occurs in AD.

that an antibody to the extracellular domain of APP

(22C11) that acts as a ligand mimetic [79] causes

activa-tion of G , argues that APP may be a G protein-coupled

o

4. Apoptosis in Alzheimer disease

receptor. As noted above, the ‘London’ mutation of APP,

V642I, causes DNA fragmentation when expressed in a

The notion that a form of cell suicide called apoptosis

neuronal cell line [113]. Notably, expression of V642I

participates in the neuropathology of AD was raised by Su

657 676

APP deleted for residues His

–Lys

in these cells did

et al. [100], when they reported evidence for DNA

not cause DNA fragmentation. Pertussis toxin (PTX), an

fragmentation in neurons in AD brain. Although other

inhibitor of G and G , blocked the DNA fragmentation

o i

groups have also detected this feature of apoptosis in AD

caused by V642I, as did co-transfection of V642I APP and

brain, many in the field have been skeptical of the idea that

a cDNA encoding a dominant negative mutant of G

a

o

, but

the neurons that die in AD undergo apoptosis, partly

not with a cDNA encoding a dominant negative mutant of

because DNA fragmentation can also be caused by

oxida-G

a

i

. Inhibition of A

b

1 – 42

production from the V642I APP

tive damage [106] or by postmortem autolysis [98].

by mutating the

g

-secretase cleavage site did not have any

However, a report from the laboratory of Mark Mattson

effect on the DNA fragmentation caused by V642I.

[34] revived interest in the possibility that apoptosis is

These data suggest that G

o

mediates the DNA frag-

operative in AD. These investigators found that levels of a

mentation caused by the V642 mutants of APP; and

marker of apoptosis, Par-4 (prostate apoptosis response-4)

indeed, a subsequent paper from Nishimoto’s group re-

protein, are increased 15–20-fold over control in

vulner-vealed that the DNA fragmentation was mediated by the

able neurons in AD brain. They also showed that Par-4

bg

complex of G

o

[29]. A

b

does not appear to play a

expression is increased in cultured neurons undergoing

causative role in inducing DNA fragmentation in this

apoptosis, and that inhibition of Par-4 expression in these

experimental paradigm. The data support the notion that

neurons blocks apoptosis.

(5)

differentially affected. PS-1 is reported to be expressed

primarily in CNS neurons in the brain, suggesting that this

protein may perform a neuron-specific function [20]. In

fact, in AD, neurons that express PS-1 antigen are less

vulnerable to the disease than are neurons that do not

express it [30], and inhibition of PS-1 expression results in

apoptosis [88], suggesting a protective role for this protein.

Although the precise role of PSs in regulation of apoptosis

in the neuron is still unclear, the evidence that they do play

a role in this pathway is strong. These data implicate both

APP and PSs in the control of apoptotic death in the brain,

and it is not unreasonable to suppose that FAD mutations

in these genes may cause dysfunction in this pathway.

It has been noted [82] that since the apoptotic process

proceeds to completion within 16–24 h, the extent of

apoptosis reported in AD brain would predict a complete

loss of neurons within a very brief period of time. Clearly,

this does not happen in AD. Cotman has suggested [15]

that the induction of compensatory responses to apoptosis

in the AD brain protects the neurons from terminal

apoptosis, and that a dynamic competition between cell

death processes and compensatory responses exists in AD

brain.

5. Cell cycle abnormalities in Alzheimer’s disease

The implication of apoptosis in AD etiology is

con-Fig. 2. The yeast two-hybrid reporter assay reveals that APP-BP1

sistent with the numerous findings of cell cycle

abnor-interacts with hUba3. Top: schematic diagram showing the deletion and

malities in AD. Apoptosis and the cell cycle are closely

point mutants used for the assay. Below: a table indicating the strength of

tied together, and the reexpression of cell cycle markers

each interaction, based on length of time for the X-gal substrate to turn

has been linked with the occurrence of certain types of

blue.

neuronal cell death [40,39,22]. One interpretation of these

findings [56] is that a neuron is committed to the

perma-APP, V642I, causes DNA fragmentation when expressed in

nent cessation of cell division, so if for any reason it is

a neuronal cell line [113]. Luo et al. [61] showed that the

forced to reenter the cell cycle after this commitment, it

same mutation, as well as two additional FAD APP

dies. Notably, ectopic expression of cell cycle proteins and

mutations, induced apoptosis in differentiated PC12 cells.

their associated kinases in AD brain have been reported

Barnes et al. [2] reported that levels of APP are increased

[84,59,108,109]. More recently, Busser et al. [7] found

in motoneurons dying of apoptosis, and that APP is

abnormal appearance of cell cycle markers in regions of

cleaved by caspase-3, a caspase activated in apoptotic

AD brain where cell death is extensive, and Chow et al.

motoneurons. Interestingly, we [6] and others [77] have

[11] found increases in expression of genes encoding cell

shown that overexpression of wild-type APP causes apo-

cycle proteins in single neurons in late-stage relative to

ptotic death of neurons, although to a lesser degree than

early-stage AD brain. The phosphoepitope S214 of the

does expression of FAD mutants of APP.

microtubule associated protein tau, that appears in the

Approximately half of inherited AD cases are caused by

neurofibrillary tangles in AD, is a prominent

phosphoryla-mutations in the presenilin genes PS1 and PS2. It has been

tion site in metaphase but not in interphase of dividing

reported that overexpression of these genes in transfected

cells expressing tau [44], supporting the view that

reactiva-cell lines can cause apoptosis [45] or result in an increased

tion of the cell cycle machinery may be involved in tau

susceptibility to apoptosis [112,32–34,16]. On the other

hyperphosphorylation in AD brain. The possibility that

hand, we have found [6] that expression of PS1 in primary

phosphorylation-dependent events occurring during the cell

neurons does not cause or enhance apoptosis; rather, it

cycle affect the normal function of APP is suggested by

protects neurons against experimentally-induced apoptosis.

the finding that regulation of the phosphorylation and

Thus, the ability of PS-1 to induce apoptosis appears to be

metabolism of this protein occurs in a cell-cycle dependent

cell type specific; and this may have important implica-

manner [103,78].

(6)

by APP may be one cause of the reactivation of cell cycle

appear to have much in common with those of ubiquitin,

proteins in AD brain. In particular, we have isolated a

but the Ubls have novel regulatory functions not

necessari-binding protein for APP, termed APP-BP1 [10], and have

ly linked to proteolysis. APP-BP1 is a member of one of

shown that it is a cell cycle protein that normally regulates

these pathways. APP-BP1 is homologous to the ubiquitin

negatively the progression of cells into the S phase and

activating enzyme E1, but lacks the catalytic site. It has

regulates positively their progression into mitosis [9]. Over

been found, by our lab [9], and others, that APP-BP1 acts

the past few years, it has emerged that eukaryotes express

in concert with a second protein that possesses an E1-like

a set of ubiquitin-like proteins (Ubls) that are significantly

catalytic site, so that the two-molecule complex behaves

diverged from ubiquitin itself yet are also ligated to other

like E1, except that this complex activates the

ubiquitin-proteins [35,46]. The reactions involving these variants

like protein NEDD8 rather than ubiquitin itself:

Fig. 3. APP-BP1 rescues the ts41 cell phenotype at the nonpermissive temperature. (A) Time course of viability of ts41 cells transfected with the vector alone or with the APP-BP1 construct at the nonpermissive temperature (408C). Data for the vector-transfected cells at the permissive temperature (348C) are shown for comparison. Counts were performed at 32, 64, and 80 h except for the vector-transfected cells at the permissive temperature, which were counted at 32 and 64 h only. Note that cells transfected with a vector expressing human wild type APP-BP1 maintained a rate of growth at 408C that was similar (albeit slightly shifted to the right) to that of cells transfected with the vector alone and maintained at 348C. Vector-transfected cells that were shifted to the nonpermissive temperature showed a decreased mitotic index and eventually died. (B) Quantification of viability of wild type and mutant APP-BP1-transfected ts41 cells grown at 408C for 81 h. Note that the d401–479 deletion mutant of APP-BP1, which does not interact with hUba3, is unable to rescue the ts41 phenotype (Scheffe post hoc t tests;[[or ** indicates P,0.01, and * indicates P,0.05;[comparison with pcDNA3; *

(7)

temperature, after which they were neutralized in 0.1 M

E1-like(Ub activating) Ubiquitin-like

sodium borate buffer (pH 8.2) for 10 min. After two 5-min

APP-BP11hUBA3 activates NEDD8→targets (e.g., cullin-4A)

washes the neurons were incubated in 5% horse serum in

NEDD8 then forms a thiol ester linkage with hUbc12, a

PBS plus 0.1% Triton-X 100 (blocking buffer) for 1 h at

human protein that has a function parallel to that of the

room temperature, after which they were incubated

over-ubiquitin-conjugating enzyme, prior to its modification of

night in a 1:1000 dilution of the anti-BrdU monoclonal

target proteins such as cullin-4A. The functions of this

antibody BU-33 (Sigma) in blocking buffer. After three

pathway (reviewed in Ref. [41]) are still unclear; but it

5-min washes in blocking buffer, the neurons were

incu-does in some cases lead to modification of ubiquitin-like

bated for 1.5 h at room temperature in a 1:200 dilution of

proteins that are linked to cell cycle regulation [46,51].

biotinylated secondary anti-mouse antibody (Vector

Lab-We have shown that APP-BP1 plays a role in the cell

oratories) in blocking buffer, washed, and visualized using

cycle [9]. APP-BP1 co-immunoprecipitates with hUba3

DAB. Stained cells were visualized under a 40

3

objective.

from mammalian cells and binds to a region between

Ten random fields were counted from each condition, and

amino acids 443 and 479 in hUba3 (Fig. 2). Wild type

the data were expressed as the percentage ratio of stained

APP-BP1 rescues the cell cycle S–M checkpoint defect in

cells to total cells.

ts41 hamster cells [9] and this rescue is dependent on the

The results (Fig. 7) indicated that expression of FAD

binding of APP-BP1 to hUba3 (Fig. 3). Dominant negative

mutants of APP in cortical neurons caused a significant

mutants of hUba3 and Ubc12 prevent the rescue. Notably,

increase over controls in the number of cells undergoing

overexpression of APP-BP1 in primary neurons (Fig. 4)

DNA synthesis. Overexpression of wild type APP in the

causes apoptosis by a pathway that also involves hUba3

cultures also caused an increase in DNA synthesis,

al-and hUbc12 (Fig. 5). We hypothesize that overexpression

though to a lesser extent.

of APP-BP1 pushes neurons into the S phase of the cell

cycle (Fig. 6), causing DNA synthesis and, for example,

aberrant expression of mitotic cdc2 / cyclin B1 kinase, as is

6. Conclusions

observed in degenerating neurons in AD brain [109].

To test this hypothesis, we expressed FAD mutants of

We propose that mechanisms other than accumulation of

APP in neurons using HSV-1 vectors. Consistent with our

A

b

may be the cause of AD neurodegeneration and

prediction, neurons expressing FAD mutants of APP

cognitive impairment. In particular, we suggest that the

showed increased expression of APP-BP1 relative to

disease may be a consequence of disruption of function of

controls (unpublished data of Y. Chen and R. Neve). We

APP. Convincing data have accumulated that support that

next evaluated DNA synthesis in the infected cultures. Ten

idea that APP is a functional receptor linked to a G

o

uM 5-bromo-2

9

-deoxyuridine BrdU (Sigma) was added to

signaling cascade. Apoptosis is induced in neuronal cells

the primary cortical cultures at the time of infection.

expressing FAD mutants of APP, and this phenotype is

Sixteen hours after infection the cells were fixed in 4%

independent of the production of A

b

1 – 42

. Expression of

paraformaldehyde in 100 mM phosphate buffer pH 7.4 for

FAD mutants of APP in neurons causes not only apoptosis

20 min. The cultures were washed twice for 5 min each in

but also intracellular accumulation of carboxyl terminal

PBS, and were treated with 2 N HCl for 10 min at room

fragments of APP. These data suggest that the FAD

(8)

Fig. 5. Schematic showing the ubiquitin-like pathway in which APP-BP1 is involved. The family of genes known as the cullins are the known targets for this pathway. APP-BP1 negatively regulates the G1 to S transition and positive regulates the S to M transition via this pathway. In dividing cells, APP-BP1 promotes cell division, but in primary neurons, it causes apoptosis.

(9)

AG12954.

References

[1] B. Allinquant, K.L. Moya, C. Bouillot, A. Prochiantz, Amyloid precursor protein in cortical neurons: coexistence of two pools differentially distributed in axons and dendrites and association with cytoskeleton, J. Neurosci. 14 (1994) 6842–6854.

[2] N.Y. Barnes, L. Li, K. Yoshikawa, L.M. Schwartz, R.W. Oppenheim, C.E. Milligan, Increased production of amyloid precursor protein provides a substrate for caspase-3 in dying motoneurons, J. Neuro-sci. 18 (1998) 5869–5880.

[3] J. Berger-Sweeney, D.L. McPhie, J.A. Arters, J. Greenan, M.L. Oster-Granite, R.L. Neve, Impairment in spatial learning accom-panied by neurodegeneration in mice transgenic for the carboxyl-terminus of the amyloid precursor protein, Mol. Brain Res. 66 (1999) 150–162.

[4] S.L. Borowicz, L.A. Dokas, Association of the amyloid precursor protein, B-50 (GAP-43), and Go in neuronal membranes, Soc. Neurosci. Abst. 21 (GAP-43) (1995) 207.

Fig. 6. Diagram depicting the stages of the cell cycle at which APP-BP1

[5] E. Brouillet, A. Trembleau, D. Galanaud, M. Volovitch, C. Bouillot, act. Note that the cdc / cyclin B1 kinase acts downstream of APP-BP1; if

C. Valenza, A. Prochiantz, B. Allinquant, The amyloid precursor dysfunction of APP-BP1 occurs in AD brain, this may explain the

protein interacts with Go heterotrimeric protein within a cell aberrant expression of cdc / cyclin B1 in neurons in AD.

compartment specialized in signal transduction, J. Neurosci. 19 (1999) 1717–1727.

[6] S. Bursztajn, R. DeSouza, D.L. McPhie, S.A. Berman, J. Shioi, N.K. Robakis, R.L. Neve, Overexpression in neurons of human

in the neuron, and that these abnormalities constitute one

presenilin-1 or a presenilin-1 familial Alzheimer disease mutant

mechanism of neurodegeneration in AD.

does not enhance apoptosis, J. Neurosci. 18 (1998) 9790–9799.

[7] J. Busser, D.S. Geldmacher, K. Herrup, Ectopic cell cycle proteins predict the sites of neuronal cell death in Alzheimer’s disease brain, J. Neurosci. 18 (1998) 2801–2807.

Acknowledgements

[8] X.D. Cai, T.E. Golde, S.G. Younkin, Release of excess amyloidb protein from a mutant amyloid b protein precursor, Science 259

We thank Dawn Morrissey for assistance in preparation

(1993) 5140516.

of the manuscript. The work from our laboratory that is

[9] Y. Chen, D.L. McPhie, J. Hirschberg, R.L. Neve, The amyloid

described in this review was funded by NIH grant

precursor protein-binding protein APP-BP1 drives the cell cycle

(10)

through the S–M checkpoint and causes apoptosis in neurons, J. [28] O. Ghiribi, L. Lapierre, M. Girard, M. Ohayon, J. Nalbantoglu, G. Biol. Chem. 275 (2000) 8929–8935. Massicotte, Hypoxia-induced loss of synaptic transmission is ex-acerbated in hippocampal slices of transgenic mice expressing C-[10] N. Chow, J.R. Korenberg, X.-N. Chen, R.L. Neve, APP-BP1, a

terminal fragments of Alzheimer amyloid precursor protein, Hip-novel protein that binds to the carboxyl-terminal region of the

pocampus 9 (1999) 201–205. amyloid precursor protein, J. Biol. Chem. 271 (1996) 11339–11346.

[29] U. Giambarella, T. Yamatsuji, T. Okamoto, T. Matsui, T. Ikezu, Y. [11] N. Chow, C. Cox, L.M. Callahan, J.M. Weimer, L. Guo, P.D.

Murayama, M.A. Levine, A. Katz, N. Gautam, I. Nishimoto, G Coleman, Expression profiles of multiple genes in single neurons of

protein bg complex-mediated apoptosis by familial Alzheimer’s Alzheimer’s disease, Proc. Natl. Acad. Sci. USA 95 (1998) 9620–

disease mutant of APP, EMBO J. 16 (1997) 4897–4907. 9625.

[30] P. Giannakopoulos, C. Bouras, E. Kovari, J. Shioi, N. Tezapsidis, [12] M. Citron, T. Oltersdorf, C. Haas, L. McConlogue, A.Y. Hung, P.

P.R. Hof, N.K. Robakis, Presenilin-1-immunoreactive neurons are Seubert, C. Vigo-Pelfrey, I. Lieberburg, D.J. Selkoe, Mutation of the

preserved in late-onset Alzheimer’s disease, Am. J. Pathol. 150 b-amyloid precursor protein in familial Alzheimer’s disease

in-(1997) 429–436. creasesb-protein production, Nature 360 (1992) 672–674.

[31] T. Golde, S. Estus, S.G. Younkin, Processing of the amyloid protein [13] M. Citron, D.B. Teplow, D.J. Selkoe, Generation of amyloid b

precursor to potentially amyloidogenic derivatives, Science 255 protein from its precursor is sequence specific, Neuron 14 (1995)

(1992) 728–730. 661–670.

[32] Q. Guo, K. Furukawa, B.L. Sopher, D.G. Pham, J. Xie, N. [14] M. Citron, T.S. Diehl, A. Capell, C. Haass, D.B. Teplow, D.J.

Robinson, G.M. Martin, M.P. Mattson, Alzheimer’s PS-1 mutation Selkoe, Inhibition of amyloidb-protein production in neural cells by

perturbs calcium homeostasis and sensitizes PC12 cells to death the serine protease inhibitor AEBSF, Neuron 17 (1996) 1–9.

induced by amyloid beta-peptide, Neuroreport 8 (1996) 379–383. [15] C.W. Cotman, Apoptosis decision cascades and neuronal

degenera-[33] Q. Guo, B.L. Sopher, K. Furukawa, D.G. Pham, N. Robinson, G.M. tion in Alzheimer’s disease, Neurobiol. Aging 19 (1998) S29–S32.

Martin, M. P Mattson, Alzheimer’s presenilin mutation sensitizes [16] G. Deng, C.J. Pike, C.W. Cotman, Alzheimer-associated presenilin-2

neural cells to apoptosis induced by trophic factor withdrawal and confers increased sensitivity to apoptosis in PC12 cells, FEBS Lett.

amyloid beta peptide: involvement of calcium and oxyradicals, J. 397 (1996) 50–54.

Neurosci. 17 (1997) 4212–4222. [17] T. Dyrks, A. Weidemann, G. Multhaup, J.M. Salbaum, H.G.

[34] Q. Guo, W. Fu, J. Xie, H. Luo, S.F. Sells, J.W. Geddes, V. Bondada, Lemaire, J. Kang, B. Muller-Hill, C.L. Masters, K. Beyreuther,

V.M. Rangnekar, M.P. Mattson, Par-4 is a mediator of neuronal Identification, transmembrane orientation and biogenesis of the

degeneration associated with the pathogenesis of Alzheimer disease, amyloid A4 precursor of Alzheimer’s disease, EMBO J. 7 (1988)

Nature 4 (1998) 957–962. 949–957.

[35] A.L. Haas, T.J. Siepmann, Pathways of ubiquitin conjugation, [18] T. Dyrks, E. Dyrks, T. Hartmann, C. Masters, K. Beyreuther,

FASEB J. 11 (1997) 1257–1268. Amyloidogenicity of bA4 and bA4-bearing amyloid protein

pre-[36] C. Haass, E. Koo, A. Mellon, A. Jung, D. Selkoe, Targeting of cursor fragments by metal-catalyzed oxidation, J. Biol.Chem. 267

cell-surface b-amyloid precursor protein to lysosomes: alternative (1992) 18210–18217.

processing into amyloid-bearing fragments, Nature 357 (1992) 500– [19] T. Dyrks, E. Dyrks, U. Monning, B. Urmoneit, J. Turner, K.

503. Beyreuther, Generation ofbA4 from the amyloid protein precursor

[37] C. Haass, A. Hung, M. Schlossmacher, D. Teplow, D. Selkoe, and fragments thereof, FEBS Lett. 335 (1993) 89–93.

b-amyloid peptide and a 3 kDa fragment are derived by distinct [20] G.A. Elder, N. Tezapsidis, J. Carter, J. Shioi, C. Bouras, H.C. Li,

cellular mechanisms, J. Biol. Chem. 268 (1993) 3021–3024. J.M. Johnson, S. Efthimiopoulos, V.L. Friedrich Jr., N.K. Robakis,

[38] J. Hardy, The Alzheimer family of diseases: many etiologies, one Identification and neuron specific expression of the PS-1 / presenilin

pathogenesis?, Proc. Natl. Acad. Sci. USA 18 (1997) 2095–2097. I protein in human and rodent brains, J. Neurosci. Res. 45 (1996)

308–320. [39] N. Heintz, Cell death and the cell cycle: a relationship between

transformation and neurodegeneration?, Trends Biochem Sci. 18 [21] S. Estus, T. Golde, T. Kunishita, D. Blades, D. Lowery, M. Eisen,

(1993) 157–159. M. Usiak, X. Qu, T. Tabira, B. Greenberg, S. Younkin, Potentially

amyloidogenic, carboxy-terminal derivatives of the amyloid protein [40] K. Herrup, J.C. Busser, The induction of multiple cell cycle events precursor, Science 255 (1992) 726–728. precedes target-related neuronal death, Development 121 (1995)

2385–2395. [22] R. Freeman, S. Estus, E. Johnson, Analysis of cell cycle-related

gene expression in postmitotic neurons: selection induction of cyclin [41] M. Hochstrasser, There’s the Rub: a novel ubiquitin-like modi-D1 during programmed cell death, Neuron 12 (1994) 343–355. fication linked to cell cycle regulation, Genes Dev. 12 (1998)

901–907. [23] K. Fukuchi, B. Sopher, G.M. Martin, Neurotoxicity of b-amyloid,

Nature 361 (1993) 122. [42] I. Hussain, D. Powell, D.R. Howlett, D.G. Tew, T.D. Meek, C.

Chapman, I.S. Gloger, K.E. Murphy, C.D. Southan, D.M. Ryan, T.S. [24] K. Fukuchi, B. Sopher, C.E. Furlong, A.C. Smith, N.T. Dang, G.M.

Smith, D.L. Simmons, F.S. Walsh, C. Dingwall, G. Christie, Martin, Selective neurotoxicity of COOH-terminal fragments of the

Identification of a novel aspartic protease (Asp2) as b-secretase, b-amyloid precursor protein, Neurosci. Lett. 154 (1993) 145–148.

Mol. Cell. Neurosci. 14 (1999) 419–427. [25] K. Fukuchi, D.D. Kunkel, P.A. Schwartzkroin, K. Kamino, C.E.

[43] N. Ida, T. Hartmann, J. Pantel, J. Schroder, R. Zerfass, H. Forstl, R. Ogburn, C.E. Furlong, G.M. Martin, Overexpression of a C-terminal

Sandbrink, C.L. Masters, K. Beyreuther, Analysis of heterogeneous portion of the b-amyloid precursor protein in mouse brains by

bA4 peptides in human cerebrospinal fluid and blood by a newly transplantation of transformed neuronal cells, Exptl. Neurol. 127

developed sensitive western blot assay, J. Biol. Chem. 271 (1996) (1994) 253–264.

22908–22914. [26] D. Games, D. Adams, R. Alessandrini, R. Barbour, P. Berthelette, C.

[44] S. Illenberger, Q. Zheng-Fischhofer, U. Preuss, K. Stamer, K. Blackwell, T. Carr, J. Clemens, T. Donaldson, F. Gillespie,

Al-Baumann, B. Trinczek, J. Biernat, R. Godemann, E.M. Mandelkow, zheimer-type neuropathology in transgenic mice overexpressing

E. Mandelkow, The endogenous and cell cycle-dependent phos-V717Fb-amyloid precursor protein, Nature 373 (1995) 523–527.

phorylation of tau protein in living cells: Implications for Alzheim-[27] J.E. Gardella, G.A. Gorgone, L. Candela, J. Ghiso, E.M. Castano, B.

er’s disease, Mol. Biol. Cell 9 (1998) 1495–1512. Frangione, P.D. Gorevic, High-level expression and in vitro

muta-[45] S. Janicki, M.J. Monteiro, Increased apoptosis arising from in-genesis of a fibrillogenic 109-amino-acid C-terminal fragment of

creased expression of Alzheimer’s disease associated presenilin-2 Alzheimer’s disease amyloid precursor protein, Biochem. J. 294

(11)

[46] P.R. Johnson, M. Hochstrasser, SUMO-1: ubiquitin gains weight, ing of cell-surface b-amyloid precursor protein: Evidence that a Trends Cell Biol. 7 (1997) 408–413. sorting intermediate participates in synaptic vesicle recycling, J.

Neurosci. 17 (1997) 140–151. [47] S.S. Jung, J. Nalbantoglu, N.R. Cashman, Alzheimer’sb-amyloid

precursor protein is expressed on the surface of immediately ex vivo [64] K. Maruyama, K. Terakado, M. Usami, K. Yoshikawa, Formation of brain cells: a flow cytometric study, J. Neurosci. Res. 46 (1996) amyloid-like fibrils in COS cells overexpressing part of the

Al-336–348. zheimer amyloid protein precursor, Nature 347 (1990) 566–569.

[48] A. Kammesheidt, F.M. Boyce, A.F. Spanoyannis, B.J. Cummings, [65] M. Mattson, B. Cheng, A. Culwell, F. Esch, I. Lieberburg, R. Rydel, M. Ortegon, C.W. Cotman, J. Vaught, R.L. Neve, Amyloid deposi- Evidence for excitoprotective and intraneuronal calcium-regulating tion and neuronal pathology in transgenic mice expressing the roles for secreted forms of theb-amyloid precursor protein, Neuron carboxyterminal fragment of the Alzheimer amyloid precursor in the 10 (1993) 243–254.

brain, Proc. Natl. Acad. Sci. USA 89 (1992) 10857–10861. [66] D.L. McPhie, R.K.K. Lee, C.B. Eckman, D.H. Olstein, S.P. Durham, [49] J. Kang, H.G. Lemaire, A. Unterbeck, J.M. Salbaum, C.L. Masters, D. Yager, S.G. Younkin, R.J. Wurtman, R.L. Neve, Neuronal K.H. Grzeschik, K. Beyreuther, B. Muller-Hill, The precursor of expression of b-amyloid precursor protein Alzheimer mutations Alzheimer’s disease amyloid A4 protein resembles a cell-surface causes intracellular accumulation of a C-terminal fragment con-receptor, Nature 325 (1987) 733–736. taining both the amyloidband cytoplasmic domains, J. Biol. Chem.

272 (1997) 24743–24746. [50] S.H. Kim, Y.H. Suh, Neurotoxicity of a carboxy-terminal fragment

of the Alzheimer’s amyloid precursor protein, J. Neurochem. 67 [67] R. Motter, C. Vigo-Pelfrey, D. Kholodenko, R. Barbour, K.

Johnson-(1996) 1172–1182. Wood, D. Galasko, L. Chang, B. Miller, C. Clark, R. Green,

Reduction of b-amyloid peptide in the cerebrospinal fluid of

[51] E.T. Kipreos, L.E. Lander, J.P. Wing, W.W. He, E.M. Hedgecock, 42

patients with Alzheimer’s disease, Ann. Neurol. 38 (1995) 643–648. Cul-1 is required for cell cycle exit in C. elegans and identifies a

novel gene family, Cell 85 (1996) 829–839. [68] K.L. Moya, L.I. Benowitz, G.E. Schneider, B. Allinquant, The amyloid precursor protein is developmentally regulated and corre-[52] J. Lang, I. Nishimoto, T. Okamoto, R. Regazzi, C. Kiraly, U. Weller,

lated with synaptogenesis, Dev. Biol. 171 (1994) 597–603. C.B. Wollheim, Direct control of exocytosis by receptor-mediated

activation of the heterotrimeric GTPases G and Gi o or by the [69] L. Mucke, E. Masliah, W.B. Johnson, M.D. Ruppe, M. Alford, E.M. expression of their active G alpha subunits, EMBO J. 14 (1995) Rockenstein, S. Forss-Petter, M. Pietropaolo, M. Mallory, C.A.

3635–3644. Abraham, Synaptotrophic effects of human amyloid b protein

precursors in the cortex of transgenic mice, Brain Res. 666 (1994) [53] A.C. LeBlanc, Increased production of 4 kDa amyloidbpeptide in

151–167. serum deprived human primary neuron cultures: possible

in-¨ ¨

volvement of apoptosis, J. Neurosci. 15 (1995) 7837–7846. [70] U. Muller, N. Cristina, A.W. Li, D.P. Wolfer, H.P. Lipp, T. Rulick, S. Brandner, A. Aguzzi, C. Weissmann, Behavioral and anatomical [54] A.C. LeBlanc, R. Xue, P. Gambetti, APP metabolism in primary cell

deficits in mice homozygous for a modified b-amyloid precursor cultures of neurons, astrocytes and microglia, J. Neurochem. 66

protein gene, Cell 79 (1994) 755–765. (1996) 2300–2310.

[71] J. Nalbantoglu, G. Tirado-Santiago, A. Lahsaini, J. Poirier, O. [55] A.C. LeBlanc, M. Papadopoulos, C. Belair, W. Chu, M. Crosato, J.

Goncalves, G. Verge, F. Momoli, S.A. Weiner, G. Massicotte, J.P. Powell, C. Goodyer, Amyloid precursor protein metabolism in

Julien, M.L. Shapiro, Impaired learning and LTP in mice expressing human neurons, astrocytes and microglia, J. Neurochem. 68 (1997)

the carboxy terminus of the Alzheimer amyloid precursor protein, 1183–1190.

Nature 387 (1997) 500–505. [56] E.Y.-H.P. Lee, C.Y. Chang, N. Hu, Y.C.J. Wang, C.C. Lai, K. Herrup,

[72] R.L. Neve, A. Kammesheidt, C.F. Hohmann, Brain transplants of W.H. Lee, A. Bradley, Mice deficient for Rb are nonviable and show

cells expressing the carboxyterminal fragment of the Alzheimer defects in neurogenesis and haematopoiesis, Nature 359 (1992)

amyloid protein precursor cause specific neuropathology in vivo, 288–294.

Proc. Natl. Acad. Sci. USA 89 (1992) 3448–3452. [57] Q.X. Li, C. Maynard, R. Cappai, C.A. McLean, R.A. Cherny, T.

[73] R.L. Neve, M.R. Kozlowski, The carboxyl-terminal 100 amino acids Lynch, J.G. Culvenor, J. Trevaskis, J.E. Tanner, K.A. Bailey, C.

of the b-amyloid protein precursor: Role in Alzheimer disease Czech, A.I. Bush, K. Benreuther, C.L. Masters, Intracellular

ac-neurodegeneration, Dev. Brain. Dysfunction. 8 (1995) 13–24. cumulation of detergent-soluble amyloidogenic Ab fragment of

Alzheimer’s disease precursor protein in the hippocampus of aged [74] R.L. Neve, N.K. Robakis, Alzheimer disease: A re-examination of transgenic mice, J. Neurochem. 72 (1999) 2479–2487. the amyloid hypothesis, Trends Neurosci. 21 (1998) 15–19. [58] Y.M. Li, M. Xu, M.T. Lai, Q. Huang, J.L. Castro, J. DiMuzio- [75] I. Nishimoto, T. Okamoto, Y. Matsuura, T. Okamoto, Y. Murayama,

Mower, T. Harrison, C. Lellis, A. Nadin, J.G. Neduvelil, R.B. E. Ogata, Alzheimer amyloid protein precursor complexes with Register, M.K. Sardana, M.S. Shearman, A.L. Smith, X.P. Shi, K.C. brain GTP-binding protein G , Nature 362 (1993) 75–79.o Yin, J.A. Shafer, S.J. Gardell, Photoactivatedg-secretase inhibitors [76] I. Nishimoto, A new paradigm for neurotoxicity by FAD mutants of directed to the active site covalently label presenilin 1, Science 405 bAPP: a signaling abnormality, Neurobiol. Aging 19 (1998) S33–

(2000) 689–694. S38.

[59] W.K. Liu, R. Williams, F. Hall, D. Dickson, S.H. Yen, Detection of a [77] I. Nishimura, T. Uetsuki, S.U. Dani, Y. Ohsawa, I. Saito, H. cdc2-related kinase associated with Alzheimer paired helical fila- Okamura, Y. Uchiyama, K. Yoshikawa, Degeneration in vivo of rat ments, Am. J. Pathol. 146 (1995) 228–238. hippocampal neurons by wild-type Alzheimer amyloid precursor [60] D.C. Lu, S. Rabizadeh, S. Chandra, R.F. Shayya, L.M. Ellerby, X. protein overexpressed by adenovirus-mediated gene transfer, J.

Ye, G.S. Salvesen, E.H. Koo, D.E. Bredesen, A second cytotoxic Neurosci. 18 (1998) 2387–2398.

proteolytic peptide derived from amyloid b-protein precursor, [78] M. Oishi, A.C. Nairn, A.J. Czernik, G.S. Lim, T. Isohara, S.E.

Nature Med. 6 (2000) 397–404. Gandy, P. Greengard, T. Suzuki, The cytoplasmic domain of

[61] J.J. Luo, W. Wallace, T. Riccioni, D.K. Ingram, G.S. Roth, J.W. Alzheimer’s amyloid precursor protein is phosphorylated at thr654, Kusiak, Death of PC12 cells and hippocampal neurons induced by ser655, and thr668 in adult rat brain and cultured cells, Mol. Med. 3 adenoviral-mediated FAD human amyloid precursor protein gene (1997) 111–123.

expression, J. Neurosci. Res. 55 (1999) 629–642. [79] T. Okamoto, S. Takeda, Y. Murayama, E. Ogata, I. Nishimoto, [62] L. Luo, T. Tully, K. White, Human amyloid precursor protein Ligand-dependent G protein coupling function of amyloid

trans-ameliorates behavioral deficit of flies deleted for Appl gene, Neuron membrane precursor, J. Biol. Chem. 270 (1995) 4205–4208.

9 (1992) 595–605. [80] M.L. Oster-Granite, D.L. McPhie, J. Greenan, R.L. Neve,

(12)

the carboxyl-terminus of the amyloid precursor protein, J. Neurosci. [96] S. Sinha, J.P. Anderson, R. Barbour, G.S. Basi, R. Caccavello, D. Davis, M. Doah, H.F. Dovey, N. Frigon, J. Hong, K. Jacobson-16 (1996) 6732–6741.

Croak, N. Jewett, P. Keim, J. Knops, I. Lieberburg, M. Power, H. [81] M. Palmert, S. Siedlak, M. Podlisny, B. Greenberg, E. Shelton, H.

Tan, G. Tatsuno, J. Tung, D. Schenk, P. Seuberg, S.M. Suomensaari, Chan, M. Usiak, D. Selkoe, G. Perry, S. Younkin, Soluble

deriva-S. Wang, D. Walker, J. Zhao, L. McConlogue, V. John, Purification tives of theb-amyloid protein precursor of Alzheimer’s disease are

and cloning of amyloid precursor protein b-secretase from human labeled by antisera to the b-amyloid protein, Biochem. Biophys.

brain, Nature 402 (1999) 537–540. Res. Commun. 165 (1989) 7533–7539.

[97] B.L. Sopher, K. Fukuchi, A.C. Smith, K.A. Leppig, C.E. Furlong, [82] G. Perry, A. Nunomura, P. Lucessen, H. Lassmann, M.A. Smith,

G.M. Martin, Cytotoxicity mediated by conditional expression of a Apoptosis and Alzheimer’s disease, Science 282 (1998) 1268–1269.

carboxyl-terminal derivative of the b-amyloid precursor protein, [83] R.G. Perez, H. Zheng, L.H.T. Van der Ploeg, E.H. Koo, The

Mol. Brain Res. 26 (1994) 207–217. b-amyloid precursor protein of Alzheimer’s disease enhances

neu-[98] C. Stadelmann, W. Bruck, C. Bancher, K. Jellinger, H. Lassmann, ron viability and modulates neuronal polarity, J. Neurosci. 17 (1997)

Alzheimer disease: DNA fragmentation indicates increased neuronal 9407–9414.

vulnerability, but not apoptosis, J. Neuropathol. Exp. Neurol. 57 [84] W. Pope, M. Lambert, B. Leypole, R. Seupaul, L. Sletten, G. Krafft,

(1998) 456–464. W. Klein, Microtubule-associated protein tau is hyperphosphorylated

[99] E. Storey, T. Spurck, J. Pickett-Heaps, K. Beyreuther, C.L. Masters, during mitosis in the human neuroblastoma cell line SH-SY5Y, Exp.

The amyloid precursor protein of Alzheimer’s disease is found on Neurol. 126 (1994) 185–194.

the surface of static but not actively motile portions of neurites, [85] W. Q Qiu, A. Ferreira, C. Miller, E.H. Koo, D.J. Selkoe,

Cell-Brain Res. 735 (1996) 59–66. surfaceb-amyloid precursor protein stimulates neurite outgrowth of

[100] J.H. Su, A.J. Anderson, B.J. Cummings, C.W. Cotman, Immuno-hippocampal neurons in an isoform-dependent manner, J. Neurosci. histochemical evidence for DNA fragmentation in neurons in the

15 (1995) 2157–2167. AD brain, NeuroReport 5 (1994) 2529–2533.

[86] J.M. Roch, E. Masliah, A.C. Roch-Levecq, M.P. Sundsmo, D.A. [101] Y.H. Suh, An etiological role of amyloidogenic carboxyl-terminal Otero, I. Veinbergs, T. Saitoh, Increase of synaptic density and fragments of the b-amyloid precursor protein in Alzheimer’s memory retention by a peptide representing the trophic domain of disease, J. Neurochem. 68 (1997) 1781–1791.

the amyloidb/A4 protein precursor, Proc. Natl. Acad. Sci. USA 91 [102] N. Suzuki, T.T. Cheung, X.D. Cai, A. Odaka, L. Otvos Jr., C.

(1994) 7450–7454. Eckman, T.E. Golde, S.G. Younkin, An increased percentage of

[87] T.T. Rohn, K.J. Ivins, B.A. Bahr, C.W. Cotman, D.H. Cribbs, A long amyloid beta protein secreted by familial amyloidbprotein monoclonal antibody to amyloid precursor protein induces neuronal precursor (bAPP717) mutants, Science 264 (1994) 1336–1340. apoptosis, J. Neurochem. 74 (2000) 2331–2342. [103] T. Suzuki, M. Oishi, D.R. Marshak, J. Czernik, A.C. Nairn, P. [88] J.P. Roperch, V. Alvaro, S. Prieur, M. Tuynder, M. Nemani, F. Greengard P, Cell cycle-dependent regulation of the phosphoryla-Lethrosne, L. Piouffre, M.C. Gendron, D. Israeli, J. Dausset, M. tion and metabolism of the Alzheimer amyloid precursor protein, Oren, R. Amson, A. Telerman, Inhibition of presenilin 1 expression EMBO J. 13 (1994) 1114–1222.

is promoted by p53 and p21WAF-1 and results in apoptosis and [104] B. Tate, K.S. Aboody-Guterman, A.M. Morris, E.C. Walcott, R.E. tumor suppression, Nature Med. 4 (1998) 835–838. Majocha, C. A Marotta, Disruption of circadian regulation by brain [89] T. Russo, R. Faraonio, G. Minopoli, P. De Candia, S. De Renzis, N. grafts that overexpress Alzheimerb/A4 amyloid, Proc. Natl. Acad.

Zambrano, Fe65 and the protein network centered around the Sci. USA 89 (1992) 7090–7094.

cytosolic domain of the Alzheimer’s b-amyloid precursor protein, [105] L.O. Tjernberg, J. Naslund, J. Thyberg, S.E. Gandy, L. Terenius,

FEBS Lett. 434 (1998) 1–7. C. Nordstedt, Generation of Alzheimer amyloidbpeptide through

[90] K. Sambamurti, J. Shioi, A.P. Anderson, M. A Pappolla, N.K. nonspecific proteolysis, J. Biol. Chem. 272 (1997) 1870–1875. Robakis, Evidence for intracellular cleavage of the Alzheimer’s [106] S.Y. Tsang, S.C. Tam, I. Bremner, M.J. Burkitt, Copper-1,10-amyloid precursor in PC12 cells, J. Neurosci. Res. 33 (1992) phenanthroline induces internucleosomal DNA fragmentation in

319–329. HepG2 cells, resulting from direct oxidation by the hydroxyl

[91] M. Sato, T. Kawarabashi, M. Shoji, T. Kobayashi, N. Tada, E. radical, Biochem. J. 317 (1996) 13–16.

Matsubara, S. Hirai, Neurodegeneration and gliosis in transgenic [107] R. Vassar, B.D. Bennett, S. Babu-Khan, S. Kahn, E.A. Mendiaz, P. mice overexpressing a carboxy-terminal fragment of Alzheimer Denis, D.B. Teplow, S. Rose, P. Amarante, R. Loeloff, Y. Lui, S. amyloid-beta protein precursor, Dement. Geriatr. Cogn. Disord. 8 Fisher, J. Fuller, S. Edenson, J. Lile, M.A. Jarosinski, A.L. Biere,

(1997) 296–307. E. Curran, T. Burgess, J.C. Louis, F. Collins, J. Treanor, G.

[92] G. Sberna, J. Saez-Valero, Q.X. Li, C. Czech, K. Beyreuther, C.L. Rogers, M. Citron, b-secretase cleavage of Alzheimer’s amyloid Masters, C.A. McLean, D.H. Small, Acetylcholinesterase is in- precursor protein by the transmembrane aspartic protease BACE, creased in the brains of transgenic mice expressing the C-terminal Science 286 (1999) 735–741.

fragment (CT100) of theb-amyloid protein precursor of Alzheim- [108] I. Vincent, M. Rosado, P. Davies, Mitotic mechanisms in Alzheim-er’s disease, J. Neurochem. 71 (1998) 723–731. er’s disease?, J. Cell Biol. 132 (1996) 413–425.

[93] G. Schettini, C. Russo, W. Sannita, P. Gambetti, Characterization of [109] I. Vincent, G. Jicha, M. Rosado, D.W. Dickson, Aberrant expression carboxy-terminal APP derivatives in Alzheimer’s disease and Down of mitotic cdc2 / cyclin B1 kinase in degenerating neurons of syndrome, Soc. Neurosci. Abstr. 25 (1999) 837. Alzheimer’s disease brain, J. Neurosci. 17 (1997) 3588–3598.

[110] T. Watanabe, J. Sukegawa, I. Sukegawa, S. Tomita, K. Iijima, S. [94] D. Scheuner, C. Eckman, M. Jensen, X. Song, M. Citron, N. Suzuki,

Oguchi, T. Suzuki, A.C. Nairn, P. Greengard, A 127-kDa protein T.D. Bird, J. Hardy, M. Hutton, W. Kukull, E. Larson, E.

Levy-(UV-DDB) binds to the cytoplasmic domain of the Alzheimer’s Lahad, M. Viitanen, E. Peskind, P. Poorkaj, G. Schellenberg, R.

amyloid precursor protein, J. Neurochem. 72 (1999) 549–556. Tanzi, W. Wasco, L. Lannfelt, D. Selkoe, S. Younkin, Secreted

[111] D. Wolf, D. Quon, Y. Wang, B. Cordell, Identification and charac-amyloidb-protein similar to that in the senile plaques of

Alzheim-terization of C-terminal fragments of the b-amyloid precursor er’s disease is increased in vivo by the presenilin 1 and 2 and APP

produced in cell culture, EMBO J. 9 (1990) 2079–2084. mutations linked to familial Alzheimer’s disease, Nat. Med. 2

[112] B. Wolozin, K. Iwasaki, P. Vito, J.K. Ganjei, E. Lacana, T. (1996) 864–870.

Sunderland, B. Zhao, J.W. Kusiak, W. Wasco, L. D’Adamio, [95] M. Simons, E. Ikonen, P.J. Tienari, A. Cid-Arregui, U. Monning, K.

Participation of presenilin 2 in apoptosis enhanced basal activity Beyreuther, C.G. Dotti, Intracellular routing of human amyloid

conferred by an Alzheimer mutation, Science 274 (1996) 1710– protein precursor: Axonal delivery followed by transport to the

(13)

[113] T. Yamatsuji, T. Okamoto, S. Takeda, H. Fukumoto, T. Iwatsubo, Gurney, Membrane-anchored aspartyl protease with Alzheimer’s N. Suzuki, A. Asami-Odaka, S. Ireland, T.B. Kinane, I. Nishimoto, diseaseb-secretase activity, Nature 402 (1999) 533–537. G protein-mediated neuronal DNA fragmentation induced by [116] B.A. Yankner, L.R. Dawes, S. Fisher, L. Villa-Komaroff, M.L. familial Alzheimer’s disease-associated mutants of APP, Science Oster-Granite, R.L. Neve, Neurotoxocity of a fragment of the

272 (1996) 1349–1352. amyloid precursor associated with Alzheimer’s disease, Science

[114] T. Yamazaki, D.J. Selkoe, E.H. Koo, Trafficking of cell surface 245 (1989) 417–420.

b-amyloid precursor protein: retrograde and transcytotic transport [117] K. Yoshikawa, Neurotoxicity of b-amyloid (reply), Nature 361 in cultured neurons, J. Cell Biol. 129 (1995) 432–442. (1993) 122–123.

[115] R. Yan, M.J. Bienkowski, M.E. Shuck, H. Miao, M.C. Tory, A.M. [118] S.G. Younkin, Evidence that Ab42 is the real culprit in Alzheim-Pauley, J.R. Brashier, N.C. Stratman, W.R. Mathews, A.E. Buhl, er’s disease, Ann. Neurol. 37 (1995) 287–288.

Gambar

Fig. 1. Schematic depicting the structural and functional domains of the amyloid precursor protein.
Fig. 2. The yeast two-hybrid reporter assay reveals that APP-BP1interacts with hUba3. Top: schematic diagram showing the deletion andpoint mutants used for the assay
Fig. 3. APP-BP1 rescues the ts41 cell phenotype at the nonpermissive temperature. (A) Time course of viability of ts41 cells transfected with the vectoralone or with the APP-BP1 construct at the nonpermissive temperature (40comparison with wild type APP-BP
Fig. 4. Schematic depicting the strategy for overexpressing APP-BP1 in primary neurons
+3

Referensi

Dokumen terkait

Namun demikian, jumlah tercatat bagi penurunan nilai adalah kerugian kumulatif yang diukur sebagai selisih antara biaya perolehan diamortisasi dan nilai wajar

Pokja Pengadaan Barang I Unit Layanan Pengadaan (ULP) Barang/Jasa Pemerintah di Lingkungan Pemerintah Provinsi Bali akan melaksanakan Pelelangan Sederhana dengan

[r]

[r]

jual rumah serta tingkat pertumbuhan ekonomi masyarakat secara keseluruhan. Berdasarkan latarbelakang tersebut, maka yang menjadi permasalahan

(2) Permohonan pembetulan sebagaimana dimaksud pada ayat (1) atas permohonan Wajib Pajak, harus disampaikan secara tertulis kepada Kepala Daerah atau pejabat yang

Penyelenggaraan PAUD yang terintegrasi dengan layanan kesehatan dan gizi dalam meningkatkan mutu layanan pembelajaran PAUD.... Hambatan yang dihadapi dalam pelaksanaan

Populasi yang dimaksud dalam suatu penelitian adalah sekelompok objek yang dapat dijadikan sumber penelitian, dapat berupa benda-benda, manusia, gejala, peristiwa, atau hal – hal