alzheimer's disease sends the wrong signals – a perspective

4
Alzheimer’s disease sends the wrong signals – a perspective RACHAEL L. NEVE McLean Hospital, Belmont, MA 02478, USA Keywords: Alzheimer’s disease, signaling, presenilin, amyloid precursor protein Abbreviations: APP ¼ amyloid precursor protein; FAD ¼ familial Alzheimer’s disease; PI3K ¼ phosphatidylinositol-3- kinase; GSK-3 ¼ glycogen synthase kinase 3; EGFR ¼ epidermal growth factor receptor Abstract Familial Alzheimer’s disease mutations in presenilin and the amyloid precursor protein (APP) are thought to cause Alzheimer’s disease (AD) neurodegeneration by increasing production and aggregation of amyloid beta (Ab). However, presenilin has functions that are distinct from its role in the g-secretase complex, while APP has signaling functions that transcend its role as the source of Ab. Three recent papers highlight the potential importance of presenilin and APP signaling in the etiology of AD. Introduction Presenilin is most commonly known as a component of the g-secretase complex. It is in this capacity that familial Alzheimer’s disease (FAD) mutations in presenilin are thought to cause Alzheimer’s disease (AD) neurodegeneration, the idea being that these mutations cause gain-of-function changes in g-secretase activity that result in increased accumula- tion of amyloid beta1–42 (Ag 1–42 ). However, pre- senilin has signaling functions that are independent of its role in g-secretase activity, raising the possibility that loss of these functions may cause at least some of the neurodegeneration and memory loss that occurs in AD. One of the first hints that this may be the case was the finding that presenilin knockout mice exhibit impairments in hippocampal memory and long-term potentiation which are followed by neurodegenera- tion and tau hyperphosphorylation [1], suggesting that normally presenilin possesses a neuronal survival function that is lost in these animals. FAD mutants of presenilin do not always cause accumulation of Ab Further buttressing the notion that there may be more to the effects of FAD mutants of presenilin than simple increases in g-secretase activity, it has been shown that certain FAD presenilin mutations actually decrease the g-secretase activity of this molecule [2–4]. Moreover, wild-type presenilin, consistent with the notion that it promotes neuronal survival, protects against FAD amyloid precursor protein (APP)-induced amyloid pathogenesis in transgenic animals [5], whereas presenilin impaired in g-secretase activity leads to exacerbated amyloid pathology in FAD APP transgenic animals [6]. These findings suggest that FAD mutations of presenilin may impair its function rather than cause gain-of-function g-secretase activity. They also, considered in the context of the Shen laboratory work cited above, suggest that we should be looking at other functions of presenilin besides its g-secretase activity to find clues as to how FAD presenilin mutants cause AD neuropathology. FAD mutants of presenilin are impaired in c-secretase-independent signaling What might these other functions consist of? Work from the Robakis laboratory [7] demonstrated that presenilin inhibits apoptosis by promoting cadherin/ phosphatidylinositol-3-kinase (PI3K) association, thereby activating PI3K/Akt cell survival signaling. Presenilin AD mutations interfere with the presenilin-dependent activation of the PI3K/Akt signaling, and this interference results in increased glycogen synthase kinase (GSK)-3 activity, overphos- phorylation of tau at AD epitopes, and activation of Correspondence: Dr. Rachael L. Neve, MRC223, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA. Tel: 617-855-2413. Fax: 617-855-3793. E-mail: [email protected] Amyloid, March 2008; 15(1): 1–4 ISSN 1350-6129 print/ISSN 1744-2818 online Ó 2008 Informa UK Ltd. DOI: 10.1080/13506120701814608 Amyloid Downloaded from informahealthcare.com by University of Otago on 05/23/13 For personal use only.

Upload: rachael-l

Post on 08-Dec-2016

219 views

Category:

Documents


1 download

TRANSCRIPT

Alzheimer’s disease sends the wrong signals – a perspective

RACHAEL L. NEVE

McLean Hospital, Belmont, MA 02478, USA

Keywords: Alzheimer’s disease, signaling, presenilin, amyloid precursor protein

Abbreviations: APP¼ amyloid precursor protein; FAD¼ familial Alzheimer’s disease; PI3K¼ phosphatidylinositol-3-kinase; GSK-3¼ glycogen synthase kinase 3; EGFR¼ epidermal growth factor receptor

AbstractFamilial Alzheimer’s disease mutations in presenilin and the amyloid precursor protein (APP) are thought to causeAlzheimer’s disease (AD) neurodegeneration by increasing production and aggregation of amyloid beta (Ab). However,presenilin has functions that are distinct from its role in the g-secretase complex, while APP has signaling functions thattranscend its role as the source of Ab. Three recent papers highlight the potential importance of presenilin and APP signalingin the etiology of AD.

Introduction

Presenilin is most commonly known as a component

of the g-secretase complex. It is in this capacity that

familial Alzheimer’s disease (FAD) mutations in

presenilin are thought to cause Alzheimer’s disease

(AD) neurodegeneration, the idea being that these

mutations cause gain-of-function changes in

g-secretase activity that result in increased accumula-

tion of amyloid beta1–42 (Ag1–42). However, pre-

senilin has signaling functions that are independent

of its role in g-secretase activity, raising the possibility

that loss of these functions may cause at least some of

the neurodegeneration and memory loss that occurs

in AD. One of the first hints that this may be the case

was the finding that presenilin knockout mice exhibit

impairments in hippocampal memory and long-term

potentiation which are followed by neurodegenera-

tion and tau hyperphosphorylation [1], suggesting

that normally presenilin possesses a neuronal survival

function that is lost in these animals.

FAD mutants of presenilin do not always

cause accumulation of Ab

Further buttressing the notion that there may be

more to the effects of FAD mutants of presenilin

than simple increases in g-secretase activity, it has

been shown that certain FAD presenilin mutations

actually decrease the g-secretase activity of this

molecule [2–4]. Moreover, wild-type presenilin,

consistent with the notion that it promotes neuronal

survival, protects against FAD amyloid precursor

protein (APP)-induced amyloid pathogenesis in

transgenic animals [5], whereas presenilin impaired

in g-secretase activity leads to exacerbated amyloid

pathology in FAD APP transgenic animals [6].

These findings suggest that FAD mutations of

presenilin may impair its function rather than cause

gain-of-function g-secretase activity. They also,

considered in the context of the Shen laboratory

work cited above, suggest that we should be looking

at other functions of presenilin besides its g-secretase

activity to find clues as to how FAD presenilin

mutants cause AD neuropathology.

FAD mutants of presenilin are impaired

in c-secretase-independent signaling

What might these other functions consist of? Work

from the Robakis laboratory [7] demonstrated that

presenilin inhibits apoptosis by promoting cadherin/

phosphatidylinositol-3-kinase (PI3K) association,

thereby activating PI3K/Akt cell survival signaling.

Presenilin AD mutations interfere with the

presenilin-dependent activation of the PI3K/Akt

signaling, and this interference results in increased

glycogen synthase kinase (GSK)-3 activity, overphos-

phorylation of tau at AD epitopes, and activation of

Correspondence: Dr. Rachael L. Neve, MRC223, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA. Tel: 617-855-2413. Fax: 617-855-3793.

E-mail: [email protected]

Amyloid, March 2008; 15(1): 1–4

ISSN 1350-6129 print/ISSN 1744-2818 online � 2008 Informa UK Ltd.

DOI: 10.1080/13506120701814608

Am

yloi

d D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Ota

go o

n 05

/23/

13Fo

r pe

rson

al u

se o

nly.

apoptotic caspase-3 [7]. Consistent with this notion,

recent data from the Koo laboratory also show that

presenilin regulates the PI3K/Akt/GSK-3 pathway

[8]. Further supporting the idea that increased

GSK-3 activity may be important in the development

of AD neurodegeneration, Lucas et al. [9] and

Hernandez et al. [10] demonstrated that transgenic

mice overexpressing GSK-3 develop AD-like neuro-

pathology. In the first of the three papers highlighted

in this article, this same group has just shown that this

pathology, which includes tau hyperphosphorylation,

reactive astrocytosis, neuronal death, and spatial

learning deficits, can be reversed by decreasing

GSK-3 expression to normal levels [11]. They used

the elegant approach of conditionally overexpressing

GSK-3 in transgenic mice. Using the Tet-off system,

they dialed down the expression of the transgene by

administering a tetracycline analog to the mice for 6

weeks. The result was a full reversal of AD-like tau

hyperphosphorylation, restoration of microtubule

binding and stabilization ability of tau, reversal of

neuronal death and reactive gliosis, and rescue of

spatial memory impairment.

Such work highlights the importance of delineat-

ing the molecular mechanisms by which impairment

of the PI3K/Akt signaling function of presenilin

might lead to specific aspects of AD neurodegenera-

tion. The second of the three papers highlighted in

this perspective [12] describes such research. The

authors hypothesized previously, based on data from

their and other laboratories, that presenilins may be

involved in regulating certain signaling receptors

in a way that could modify the state of tau

phosphorylation and neuronal viability via the

PI3K/Akt pathway. In their current paper [12], they

present data in support of this hypothesis. They show

that the levels of epidermal growth factor receptor

(EGFR) are dramatically elevated in cells deficient in

presenilin, and in brains of presenilin conditional

knockout mice. Evidence is presented that the

elevation of EGFR in presenilin-deficient cells is

caused by impaired trafficking of EGFR from

endosomes to lysosomes. This phenotype was

rescued by expression of wild-type presenilin, but

not of FAD presenilin mutants, in the cells.

Importantly, several of these FAD presenilin mutants

retain their g-secretase activity. Further, g-secretase

inhibitors failed to mimic presenilin deficiency by

increasing EGFR levels, suggesting that the preseni-

lin g-secretase function is independent of the role of

presenilin in EGFR signaling.

FAD mutants of APP also send mixed signals

These two recent papers build an increasingly

strong case for the notion that impairment of

presenilin signaling, rather than gain-of-function

of its g-secretase cleavage activity, may be critical

for the development of AD neurodegeneration

(Figure 1). Does this mean that we should be

looking at alternative functions for the amyloid

precursor protein (APP) as well, beyond its role as

the source of the beta-amyloid peptides that

accumulate in the brains of patients with AD?

Probably. A multitude of cytosolic proteins that

interact with the APP cytodomain have been

Figure 1. Presenilin 1 possesses g-secretase-independent signaling functions. In a g-secretase-independent pathway, wild-type presenilin 1

promotes cadherin-mediated activation of the PI3K-Akt pathway, thereby suppressing apoptosis and GSK-3 activity and preventing GSK-3-

mediated overphosphorylation of tau. FAD mutants of presenilin 1 appear to interfere with this function of presenilin 1, leading to

suppression of PI3K/Akt signaling, overactivation of GSK-3, hyperphosphorylation of tau and neuronal death [7–9,12]. Presenilin 1 also

participates in g-secretase-dependent signaling pathways that regulate transcription [reviewed in ref. 20].

2 R. L. Neve

Am

yloi

d D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Ota

go o

n 05

/23/

13Fo

r pe

rson

al u

se o

nly.

described [reviewed in ref. 13] suggesting that APP

has versatile signaling roles.

All but one of the binding proteins for the APP

cytodomain interact with APP within the last 31

amino acids of this domain (Figure 2). Why is this

significant? For one thing, C31 can be generated

from APP by caspase cleavage [14,15]. Furthermore,

this cleavage has functional significance: expression

of C31 alone has been shown to cause neuronal cell

cycle entry and apoptosis [15–17]. Most recently, in

the third paper highlighted in this commentary,

inhibition of C31-producing caspase cleavage of APP

prevented the development of AD-like pathology and

behavior caused by the Indiana and Swedish FAD

mutations of APP [18]. The authors of this paper

introduced the D664A mutation (which prevents

the generation of C31) into the background of a

human APP minigene carrying the K670N/M671L

(Swedish) and V717F (Indiana) mutations. Both the

original FAD mutant minigene (PDAPP) and also

the D664A version of it [PDAPP(D664A)] were

expressed in transgenic mice under the control of the

PDGF B-chain promoter.

The D664A mutation did not alter the net

in vivo production of Ab40 and Ab42 in the brains

of the mice, nor did it affect the extent of amyloid

plaque deposition in PDAPP(D664A) mice com-

pared with PDAPP mice. However, the D664A

mutation did have an effect on neurodegeneration

and on behavior. While the PDAPP mice displayed

decreased hippocampal presynaptic density num-

ber, increased GFAP immunoreactivity in the

hippocampus, loss of dentate gyrus volume, and

impaired spatial learning, relative to controls, the

PDAPP(D664A) mice were indistinguishable from

controls in these parameters. A subsequent study

[19] showed that PDAPP mice have impaired

synaptic transmission, synaptic plasticity, and

learning; and that the PDAPP(D664A) mutation

rescued these abnormalities despite elevated levels

of Ab42 and plaque accumulation in these trans-

genic mice.

From these data, it can be inferred that if C31

cannot be generated from APP carrying FAD

mutations, multiple aspects of neuropathology, im-

paired learning, and impaired deficits in synaptic

transmission that are normally caused by these

mutations do not occur. What are the implications

of these findings? First of all, note that Asp664

selectively rescues the neurodegeneration and the

learning abnormalities of the PDAPP mice without

decreasing the production of Ab40 or Ab42. Thus, the

rescue is independent of the production of Ab.

Figure 2. All but one of the APP cytodomain-binding proteins that have been identified bind to the C-terminal 31 amino acids of APP

[reviewed in ref. 13]. These include the p21-activated kinase 3 (PAK3) [15], APP-binding protein 1 (APP-BP1) [16], the heterotrimeric G

protein Go [21], and multiple intracellular adaptor proteins [reviewed in ref. 22].

Figure 3. C31, when released from FAD APP by caspase cleavage,

may abnormally activate or disrupt signaling pathways mediated by

APP. In the scenario shown, both APP-BP1 and a complex that

includes Go and PAK3 bind to the C-terminus of APP, not

necessarily simultaneously. Also interacting with the C-terminus

of APP but not shown are a number of intracellular adaptor

proteins [reviewed in ref. 20]. Caspase cleavage of FAD APP,

which occurs preferentially on the b-secretase cleavage product,

C99 [14], releases C31, which has been shown to cause abnormal

neuronal DNA synthesis and apoptosis mediated by PAK3 and

APP-BP1 [15,16].

Faulty signaling in Alzheimer’s disease 3

Am

yloi

d D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Ota

go o

n 05

/23/

13Fo

r pe

rson

al u

se o

nly.

Secondly, the C31 region of APP encompasses the

binding sites for nearly all of the signaling proteins

that have been shown to bind to the intracellular

domain of APP. The data suggest a scenario in which

C31, when removed from APP, may abnormally

activate or disrupt signaling pathways mediated by

APP (Figure 3). Such a scenario is supported by the

findings that caspase-cleaved APP and activated

caspases are present in the brains of AD patients

but not in control brains [15], and that expression of

C31 alone causes apoptosis [15,16].

Concluding remarks

The trio of papers discussed in this commentary,

taken together with the body of work preceding

them, make the case that not only Ab production,

but also presenilin and/or APP signaling is likely to

be important in the etiology of AD. Studies that

delineate the normal signaling pathways mediated by

presenilin and APP are to be encouraged, for they are

certain to help us understand how signal transduc-

tion by these proteins goes awry in AD.

Acknowledgements

I thank Nikolaos Robakis for numerous helpful

discussions. I also thank Lia Baki for the Figure 1

artwork and Donna McPhie for designing Figures 2

and 3.

References

1. Saura CA, Choi SY, Beglopoulos V, Malkani S, Zhang D,

Shankaranarayana Rao BS, Chattarji S, Kelleher RJ 3rd,

Kandel ER, Duff K, Kirkwood A, Shen J. Loss of presenilin

function causes impairments of memory and synaptic plasti-

city followed by age-dependent neurodegeneration. Neuron

2004;42:23–36.

2. Marambaud P, Wen PH, Dutt A, Shioi J, Takashima A,

Siman R, Robakis NK. A CBP binding transcriptional

repressor produced by the PS1/epsilon-cleavage of N-cadherin

is inhibited by PS1 FAD mutations. Cell 2003;114:635–645.

3. Bentahir M, Nyabi O, Verhamme J, Tolia A, Horre K,

Wiltfang J, Esselmann H, De Strooper B. Presenilin clinical

mutations can affect g-secretase activity by different mechan-

isms. J Neurochem 2006;96:732–742.

4. Kumar-Singh S, Theuns J, Van Broeck B, Pirici D,

Vannekens K, Corsmit E, Cruts M, Dermaut B, Wang R,

Van Broeckhoven C. Mean age-of-onset of familial Alzheimer

disease caused by presenilin mutations correlates with both

increased Abeta42 and decreased Abeta40. Hum Mutat

2006;27:686–695.

5. Wang R, Wang B, He W, Zheng H. Wild-type presenilin 1

protects against Alzheimer disease mutation-induced amyloid

pathology. J Biol Chem 2006;281:15330–15336.

6. Deng Y, Tarassishin L, Kallhoff V, Peethumnongsin E, Wu L,

Li YM, Zheng H. Deletion of presenilin 1 hydrophilic loop

sequence leads to impaired g-secretase activity and exacer-

bated amyloid pathology. J Neurosci 2006;26:3845–3854.

7. Baki L, Shioi J, Wen P, Shao Z, Schwarzman A,

Gama-Sosa M, Neve R, Robakis NK. PS1 activates PI3K

thus inhibiting GSK-3 activity and tau overphosphorylation:

effects of FAD mutations. EMBO J 2004;23:2586–2596.

8. Kang DE, Yoon IS, Repetto E, Busse T, Yermian N, Ie L,

Koo EH. Presenilins mediate phosphatidylinositol 3-kinase/

AKT and ERK activation via select signaling receptors.

Selectivity of PS2 in platelet-derived growth factor signaling.

J Biol Chem 2005;280:31537–31547.

9. Lucas JJ, Hernandez F, Gomez-Ramos P, Moran MA, Hen R,

Avila J. Decreased nuclear beta-catenin, tau hyperphosphor-

ylation and neurodegeneration in GSK-3beta conditional

transgenic mice. EMBO J 2001;20:27–39.

10. Hernandez F, Borrell J, Guaza C, Avila J, Lucas JJ. Spatial

learning deficit in transgenic mice that conditionally over-

express GSK-3beta in the brain but do not form tau filaments.

J Neurochem 2002;83:1529–1533.

11. Engel T, Hernandez F, Avila J, Lucas JJ. Full reversal of

Alzheimer’s disease-like phenotype in a mouse model with

conditional overexpression of glycogen synthase kinase-3.

J Neurosci 2006;26:5083–5090.

12. Repetto E, Yoon I-S, Zheng H, Kang DE. Presenilin-1

regulates epidermal growth factor receptor turnover and

signaling in the endosomal-lysosomal pathway. J Biol Chem

2007;282:31504–31516.

13. Neve RL, McPhie DL. The cell cycle as a therapeutic target

for Alzheimer’s disease. Pharmacol Ther 2006;111:99–113.

14. Gervais FG, Xu D, Robertson GS, Vaillancourt JP, Zhi Y,

Huang J, LeBlanc A, Smith D, Rigby M, Shearman MS,

Clarke EE, Zheng H, Van Der Ploeg LH, Ruffolo SC,

Thornberry NA, Xanthoudakis S, Zamboni RJ, Roy S,

Nicholson DW. Involvement of caspases in proteolytic

cleavage of Alzheimer’s amyloid-beta precursor protein and

amyloidogenic Ab peptide formation. Cell 1999;97:395–406.

15. Lu DC, Rabizadeh S, Chandra S, Shayya RF, Ellerby LM,

Ye X, Salvesen GS, Koo EH, Bredesen DE. A second

cytotoxic proteolytic peptide derived from amyloid beta-

protein precursor. Nat Med 2000;6:397–404.

16. McPhie DL, Coopersmith R, Hines-Peralta A, Chen Y,

Ivins KJ, Manly SP, Kozlowski MR, Neve KA, Neve RL.

DNA synthesis and neuronal apoptosis caused by familial

Alzheimer disease mutants of the amyloid precursor protein

are mediated by the p21 activated kinase PAK3. J Neurosci

2003;23:6914–6927.

17. Chen Y, Liu W, McPhie DL, Hassinger L, Neve RL. APP-

BP1 mediates APP-induced apoptosis and DNA synthesis

and is increased in Alzheimer’s disease brain. J Cell Biol

2003;163:27–33.

18. Galvan V, Gorostiza OF, Banwait S, Ataie M, Logvinova AV,

Sitaraman S, Carlson E, Sagi SA, Chevallier N, Jin K, Greenberg

DA, Bredesen DE. Reversal of Alzheimer’s-like pathology and

behavior in human APP transgenic mice by mutation of Asp664.

Proc Natl Acad Sci USA 2006;103:7130–7135.

19. Saganich MJ, Schroeder BE, Galvan V, Bredesen DE, Koo EH,

Heinemann SF. Deficits in synaptic transmission and learning

in amyloid precursor protein (APP) transgenic mice require C-

terminal cleavage of APP. J Neurosci 2006;26:13428–13426.

20. Marambaud P, Robakis NK. Genetic and molecular aspects of

Alzheimer’s disease shed light on new mechanisms of

transcriptional regulation. Genes Brain Behav 2005;4:134–146.

21. Nishimoto I, Okamoto T, Matsuura Y, Takahashi S,

Okamoto T, Murayama Y, Ogata E. Alzheimer amyloid

protein precursor complexes with brain GTP-binding protein

G(o). Nature 1993;362:75–79.

22. Russo C, Venezia V, Repetto E, Nizzari M, Violani E, Carlo

P, Schettini G. The amyloid precursor protein and its network

of interacting proteins: physiological and pathological im-

plications. Brain Res Rev 2005;48:257–264.

4 R. L. Neve

Am

yloi

d D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Ota

go o

n 05

/23/

13Fo

r pe

rson

al u

se o

nly.