You are on page 1of 9

articles

β-arrestin 2 regulates Aβ generation and γ-secretase


activity in Alzheimer’s disease
Amantha Thathiah1,2, Katrien Horré1,2, An Snellinx1,2, Elke Vandewyer1,2, Yunhong Huang1,2,
Marta Ciesielska1,2, Gerdien De Kloe1,3, Sebastian Munck1,2 & Bart De Strooper1,2

b-arrestins are associated with numerous aspects of G protein–coupled receptor (GPCR) signaling and regulation and accordingly
influence diverse physiological and pathophysiological processes. Here we report that b-arrestin 2 expression is elevated in two
independent cohorts of individuals with Alzheimer’s disease. Overexpression of b-arrestin 2 leads to an increase in amyloid-b
(Ab) peptide generation, whereas genetic silencing of Arrb2 (encoding b-arrestin 2) reduces generation of Ab in cell cultures and
© 2013 Nature America, Inc. All rights reserved.

in Arrb2−/− mice. Moreover, in a transgenic mouse model of Alzheimer’s disease, genetic deletion of Arrb2 leads to a reduction
in the production of Ab40 and Ab42. Two GPCRs implicated previously in Alzheimer’s disease (GPR3 and the b2-adrenergic
receptor) mediate their effects on Ab generation through interaction with b-arrestin 2. b-arrestin 2 physically associates with the
Aph-1a subunit of the g-secretase complex and redistributes the complex toward detergent-resistant membranes, increasing the
catalytic activity of the complex. Collectively, these studies identify b-arrestin 2 as a new therapeutic target for reducing amyloid
pathology and GPCR dysfunction in Alzheimer’s disease.

Considerable insight into the genetic and molecular biological basis of the potential of Aβ-modulating agents in the early treatment of
Alzheimer’s disease has yet to be translated into a new medication for Alzheimer’s disease3.
the disease. Removal of tau1,2 or the Aβ peptide3–5 from the brain by GPCRs, also called seven transmembrane receptors (7TMRs),
various vaccination strategies, inhibition or modulation of the β- and comprise the largest family of membrane proteins12 and are the most
γ-secretases, which cleave the β-amyloid precursor protein (APP) to common target for therapeutic drugs13. Over 370 nonsensory GPCRs
generate the Aβ peptide, or alterations in apolipoprotein E (APOE) have been identified14, of which more than 90% are expressed in the
expression6 are the primary therapeutic avenues that are currently brain, where they have important roles in cognition, mood, appe-
pursued. Unfortunately, many therapeutic agents have failed at several tite, pain and synaptic transmission15. In the context of Alzheimer’s
stages of development as a result of poor blood-brain barrier penetra- disease, GPCRs are associated with multiple stages of APP proteo­
tion or severe side effects3–5,7. Moreover, new findings suggest the lysis, including modulation of processing of APP by the α-, β- and
npg

need for very early symptomatic or presymptomatic treatment, which γ-secretases and the regulation of Aβ degradation and Aβ-mediated
will require the identification of alternative disease-modifying targets toxicity16; however, the signaling mechanisms mediating these effects
to safely modulate the pathogenesis of Alzheimer’s disease3–5,7. remain at best only partially understood, and the hypothesized regula-
Central to many Aβ-directed therapeutic strategies is the tion of the γ-secretase complex by GPCRs remains unexplained.
γ-secretase complex, which is a multimeric aspartyl protease com- From a drug discovery perspective, GPCRs are very versatile
posed of four subunits: presenilin 1 or 2 (PS1 or PS2), nicastrin (NCT), targets. Drugs that directly target a GPCR have been classically
APH-1A or APH-1B and PEN 2 (ref. 8). The majority of cases of described as either agonists or antagonists for G protein signaling.
early onset familial Alzheimer’s disease are attributed to mutations The binding of an agonist to a GPCR promotes a conformational
in the PS1 (Presenilin 1), PS2 (Presenilin 2) and APP genes9, pro- change that results in the activation of receptor-associated hetero­
viding the rationale for the therapeutic targeting of the proteins trimeric G proteins and consequent downstream signaling. However,
they encode. However, familial Alzheimer’s disease accounts for a small family of multifunctional GPCR regulatory or adaptor pro-
less than 0.5% of all Alzheimer’s disease cases10. The vast major- teins known as the β-arrestins, which have an almost universal role
ity of late-onset Alzheimer’s disease cases are sporadic and include in facilitating traditional GPCR desensitization, is also capable
environmental and genetic risk factors, among which the ε4 allele of of initiating distinct, independent signaling events17. These signals
APOE accounts for more than 25% of the genetic risk. Interestingly, are often both spatially and temporally different from the classic
APOE4 also seems to modulate Aβ accumulation11, strengthening G protein signals and result in unique cellular and physiological or

1Vlaams Instituut voor Biotechnologie (VIB) Center for the Biology of Disease, Leuven, Belgium. 2Center for Human Genetics and Leuven Institute for Neuroscience

and Disease (LIND), University of Leuven (KU Leuven), Leuven, Belgium. 3Neuroscience Department, Johnson & Johnson Pharmaceutical Research and
Development, Janssen Pharmaceutica, Beerse, Belgium. Correspondence should be addressed to A.T. (amantha.thathiah@cme.vib.kuleuven.be) or
B.D.S. (bart.destrooper@cme.vib-kuleuven.be).

Received 17 September; accepted 7 November; published online 2 December 2012; doi:10.1038/nm.3023

nature medicine  VOLUME 19 | NUMBER 1 | JANUARY 2013 43


Articles

a 3.0 β-arrestin 2 b 2.0 β-arrestin 1 c Vector d Vector e Aβ40 f


β-arrestin 2 β-arrestin 2 Aβ42

nonsilencing siRNA (Aβ)


2.0 2.0 1.5

Normalized to vector

Normalized to vector
** ** *

in
Normalized to

st
2.5 1.5 1.5

re
or
1.0
Fold change (relative to control)

ar
ct
Fold change (relative to control)

(Aβ40)

(Aβ42)

Ve

β-
1.5 1.0 1.0 **
2.0 0.5 ** sAPPtotal
0.5 0.5
*** *** ** ***
sAPPα
***
1.5 1.0

5, le

5, le
L- eh 8

5, e
L- ehi 8
5, e
8

β- ilen 2 s A

si A
re ing NA

A
V 5

45

68 icl
V 5
68 cl
45
68 ic

68 ic

2 N
N
4

4
L- Veh

on tin iR

in iR
ar c iR

R
L- eh

N res g s

st s
V

ar in
β- lenc
1.0

si

s
on
0.5

N
0.5
g h i Non
silencing β-arrestin 2
Vector β-arrestin siRNA siRNA
L-685,458: – – + – – +
Non β-arrestin 2
Control Alzheimer’s Control Alzheimer’s β-arrestin 2
silencing β-arrestin 2 ADAM10
disease disease siRNA siRNA ADAM10
β-secretase
sAPPtotal β-secretase
NCT
sAPPα NCT
PS1-NTF
PS1-NTF
PS1-CTF
Figure 1  Expression of β-arrestin 2 is elevated in individuals with Alzheimer’s disease, PS1-CTF
APH-1A
and overexpression and silencing of β-arrestin 2 differentially regulate Aβ accumulation. APH-1A
PEN 2
(a,b) Expression of β-arrestin 2 (a) and β-arrestin 1 (b) assessed by quantitative PCR (qPCR) in PEN 2
APP-FL
brain samples from control individuals and individuals with Alzheimer’s disease. **P = 0.0089, APP-FL
APP-CTF
© 2013 Nature America, Inc. All rights reserved.

***P = 0.0002 by unpaired Student’s t test. n = 20 control individuals, n = 18 individuals APP-CTF


with Alzheimer’s disease. (c,d) Amounts of Aβ40 (c) and Aβ42 (d) assessed by ELISA in culture β-actin
β-actin
supernatants from HEK293-APP695 cells after overexpression of β-arrestin 2 and treatment
with the γ-secretase inhibitor L-685,458. *P < 0.05, **P < 0.01, ***P < 0.001 by one-way analysis of variance (ANOVA) and Dunnett’s multiple
comparisons post test. n = 4 experiments performed in triplicate. Error bars, s.e.m. (e) Amounts of Aβ40 (black bars) and Aβ42 (red bars) determined
by ELISA after silencing of β-arrestin 2. **P < 0.01 by ANOVA and Dunnett’s post test. n = 5–6 experiments performed in triplicate. Error bars, s.e.m.
(f,g) Amounts of total soluble APP (sAPPtotal) and the α-secretase–mediated cleavage product of APP (sAPPα) from HEK293-APP695 cell culture
supernatants determined by immunoblot analysis after overexpression (f) or silencing (g) of β-arrestin 2. (h) Expression of ADAM10, β-secretase,
the γ-secretase complex, APP-FL and APP-CTF evaluated by Immunoblot analysis after overexpression of β-arrestin 2 and treatment with L-685,458.
(i) Expression of ADAM10, β-secretase, the γ-secretase complex, APP-FL and APP-CTF evaluated by immunoblot analysis after silencing of β-arrestin 2.

pathophysiological consequences18 that can be exploited for the effectively abolished by the addition of L-685,458, a highly selective
therapeutic development of G protein− or β-arrestin–biased drugs. γ-secretase inhibitor22. Furthermore, the release of Aβ40 and Aβ42
Differential regulation of β-arrestins is implicated in type 2 diabetes was not diminished in cells with reduced β-arrestin 1 expression
and psychiatric disorders19,20, for which pharmacological manipula- (Supplementary Fig. 1c–e). In contrast, silencing of β-arrestin 2
tion of selective β-arrestin–dependent complexes may provide thera- efficiently suppressed Aβ secretion to 50% below that of control
peutic benefits21. As mediators of GPCR desensitization, trafficking transfected cells (Fig. 1e). Furthermore, the amounts of total soluble
and cell signaling, the β-arrestins could provide a putative basis to APP and the α-secretase–mediated cleavage product of APP
understand GPCR dysfunction in Alzheimer’s disease. Nevertheless, (Fig. 1f,g) and expression of the α- and β-secretases were unchanged
npg

no study so far has documented a role for β-arrestins in Alzheimer’s after expression or silencing of β-arrestin 2 (Fig. 1h,i), indicating
disease progression. that the modulation of Aβ generation by β-arrestin 2 occurs down-
stream of β-secretase activity. Expression of the NCT, PS1, APH-1A
RESULTS and PEN 2 subunits of the γ-secretase complex was also unaffected
b-arrestin 2 in Alzheimer’s disease and effects on Ab generation by expression or silencing of β-arrestin 2 (Fig. 1h,i). Notably,
We compared the expression of β-arrestins 1 and 2 in samples from accumulation of the APP C-terminal fragment (APP-CTF), which is
the hippocampus and entorhinal cortex of autopsied human brains typically observed after a reduction in γ-secretase activity, was only
with Alzheimer’s disease (N = 18) and age-matched control subjects modestly detectable, raising the question of how β-arrestin 2 affects
(N = 20). Levels of β-arrestin 2 mRNA were significantly increased Aβ generation (Fig. 1h,i).
(Fig. 1a), whereas levels of β-arrestin 1 mRNA were decreased, in To assess the physiological relevance of genetic silencing of
the Alzheimer’s disease samples compared to the control samples β-arrestins 1 and 2, we isolated embryonic neuronal cultures from
(Fig. 1b). We confirmed these observations in an independent group wild-type Arrb1+/+ (encoding endogeneous β-arrestin 1), Arrb2+/+,
of Braak-staged human brain samples (Braak 0–2 compared to Braak Arrb1−/− (ref. 23) and Arrb2−/− (ref. 24) mice (Fig. 2a,b). The amounts
5–6) (Supplementary Fig. 1a,b). Although there are limitations in of endogenous Aβ40 and Aβ42 were substantially reduced in Arrb2−/−
using postmortem tissue, such as mRNA degradation and cellular but not Arrb1−/− neurons, demonstrating that β-arrestin 2 is involved
alterations during disease progression that complicate the interpreta- endogenously in the modulation of neuronal Aβ production.
tion of results, these data suggest that β-arrestins 1 and 2 are differen-
tially regulated in brain areas affected in Alzheimer’s disease. GPCRs regulate Ab generation through b-arrestin 2
We next determined whether the expression of β-arrestins 1 and β-arrestins have traditionally been associated with the termination
2 directly affects Aβ generation in a cellular context. Expression of of GPCR signaling and receptor desensitization 25,26. In parallel,
β-arrestin 2 in the HEK293-APP695 cell line led to a significant β-arrestins can initiate a second set of G protein–independent signals,
increase in Aβ40 and Aβ42 release (Fig. 1c,d), an effect that was resulting in receptor desensitization, endocytosis and various cellular

44 VOLUME 19 | NUMBER 1 | JANUARY 2013  nature medicine


articles

a b c d e Arrb2+/+ Arrb2–/–
Aβ40 Aβ40 Ad-GFP Ad-GFP
1.5

GPR3
GPR3
1.5 ** * 1.5 0.3

GFP
GFP
Aβ42 Aβ42 Ad-GPR3 Ad-GPR3
NS NS ** * ** **
Normalized value

Normalized value

Aβ42 (ng ml–1)


1.0 Nct

Aβ40 (ng ml–1)


1.0 1.0 0.2
Ps1-CTF
NS Pen 2
NS
0.5 0.5 0.5 0.1 App-FL
App-CTF
GPR3
β-actin
+

+


+

+


–/

–/
+/

+/

–/
+/
+/
–/

+/
–/
+/

+/

–/
+/
1

1
Ar 1

Ar 1

2
2
2

2
Ar b2

Ar b2
Ar b2

Ar 2

2
2
rb

rb
rb

rb

rb
rb
rb

rb
rb

rb
rb
r

r
r
Ar

Ar

Ar
Ar
Ar

Ar

Ar
Ar
Figure 2  Endogenous Aβ generation is reduced in β-arrestin 2–deficient mice. (a) Amounts of Aβ40 and Aβ42 measured by ELISA in culture supernatant
samples from primary cortical neuronal cultures from wild-type Arrb1+/+ and Arrb1−/− embryonic day 14 fetal mice. Data are normalized to the amount
of Aβ secreted from the wild-type cultures (Arrb1+/+ or Arrb2+/+, respectively). NS, not significant by unpaired Student’s t test. n = 19 Arrb1+/+,
n = 20 Arrb1−/−. Error bars, s.e.m. (b) Amounts of Aβ40 and Aβ42 measured in Arrb2+/− and Arrb2−/− primary neuronal cultures. *P < 0.05, **P < 0.01
by ANOVA and Dunnett’s post test. n = 11 wild-type Arrb2+/+, n = 18 Arrb2+/−, n = 20 Arrb2−/−. Error bars, s.e.m. (c,d) Endogenous release of Aβ40 (c)
and Aβ42 (d) measured in culture supernatant samples from primary cortical neuronal cultures transduced with Ad-GPR3 (red bars) or Ad-GFP control
(black bars). **P < 0.01, NS, not significant by ANOVA and Dunnett’s post test. n = 18 Arrb2+/+, n = 18 Arrb2−/−. Error bars, s.e.m. (e) Expression of
the γ-secretase complex components in primary cortical neuronal cultures, App-FL and App-CTF determined by immunoblot analysis in the absence of
β-arrestin 2 or after transduction with GPR3.
© 2013 Nature America, Inc. All rights reserved.

responses17,27,28. Thus far, two GPCRs have been associated with the in the release of Aβ40 and Aβ42 (Supplementary Fig. 2c). We used
γ-secretase–mediated processing of APP and Aβ generation: GPR3 APP-C99, which is directly cleaved by the γ-secretase to yield Aβ40
(ref. 29) and the β2-adrenergic receptor (β2-AR)30. In addition, the and Aβ42, in these assays, further demonstrating that the β-arrestin
δ-opioid receptor has been implicated in modulation of the β-secretase– 2–mediated effect on Aβ generation is downstream of the α- and
mediated and subsequent γ-secretase–mediated proteolysis of APP31. β-secretases. This result is similar to the previously described effects of
All three GPCRs seem to modulate Aβ generation independently of GPR3 on APP processing29. β2-AR has also been reported to modulate
G protein signaling29–31. Consequently, we hypothesized that the γ-secretase–mediated Aβ release30. Therefore, we used the CHO-K1
β-arrestins could regulate Aβ generation through modulation of their cell line, which stably expresses β-arrestin 2 and the β2-AR, to perform
interaction with these specific GPCRs. We used the CHO-K1 cell line, a similar series of experiments. Silencing of β-arrestin 2 expression
which stably expresses β-arrestin 2 and GPR3, to monitor the direct in this cell line resulted in reduced interaction between β-arrestin 2
interaction between β-arrestin 2 and GPR3 using the PathHunter and the β2-AR after treatment with the β2-AR agonist isoproterenol as
technology32 (Supplementary Fig. 2a). In this cell line, silencing of monitored with the PathHunter assay (Supplementary Fig. 2d), and
β-arrestin 2 expression (Supplementary Fig. 2b) led to a reduction a reduction in Aβ generation (Supplementary Fig. 2e,f).

a b c Aβ40
d e Aβ40
npg

15,000 ** 4.0 Aβ42 10,000 2.5 Aβ42


Aβ generation normalized to C99

Aβ generation normalized to C99

**
*** 8,000 2.0 * ** ** *
Extracellular
3.0 ***
10,000
domain 6,000 1.5
* **
RLU

RLU

2.0
4,000 1.0
Plasma 5,000
membrane
1.0
2,000 0.5

Intracellular
domain
D + e
+ 9
99

D + ne
+ 9
9
D + ne
+ 9
99

se + ne
e 99
99

rin + ne
G T C +C 9
3 R al 9
rin + e
+ 9
99
3 R3 lon
Y 9

Y 9
G G C9 C9

Y 9

e C9
PR G 99 9
se 3 on
e C9
R C
C

3 R3 alo
R C

3 R3 alo
R C
C

3 R3 alo
rin C
C

se 3 lo

C
PR P a

3 PR 9 a
+
G G 99

PR P 9

PR P 9

PR P 9
G T G C9

G T G C9

PR G 9
W C

G T C

P
T

T
W

Figure 3  The C-terminal domain of GPR3 modulates the interaction with β-arrestin 2 and Aβ generation. (a) Schematic representation of GPR3.
The single-letter amino acid residue codes are used. The DRY putative binding site for G proteins is indicated in red and was mutated to AAY (GPR3
DRY/AAY mutant). Putative phosphorylation sites for G protein-coupled receptor kinases (GRKs) in the cytoplasmic tail are indicated in red. These
serine residues were mutated to alanine to obtain the GPR3 serine mutant that does not interact with β-arrestin 2. (b) Interaction of WT GPR3 and the
GPR3 DRY/AAY mutant (GPR3 DRY) with β-arrestin 2 in the CHO-K1 β-arrestin 2 cell line measured with the PathHunter assay as relative light units
(RLU). (c) Generation of Aβ40 (black bars) and Aβ42 (red bars) measured by ELISA in CHO-K1 β-arrestin 2 cells that express APP-C99 (C99) alone,
WT GPR3 + APP-C99 or the GPR3 DRY/AAY mutant + APP-C99. (d) Interaction of WT GPR3 and the GPR3 serine mutant with β-arrestin 2 in the
CHO-K1 β-arrestin 2 cell line measured with the PathHunter assay. (e) Generation of Aβ40 (black bars) and Aβ42 (red bars) measured by ELISA in
CHO-K1 β-arrestin 2 cells that express APP-C99 alone, WT GPR3 + APP-C99 or the GPR3 serine mutant + APP-C99. *P < 0.05, **P < 0.01,
***P < 0.001 by ANOVA and Dunnett’s post test. Data are the mean ± s.e.m. of four to six independent experiments performed in triplicate.

nature medicine  VOLUME 19 | NUMBER 1 | JANUARY 2013 45


Articles

a Vehicle Lactacystin b c d

(normalized to nonsilencing siRNA)


2.0 Vehicle Lactacystin 2.0
ed
** ***

ed
+/+ –/– +/+ –/–

(normalized to Arrb2+/+)
APP-CTF accumulation

APP-CTF accumulation
Arrb2 Arrb2 Arrb2 Arrb2
g

g
in

in
1

2
ct

ct
nc

nc
fe

fe
in

in

in

in
1.5 1.5
st

st

st

st
ns

ns
le

le
re Nct

re

re

re
si

si
ra

ra
ar

ar

ar

ar
on

on
nt

nt
NS
β-

β-

β-

β-
U

N
Ps1-NTF
NCT 1.0 1.0
PS1-NTF Ps1-CTF

PS1-CTF 0.5 0.5


Pen 2
PEN 2
App-FL
APP-FL


App-CTF

re 1 NA
2 NA

+/

–/
N
β- rest siR

2
in R
R

2
APP-CTF

rb
rb
st si
si
β-actin

Ar
Ar
ar ing
ar in
β-actin

β- nc
le
si
on
N
JC-8
e Vector β-arrestin 2 f Vector g Input Vector β-arrestin 2 h JC-8 + X
2.5 β-arrestin 2 4
***

Normalized intensity
Normalized intensity (to vector)
* *

β-arrestin 2
Fractions:
10
11
12
13

10
11
12
13
1
2
3
4
5
6
7
8
9

1
2
3
4
5
6
7
8
9

JC-8 + X

JC-8 + X

(to vector)
NCT 2.0

Vector
* *

JC-8

JC-8
β-arrestin 2 2
PS1-NTF 1.5 ***
PS1-CTF 1
Raft PS1-CTF
APH-1A 1.0
PEN 2

or

2
Non-raft PS1-CTF

in
ct
Caveolin-1 0.5

st
Ve

re
© 2013 Nature America, Inc. All rights reserved.

GM130

ar
β-
EEA1
T
TF

2
C

-1

N
C
N

PE
1-

AP
PS

Figure 4  β-arrestin 2 and the active γ-secretase complex are enriched in DRMs. (a) Immunoblot analysis of the γ-secretase complex and APP-FL
in HEK293-APP695 cells after silencing β-arrestin 1 or 2, treatment with lactacystin or both. (b) Quantification of APP-CTF accumulation after
lactacystin treatment normalized to nonsilencing siRNA. n = 3 independent experiments performed in triplicate. (c) Immunoblot analysis of the
γ-secretase complex and App-FL in mouse neuronal cultures after treatment with vehicle or lactacystin. (d) Quantification of APP-CTF accumulation
after lactacystin treatment normalized to Arrb2+/+ neuronal cultures. n = 3 independent experiments with four to six brain cultures per experiment.
(e) Sucrose gradient fractionation of HEK293 cells transfected with empty vector or β-arrestin 2–GFP complementary DNA (cDNA). Equivalent volumes
were assessed by immunoblot using antibodies that recognize γ-secretase complex subunits, β-arrestin 2 or the organelle-specific markers GM130
(cis-Golgi), EEA1 (early endosomes) and Caveolin-1 (caveolae). (f) Quantification of the expression of the γ-secretase complex subunits in pooled
fractions 3 and 4 from HEK293 cells transfected with empty vector or β-arrestin 2–GFP cDNA normalized to Caveolin 1 expression. n = 4–6
independent experiments. (g) Immunoblot analysis using a PS1-CTF–specific antibody of pooled raft and non-raft fractions incubated with JC-8
or JC-8 + L-685,458 (JC-8 + X) followed by photoaffinity crosslinking. (h) Quantification of the expression of the PS1-CTF in the raft and non-raft
fractions normalized to input. n = 3 independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001, NS, not significant by ANOVA and the Dunnett’s
post test. Error bars, s.e.m.

To genetically establish the requirement of β-arrestin 2 for the cellular responses33–35 (Fig. 3a). The GPR3 DRY/AAY mutant did
npg

modulation of Aβ generation by GPR3, we transduced wild-type not activate G protein signaling, as measured by reduced intracel-
Arrb2+/+ and Arrb2−/− neuronal cultures with a GPR3 adenoviral lular cyclic AMP (cAMP) generation (Supplementary Fig. 3). The
vector (Ad-GPR3). Notably, expression of Ad-GPR3 in the Arrb2−/− expression and cellular distribution of the GPR3 DRY/AAY mutant
neuronal cultures failed to increase Aβ generation, in contrast to the were similar to those of wild-type (WT) GPR3 (data not shown),
elevation in Aβ generation observed in Arrb2+/+ neuronal cultures establishing the involvement of this conserved motif in the activation
(Fig. 2c,d). After GPR3 transduction, GPR3 expression was equiva- of GPR3. Notably, mutation of the DRY motif also led to an increase
lent in the Arrb2+/+ and Arrb2−/− neuronal cultures, and expression in β-arrestin 2 recruitment to GPR3 (Fig. 3b) and release of Aβ40
of the γ-secretase complex components and APP was unaffected by and Aβ42 (Fig. 3c).
the absence of β-arrestin 2 or transduction with GPR3 (Fig. 2e). We then mutated conserved serine residues in the C terminus of
Consistent with our previous work29, overexpression of GPR3 resulted GPR3, which could serve as putative binding sites for β-arrestin 2
in a modest decrease in the amount of APP-CTF, whereas deficiency (Fig. 3a). The GPR3 serine mutant was defective in its ability to inter-
of Arrb2 tended to increase accumulation of APP-CTF (Fig. 2e). act with β-arrestin 2, as measured with the PathHunter complementa-
tion assay (Fig. 3d). The expression and cellular distribution of the
The C terminus of GPR3 is required for enhanced Ab generation GPR3 serine mutant and WT GPR3 were similar (data not shown).
We further investigated the mechanism of the β-arrestin 2–mediated Mutation of these serine residues also resulted in a reduction in
effect on Aβ generation and γ-secretase activation by generating the generation of Aβ40 and Aβ42 (Fig. 3e), providing validation that
mutations in GPR3 that alter either G protein activation or β-arrestin β-arrestin 2 recruitment to GPR3 is required for Aβ generation.
recruitment29. Alanine substitution of the first two amino acids of
a conserved motif in the second intracellular loop of GPR3 (the Localization of b-arrestin 2 and the g-secretase complex in DRMs
AspArgTyr (DRY) motif mutated to AlaAlaTyr (AAY), termed here The experiments described above indicate that the effect of β-arrestin 2
the GPR3 DRY/AAY mutant) renders a GPCR incapable of activating on Aβ generation is downstream of cleavage of APP by the α- and
G proteins while retaining its ability to activate β-arrestin–mediated β-secretases. Previous observations also indicated that both GPR3

46 VOLUME 19 | NUMBER 1 | JANUARY 2013  nature medicine


articles

a IP: Ps1-CTF IP: Aph-1a


c Input Bound
e IP: Aph-1a IP: β-arrestin 2
Input Unbound Bound Input Unbound Bound Input Bound Input Bound

2
tin
Ps1 Control Ps1 Control Aph-1a Control Aph-1a Control

2 –/–

2 –/–

2 –/–

2 –/–
s
l

3 –/–

3 –/–

3 –/–

3 –/–
tro

re
Nct

ar
on
IP:

rb

rb

rb

rb
pr

pr

pr

pr
T

T
β-

Ar

Ar

Ar

Ar
W

W
C

G
Ps1-NTF Nct
Nct
Ps1-CTF Aph-1a
Ps1-NTF
Aph-1a β-arrestin 2
Pen 2 Ps1-CTF

β-arrestin 2 Aph-1a

β-arrestin 2

b IP: Ps1-CTF IP: Aph-1a


d Input Bound
f IP: Aph-1a IP: β-arrestin 2
Input Unbound Bound Input Unbound Bound Input Bound Input Bound

2
in
st
Ps1 Control Ps1 Control Aph-1a Control Aph-1a Control

l
tro

/–

/–
re

3 –/–

3 –/–
–/

–/
2 –

2 –
–/

–/
ar

on

2
3

3
Nct IP:

rb

rb

rb

rb
pr

pr

pr

pr
β-

T
C

Ar

Ar

Ar

Ar
W

W
G

G
Ps1-NTF Nct Nct
Ps1-CTF Aph-1a
Ps1-NTF
Aph-1a β-arrestin 2
Ps1-CTF
Pen 2
β-arrestin 2 Aph-1a
© 2013 Nature America, Inc. All rights reserved.

β-arrestin 2

Figure 5  β-arrestin 2 interacts with the Aph-1a subunit of the γ-secretase complex. (a,b) Immunoprecipitation (IP) of cell lysates from the N2a
neuroblastoma cell line after extraction in 1% CHAPSO-containing (a) or 1% TX-100–containing (b) buffer with antibodies to Ps1-CTF (left), Aph-1a
(B80.3; right) or negative control antibodies and immunoblot analysis with the indicated antibodies to the γ-secretase complex subunits or β-arrestin 2.
(c,d) Immunoprecipitation of cell lysates from the N2a neuroblastoma cell line after extraction in 1% CHAPSO-containing (c) or 1% TX-100–containing
(d) buffer with β-arrestin 2–specific or negative control antibodies and immunoblot analysis with the indicated antibodies to the γ-secretase complex
subunits or β-arrestin 2. (e,f) Immunoprecipitation of cortical brain samples from WT, Gpr3−/− and Arrb2−/− mice after extraction in 1% CHAPSO-
containing (e) or 1% TX-100–containing (f) buffer with antibodies to Aph-1a (B80.3; left) or β-arrestin 2 (right) and immunoblot analysis with the
indicated antibodies to the γ-secretase complex subunits or β-arrestin 2.

and β2-AR affect γ-secretase–mediated proteolysis of APP29,30. Classic We then determined whether increased localization of the indi-
inhibition of γ-secretase activity36 leads to accumulation of APP-CTF; vidual γ-secretase subunits in DRMs also coincided with the presence
however, genetic silencing of β-arrestin 2 leads to only a limited or of a more active γ-secretase complex in DRMs. JC-8 is a photoreactive,
barely observable accumulation of APP-CTF (Figs. 1i and 4). To biotinylated derivative of the highly specific and potent transition-
determine whether β-arrestin 2 affects turnover of the APP-CTF, state analog inhibitor L-685,458 (ref. 43) that only binds the cata-
we treated HEK293-APP695 cells (after silencing β-arrestin 2) and lytically active γ-secretase complex44. We combined DRM fractions
Arrb2−/− neuronal cultures with the proteasome inhibitor lactacystin 2–4 and the non–lipid raft fractions (pooled fractions 9–12) from
npg

and found an accumulation of the APP-CTF under these conditions empty vector–transfected control and β-arrestin 2–transfected cells.
(Fig. 4a–d), suggesting that downregulation of β-arrestin 2 leads not We normalized the fractions for γ-secretase expression and incubated
only to inhibition of γ-secretase activity but also to increased APP- these fractions with JC-8. We used unlabeled L-685,458 as a control
CTF turnover through the proteasome. Given the complexity of the to compete with the biotinylated inhibitor and photoactivation to
signaling pathways modulated by the β-arrestins, an effect on Aβ crosslink the biotinylated inhibitor to the active γ-secretase complex45.
generation at multiple levels is not surprising. Nevertheless, from a Subsequent recovery of the biotinylated polypeptides revealed that the
therapeutic point of view, stimulation of APP-CTF turnover could photoprobe readily labeled the PS1-CTF in the lipid raft DRM frac-
be advantageous given that accumulation of the APP-CTF correlates tions (Fig. 4g). We were not able to detect labeling in the non–lipid
with cognitive deficits in mice37. raft fractions (Fig. 4g). Overexpression of β-arrestin 2 resulted in a
Previous studies indicated that GPCRs, the γ-secretase complex twofold to threefold increase of the active γ-secretase complex pool
and Aβ generation are localized in DRMs29,38–42. Therefore, we in the DRMs (Fig. 4h).
assessed the distribution of the γ-secretase complex subunits and
β-arrestin 2 in lipid raft and non–lipid raft domains by differential b-arrestin 2 interacts with the Aph-1a g-secretase subunit
flotation after sucrose density gradient centrifugation. The γ-secretase To determine whether β-arrestin 2 physically associates with
complex subunits co-distributed in low-density fractions 3 and 4. the γ-secretase complex, we performed coimmunoprecipitation exper-
Notably, GPR3 (ref. 29) and β-arrestin 2 also accumulated in iments in untransfected N2a neuroblastoma cells. The γ-secretase
DRMs (Fig. 4e). Moreover, overexpression of β-arrestin 2 (Fig. 4e,f), subunits Nct, Ps1, Aph-1a and Pen 2 coimmunoprecipitated with
led to an enrichment of the γ-secretase subunits NCT, PS1-CTF, β-arrestin 2 in a 3-[(3-cholamidopropyl) dimethylammonio]-
APH-1A and PEN 2 in the detergent-resistant, buoyant fractions 2-hydroxy-1-propanesulfonate (CHAPSO)-containing buffer (Fig. 5a),
(Fig. 4f). In contrast, silencing of β-arrestin 2 led to an appreciable which maintains the integrity of the intact γ-secretase complex46,47.
decrease in the distribution of the γ-secretase complex in DRMs In contrast, the detergent Triton X-100 (TX-100) dissociates the
(Supplementary Fig. 4a,b). γ-secretase complex48. In the presence of TX-100, β-arrestin 2 only

nature medicine  VOLUME 19 | NUMBER 1 | JANUARY 2013 47


Articles

a b c implications for the pathogenesis of Alzheimer’s disease. β-arrestin 2


+/

+/

–/
APP/PS1 Arrb2+/+ APP/PS1 Arrb2+/+

2
2

2
induces the redistribution of an inactive γ-secretase complex

rb
rb

rb
+/–
+/– APP/PS1 Arrb2

Ar
APP/PS1 Arrb2–/–

Ar

Ar
APP/PS1 Arrb2–/–

1
1

1
APP/PS1 Arrb2
toward a DRM-associated, active γ-secretase pool through direct

PS
PS

PS
(to APP/PS1 Arrbb2+/+)

(to APP/PS1 Arrbb2+/+)

P/
P/

P/
1.5 1.5

Aβ42 normalized value


Aβ40 normalized value

interaction with Aph-1a. We corroborated the overexpression studies

AP
AP

AP
NCT
1.0 1.0
PS1-NTF
with several loss-of-function experiments that confirmed the overall
** * ** PS1-CTF effects of β-arrestin 2 on γ-secretase–mediated Aβ generation.
0.5 * 0.5
+ *** ++ APP-FL Our data also suggest additional mechanisms, including increased
+ APP-CTF APP-CTF turnover, by which APP metabolism is affected by
β-actin
β-arrestin 2 downregulation.
s

x
pu

te

pu

te
or

or
am

am

This study provides insight into the mechanism of γ-secretase regu-


C

C
oc

oc
p

lation by GPR3 (ref. 29) and β2-AR30, two GPCRs that have previously
ip

ip
H

Figure 6  β-arrestin 2 contributes to Aβ generation in an Alzheimer’s been implicated in the pathogenesis of Alzheimer’s disease. Most nota-
disease transgenic mouse model. (a,b) Hippocampal and cortical bly, we have determined that β-arrestins are aberrantly expressed in
concentrations of soluble Aβ40 (a) and Aβ42 (b) in 3-month-old APP/PS1 the brain of individuals with Alzheimer’s disease and genetic deletion
transgenic mice crossed with wild-type Arrb2+/+, Arrb2+/− or Arrb2−/− mice of β-arrestin 2 reduces the accumulation of endogenous mouse Aβ.
determined by ELISA. *P < 0.05 relative to Arrb2+/+, +P < 0.01 relative Furthermore, in an Alzheimer’s disease mouse model, we have shown
to Arrb2+/+ for hippocampal Aβ40; **P < 0.001 relative to Arrb2+/+,
that β-arrestin 2 is also involved in Aβ generation, which provides
***P < 0.0001 relative to Arrb2+/+ for cortical Aβ40; *P < 0.05 relative
to Arrb2+/+, +P < 0.01 relative to Arrb2+/+ for hippocampal Aβ42; evidence for the therapeutic potential of β-arrestins in Alzheimer’s
**P < 0.005 relative to Arrb2+/+,++P < 0.001 relative to Arrb2+/+ for disease. Ligands that show bias for either G protein–mediated
cortical Aβ42 by ANOVA and Dunnett’s post test. n = 6 independent (G protein–biased) or β-arrestin–mediated (β-arrestin–biased) sig-
female mice per cross. Error bars, s.e.m. (c) Immunoblot of the expression naling are being intensively investigated because they could selectively
© 2013 Nature America, Inc. All rights reserved.

of the γ-secretase complex components and APP in brain samples from promote beneficial signaling and even block or negate detrimental or
APP/PS1 Arrb2+/+, APP/PS1 Arrb2+/− and APP/PS1 Arrb2−/− mice.
unwanted actions of receptor activation (for example, side effects, toxicity
or tolerance). Recently, the GPCR M3-muscarinic receptor–dependent
coimmunoprecipitated with Aph-1a (Fig. 5b). In the reciprocal regulation of learning and memory has been shown to require receptor
assay, the interaction between β-arrestin 2 and the intact γ-secretase phosphorylation and β-arrestin recruitment independent of G protein
complex was preserved in a CHAPSO-containing buffer (Fig. 5c); signaling50. These data suggest that the development of biased ligands
however, β-arrestin 2 only coimmunoprecipitated with the Aph-1a could be beneficial for both learning and memory and, potentially, the
subunit (Fig. 5d) in the presence of a TX-100 solubilization treatment of cognitive disorders such as Alzheimer’s disease.
buffer. We confirmed this interaction in vivo, showing that in the Arrb2−/− mice develop normally in the absence of an apparent
presence of either CHAPSO (Fig. 5e) or TX-100 detergent (Fig. 5f), Notch-deficiency phenotype. Instead, they show increased analge-
β-arrestin 2 coimmunoprecipitated with the Aph-1a subunit of sia in response to morphine24 because of misregulated internali-
the γ-secretase complex in extracts of cortical brain samples from WT zation and desensitization of the µ-opioid receptor51. The current
but not Gpr3−/− or Arrb2−/− mice. Furthermore, we observed colocali- study indicates that APP-CTF does not accumulate in APP/PS1
zation of β-arrestin and APH-1A after coexpression of β-arrestin 2 Arrb2−/− mice, which has been suggested to occur after treatment
and APH-1A in HeLa cells (Supplementary Fig. 5). Collectively, with certain γ-secretase inhibitors37. Therefore, a physiologically
these data suggest that expression of β-arrestin 2 regulates Aβ gen- relevant regulatory mechanism of the modulation of Aβ generation
eration through interaction with the γ-secretase complex and redis- by the γ-secretase, potentially mediated through β-arrestin 2, could
npg

tribution and accumulation of the active γ-secretase complex in be beneficial in preventing the adverse side effects associated with
DRM domains. direct γ-secretase inhibition, such as interference with Notch signal-
ing52 or APP-CTF accumulation37. As it becomes increasingly evident
Genetic deletion of b-arrestin 2 reduces Ab generation that presymptomatic and/or very early symptomatic treatments are
We then established the in vivo consequence of the absence of necessary to prevent the onset of dementia, the work here suggests
β-arrestin 2 on Aβ generation in the APP/PS1 transgenic mouse a previously unexplored avenue involving β-arrestin 2 inhibition for
model for Alzheimer’s disease. This model coexpresses two familial therapeutic intervention and prevention in Alzheimer’s disease.
Alzheimer’s disease–linked mutations, APP with the KM670/671NL
‘Swedish’ mutation and PS1 with the L166P mutation49. Both het- Methods
erozygosity and complete genetic ablation of β-arrestin 2 expression Methods and any associated references are available in the online
resulted in a marked reduction in Aβ40 and Aβ42 generation in the version of the paper.
hippocampus and cortex of 3-month-old APP/PS1 Arrb2+/− and
Note: Supplementary information is available in the online version of the paper.
APP/PS1 Arrb2−/− mice (Fig. 6a,b) with no effects on the expression
of NCT, PS1 or APP-FL (Fig. 6c), providing in vivo evidence for
the involvement of endogenous β-arrestin 2 in Aβ generation in an Acknowledgments
We are grateful to R.J. Lefkowitz and S. Ahn (Duke University Medical Center,
Alzheimer’s disease mouse model. Durham, North Carolina, USA) for the generous gift of the β-arrestin 2 wild-type
and knockout mouse embryonic fibroblasts, the Arrb1−/− and Arrb2−/− mice,
DISCUSSION the β-arrestin 2–GFP-Flag cDNA and helpful discussion. We thank M. Jucker
β-arrestins are scaffolding proteins that are intimately involved (University of Tübingen, Germany) for the gift of APP/PS1 transgenic mice. We
greatly appreciate the kind gift of human control and Alzheimer’s disease brain
in numerous aspects of GPCR signaling and regulation. Here we samples from K. Bossers and D.F. Swaab (Netherlands Institute for Neuroscience,
present evidence implicating this class of proteins in the regulation Amsterdam, The Netherlands) and C. Troakes (the London Neurodegenerative
of γ-secretase proteolytic activity and Aβ generation, with potential Diseases Brain Bank, London, UK). We thank M. Mercken (Johnson & Johnson

48 VOLUME 19 | NUMBER 1 | JANUARY 2013  nature medicine


articles

Pharmaceuticals Research and Development, Beerse, Belgium) for the antibodies 23. Conner, D.A. et al. β-arrestin1 knockout mice appear normal but demonstrate
to Aβ. We are grateful to Y. Li (Memorial Sloan Kettering Cancer Center, New altered cardiac responses to β-adrenergic stimulation. Circ. Res. 81, 1021–1026
York, USA) for the kind initial gift of JC-8. This work was supported by a Mentored (1997).
24. Bohn, L.M. et al. Enhanced morphine analgesia in mice lacking β-arrestin 2. Science
New Investigator Research grant from the Alzheimer’s Association to A.T., the
286, 2495–2498 (1999).
Fund for Scientific Research Flanders, KU Leuven, a Methusalem grant from 25. Ferguson, S.S. et al. Role of β-arrestin in mediating agonist-promoted G protein–
the KU Leuven and the Flemish government, and the Foundation for Alzheimer coupled receptor internalization. Science 271, 363–366 (1996).
Research (SAO/FRMA) to B.D.S. B.D.S. is the Arthur Bax and Anna Vanluffelen 26. Lohse, M.J., Lefkowitz, R.J., Caron, M.G. & Benovic, J.L. Inhibition of β-adrenergic
chair for Alzheimer’s disease. receptor kinase prevents rapid homologous desensitization of β 2-adrenergic
receptors. Proc. Natl. Acad. Sci. USA 86, 3011–3015 (1989).
AUThOR CONTRIBUTIONS 27. Ahn, S., Shenoy, S.K., Wei, H. & Lefkowitz, R.J. Differential kinetic and spatial
patterns of β-arrestin and G protein–mediated ERK activation by the angiotensin II
A.T. and B.D.S. designed the experiments and wrote the manuscript. A.T., K.H.,
receptor. J. Biol. Chem. 279, 35518–35525 (2004).
A.S., E.V. and Y.H. conducted the experiments. M.C. conducted the qPCR 28. Luttrell, L.M. et al. β-arrestin–dependent formation of β2 adrenergic receptor-Src
experiments. G.D.K. synthesized JC-8. S.M. conducted the immunofluorescence protein kinase complexes. Science 283, 655–661 (1999).
image analysis. 29. Thathiah, A. et al. The orphan G protein–coupled receptor 3 modulates amyloid-β
peptide generation in neurons. Science 323, 946–951 (2009).
COMPETING FINANCIAL INTERESTS 30. Ni, Y. et al. Activation of β2-adrenergic receptor stimulates γ-secretase activity and
The authors declare competing financial interests: details are available in the online accelerates amyloid plaque formation. Nat. Med. 12, 1390–1396 (2006).
version of the paper. 31. Teng, L., Zhao, J., Wang, F., Ma, L. & Pei, G.A. GPCR/secretase complex regulates
β- and γ-secretase specificity for Aβ production and contributes to AD pathogenesis.
Cell Res. 20, 138–153 (2010).
Published online at http://www.nature.com/doifinder/10.1038/nm.3023. 32. Olson, K.R. & Eglen, R.M. β galactosidase complementation: a cell-based
Reprints and permissions information is available online at http://www.nature.com/ luminescent assay platform for drug discovery. Assay Drug Dev. Technol. 5, 137–144
reprints/index.html. (2007).
33. Gáborik, Z. et al. The role of a conserved region of the second intracellular loop in
AT1 angiotensin receptor activation and signaling. Endocrinology 144, 2220–2228
1. Ballatore, C., Lee, V.M. & Trojanowski, J.Q. Tau-mediated neurodegeneration (2003).
in Alzheimer’s disease and related disorders. Nat. Rev. Neurosci. 8, 663–672 34. Shenoy, S.K. et al. β-arrestin–dependent, G protein–independent ERK1/2 activation
© 2013 Nature America, Inc. All rights reserved.

(2007). by the β2 adrenergic receptor. J. Biol. Chem. 281, 1261–1273 (2006).


2. Götz, J., Ittner, A. & Ittner, L.M. Tau-targeted treatment strategies in Alzheimer’s 35. Wei, H. et al. Independent β-arrestin 2 and G protein–mediated pathways for
disease. Br. J. Pharmacol. 165, 1246–1259 (2012). angiotensin II activation of extracellular signal-regulated kinases 1 and 2. Proc.
3. Holtzman, D.M., Goate, A., Kelly, J. & Sperling, R. Mapping the road forward in Natl. Acad. Sci. USA 100, 10782–10787 (2003).
Alzheimer’s disease. Sci. Transl. Med. 3, 114ps148 (2011). 36. De Strooper, B. et al. Deficiency of presenilin-1 inhibits the normal cleavage of
4. Golde, T.E., Schneider, L.S. & Koo, E.H. Anti-aβ therapeutics in Alzheimer’s disease: amyloid precursor protein. Nature 391, 387–390 (1998).
the need for a paradigm shift. Neuron 69, 203–213 (2011). 37. Mitani, Y. et al. Differential effects between γ-secretase inhibitors and modulators
5. Karran, E., Mercken, M. & De Strooper, B. The amyloid cascade hypothesis for on cognitive function in amyloid precursor protein–transgenic and nontransgenic
Alzheimer’s disease: an appraisal for the development of therapeutics. Nat. Rev. mice. J. Neurosci. 32, 2037–2050 (2012).
Drug Discov. 10, 698–712 (2011). 38. Chini, B. & Parenti, M. G-protein coupled receptors in lipid rafts and caveolae:
6. Cramer, P.E. et al. ApoE-directed therapeutics rapidly clear β-amyloid and reverse how, when and why do they go there? J. Mol. Endocrinol. 32, 325–338 (2004).
deficits in AD mouse models. Science 335, 1503–1506 (2012). 39. Vetrivel, K.S. et al. Association of γ-secretase with lipid rafts in post-Golgi and
7. Selkoe, D.J. Resolving controversies on the path to Alzheimer’s therapeutics. endosome membranes. J. Biol. Chem. 279, 44945–44954 (2004).
Nat. Med. 17, 1060–1065 (2011). 40. Wada, S. et al. γ-secretase activity is present in rafts but is not cholesterol-
8. De Strooper, B. Aph-1, Pen-2, and Nicastrin with Presenilin generate an active dependent. Biochemistry 42, 13977–13986 (2003).
γ-Secretase complex. Neuron 38, 9–12 (2003). 41. Wahrle, S. et al. Cholesterol-dependent γ-secretase activity in buoyant cholesterol-
9. Bertram, L., Lill, C.M. & Tanzi, R.E. The genetics of Alzheimer disease: back to rich membrane microdomains. Neurobiol. Dis. 9, 11–23 (2002).
the future. Neuron 68, 270–281 (2010). 42. Yagishita, S., Morishima-Kawashima, M., Ishiura, S. & Ihara, Y. Aβ46 is processed
10. Bekris, L.M., Yu, C.E., Bird, T.D. & Tsuang, D.W. Genetics of Alzheimer disease. to Aβ40 and Aβ43, but not to Aβ42, in the low density membrane domains.
J. Geriatr. Psychiatry Neurol. 23, 213–227 (2010). J. Biol. Chem. 283, 733–738 (2008).
11. Morris, J.C. et al. APOE predicts amyloid-β but not tau Alzheimer pathology in 43. Chun, J., Yin, Y.I., Yang, G., Tarassishin, L. & Li, Y.M. Stereoselective synthesis of
cognitively normal aging. Ann. Neurol. 67, 122–131 (2010). photoreactive peptidomimetic γ-secretase inhibitors. J. Org. Chem. 69, 7344–7347
12. Fredriksson, R. & Schioth, H.B. The repertoire of G-protein–coupled receptors in (2004).
fully sequenced genomes. Mol. Pharmacol. 67, 1414–1425 (2005). 44. Chau, D.M., Crump, C.J., Villa, J.C., Scheinberg, D.A. & Li, Y.M. Familial Alzheimer
npg

13. Gudermann, T., Nurnberg, B. & Schultz, G. Receptors and G proteins as primary disease presenilin-1 mutations alter the active site conformation of γ-secretase.
components of transmembrane signal transduction. Part 1. G-protein–coupled J. Biol. Chem. 287, 17288–17296 (2012).
receptors: structure and function. J. Mol. Med. 73, 51–63 (1995). 45. Vetrivel, K.S. et al. Spatial segregation of γ-secretase and substrates in distinct
14. Watson, S.A.S. The G Protein-Coupled Receptor Factors Book (Academic, San Diego, membrane domains. J. Biol. Chem. 280, 25892–25900 (2005).
1994). 46. Li, Y.M. et al. Presenilin 1 is linked with γ-secretase activity in the detergent
15. Vassilatis, D.K. et al. The G protein–coupled receptor repertoires of human and solubilized state. Proc. Natl. Acad. Sci. USA 97, 6138–6143 (2000).
mouse. Proc. Natl. Acad. Sci. USA 100, 4903–4908 (2003). 47. Esler, W.P. et al. Activity-dependent isolation of the presenilin–γ-secretase complex
16. Thathiah, A. & De Strooper, B. The role of G protein–coupled receptors in the reveals nicastrin and a γ substrate. Proc. Natl. Acad. Sci. USA 99, 2720–2725
pathology of Alzheimer’s disease. Nat. Rev. Neurosci. 12, 73–87 (2011). (2002).
17. DeWire, S.M., Ahn, S., Lefkowitz, R.J. & Shenoy, S.K. β-arrestins and cell signaling. 48. Fraering, P.C. et al. Detergent-dependent dissociation of active γ-secretase reveals
Annu. Rev. Physiol. 69, 483–510 (2007). an interaction between Pen-2 and PS1-NTF and offers a model for subunit
18. Whalen, E.J., Rajagopal, S. & Lefkowitz, R.J. Therapeutic potential of β-arrestin– organization within the complex. Biochemistry 43, 323–333 (2004).
and G protein–biased agonists. Trends Mol. Med. 17, 126–139 (2011). 49. Radde, R. et al. Aβ42-driven cerebral amyloidosis in transgenic mice reveals early
19. Beaulieu, J.M. et al. An Akt/β-arrestin 2/PP2A signaling complex mediates and robust pathology. EMBO Rep. 7, 940–946 (2006).
dopaminergic neurotransmission and behavior. Cell 122, 261–273 (2005). 50. Poulin, B. et al. The M3-muscarinic receptor regulates learning and memory in
20. Luan, B. et al. Deficiency of a β-arrestin–2 signal complex contributes to insulin a receptor phosphorylation/arrestin-dependent manner. Proc. Natl. Acad. Sci. USA
resistance. Nature 457, 1146–1149 (2009). 107, 9440–9445 (2010).
21. Beaulieu, J.M. et al. A β-arrestin 2 signaling complex mediates lithium action on 51. Bohn, L.M., Gainetdinov, R.R., Lin, F.T., Lefkowitz, R.J. & Caron, M.G. Mu-opioid
behavior. Cell 132, 125–136 (2008). receptor desensitization by β-arrestin–2 determines morphine tolerance but not
22. Shearman, M.S. et al. L-685,458, an aspartyl protease transition state mimic, is dependence. Nature 408, 720–723 (2000).
a potent inhibitor of amyloid β-protein precursor γ-secretase activity. Biochemistry 39, 52. De Strooper, B. et al. A presenilin-1–dependent γ-secretase–like protease mediates
8698–8704 (2000). release of Notch intracellular domain. Nature 398, 518–522 (1999).

nature medicine  VOLUME 19 | NUMBER 1 | JANUARY 2013 49


ONLINE METHODS ­ easurements. Cells were harvested in 1× PBS containing complete protease
m
Reagents. Unless otherwise indicated, chemicals were purchased from inhibitors (Roche), centrifuged at 800g for 10 min and lysed in 150 mM NaCl,
Sigma-Aldrich. 50 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES), pH 7.4,
and 1% TX-100 supplemented with complete protease inhibitors for 30 min at
Antibodies and compounds. Rabbit polyclonal antibodies to human PS1-NTF 4 °C. After centrifugation at 16,000g for 15 min, the cleared cell extracts were
(B14.5, 1:1,000), mouse PS1-NTF (B19.2, 1:5,000), APH-1AL (B80.3, 1:1,000), separated on 4–12% Bis-Tris gels (Invitrogen), transferred to nitrocellulose
PEN 2 (B126.2, 1:1,000) and the APP C terminus (B63.5, 1:5,000) and the membranes, blocked and probed with the indicated antibodies for western blot
monoclonal antibody 9C3 (1:3,000) directed against the C terminus of NCT analysis. Immunodetection was performed using horseradish peroxidase (HRP)-
have been previously described and were generated in house53,54. ADAM10 coupled secondary antibodies (Bio-Rad) and the chemiluminescent detection
was detected using a polyclonal antiserum (B42.1, 1:1,000) generated against reagent Renaissance (PerkinElmer Life Sciences).
the 17 C-terminal amino acid residues of ADAM10 and was generated in house.
Antibodies to the following were purchased: Flag (M2, Sigma, 1:1,000), 22C11 Patient samples. Frozen hippocampal and entorhinal cortex tissue samples
(Chemicon, 1:1,000), 6E10 (Sigma-Aldrich, 1:1,000), β-secretase (D10E5, were obtained from the London Neurodegenerative Diseases Brain Bank. These
Cell Signaling, 1:1,000), PK (DiscoveRx, 1:500), β-arrestin (BD Biosciences, samples were pathologically confirmed but not further categorized according
1:500), β-arrestin 2 (Cell Signaling, 1:250), β-arrestin 1/2 (Santa Cruz, 1:500), to Braak stage. The second cohort of patient samples consisted of snap-frozen
Myc (9E10, 1:1,000), PS1-NTF (MAB1563, Chemicon), PS1-CTF (MAB5232, human medial frontal gyrus brain samples obtained from the Netherlands Brain
Chemicon), hemagglutinin (HA) (3F10, Roche), caveolin-1 (Santa Cruz), Bank, Amsterdam (NBB). For these samples, individual, neuropathological
calnexin, GM130 and EEA1 (Transduction lab) and β-actin (Sigma). L-685,458 reports, including Braak staging for neurofibrillary changes (NFC) and neuritic
was purchased from Calbiochem. plaques56, and clinical reports were available. For each of the six Braak stages,
seven individuals were included. Furthermore, seven individuals without any
Plasmid construction. The human GPR3 plasmid was purchased from NFC pathology were included as Braak stage 0. Samples were matched as closely
DiscoveRx. All mutations in GPR3 were generated with the XL Site-Directed as possible for sex, age, postmortem interval, cerebrospinal fluid pH and APOE
Mutagenesis Kit (Stratagene) and confirmed by DNA sequence analysis. The genotype. Only samples with a relatively high RNA integrity number (>7) were
© 2013 Nature America, Inc. All rights reserved.

β-arrestin 2–GFP-Flag plasmid was the kind gift of R.J. Lefkowitz (Duke included. Tissue dissection was performed as previously described57. Total RNA
University Medical Center, Durham, North Carolina, USA). was isolated from both the London and the Netherlands Brain Bank samples
using a combination of TRIzol-based and mirVana RNA isolation methods.
Cell lines. The CHO-K1, CHO-K1 GPR3 and CHO-K1 ADRB2 β-arrestin cell Briefly, samples were homogenized in ice-cold TRIzol (Life Technologies). After
lines were purchased from DiscoveRx. The WT HEK293, N2a and HeLa cell phase separation by the addition of chloroform, the aqueous phase was mixed
lines were purchased from American Type Culture Collection. with an equal volume of 70% RNase-free ethanol. Samples were then applied to
a mirVana filter cartridge (Ambion/Life Technologies) and processed according
Mice. The Arrb1−/− and Arrb2−/− mice (C57BL/6J background) were kindly to the manufacturer’s instructions. RNA yields and purities were determined
provided by R.J. Lefkowitz (Duke University Medical Center, Durham, North using a NanoDrop ND-1000 spectrophotometer (NanoDrop Technologies).
Carolina, USA). Wild-type C57BL/6J mice or littermate mice were used as RNA integrity was determined by the RNA integrity number as measured by
controls for all of the experiments. The APP/PS1 transgenic mice (C57BL/6J the Agilent 2100 bioanalyzer (Agilent Technologies). All brain samples were col-
background) were the kind gift of M. Jucker (University of Tübingen, Tübingen, lected according to legislation and ethical boards of the respective Brain Banks.
Germany)49. The APP/PS1 transgenic mice were crossed with Arrb2+/− and The human study was evaluated and approved by ethical committees of Leuven
Arrb2−/− mice to obtain APP/PS1 Arrb2+/− and APP/PS1 Arrb2−/− mice, University and UZ Leuven (ML5919).
respectively. Only female mice were used for the studies. The mouse studies
were approved by ethical committees of Leuven University and UZ Leuven Ab ELISA screen. Ninety-six-well plates were coated and incubated overnight
(LA1210231). with the capture antibodies previously described29 and detected using the HRP-
labeled detection antibodies29. Brains of the mice with the respective genotypes
qPCR. qPCR was performed with the SYBR Green PCR Master Mix (Exiqon) of the ages indicated (±2 weeks) were dissected after transcardial perfusion with
npg

and the LightCycler 480 Real-Time PCR System (Roche Applied Science) ice-cold PBS. The hippocampus and the cerebral cortex were removed separately
following the manufacturer’s protocol. The primers for human β-arrestin 1 and homogenized in Tissue Protein Extraction reagent (Pierce) supplemented
used were 5′-TGTTGAGGGAAGGTGCCAACCG-3′ and 5′-GATGCAAGA with complete protease inhibitor and phosphatase inhibitor tablets (Roche
TCTCCCAACAGGCCG-3′. The primers for human β-arrestin 2 used were Applied Science). The homogenized samples were briefly sonicated to shear the
5′-CCTGTAGATGGCGTGGTGCTTG-3′ and 5′-CCAGGTCTTCACGGCCA DNA and centrifuged at 4 °C for 1 h at 100,000g. The supernatant was used for
TAGCG-3′. The housekeeping primers used were to hypoxanthine guanine immunoblot analysis and Aβ ELISA measurements. Alternately, the Aβ peptides
phosphoribosyl transferase (HPRT) (5′-TGACACTGGCAAAACAATGCA-3′ were allowed to accumulate in the absence of serum in the culture supernatants
and 5′-GGTCCTTTTCACCAGCAAGCT-3′), ribosomal protein L13a (RPL13A) from HEK293, HEK293-APP695, CHO-K1 GPR3 β-arrestin or CHO-K1 ADRB2
(5′-CCTGGAGGAGAAGAGGAAAGAGA-3′ and 5′-TTGAGGACCTCTG β-arrestin cells and neuronal cultures for 16–18 h before Aβ40 and Aβ42 ELISA
TGTATTTGTCAA-3′) and β2 microglobulin (β2M) (5′-TGCTGTCTCCA analysis of the culture supernatant samples.
TGTTTGATGTATC-3′ and 5′-TCTCTGCTCCCCACCTCTAAGT-3′).
siRNA-mediated knockdown experiments. The CHO-K1 GPR3 β-arrestin,
Neuronal cultures. Primary mouse cortical or hippocampal cultures were estab- CHO-K1 ADRB2 β-arrestin or WT HEK293 cell lines were transfected with
lished from the brains of embryonic day 14 or 17 fetal mice, respectively, as siRNA directed against β-arrestin 1 or β-arrestin 2 or with control siRNA
previously described55. Briefly, the dissected brain cortices or hippocampi were using GeneSilencer (Genlantis) according to the manufacturer’s instruc-
suspended in HBSS supplemented with 0.25% trypsin and incubated at 37 °C tions as described27. Chemically synthesized, double-stranded siRNAs with
for 15 min. The tissues were then transferred to HBSS supplemented with 10% 19-nucleotide duplex RNA and 2-nucleotide 3′-dTdT overhangs were pur-
(v/v) horse serum and dissociated by repeated trituration. The dispersed cells chased from Qiagen. The siRNA sequences targeting human β-arrestin 1
were counted and plated on poly-d-lysine–coated six-well cell culture plates and β-arrestin 2 were 5′-AAAGCCUUCUGCGCGGAGAAU-3′ and 5′-AAG
in Neurobasal medium supplemented with B27 (Invitrogen). Forty-eight to GACCGCAAAGUGUUUGUG-3′, corresponding to positions 439–459 and
72 h after plating, the cells were transduced with the recombinant adenoviral 148–168 relative to the start codon, respectively. A nonsilencing RNA duplex
vectors for 24 h. The infection medium was replaced with fresh Neurobasal (5′-AAUUCUCCGAACGUGUCACGU-3′) was used as a control. Briefly, the
medium, and cells were maintained in culture for an additional 24 h. Cell culture cells were seeded on six-well plates at a cell density of 250,000 cells per well. One
supernatants were collected, centrifuged for 10 min at 800g and used for ELISA day after seeding, the cells were transfected with the siRNAs. Two days after

nature medicine doi:10.1038/nm.3023


seeding, the cells were infected with an adenoviral vector expressing enhanced MES, pH 6.5, and 150 mM NaCl) containing 1% CHAPSO and supplemented
GFP (eGFP) or APP-C99. On day 5, the medium was refreshed with serum-free with a complete protease inhibitor cocktail. Cells were lysed by sequential pas-
medium, and the cells were allowed to accumulate Aβ peptides in the condi- sage through 18-gauge (five times) and 26-gauge (ten times) needles and then
tioned medium for 16–18 h before analysis of the culture supernatant samples placed on ice for 1 h. After the removal of insoluble material by centrifugation
with the ELISA described above. at 15,000g for 15 min, the lysates were adjusted to a 45% sucrose concentration
and transferred to ultracentrifugation tubes. A discontinuous sucrose gradient
b-arrestin assay. The CHO-K1 GPR 3 β-arrestin or CHO-K1 ADRB2 β-arrestin was prepared by layering 35% and 5% sucrose in MES buffer. Samples were
cell lines were seeded on 96-well plates at a cell density of 20,000 cells per well. centrifuged at 100,000g for 16–18 h. Thirteen 960-µl fractions were collected
One day after seeding, cells were transfected with the siRNAs as described above. from the top of the gradient and used for western blot analysis.
Two days after seeding, the cells were infected with an adenoviral vector express-
ing eGFP or APP-C99. On day 5, the medium was refreshed with serum-free Photoaffinity probe and crosslinking. JC-8 was synthesized according to a
medium, and the cells were allowed to accumulate Aβ peptides in the condi- previously described procedure43. Fractions 2–4 and 9–12 from the subcellular
tioned medium for 24 h before analysis with the PathHunter β-arrestin assay fractionation experiments described above were combined, and the crosslinking
from DiscoveRx according to the manufacturer’s protocol. studies were performed as previously described45.

Coimmunoprecipitation experiments. N2a cells were seeded in 10-cm plates Structured illumination microscopy and immunofluorescence. HeLa cells
at a density of 2,000,000 cells per dish. Cell lysates were prepared 48 h after seed- were plated on glass coverslips and transfected with β-arrestin 2–GFP and APH-
ing in PBS with 1% TX-100 or 1% CHAPSO and protease inhibitors (Complete 1A. Twenty-four hours after transfection, the cells were fixed (4% paraformalde-
Protease Inhibitor Cocktail Tablets, Roche). Cell lysates were precleared with hyde, 10 min), blocked (2% BSA, 2% FBS and 1% gelatin in PBS supplemented
protein G sepharose for 1 h at 4 °C followed by centrifugation at 10,000g for with 5% serum), incubated with primary antibody (4 °C, overnight), washed
15 min. Immunoprecipitations were conducted overnight at 4 °C using the (PBS), incubated with fluorophore-conjugated secondary antibody (room tem-
appropriate antibodies or a negative control antibody (9E10). The beads were perature, 1 h), rinsed and mounted in Mowiol-containing medium. The second-
washed four times with PBS with 1% TX-100 or 1% CHAPSO and once with ary antibodies were conjugated with the following fluorophores: Alexa-546 and
© 2013 Nature America, Inc. All rights reserved.

PBS. Proteins were eluted with 1× lithium dodecyl sulfate (LDS) loading buffer. -647. Images were captured with a structured illumination microscope (Elyra
Microsomal membranes were prepared from fresh or frozen postmortem mouse S1 (Carl Zeiss, Jena, Germany) equipped with a 63× oil objective lens and a
cortical brain samples first by homogenization using a Teflon homogenizer in 1.4 numerical aperture (NA) and an Andor iXon 885 EMCCD camera) that
0.5 M sucrose PKM buffer (100 mM potassium phosphate, 5 mM MgCl2 and is capable of revealing information beyond the diffraction barrier that limits
3 mM KCl, pH 6.5) followed by centrifugation for 10 min at 8,000 r.p.m. The conventional widefield and confocal microscopes. β-arrestin 2 and APH-1A
protein concentration was determined by the Bradford dye-binding procedure were imaged at resolutions of ~110 nm and ~134 nm, respectively.
(Bio-Rad), and the samples were centrifuged at 100,000g for 1 h. The mem-
brane pellets were each solubilized with the addition of an equal volume of Statistical analyses. The data are presented as means ± s.e.m. The data were
buffer containing 2% CHAPSO (Pierce) and incubated on ice for 1 h. After analyzed using two-tailed Student’s t tests. ANOVA was performed with the
centrifugation at 100,000g for 1 h, the immunoprecipitation was performed as GraphPad Prism 5 software.
previously described58.
53. Annaert, W.G. et al. Interaction with telencephalin and the amyloid precursor protein
Endogenous cAMP assessment. CHO-K1 β-arrestin cells were transfected with predicts a ring structure for presenilins. Neuron 32, 579–589 (2001).
empty vector, WT GPR3 or the GPR3 mutant cDNA constructs (X-tremeGENE 54. Esselens, C. et al. Presenilin 1 mediates the turnover of telencephalin in hippocampal
HP, Roche). Forty-eight hours after transfection, the culture medium was replaced neurons via an autophagic degradative pathway. J. Cell Biol. 166, 1041–1054
(2004).
with serum-free DMEM, and the cells were treated with 100 µM forskolin 55. Cai, H. et al. BACE1 is the major β-secretase for generation of Aβ peptides by
in the presence or absence of 25 µM IBMX, a phosphodiesterase inhibitor, for neurons. Nat. Neurosci. 4, 233–234 (2001).
30–45 min. Intracellular cAMP levels were then measured in cells using a spe- 56. Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes.
cific cAMP assay kit (R&D Systems) according to the manufacturer’s protocol. Acta Neuropathol. 82, 239–259 (1991).
npg

57. Bossers, K. et al. Concerted changes in transcripts in the prefrontal cortex precede
neuropathology in Alzheimer’s disease. Brain 133, 3699–3723 (2010).
Subcellular fractionation. WT HEK293 cells were rinsed twice with ice-cold 1× 58. Hébert, S.S. et al. Coordinated and widespread expression of γ-secretase in vivo:
PBS, solubilized in 2-(N-morpholino)ethanesulfonic acid (MES) buffer (25 mM evidence for size and molecular heterogeneity. Neurobiol. Dis. 17, 260–272 (2004).

doi:10.1038/nm.3023 nature medicine

You might also like