ArticlePDF Available

Abstract and Figures

A long-held tenet of molecular pharmacology is that canonical signal transduction mediated by G-protein-coupled receptor (GPCR) coupling to heterotrimeric G proteins is confined to the plasma membrane. Evidence supporting this traditional view is based on analytical methods that provide limited or no subcellular resolution. It has been subsequently proposed that signalling by internalized GPCRs is restricted to G-protein-independent mechanisms such as scaffolding by arrestins, or GPCR activation elicits a discrete form of persistent G protein signalling, or that internalized GPCRs can indeed contribute to the acute G-protein-mediated response. Evidence supporting these various latter hypotheses is indirect or subject to alternative interpretation, and it remains unknown if endosome-localized GPCRs are even present in an active form. Here we describe the application of conformation-specific single-domain antibodies (nanobodies) to directly probe activation of the β2-adrenoceptor, a prototypical GPCR, and its cognate G protein, Gs (ref. 12), in living mammalian cells. We show that the adrenergic agonist isoprenaline promotes receptor and G protein activation in the plasma membrane as expected, but also in the early endosome membrane, and that internalized receptors contribute to the overall cellular cyclic AMP response within several minutes after agonist application. These findings provide direct support for the hypothesis that canonical GPCR signalling occurs from endosomes as well as the plasma membrane, and suggest a versatile strategy for probing dynamic conformational change in vivo.
Content may be subject to copyright.
LETTER doi:10.1038/nature12000
Conformational biosensors reveal GPCR signalling
from endosomes
Roshanak Irannejad
1
, Jin C. Tomshine
1
, Jon R. Tomshine
1
, Michael Chevalier
2
, Jacob P. Mahoney
3
, Jan Steyaert
4,5
,
Søren G. F. Rasmussen
6
, Roger K. Sunahara
3
, Hana El-Samad
2
, Bo Huang
2,7
& Mark von Zastrow
1,8
A long-held tenet of molecular pharmacology is that canonical
signal transduction mediated by G-protein-coupled receptor (GPCR)
coupling to heterotrimeric G proteins is confined to the plasma
membrane. Evidence supporting this traditional view is based on
analytical methods that provide limited or no subcellular resolu-
tion
1
. It has been subsequently proposed that signalling by inter-
nalized GPCRs is restricted to G-protein-independent mechanisms
such as scaffolding by arrestins
2,3
, or GPCR activation elicits a dis-
crete form of persistent G protein signalling
4–9
, or that internalized
GPCRs can indeed contribute to the acute G-protein-mediated
response
10
. Evidence supporting these various latter hypotheses is
indirect or subject to alternative interpretation, and it remains
unknown if endosome-localized GPCRs are even present in an
active form. Here we describe the application of conformation-
specific single-domain antibodies (nanobodies) to directly probe
activation of the b
2
-adrenoceptor, a prototypical GPCR
11
, and its
cognate G protein, G
s
(ref. 12), in living mammalian cells. We show
that the adrenergic agonist isoprenaline promotes receptor and G
protein activation in the plasma membrane as expected,but also in
the early endosome membrane, and that internalized receptors con-
tribute to the overall cellular cyclic AMP response within several
minutes after agonist application. These findings provide direct
support for the hypothesis that canonical GPCR signalling occurs
from endosomes as well as the plasma membrane, and suggest a
versatile strategy for probing dynamic conformational change
in vivo
.
Ligand binding to the extracellular surface of the b
2
-adrenoceptor
(b
2
-AR) stabilizes an activating conformational change in the receptor
that promotes guanine nucleotide dissociation from the cytoplasmic
GTP-binding protein Ga
s
; this represents the critical biochemical event
initiating classical GPCR signal transduction (Fig. 1a)
13
. Activated b
2
-
ARs are substrates for phosphorylation and binding of b-arrestins,
events which inhibit interaction with G proteins and promote endocy-
tosis of receptors via clathrin-coated pits (CCPs)
14,15
.Acuteb
2
-AR Ga
s
signalling is thus traditionally thought to be restricted to the plasma
membrane
14,16,17
. However, to our knowledge, this assumption has not
been directly tested. To do so, we generated a biosensor of activatedb
2
-
AR based on a conformation-specific single-domain camelid antibody
(Nb80) used in recent structural studies
18,19
. We reasoned that this
nanobody, which selectively binds the agonist-occupied b
2
-AR and
is able to stabilize an activated receptor conformation when present
in vitro at high concentration, might act as a sensor of receptor activa-
tion when expressed at relatively low concentration in intact cells
(Fig. 1b). This proved to be the case; in cells maintained in the absence
of agonist, Nb80 fused to enhanced green fluorescent protein (Nb80–
GFP) localized to the cytoplasm and not with b
2
-ARs present in the
plasma membrane (Fig. 1c, 0 min, top; Pearson’s coefficient50.135).
Line scan analysis verified the cytoplasmic distribution of Nb80–
GFP before b
2
-AR activation (Fig. 1d, top) as expected because the
cytoplasmic concentration of Nb80–GFP achieved in our experiments
(approximately 20 nM) was considerably lower than the equilibrium
dissociation constant estimated in vitro for Nb80 binding to purified
b
2
-ARs in the absence of agonist (0.76 60.14 mM; Supplementary Fig.
1a–d). After application of the adrenergic agonist isoprenaline (10 mM),
Nb80–GFP was rapidly recruited to the plasma membrane and co-
localized there with b
2
-ARs (Fig. 1c, middle; Pearson’s coefficient 5
0.625). Line scan analysis verified robust Nb80–GFP recruitment to
the plasma membrane and concomitant depletion from the cytoplasm
(Fig. 1d, middle), consistent with the much higher affinity of Nb80 for
isoprenaline-activated b
2
-ARs (2.9 60.5 nM; Supplementary Fig. 1d).
Agonist-induced membrane recruitment of Nb80–GFP was specific
because the D1 dopamine receptor (DRD1), which is also G
s
-coupled
but does not bind Nb80 in vitro (data not shown), failed to recruit
Nb80–GFP to the plasma membrane in response to dopamine (10 mM)
application (Supplementary Fig. 2). Furthermore, b
2
-AR–cyan fluor-
escent protein (CFP) and Nb80–yellow fluorescent protein (YFP) gene-
rated a pronounced fluorescence (Fo¨rster) resonance energy transfer
(FRET) signal after isoprenaline application whereas DRD1–CFP did
not (Supplementary Fig. 3a, b).
b
2
-AR internalization began 1 to 2min after Nb80–GFP recruit-
ment to the plasma membrane, indicated by the emergence of surface-
labelled b
2
-AR in peripheral cytoplasmic vesicles. Nb80–GFP did not
co-localize with b
2
-AR-containing endocytic vesicles upon first
appearance (Fig. 1c, middle, arrow in merged image points to an
example) but was recruited at later time points (Fig. 1c, bottom,
Pearson’s coefficient 50.702; examples are indicated by arrowheads).
Endosome recruitment of Nb80–GFP was evident by line scan analysis
(Fig. 1d, bottom; line scans are from the representative individual
examples with further quantification in legend) and localized to
EEA1-marked early endosomes (Pearson’s coefficient 50.846;
Supplementary Fig. 4) through which b
2
-ARs iteratively cycle in the
presence of agonist
20
.b
2
-AR-containing endosomes were initially
devoid of Nb80–GFP and later acquired Nb80–GFP during their
movement (Supplementary Videos 1 and 2). Interaction at endosomes
was verifiedby b
2
-AR–CFP andNb80–YFP normalizedFRET (nFRET)
(Supplementary Fig. 3c). These results suggest that b
2
-AR activation
initiates a precisely choreographed series of events: Nb80–GFP is first
recruited from the cytoplasm to the plasma membrane, then b
2
-ARs
internalize devoid of Nb80–GFP, followed by a second phase of Nb80–
GFP recruitment to the internalized b
2
-ARs.
Nb80–GFP recruitment to endosomes required b
2
-ARs because a
phosphorylation-deficient mutant version of the b
2
-AR (b
2
-AR-3S,
with three serine mutations) that couples to Ga
s
but is impaired in
agonist-induced endocytosis
21
recruited Nb80–GFP to the plasma mem-
brane but produced much less recruitment to endosomes (Fig. 1e, top).
Nb80–GFP co-localized with b
2
-AR-3S after agonist-induced activation
(Pearson’s coefficient 50.674) but this was largely restricted to the
1
Department of Psychiatry, University of California, San Francisco, California 94158, USA.
2
Department of Biochemistry & Biophysics, University of California, San Francisco, California 94158, USA.
3
Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.
4
Department of Molecular and Cellular Interactions, Vrije Universiteit Brussel, B-1050 Brussels,
Belgium.
5
Structural Biology Research Centre, VIB, B-1050 Brussels, Belgium.
6
Department of Neuroscience and Pharmacology, The Panum Institute, University of Copenhagen, 2200 Copenhagen N,
Denmark.
7
Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, USA.
8
Department of Cellular & Molecular Pharmacology, University of California, San
Francisco, California 94158, USA.
534 | NATURE | VOL 495 | 28 MARCH 2013
Macmillan Publishers Limited. All rights reserved
©2013
20 min Iso
20 min Iso
+
5 min CGP
ab
cd
ef
Normalized uorescence intensity
20 min Iso
+
20 min CGP
β2-AR-3S
β2-AR
Merged
Merged
Iso
0 min
3.5 min
20 min
Nb80–GFP
Nb80–GFP
Distance
(p
ixels
)
)
g
1
Agonist
binding
2
Receptor
activation
3
G protein
activation
4
cAMP
production
RR* R*
αα αα
ββ β
γγ γ
AC
GTP ATP
cAMP
GDPGTP
GDP
GDP
R*R
Blosensor of
receptor activation
Nb80
GFP
Nb80–GFP
0 min
3.5 min
20 min
120
110
100
90
80
70
60
50
120
110
100
90
80
70
60
50
120
110
100
90
80
70
60
50
Normalized uorescence intensity
0
40
80
120
160
200
20
60
100
140
180
220
Distance (pixels)
120
110
100
90
80
70
60
50
120
110
100
90
80
70
60
50
120
110
100
90
80
70
60
50
0
20
40
60
80
100
120
140
400
300
200
100
0 250 500 750 1,000 1,250 1,500
Time (s)
Cytoplasmic Nb80 (% of initial)
+ CGP
Figure 1
|
Nb80–GFP detects activatedb
2
-ARs in
the plasma membrane and endosomes. a,The
main events in b
2
-AR cAMP signalling include
agonist binding (step 1), conformational activation
of the receptor (step 2) that is coupled to
conformational activation of G
s
(step 3) that
produces guanine nucleotide exchange on G
s
and
subsequent activation of adenylyl cyclase (AC)
(step 4). b, Scheme for detecting conformational
activation of b
2
-AR with Nb80–GFP.
c, Representative Nb80–GFP (green) and b
2
-AR
(red) localization at the indicated time (left) after
10 mM isoprenaline addition (.30 Nb80–GFP
positive endosomes per cell observed at 20 min;
n529 cells, 10 experiments). d, Representative
individual Nb80–GFP line scans (shown at the
same magnification as panel c). e, Representative
Nb80–GFP (green) and b
2
-AR-3S (red)
localization after 20 min of isoprenaline treatment
(top) followed by reversal with 50 mM CGP-12177
for the indicated times (6.4 Nb80–GFP positive
endosomes per cell; n540 cells, 3 experiments).
f, Representative individual Nb80–GFP line scans.
g, Recovery of cytoplasmic Nb80–GFP
fluorescence (black) or bleaching control of the
plasma membrane b
2
-AR-3S (grey)
(mean 6s.e.m., n55 experiments). Scale bars,
10 mm.
Surface Nb80–GFP
(% of maximum)
390
345
β2-AR β2-AR
e
β2-AR
Merged
Merged
Nb80–mRuby
Nb80–GFP Nb80–GFP
372 375 378 381 384 387
273
c
b
a
Time (s) 321 324 327 330 d318
Iso 0 min 2 min 2.5 min 10 min
Nb80–GFP
β2-AR
Merged
100
75
50
25
0
0 2 2.5 10
β2-AR
Nb80–GFP
Time (s)
Time
Time
Time (min)
Figure 2
|
Nb80–GFP accumulates on b
2
-AR-
containing endosomes after their formation.
a,b
2
-AR (red) and Nb80–GFP (green) at the
indicated times after isoprenaline addition. Scale
bar, 10 mm. b, Average Nb80–GFP fluorescence
measured in the TIRF illumination field at the
indicated times (mean 6s.e.m., n57 cells).
c, TIRF image series showing b
2
-AR (red) and
Nb80–GFP (green) in sequential frames.
d, Kymograph of an individual b
2
-AR-containing
endosome (red, Alexa555) showing Nb80–GFP
(green) acquisition over 4 min. e, Kymograph of
b
2
-AR (green, Alexa488) and Nb80–mRuby (red)
over 6 min.
LETTER RESEARCH
28 MARCH 2013 | VOL 495 | NATURE | 535
Macmillan Publishers Limited. All rights reserved
©2013
plasma membrane (line scan analysis in Fig. 1f is representative; further
quantification in the figurelegend). Nb80–GFP membrane recruitment
was also reversible because the biosensor returned to a cytoplasmic
distribution after addition of the competitive antagonist CGP-12177
(Fig. 1e, f, middle and bottom rows; Pearson’s coefficient50.106),
verified by recovery of Nb80–GFP fluorescence intensity in the cyto-
plasm (Fig. 1g, see also Supplementary Video 3). Thus, Nb80–GFP
detected activated b
2
-ARs both in the plasma membrane and endo-
somes after acute agonist application.
Discrete Nb80–GFP recruitment phases were clearly resolved by total
internal reflection fluorescence (TIRF) microscopy that selectively detects
events occurring in the plasma membrane and extending approximately
100 nm into the peripheral cytoplasm
22
.First,within2minafteragonist
application, Nb80–GFP was progressively recruited to the plasma mem-
brane in a diffuse distribution (Fig. 2a, compare first and second columns
fromleft;quantificationinFig.2b).Nb80GFPdidnotco-localizewith
b
2
-ARs when they clustered in relatively static punctae characteristic of
receptor-containing CCPs
23
(Fig. 2a, third column). Second, over a period
of several additional minutes, Nb80–GFP was recruited to a discrete
population of highly mobile punctae (Fig. 2a, fourth column) represent-
ing peripheral b
2
-AR-containing endocytic vesicles
23
.
Sequential TIRF imaging emphasized the distinction between rela-
tively static b
2
-AR puncta not co-localizing with Nb80–GFP (Fig. 2c,
arrowhead indicates an example) and mobile endosomes that did
(Fig. 2c, arrow indicates a representative example; many are visible
in Supplementary Video 4). Later recruitment of Nb80–GFP occurred
rather suddenly, typically within approximately 5 s (Supplementary
Video 5), as also evident in kymographs of individual endosome trajec-
tories (Fig. 2d). Ruling out potential artefacts of wavelength-dependent
differences in TIRF microscopy illumination depth, later recruitment of
the biosensor to endosomes was similarly observed when the excitation
wavelengths used to detect receptor and biosensor were reversed (Fig. 2e).
Nb80–GFP did not detectably concentrate in CCPs labelled with
either of two independent markers, the adaptor protein b-arrestin-2
(Fig. 3a and Supplementary Video 6) or the coat protein component
clathrin light chain (Fig. 3b and Supplementary Video 7). Represen-
tative examples are shown and this was verified across multiple cells
and experiments (Pearson’s coefficient 50.365 and 0.319, respect-
ively, with numbers of replicates specified in the legend). In contrast,
and as expected based on previous work, extensive co-localization
of b
2
-AR with b-arrestin was observed under the same conditions
(Pearson’s coefficient 50.677). Separation of Nb80–GFP localization
from that of either b-arrestin or clathrin was clear in line scans(bottom
panels of Fig. 3a, b were derived from the images shown and are
representative, further quantification is in the legend).
A simple interpretation is that Nb80–GFP associates with activated
b
2
-ARs in the plasma membrane but then dissociates before receptors
cluster in CCPs and internalize. This was surprising because b
2
-AR
clusteringin CCPs occurs much more rapidly
23
than the overallreversal
rate of Nb80–GFP recruitment observed after agonist washout in
intact cells (Fig. 1g), or the kinetics of Nb80 dissociation from purified
b
2
-ARs in vitro (Supplementary Fig. 1b, e). One possibility is that
Nb80–GFP dissociation is accelerated during the clustering process,
by mechanisms such as receptor phosphorylation or steric exclusion
mediated by b-arrestins or other CCP-associated components. An
alternative possibility is that the fraction of b
2
-ARs that have bound
Nb80–GFP in the plasma membrane by the time of the clustering
reaction are unable to enter CCPs, and only those b
2
-ARs not initially
bound to Nb80–GFP in the plasma membrane are able to cluster in
CCPs and subsequently internalize. In either case, the data clearlyindi-
cate that Nb80–GFP associateswith b
2
-ARs after endocytosis, and after
uncoating of the endocytic vesicle has occurred (Fig. 3c). Accordingly,
Nb80–GFP recruitment to b
2
-AR-containing endosomes cannot repre-
sent an artefact of persistent nanobody binding from the plasma
membrane; instead, this observation reveals that b
2
-ARs present in
early endosomes are in an activated conformation.
Normalized uorescence
intensity
Distance (pixels)
Iso β-arrestin Nb80–GFP MergedIso
0 min
5 min
10 min
β-arrestin
Nb80
50
40
30
20
10
00 10 20 30
Normalized uorescence
intensity
Clathrin
Nb80
Nb80–GFP MergedIso Clathrin
0 min
5 min
10 min
0
20
40
60
80
0 10 20 30 40
Distance (pixels)
TIRF illumination eld (~100 nm)
RR*
Nb80
GFP R*
Arrestin
Clathrin/AP-2 coat
Recruitment phase 1
Recruitment
p
hase 2
R*
R*
Arrestin Nb80
GFP
Arrestin
Endosome
Plasma membrane
a
b
c
Figure 3
|
Nb80–GFP does not accumulate in clathrin-coated pits or
vesicles. a, Representative TIRF microscopy frames showing Nb80–GFP
(green) and b-arrestin-2–mCherry (red) before and after agonist addition.
Fluorescence intensity profiles are shown below for the indicated region and
path; representative of n539 cells, 5 experiments and 4,849 punctae.
b, Equivalent analysis comparing Nb80–GFP (green) to clathrin light chain-
dsRed (red); representative of n526 cells, 3 experiments and 3,965 punctae.
Arrowheads indicate examples of Nb80–GFP labelled endosomes. Scale bars,
5mm. c, Model for two phases of Nb80–GFP recruitment by the activated b
2
-
AR, first at the plasma membrane and then at endosomes.
RESEARCH LETTER
536 | NATURE | VOL 495 | 28 MARCH 2013
Macmillan Publishers Limited. All rights reserved
©2013
Because endosomes contain activated b
2
-ARs, we next asked if
receptors engage their cognate G protein from this compartment.
Heterotrimeric G proteins and adenylyl cyclase can be observed in
endosomes as well as at the plasma membrane, supporting the concept
of endosome-based G protein signalling
4,6,10,24
. To directly investigate
the subcellular location of G protein activation, we developed a dis-
tinct biosensor based on another nanobody, Nb37, which specifically
recognizes the guanine-nucleotide-free form of Ga
s
representing the
catalytic intermediate of G protein activation (Fig. 4a)
25
. We hoped
that, because Nb37 binds a surface of the alpha-helical domain that is
accessible only in the nucleotide-free form, we would be able to detect
production of this critical butfleeting activation intermediate in living
cells. This was indeed the case because Nb37–GFP localized in the
cytoplasm of untreated cells and was rapidly recruited from the cyto-
plasm to the plasma membrane in response to isoprenaline application
(Fig. 4b, yellow arrowhead, Pearson coefficient50.627). Membrane
recruitment of Nb37–GFP was agonist-dependent even in cells over-
expressing Ga
s
and, notably, the cytoplasmic concentration of Nb37–
GFP achieved in our experiments (also approximately 20nM) was
substantially lower than that producing detectable inhibition of G
protein activation in vitro (Supplementary Fig. 5). Together, these
observations suggest that Nb37–GFP indeed functions as a specific
biosensor of G
s
activation under the experimental conditions used.
Nb37–GFP was recruited not only to the plasma membrane (Fig. 4b,
yellow arrowhead) but also to b
2
-AR-containing endosomes. Notably,
endosome recruitment of Nb37–GFP occurred after the appearance of
b
2
-ARs in the endosome. Such later recruitment was evident in dual
label confocal image series (Fig. 4b shows an example: white arrowhead
indicates recently internalized b
2
-AR, white arrows indicate Nb37–GFP
recruitment, Pearson coefficient 50.710; see also Supplementary Video
8). Nb37–GFP recruitment was specifically dependent on receptor
activation because a mutant b
2
-AR that is defective in G-protein coup-
ling (b
2
-AR-Cys 341 Gly)
26
did not produce detectable recruitment
(Supplementary Fig. 6), whereas the distinct G
s
-coupled DRD1
recruited Nb37–GFP to both the plasma membrane and endo-
somes (data not shown). Nb37–GFP localized uniformly on b
2
-AR-
containing endosomes in cells overexpressing Ga
s
(Fig. 4b and
Supplementary Video 8) but was recruited non-uniformly in cells
expressing endogenous levels (Fig. 4c, arrowhead indicates an
example). Moreover, Nb37–GFP recruitment to endosomes had a
scintillating appearance, fluctuating rapidly when viewed in live image
series (Supplementary Video 9), suggesting that G protein activation
detected by Nb37–GFP occurs dynamically from limited regions of the
endosome membrane.
We then asked if active b
2
-AR G
s
coupling from endosomes con-
tributes to the cellular response, focusing on cAMP as a classical
second messenger carrying the downstream signal, using a previously
described real-time assay of cAMP accumulation in living cells and
normalizing to the signal produced by receptor-independent activa-
tion of adenylyl cyclase with forskolin (5 mM)
27
. Isoprenaline produced
a rapid increase in cytoplasmic cAMP accumulation, reaching a maxi-
mum within approximately 10min (Fig. 4d, blue line). Dyngo-4a, a
dynamin inhibitor that blocks b
2
-AR endocytosis by inhibiting CCP
function
28,29
, did not detectably affect the earliest portion of the
forskolin-normalized cAMP accumulation curve but significantly
reduced its later rise (Fig. 4d, green line, grey dots indicate Pvalues),
consistent with the two phases of endosome-localized activation
detected by the nanobody-derived biosensors. A similar inhibition
was observed in cells expressing a mutant b
2
-AR with a carboxy-
terminal alanine residue added that prevents efficient recycling
β2-AR
β2-AR Merged
c
e
Nb37–GFP
bIso
f
d
g
R
GFP
Nb37
α
0 min 6 min 6.5 min 7 min 7.5 min 8 min 8.5 min
125
100
75
50
25
0
Luminescence
(% of max Fsk)
0246810
Time (min)
0.25
0.20
0.15
0.10
0.05
0.00
P value
β2-AR-3S + 5 μM Iso
β2-AR-3S + 5 μM Iso + 30 μM Dyngo-4a
125
100
75
50
25
0
Luminescence
(% of max Fsk)
0246810
Time (min)
β2-AR β2-AR-3S
Internalization (%)
50
40
30
20
10
0
Agonist binding
G protein activation
from plasma
membrane
Arrestin binding Clustering in CCP
Recycling Endocytosis
Uncoating
G protein activation
from endosome
membrane
Signalling phase 1
Signalling phase 2
RR* R*
R*
R*
R*
αα
α
GDP ββ
β
γγ
γ
GDP GTP
Arrestin
GDP
GTP
Nb37–GFP
a
β2-AR + 5 μM Iso
β2-AR + 5 μM Iso + 30 μM Dyngo-4a e
Biosensor of G
protein activation
Figure 4
|
Internalized b
2
-ARs contribute to the
acute cAMP response. a, Scheme for detecting
conformational activation of G
s
with Nb37–GFP.
b, Confocal image frames showing Nb37–GFP
(green) and b
2
-AR (red) at the indicated time
points after isoprenaline addition (representative
of n514 cells; estimated Nb37–GFP recruitment
delay ranged from 0.7 to 2.65 min).Scale bar, 5 mm.
Yellow arrowhead indicates Nb37–GFP
recruitment to the plasma membrane. White
arrowhead indicates an endosome containing
recently internalized b
2
-AR and not associated
with Nb37–GFP, white arrows indicate Nb37–GFP
recruitment. c, Representative confocal frames
showing a discrete endosomal structure labelled
with Nb37–GFP (green) and b
2
-AR (red) at higher
magnification. Scale bar, 2 mm. Arrowhead
indicates non-uniform localization of Nb37–GFP
to the endosome. d, Forskolin-normalized b
2
-AR-
mediated cAMP response in the absence or
presence of 30 mM Dyngo-4a (mean 6s.e.m.,
n510 experiments, Pvalues in grey).
e, Isoprenaline (20 min)-induced b
2
-AR and b
2
-
AR-3S internalization measured by flow cytometry
(n54 experiments). f, Forskolin-normalized b
2
-
AR-3S-mediated cAMP response in the absence or
presence of 30 mM Dyngo-4a (n58 experiments;
P50.1192). g, Model for two phases of b
2
-AR G
s
activation, first at the plasma membrane and then
at endosomes, separated by the endocytic event.
LETTER RESEARCH
28 MARCH 2013 | VOL 495 | NATURE | 537
Macmillan Publishers Limited. All rights reserved
©2013
(b
2
-AR-Ala)
30
(Supplementary Fig. 7), suggesting that the Dyngo-
sensitive component of the cAMP response did not represent a secon-
dary consequence of resensitization by receptor recycling
17
. Dyngo-4a
did not produce the same effect on cAMP accumulation elicited by
the internalization-defective b
2
-AR-3S mutant receptor
21
(Fig. 4e, f),
further supporting the conclusion that the second signalling phase
indeed requires receptor localization in endosomes.
The present findings revise a long-held tenet of molecular phar-
macology, that acute signal transduction mediated by the canoni-
cal b
2
-AR G
s
signal transduction mechanism is plasma membrane
delimited. The results provide direct evidence supporting the hypo-
thesis that b
2
-AR endocytosis contributes to a second phase of the
acute cellular cAMP response, which represents a significant com-
ponent of the overall biochemical signal developed within several
minutes after the initial agonist application. Thus, although it remains
clear that b
2
-ARs can elicit G
s
-mediated signal transduction from the
plasma membrane, the present data reveal a discrete component of the
acute signalling response that is initiated from endosomes (Fig. 4g). It
remains unknown if b
2
-ARs are continuously bound by agonist in
endosomes, as depicted in the figure for simplicity, but conforma-
tional activation of G
s
in endosomes is both receptor- and agonist-
dependent. Unambiguous detection of endosome-based activation of
acute G-protein-linked signalling is presently limited to the b
2
-AR
G
s
system for which the critical nanobodies are available. However,
endosome-based contribution to the acute signalling response is pro-
bably widespread in the GPCR superfamily because the b
2
-AR belongs
to the largest group (family A) of GPCRs and is often considered a
prototype. We also suggest, more generally, that nanobody-based bio-
sensors represent a versatile strategy for probing other types of dyna-
mic conformational change with high spatiotemporal resolution in
living cells.
METHODS SUMMARY
Experiments were carried out using human embryonal kidney HEK293 cells
(ATCC) expressing the indicated receptor constructs and nanobodies fused to
enhanced GFP. Opticalimaging was carried out at 37 uC in DMEM not containing
phenol red and supplemented with 30 mM HEPES. Live cell cAMP accumulation
was assessed at 37 uC using pGlosensor-20F (Promega). Flow cytometry was
carried out using Alexa647 (Invitrogen)-conjugated M1 anti-Flag monoclonal
antibody (Sigma) and a FACSCalibur instrument (Becton Dickinson).
Full Methods and any associated references are available in the online version of
the paper.
Received 13 August 2012; accepted 8 February 2013.
Published online 20 March 2013.
1. Lohse, M. J., Nuber, S. & Hoffmann, C. Fluorescence/bioluminescence resonance
energy transfer techniques to study G-protein-coupled receptor activation and
signaling. Pharmacol. Rev. 64, 299–336 (2012).
2. Shenoy, S. K. & Lefkowitz, R. J. Seven-transmembranereceptor signaling through
b-arrestin. Sci. STKE 2005, cm10 (2005).
3. Murphy, J. E., Padilla,B. E., Hasdemir, B.,Cottrell, G. S. & Bunnett,N. W. Endosomes:
a legitimate platform for the signaling train. Proc. Natl Acad. Sci. USA 106,
17615–17622 (2009).
4. Ferrandon, S. et al. Sustained cyclic AMP production by parathyroid hormone
receptor endocytosis. Nature Chem. Biol. 5, 734–742 (2009).
5. Feinstein, T. N. et al. Retromer terminates the generation of cAMP by internalized
PTH receptors. Nature Chem. Biol. 7, 278–284 (2011 ).
6. Calebiro, D. et al. Persistent cAMP-signals triggered by internalized G-protein-
coupled receptors. PLoS Biol. 7, e1000172 (2009).
7. Werthmann, R. C., Volpe, S., Lohse,M. J. & Calebiro, D. PersistentcAMP signaling by
internalized TSH receptors occurs in thyroid but not in HEK293 cells. FASEB J. 26,
2043–2048 (2012).
8. Mullershausen, F. et al. Persistent signaling induced by FTY720-phosphate is
mediated by internalized S1P1 receptors. Nature Chem. Biol. 5, 428–434 (2009).
9. Calebiro, D., Nikolaev, V. O. & Lohse, M. J. Imaging of persistent cAMP signaling by
internalized G protein-coupled receptors. J. Mol. Endocrinol. 45, 1–8 (2010).
10. Kotowski, S. J., Hopf, F. W., Seif, T., Bonci, A. & von Zastrow, M. Endocytosis
promotes rapid dopaminergic signaling. Neuron 71, 278–290 (2011).
11. Lefkowitz, R. J. Seven transmembrane receptors: something old, something new.
Acta Physiol. (Oxf.) 190, 9–19 (2007).
12. Gilman, A. G. Transmembrane signaling, G proteins, and adenylyl cyclase. Harvey
Lect. 85, 153–172 (1989).
13. Rasmussen, S. G. et al. Crystal structure of the b
2
adrenergic receptor–Gs protein
complex. Nature 477, 549–555 (2011).
14. Moore, C. A., Milano, S. K. & Benovic,J. L. Regulation of receptortrafficking by GRKs
and arrestins. Annu. Rev. Physiol. 69, 451–482 (2007).
15. Sorkin, A. & von Zastrow, M. Endocytosis and signalling: intertwining molecular
networks. Nature Rev. Mol. Cell Biol. 10, 609–622 (2009).
16. Luttrell, L. M. & Lefkowitz, R. J. The role of b-arrestins in the termination and
transduction of G-protein-coupled receptor signals. J. Cell Sci. 115, 455–465 (2002).
17. Gainetdinov, R. R., Premont, R. T., Bohn, L. M., Lefkowitz, R. J. & Caron, M. G.
Desensitization of G protein-coupled receptors and neuronalfunctions. Annu. Rev.
Neurosci. 27, 107–144 (2004).
18. Rasmussen, S. G. et al. Structure of a nanobody-stabilized active state of the b
2
adrenoceptor. Nature 469, 175–180 (2011).
19. Steyaert, J. & Kobilka, B. K. Nanobody stabilization of G protein-coupled receptor
conformational states. Curr. Opin. Struct. Biol. 21, 567–572 (2011).
20. Lauffer, B. E. et al. SNX27 mediates PDZ-directed sorting from endosomes to the
plasma membrane. J. Cell Biol. 190, 565–574 (2010).
21. Hausdorff, W. P. et al. A small region of the b-adrenergic receptor is selectively
involved in its rapid regulation. Proc. Natl Acad. Sci. USA 88, 2979–2983 (1991).
22. Steyer, J. A. & Almers, W. A real-time view of life within 100 nm of the plasma
membrane. Nature Rev. Mol. Cell Biol. 2, 268–275 (2001).
23. Puthenveedu, M. A. & von Zastrow, M. Cargo regulates clathrin-coated pit
dynamics. Cell 127, 113–124 (2006).
24. Slessareva, J.E., Routt, S. M., Temple, B.,Bankaitis, V. A. & Dohlman,H. G. Activation
of the phosphatidylinositol 3-kinase Vps34 by a G protein asubunit at the
endosome. Cell 126, 191–203 (2006).
25. Westfield, G. H. et al. Structural flexibility of the G
as
a-helical domain in the b
2
-
adrenoceptor G
s
complex. Proc. Natl Acad. Sci. USA 108, 16086–16091 (2011).
26. Campbell, P. T. et al. Mutations of the human b
2
-adrenergic receptor that impair
coupling to G
s
interfere withreceptor down-regulationbut not sequestration. Mol.
Pharmacol. 39, 192–198 (1991).
27. Violin, J. D. et al. b
2
-adrenergic receptor signaling and desensitization elucidated
by quantitative modeling of real time cAMP dynamics. J. Biol. Chem. 283,
2949–2961 (2008).
28. Harper, C.B. et al. Dynamin inhibition blocks botulinum neurotoxin ty pe A endocytosis
in neurons and delays botulism. J. Biol. Chem. 286, 35966–35976 (2011).
29. Howes, M. T. et al. Clathrin-independent carriers form a high capacity endocytic
sorting system at the leading edge of migrating cells. J. Cell Biol. 190, 675–691 (2010).
30. Cao, T. T., Deacon, H. W., Reczek, D., Bretscher, A. & von Zastrow, M. A kinase-
regulated PDZ-domain interaction controls endocytic sorting of theb
2
-adrenergic
receptor. Nature 401, 286–290 (1999).
Supplementary Information is available in the online version of the paper.
Acknowledgements We thank B. Kobilka, P. Robinson, A. Kruse, E. Pardon, P. Temkin,
M. Puthenveedu, A. Henry,A. Marley and K. Thorn for assistance, adviceand discussion.
These studies were supported by the National Institute on Drug Abuse of the US
National Institutes of Health (DA010711and DA012864 to M.v.Z. and F32 DA029993
to J.C.T.). R.I. is supported by the American Heart Association. R.K.S. and J.P.M. are
supported by the National Institute of General Medical Sciences (GM083118 to R.K.S.
and T32 GM007767 to J.P.M.). S.G.F.R. is supported by the Lundbeck Foundation. J.S.
is supported by FWO-Vlaanderen grants (FWO551 and FWO646) and
Innoviris-Brussels (BRGEOZ132). B.H. is supported by a Packard Fellowship for
Science and Engineering.
Author Contributions R.I.constructed and validated the nanobodybiosensors, carried
out most of the cell biological experiments and analysis, contributed to overall
experimental strategy and took a lead role in writing the manuscript. J.C.T. carried out
early experiments identifying endocytic inhibitor effects on cellular cAMP signalling,
and contributed to initial project planning. J.R.T. built the luminometer system,
developed software for analysis of luminometry data, and contributed to early
experiments on cellular cAMP signalling. M.C. contributed to experimental design and
data analysis, and modelled effects of endocytic inhibitors on the cellular cAMP
response. J.P.M. contributed to the production of receptor-containing rHDL particles
and carried out in vitro studies of Nb80 binding and dissociation. J.S. developed the
nanobody reagents used as the basis for the biosensors described in this study and
advised on biosensor design and expression. S.G.F.R. contributed to developing and
screeningthe initial nanobodyreagents, and carriedout in vitro studies of Nb80binding
and dissociation in rHDL particles reconstituted with bimane-labeled receptors. R.K.S.
contributed to overall experimental interpretation, supervised J.P.M. in carrying out in
vitro studies of Nb80 binding to receptors, and performed in vitro experiments
evaluating Nb37 effects on G protein activation. H.E.-S. contributed to experimental
design anddata interpretation,and supervisedefforts to model endocytic effectson the
cellular cAMP response. B.H. contributed to overall experimental design and
interpretation, implementation of biosensorsand advised on imageanalysis. M.v.Z. was
responsible for overall project strategy, carried out some of the imaging experiments,
and drafted the manuscript together with R.I.
Author Information Reprints and permissions information is available at
www.nature.com/reprints. The authors declare no competing financial interests.
Readers are welcome to comment on the online version of the paper. Correspondence
and requests for materials should be addressed to M.v.Z.
(Mark.VonZastrow@ucsf.edu).
RESEARCH LETTER
538 | NATURE | VOL 495 | 28 MARCH 2013
Macmillan Publishers Limited. All rights reserved
©2013
METHODS
Cell Culture, cDNA constructs and transfection. HEK293 cells were grown in
DMEM supplemented with 10% FBS (UCSF Cell Culture Facility) without anti-
biotics. Stably transfected HEK293 cell clones expressing Flag-tagged b
2
-AR-3S
were created using previously described Flag-tagged b
2
-AR
30
. A plasmid encoding
a cyclic permuted luciferase reporter construct,based on a mutated RIIbB cAMP-
binding domain from PKA(pGloSensor-20F, Promega). Nb80–eGFP and Nb37–
eGFP were created by amplifying Nb80 and Nb37 nanobody complementary
DNAs using 59-CTTGAAAAGCTTGCCGCCACCATGGGACAGGTGCAGCT
GCA-39;59-TTCAAGGGATCCATGTGATGGTGATGGTGGTGTGAGGAGA
CGGT-39and 59-CTTGAAAAGCTTGCCGCCACCATGGGACAGGTGCAGC
TGCA-39;59-TTCAAGGGATCCATGTGATGGGCTTCAGGTTCGTGATGG
TGATG-39primers, respectively, and cloning into the peGFP-N1 vector using
HindIII and BamH1. b-arrestin-2–GFP, clathrin–DsRed, EEA1–DsRed and Ga
s
HA were gifts from M. Caron, W. Almers, K. Mostov and P. Wedegaertner,
respectively. b-arrestin-2–mCherry was generated by subcloning b-arrestin-2 to
pmCherry (Clontech)and b
2
-AR–CFP was generated fromthe Flag-tagged b
2
-AR
construct. Transfections were performed using Lipofectamine 2000 (Invitrogen)
according to the manufacturer’s instructions. Flag-tagged human b
2
-AR and b
2
-
AR-3S (Ser 355 Gly, Ser 356 Gly and Ser 364 Gly were mutated simultaneously)
constructs were labelled with Alexa555- or Alexa488-conjugated M1 anti-Flag
monoclonal antibody (Sigma) as described previously
31
.
Live-cell confocal imaging. Live cell imaging was carried out using Yokagawa
CSU22 spinning disk confocal microscope with a 3100, 1.4 numerical aperture,
oil objective and a CO
2
and 37 uC temperature-controlled incubator. A 488 nm
argon laser and a 568nm argon/krypton laser (Melles Griot) were used as light
sources for imaging GFP and Flag signals, respectively. Cells expressing both Flag-
tagged receptor and the indicated nanobody–GFP were plated onto glass cover-
slips. Receptors were surface labelled by addition of M1 anti-Flag antibody
(1:1,000, Sigma) conjugated to Alexa 555 (A10470, Invitrogen) to the media for
30 min, as described previously
32
. Indicatedagonist (isoprenaline,Sigma) or antago-
nist (CGP-12177,Tocris) were added and cells were imaged every 3 s for 20min in
DMEM without phenolred supplemented with 30 mM HEPES, pH 7.4 (UCSF Cell
Culture Facility). Time-lapse images were acquired with a Cascade II EM charge-
coupled-device (CCD) camera (Photometrics) driven by Micro-Manager 1.4
(http://www.micro-manager.org).
Live cell TIRF microscopy. TIRF imaging was carried out as described
previously
33
. Briefly, HEK293 cells co-expressing either Nb80–eGFP and
b-arrestin-2–mCherry or clathrin light chain–DsRed, were imaged in DMEM
without phenol red supplemented with 30 mM HEPES, pH 7.4 (UCSF Cell
Culture Facility). Imaging was carried out using a Nikon TE-2000E inverted
microscope with a 3100, 1.49 numerical aperture TIRF objective, equipped for
through-the-objective TIRF illumination, a 37 uC temperature-controlled stage
(Bioscience Tools) and an objective warmer (Bioptechs). A 488nm argon laser
(Melles Griot) and a 543 nm helium-neonlaser (Spectra Physics) were used as light
sources. Time-lapse sequences were acquired with a C9100-12 camera
(Hamamatsu Photonics) driven by iQ software (Andor). Cells were imaged every
3 s for 20 min.
Image analysis and statistical analysis. Images were saved as 16-bit TIFF files.
Quantitative image analysis was carried out on unprocessed images using ImageJ
software (http://rsb.info.nih.gov/ij). Co-localization analysis wasestimated by cal-
culating the Pearson’s correlation coefficient between the indicated image chan-
nels using the co-localization plug-in for ImageJ. Analysis of Nb80–GFP intensity
profile along the straight line and Nb80–GFP/b-arrestin or Nb80–GFP/clathrin
along the segmented line were carried out using the ImageJ plot profile function.
For estimating changes in Nb80–GFP surface fluorescence over time in TIRF
images, individual cells were selected manually and fluorescence values measured
over the entire stack. A blank area of the image lacking cells was used to estimate
background fluorescence. Average fluorescence intensity was measured in each
frame, background-subtracted and normalizedto the maximum value. Pvalues are
from one-tailedunpaired Student’s t-tests. For visual presentation(but not quanti-
tative analysis), image series were processed using Kalman stack filter in ImageJ.
Luminescence-based rapid cAMP assay. HEK293 cells were transfected with a
plasmid encoding a cyclic-permuted luciferase reporter construct, based on a
mutated RIIbB cAMP-binding domain from PKA (pGloSensor-20F, Promega),
which produces rapid and reversible cAMP-dependent activation of luciferase
activity in intactcells. Cells were plated in 24-well dishes containingapproximately
200,000 cells per well in 500 ml DMEM without phenol red and no serum and
equilibrated to 37 uC in a light-proof cabinet. An image of the plate was focused
on a 512 3512 pixel electron multiplying CCD sensor (Hamamatsu C9100-13),
cells were equilibrated for 1 h in the presence of 250mgml
21
luciferin (Biogold),
and sequential luminescence images were collected every 10s to obtain basal
luminescence values. The camera shutter was closed, the cabinet opened and
the indicated concentration of isoprenaline was bath applied, with gentle manual
rocking before replacing in the dark cabinet and resuming luminescence image
acquisition. In endocytic manipulation experiments, cells were pre-incubated
with 30 mM Dyngo-4a (abcam Biochemicals) for 15 min. Every 10 s, sequential
images were acquired using Micro-Manager (http://www.micro-manager.org)
and integrated luminescence intensity detected from each well wascalculated after
background subtraction and correction for vignetting using scripts written in
MATLAB (MathWorks). In each multiwell plate, and for each experimental con-
dition, a reference value of luminescence was measured in the presence of 5 mM
forskolin, a manipulation that stimulates a moderate amount of receptor-inde-
pendent activation of adenylyl cyclase. The average luminescence value—mea-
sured across duplicate wells—was normalized to the maximum luminescence
value measured in the presence of 5 mM forskolin.
FRET imaging. FRET imaging was carried out as described previously
10
. Briefly,
HEK293 cells co-expressing b
2
-AR–CFP or DRD1–CFP and Nb80–YFP were
imaged in wide field at 37 uC using a shuttered mercury arc lamp and standard
CFP excitation (ET430/243) and YFP emission (ET500/203) band pass filters
(Chroma). YFP emission was collected using a 535/30 m filter, and CFP emission
was collected through a 470/24 m filter. Corrected FRET ratios were obtained
using the following equation: NFRET 5[(I
FRET
2BG
FRET
)2(I
CFP
2BG
CFP
)3
BT
DONOR
2(I
YFP
2BG
YFP
)3DE
ACCEPTOR
]/I
CFP
.BT
DONOR
, donor bleed
through; DE
ACCEPTOR
, direct excitation of the acceptor; BG
X
, background fluor-
escence; and I
X
, integrated fluorescence intensity measured in a given channel.
Flow cytometric assay of receptor endocytosis. Surface fluorescence ofFlag–b
2
-
AR or Flag–b
2
-AR-3S expressing HEK293 cells was used to measure receptor
endocytosis. Cells were incubated with 10mM isoprenaline for 20 min at 37 uC
to drive receptor internalization to steady state and were subsequently rinsed 3
times with ice-cold PBS, then mechanically lifted and incubated with 1mgml
21
Alexa647 (Invitrogen)-conjugated M1 anti-Flag monoclonal antibody (Sigma)
at 4 uC for 1 h. Mean fluorescence intensity of 10,000 cells was measured using
a FACSCalibur instrument (Becton Dickinson). Each condition was performed
in triplicate.
Enhanced GFP calibration. Recombinant eGFP (BioVision) was used for cal-
ibrating average fluorescence intensity of the biosensors, imaged in confocal
optical sections through the cytoplasm of cells not exposed to agonist (to achieve
diffuse cytoplasmic distribution of the biosensors). eGFP was diluted in Hank’s
balanced salt solution andconfocal sections were imaged through droplets of each
using the same illumination and acquisition parameters as for imaging the bio-
sensors in cells. For each cell, a background fluorescence value was determined
by average fluorescence intensity of a blank region in the same image. The cyto-
plasmic concentration of biosensors was estimated by interpolation of the back-
ground-subtracted value using a linear least-squares fit to the standard plot.
Generation of b
2
-AR-rHDL nanoparticles. Apolipoprotein-AI (Apo-AI) was
biotinylated using NHS-PEG4-biotin (Pierce Biotechnology) at a 1:1 molar ratio.
Following a 30-min biotinylation reaction at room temperature, the sample was
dialysed to remove free biotin. Flag-tagged b
2
-AR was incorporated into recom-
binant high density lipoprotein (rHDL) particles as previously described
34,35
using
biotinylated Apo-AI. Receptor-containing particles were then purified by M1
anti-Flag immunoaffinity chromatography
36
. Particles containingpurified mono-
bromobimane-labelled b
2
-AR were generated similarly except not using biotiny-
lated HDL, with receptor labelling and fluorescence analysis carried out as
described previously
18
.
Assessing Nb80 binding to immobilized b
2
AR-rHDL. Nb80 binding to unli-
ganded and agonist-occupied b
2
-AR was measured using the OctetRED biolayer
interferometrysystem (Pall Forte
´Bio). In this assay, a targetprotein is immobilized
on the functionalized tip of a fibre optic probe that is dipped into an analyte
solution to observe analyte association to the target protein. A dissociation step
is then performed by transferring the biosensor into buffer lacking analyte.
Analyte association/dissociation is measured by monitoring changes in the inter-
ference pattern of a light beam reflected from the biosensor tip as the total mass
bound at the tip surface changes
37
. Streptavidin-coated biosensors (Pall Forte
´Bio)
were loaded with biotinylated b
2
-AR-rHDL particles for 15min at room temper-
ature and the biosensors were transferred to the OctetRED instrument. Sensors
were placed into assay buffer (20mM HEPES, pH7.7, 100mM NaCl, 1mM
EDTA, 0.02% (w/v) ascorbic acid, 0.05% (w/v) BSA) with or without 100mM
isoprenaline for 30min. To measure Nb80 association, the sensor was transferred
to assay buffer with Nb80 (at indicated concentrations) for 5 min, followed by a
30 min dissociationstep in assay buffer. Isoprenaline (100 mM) was includedin the
association and dissociation steps when measuring Nb80 binding to agonist-occu-
pied receptor. All experiments were carried out at 25 uC with the assay plate
shaking at 1,000r.p.m. Buffer-only controls were included in each experiment
to monitor for baseline drift, and nonspecific Nb80 binding was measured in a
parallel assay using sensors loaded with empty rHDL particles. Raw data were
LETTER RESEARCH
Macmillan Publishers Limited. All rights reserved
©2013
processed to remove baseline and nonspecific binding using Octet Data Ana-
lysis 7.0 software (Pall Forte
´Bio) and exported to Prism 5 (GraphPad) for curve
fitting. All association and dissociation curves were fit using a single-phase
exponential association or decay curves, respectively. Equilibrium binding
affinity of Nb80 for b
2
-AR in the presence or absence of the agonist isoprenaline
was assessed by monitoring the maximal interference shift generated by Nb80
binding (at varying Nb80 concentrations) to the probe containing b
2
-AR recon-
stituted in rHDL. The maximal shift was plotted against the Nb80 concen-
tration and fitted by nonlinear regression in Prism 5 (GraphPad) to generate
the apparent affinity.
Inhibition of bodipy-GTPcS-FL binding by Nb37. The effect of Nb37 on GTP
loading of purified G proteins was measured using 100nM bodipy-GTPcS-FL
(Invitrogen) essentially as described
25
. In this assay we used the fluorescence
emission of bodipy-GTPcS-FL (l
ex
,470 nm, l
em
,515 nm) that accompanies
binding of the labelled nucleotide to G protein
38
. Briefly, the fluorescence of
100 nM bodipy-GTPcS-FL was measured in the presence of 1 mM of theindicated
G protein using a96-well microtitre plate format on a M5fluorescence plate reader
(Molecular Devices). Nb37 was added together with bodipy-GTPc
s
-FL and the
binding reaction was initiated by the addition of G protein (1 mM) in 20 mM Tris-
HCl, pH 8.0, 3 mM MgCl
2
, 1 mM dithiothreitol in a final volume of 200 ml.
Bodipy-GTPcS-FL binding to heterotrimeric G protein included 0.1% dodecyl-
maltoside (final). Ga
s
was purified as described
39
.Ga
s
bc was purified as
described
13
. Myristoylated Ga
i
was purified as described
40
. The time scans were
limited to 240s to minimize the accumulation of hydrolysis of the product of
bodipy-GTPcS-FL, bodipy-phosphate
41
.
31. Gage, R. M., Matveeva,E. A., Whiteheart, S. W. & von Zastrow, M. Type I PDZ ligands
are sufficient to promote rapid recycling of G protein-coupled receptors
independent of binding to N-ethylmaleimide-sensitive factor. J. Biol. Chem. 280,
3305–3313 (2005).
32. Puthenveedu, M. A. et al. Sequence-dependent sorting of recycling proteins by
actin-stabilized endosomal microdomains. Cell 143, 761–773 (2010).
33. Yudowski, G. A., Puthenveedu, M. A., Henry, A. G. & von Zastrow, M. Cargo-mediated
regulation of a rapid Rab4-dependent recycling pathway. Mol. Biol. Cell 20,
2774–2784 (2009).
34. Whorton, M. R. et al. A monomeric G protein-coupled receptor isolated in a high-
density lipoprotein particle efficiently activates its G protein. Proc. Natl Acad. Sci.
USA 104, 7682–7687 (2007) .
35. Kuszak, A. J. et al. Purification and functionalreconstitution of monomericm-opioid
receptors: allosteric modulation of agonist binding by Gi
2
.J. Biol. Chem. 284,
26732–26741 (2009).
36. Yao, X. et al. Coupling ligand structure to specific conformational switches in the
b
2
-adrenoceptor. Nature Chem. Biol. 2, 417–422 (2006).
37. Abdiche, Y., Malashock, D., Pinkerton, A. & Pons, J. Determining kinetics and
affinities of protein interactions using a parallel real-time label-free biosensor, the
Octet. Anal. Biochem. 377, 209–217 (2008).
38. McEwen, D. P., Gee, K. R., Kang, H. C. & Neubig, R. R. Fluorescent BODIPY-GTP
analogs: real-time measurement of nucleotide binding to G proteins. Anal.
Biochem. 291, 109–117 (2001).
39. Sunahara, R. K., Tesmer, J. J., Gilman, A. G. & Sprang, S. R. Crystal structure of the
adenylyl cyclase activator G
sa
.Science 278, 1943–1947 (1997).
40. Lee, E., Linder, M. E. & Gilman, A. G. Expression of G-protein asubunits in
Escherichia coli.Methods Enzymol. 237, 146–164 (1994).
41. Jameson, E. E. et al. Real-time detection of basal and stimulated G protein
GTPase activity using fluorescent GTP analogues. J. Biol. Chem. 280, 7712–7719
(2005).
RESEARCH LETTER
Macmillan Publishers Limited. All rights reserved
©2013
... Canonical G protein signaling occurs when a cell surface GPCR is activated by an extracellular ligand, which in turn promotes activation of plasma membrane G protein heterotrimers and downstream effectors. Recently, it has become clear that GPCRs can also signal from intracellular compartments (Calebiro et al., 2010;Eichel and von Zastrow, 2018), most notably endosomes and the Golgi apparatus (Calebiro et al., 2009;Ferrandon et al., 2009;Irannejad et al., 2017;Irannejad et al., 2013;Mullershausen et al., 2009). Signaling from endosomes is often a continuation of signaling that starts at the plasma membrane and persists as (or resumes after) active receptors are endocytosed (Tsvetanova et al., 2015). ...
... However, these processes reverse quickly when activation ceases (Akgoz et al., 2004;Martin and Lambert, 2016), meaning that activationdependent translocation of free Gα subunits and Gβγ dimers would be an inefficient mechanism to deliver inactive heterotrimers to intracellular membranes. G proteins have been detected on the surface of endosomes and other intracellular compartments using a variety of approaches (Hewavitharana and Wedegaertner, 2012;Irannejad et al., 2013;Scarselli and Donaldson, 2009;Wedegaertner, 2012). However, there has been no quantitative comparison of G protein distribution across identified subcellular compartments. ...
... To test this possibility, we performed similar imaging experiments with mNG-β 1 cells transfected with SNAP-tagged β 2 adrenergic receptors (SNAPf-β 2 AR). This receptor is often used as a model of activity-dependent GPCR internalization (Benovic et al., 1988;von Zastrow and Kobilka, 1992) and has been shown to activate G proteins on endosomes (Bowman et al., 2016;Irannejad et al., 2013). We labeled SNAPf-β 2 AR with a membrane-impermeant SNAP ligand (AF 647) at room temperature to prevent constitutive endocytosis, then incubated cells with FM4-64 and the agonist isoproterenol for 15 min at physiological temperature to promote receptor endocytosis. ...
Article
Full-text available
Classical G-protein-coupled receptor (GPCR) signaling takes place in response to extracellular stimuli and involves receptors and heterotrimeric G proteins located at the plasma membrane. It has recently been established that GPCR signaling can also take place from intracellular membrane compartments, including endosomes that contain internalized receptors and ligands. While the mechanisms of GPCR endocytosis are well understood, it is not clear how well internalized receptors are supplied with G proteins. To address this gap, we use gene editing, confocal microscopy, and bioluminescence resonance energy transfer to study the distribution and trafficking of endogenous G proteins. We show here that constitutive endocytosis is sufficient to supply newly internalized endocytic vesicles with 20–30% of the G protein density found at the plasma membrane. We find that G proteins are present on early, late, and recycling endosomes, are abundant on lysosomes, but are virtually undetectable on the endoplasmic reticulum, mitochondria, and the medial-trans Golgi apparatus. Receptor activation does not change heterotrimer abundance on endosomes. Our findings provide a subcellular map of endogenous G protein distribution, suggest that G proteins may be partially excluded from nascent endocytic vesicles, and are likely to have implications for GPCR signaling from endosomes and other intracellular compartments.
... This engineered receptor has the key advantage of leaving the intracellular domains of B2AR unmodified. We and others have extensively characterized the trafficking, signaling, and biochemical characteristics of this tagged B2AR (Cao et al., 1999;Lauffer et al., 2010;Puthenveedu et al., 2010;Irannejad et al., 2013). Consistent with previous data, live imaging of surface FLAG-B2AR with an M1 anti-FLAG antibody conjugated to Alexa Fluor 647 (M1-647) showed agonist-mediated internalization upon addition of 10 µM isoproterenol and localized primarily at endosomes within ten minutes (Fig. 1A). ...
Article
The emerging picture of G protein-coupled receptor function suggests that the global signaling response is an integrated sum of a multitude of individual receptor responses, each regulated by their local protein environment. The beta 2 adrenergic receptor (B2AR) has long served as an example receptor in the development of this model. But the mechanism and the identity of the protein-protein interactions that govern the availability of receptors competent for signaling remains incompletely characterized. To address this question, we characterized the interactome of agonist-stimulated B2AR in HEK293 cells using FLAG co-immunoprecipitation coupled to SILAC labeling and mass spectrometry. Our B2AR cross-linked interactome identified 190 high-confidence proteins, including almost all known interacting proteins and six out of seven isoforms of the 14-3-3 family of scaffolding proteins. Inhibiting 14-3-3 proteins with the peptide difopein enhanced isoproterenol-stimulated adrenergic signaling via cAMP approximately three-fold, and increased both miniGs and arrestin recruitment to B2AR more than two fold each, without noticeably changing EC50 with respect to cAMP signaling or effector recruitment upon stimulation. Our results show that 14-3-3 proteins negatively regulate downstream signaling by inhibiting access of B2AR to effector proteins. We propose that 14-3-3 proteins maintain a dynamic pool of B2AR that has reduced signaling efficacy in response to acute agonist stimulation, limiting the amount of signaling-competent receptors at the plasma membrane. Significance Statement This study presents a new interactome of the agonist-stimulated beta 2 adrenergic receptor (B2AR), a paradigmatic GPCR that is both a model system for members of this class and an important signaling protein in respiratory, cardiovascular, and metabolic regulation. We identify 14-3-3 proteins as responsible for restricting B2AR access to signaling effectors and maintaining a receptor population that is insensitive to acute stimulation by agonists.
... Gs-coupled GPCRs are thought to increase G protein activity on endosomes by internalizing and mediating a second round of allosteric coupling to cognate G proteins on the endosome limiting membrane. This model is supported by experiments that demonstrate that endosomal G protein-mediated signaling requires receptor internalization (9)(10)(11)(12)(13)(14)(15) and that its strength depends on the degree or duration of receptor activation in the endosome limiting membrane (16)(17)(18)(19)(20). ...
Preprint
Full-text available
Many GPCRs initiate a second phase of G protein-mediated signaling from endosomes, which inherently requires an increase in G protein activity on the endosome surface. Gs-coupled GPCRs are thought to achieve this by internalizing and allosterically activating cognate G proteins again on the endosome membrane. Here we demonstrate that the μ-opioid receptor (MOR), a Gi- coupled GPCR, increases endosomal G protein activity in a different way. Leveraging conformational biosensors, we resolve the subcellular activation dynamics of endogenously expressed MOR and Gi/o-subclass G proteins. We show that MOR activation triggers a transient increase of active-state Gi/o on the plasma membrane that is followed by a prolonged increase on endosomes. Contrary to the Gs-coupled GPCR paradigm, however, we show that the MOR- induced increase of active-state Gi/o on endosomes requires neither internalization of MOR nor activation of MOR in the endosome membrane. We propose a distinct and additional cellular mechanism for GPCR-triggered elevation of G protein activity on endosomes that is mediated by regulated trafficking of the activated G protein rather than its activating GPCR.
... For example, Nbs were used as crystallization chaperones or as fiducial markers in high-resolution structural studies [9][10][11][12] . Conformation-selective Nbs were also repurposed into biosensors to report GPCR activity in living cells 13,14 . ...
Article
Full-text available
The μ-opioid receptor (μOR), a prototypical G protein-coupled receptor (GPCR), is the target of opioid analgesics such as morphine and fentanyl. Due to the severe side effects of current opioid drugs, there is considerable interest in developing novel modulators of μOR function. Most GPCR ligands today are small molecules, however biologics, including antibodies and nanobodies, represent alternative therapeutics with clear advantages such as affinity and target selectivity. Here, we describe the nanobody NbE, which selectively binds to the μOR and acts as an antagonist. We functionally characterize NbE as an extracellular and genetically encoded μOR ligand and uncover the molecular basis for μOR antagonism by determining the cryo-EM structure of the NbE-μOR complex. NbE displays a unique ligand binding mode and achieves μOR selectivity by interactions with the orthosteric pocket and extracellular receptor loops. Based on a β-hairpin loop formed by NbE that deeply protrudes into the μOR, we design linear and cyclic peptide analogs that recapitulate NbE’s antagonism. The work illustrates the potential of nanobodies to uniquely engage with GPCRs and describes lower molecular weight μOR ligands that can serve as a basis for therapeutic developments.
... The β2-adrenergic receptor (β2AR), another GPCR capable of signaling from endosomes 43 , also accumulated in endosomes stably containing AC9 following activation by the β-adrenergic receptor agonist isoproterenol ( Supplementary Fig. 2e, f). Altogether, these results indicate that D1Rs can populate each of the subcellular membrane domains in which striatal AC isoforms distribute: D1R localizes in the plasma membrane, both to the ciliary microdomain containing AC3 and AC5 and to the extraciliary domain containing AC5 and AC9. ...
Article
Full-text available
The cAMP cascade is increasingly recognized to transduce physiological effects locally through spatially limited cAMP gradients. However, little is known about how adenylyl cyclase enzymes that initiate cAMP gradients are localized. Here we address this question in physiologically relevant striatal neurons and investigate how AC localization impacts downstream signaling function. We show that the major striatal AC isoforms are differentially sorted between ciliary and extraciliary domains of the plasma membrane, and that one isoform, AC9, is uniquely concentrated in endosomes. We identify key sorting determinants in the N-terminal cytoplasmic domain responsible for isoform-specific localization. We further show that AC9-containing endosomes accumulate activated dopamine receptors and form an elaborately intertwined network with juxtanuclear PKA stores bound to Golgi membranes. Finally, we provide evidence that endosomal localization enables AC9 to selectively elevate PKA activity in the nucleus relative to the cytoplasm. Together, these results reveal a precise spatial landscape of the cAMP cascade in neurons and a key role of AC localization in directing downstream PKA signaling to the nucleus.
Article
Gprotein-coupled receptors (GPCRs) regulate several physiological and pathological processes and represent the target of approximately 30% of Food and Drug Administration-approved drugs. GPCR-mediated signaling was thought to occur exclusively at the plasma membrane. However, recent studies have unveiled their presence and function at subcellular membrane compartments. There is a growing interest in studying compartmentalized signaling of GPCRs. This requires development of tools to separate GPCR signaling at the plasma membrane from the ones initiated at intracellular compartments. We leveraged the structural and pharmacological information available for β-adrenergic receptors (βARs) and focused on β1AR as exemplary GPCR that functions at subcellular compartments, and rationally designed spatially restricted antagonists. We generated a cell-impermeable βAR antagonist by conjugating a suitable pharmacophore to a sulfonate-containing fluorophore. This cell-impermeable antagonist only inhibited β1AR on the plasma membrane. In contrast, a cell-permeable βAR antagonist containing a nonsulfonated fluorophore efficiently inhibited both the plasma membrane and Golgi pools of β1ARs. Furthermore, the cell-impermeable antagonist selectively inhibited the phosphorylation of PKA downstream effectors near the plasma membrane, which regulate sarcoplasmic reticulum (SR) Ca ²⁺ release in adult cardiomyocytes, while the β1AR Golgi pool remained active. Our tools offer promising avenues for investigating compartmentalized βAR signaling in various contexts, potentially advancing our understanding of βAR-mediated cellular responses in health and disease. They also offer a general strategy to study compartmentalized signaling for other GPCRs in various biological systems.
Article
Cellular communication is regulated at the plasma membrane by the interactions of receptor, adhesion, signaling, and endocytic proteins. Yet, the composition and control of these complexes in response to external cues remain unclear. We use high-resolution and high-throughput fluorescence imaging to map the localization of growth factor receptors and related proteins at single clathrin-coated structures in human squamous HSC3 cells. We find distinct protein signatures between control cells and cells stimulated with growth factors. Clathrin sites at the plasma membrane are preloaded with some receptors but not others. Stimulation with epidermal growth factor induces capture and concentration of epidermal growth factor-, fibroblast growth factor-, and low-density lipoprotein-receptors (EGFR, FGFR1, and LDLR). Regulatory proteins including ubiquitin ligase Cbl, the scaffold Grb2, and the mechanoenzyme dynamin2 are also recruited. Disrupting FGFR or EGFR activity with drugs prevents the recruitment of both EGFR and FGFR1. EGF was able to activate FGFR1 phosphorylation. Our data reveals novel co-clustering and activation of receptors and regulatory factors at clathrin-coated sites in response to stimulation by a single growth factor, EGF or FGF. This behavior integrates growth factor signaling and allows for complex responses to extracellular cues and drugs at the plasma membrane of human cells.
Article
Full-text available
The active-state complex between an agonist-bound receptor and a guanine nucleotide-free G protein represents the fundamental signaling assembly for the majority of hormone and neurotransmitter signaling. We applied single-particle electron microscopy (EM) analysis to examine the architecture of agonist-occupied β(2)-adrenoceptor (β(2)AR) in complex with the heterotrimeric G protein Gs (Gαsβγ). EM 2D averages and 3D reconstructions of the detergent-solubilized complex reveal an overall architecture that is in very good agreement with the crystal structure of the active-state ternary complex. Strikingly however, the α-helical domain of Gαs appears highly flexible in the absence of nucleotide. In contrast, the presence of the pyrophosphate mimic foscarnet (phosphonoformate), and also the presence of GDP, favor the stabilization of the α-helical domain on the Ras-like domain of Gαs. Molecular modeling of the α-helical domain in the 3D EM maps suggests that in its stabilized form it assumes a conformation reminiscent to the one observed in the crystal structure of Gαs-GTPγS. These data argue that the α-helical domain undergoes a nucleotide-dependent transition from a flexible to a conformationally stabilized state.
Article
Full-text available
The botulinum neurotoxins (BoNTs) are di-chain bacterial proteins responsible for the paralytic disease botulism. Following binding to the plasma membrane of cholinergic motor nerve terminals, BoNTs are internalized into an endocytic compartment. Although several endocytic pathways have been characterized in neurons, the molecular mechanism underpinning the uptake of BoNTs at the presynaptic nerve terminal is still unclear. Here, a recombinant BoNT/A heavy chain binding domain (Hc) was used to unravel the internalization pathway by fluorescence and electron microscopy. BoNT/A-Hc initially enters cultured hippocampal neurons in an activity-dependent manner into synaptic vesicles and clathrin-coated vesicles before also entering endosomal structures and multivesicular bodies. We found that inhibiting dynamin with the novel potent Dynasore analog, Dyngo-4aTM, was sufficient to abolish BoNT/A-Hc internalization and BoNT/A-induced SNAP25 cleavage in hippocampal neurons. Dyngo-4a also interfered with BoNT/A-Hc internalization into motor nerve terminals. Furthermore, Dyngo-4a afforded protection against BoNT/A-induced paralysis at the rat hemidiaphragm. A significant delay of >30% in the onset of botulism was observed in mice injected with Dyngo-4a. Dynamin inhibition therefore provides a therapeutic avenue for the treatment of botulism and other diseases caused by pathogens sharing dynamin-dependent uptake mechanisms.
Article
Full-text available
G protein-coupled receptors (GPCRs) are responsible for the majority of cellular responses to hormones and neurotransmitters as well as the senses of sight, olfaction and taste. The paradigm of GPCR signalling is the activation of a heterotrimeric GTP binding protein (G protein) by an agonist-occupied receptor. The β(2) adrenergic receptor (β(2)AR) activation of Gs, the stimulatory G protein for adenylyl cyclase, has long been a model system for GPCR signalling. Here we present the crystal structure of the active state ternary complex composed of agonist-occupied monomeric β(2)AR and nucleotide-free Gs heterotrimer. The principal interactions between the β(2)AR and Gs involve the amino- and carboxy-terminal α-helices of Gs, with conformational changes propagating to the nucleotide-binding pocket. The largest conformational changes in the β(2)AR include a 14 Å outward movement at the cytoplasmic end of transmembrane segment 6 (TM6) and an α-helical extension of the cytoplasmic end of TM5. The most surprising observation is a major displacement of the α-helical domain of Gαs relative to the Ras-like GTPase domain. This crystal structure represents the first high-resolution view of transmembrane signalling by a GPCR.
Article
The crystal structure of Gs α, the heterotrimeric G protein α subunit that stimulates adenylyl cyclase, was determined at 2.5 Å in a complex with guanosine 5′-O-(3-thiotriphosphate) (GTPγS). Gs α is the prototypic member of a family of GTP-binding proteins that regulate the activities of effectors in a hormone-dependent manner. Comparison of the structure of Gs α·GTPγS with that of Gi α·GTPγS suggests that their effector specificity is primarily dictated by the shape of the binding surface formed by the switch II helix and the α3-β5 loop, despite the high sequence homology of these elements. In contrast, sequence divergence explains the inability of regulators of G protein signaling to stimulate the GTPase activity of Gs α. The βγ binding surface of Gs α is largely conserved in sequence and structure to that of Gi α, whereas differences in the surface formed by the carboxyl-terminal helix and the α4-β6 loop may mediate receptor specificity.
Article
The finely balanced activity of enzymes and their regulators keeps the cell-division cycle under control. A newly discovered molecule that ensures the timely destruction of one regulator is mutated in some cancer cells.
Article
G-protein-coupled receptors (GPCRs) have long been believed to activate G proteins only on the cell surface. However, we have recently shown that, in thyroid cells, the GPCR for the thyroid-stimulating hormone (TSH) can continue stimulating cAMP production after cointernalization with TSH. cAMP signaling by internalized TSH receptors (TSHRs) was persistent, whereas that by cell-surface TSHRs was apparently transient, but the reasons for the transient signaling by cell-surface TSHRs were not investigated. Here, we developed and used fluorescence resonance energy transfer (FRET)-based methods to precisely compare the kinetics of TSH binding and dissociation from cell-surface TSHRs with those of the subsequent termination of cAMP signaling directly in living cells. Our results indicate that both TSH binding to human TSHRs expressed in a human embryonic kidney cell line (HEK 293) and the ensuing cAMP signals are rapidly and fully reversible (t(1/2,off)=2.96±1.04 and 2.70±0.73 min, respectively). The FRET measurement of TSH binding was specific, as shown by the lack of a detectable interaction between TSH and the β(2)-adrenergic receptor expressed in control cells. Enhancing TSHR internalization by β-arrestin 2 overexpression did not modify the reversibility of TSHR-cAMP signaling. These findings strengthen the view that the cointernalization of TSH-TSHR complexes to a signaling compartment present in thyroid, but not in HEK 293 cells, is responsible for persistent cAMP signaling.
Article
Fluorescence and bioluminescence resonance energy transfer (FRET and BRET) techniques allow the sensitive monitoring of distances between two labels at the nanometer scale. Depending on the placement of the labels, this permits the analysis of conformational changes within a single protein (for example of a receptor) or the monitoring of protein-protein interactions (for example, between receptors and G-protein subunits). Over the past decade, numerous such techniques have been developed to monitor the activation and signaling of G-protein-coupled receptors (GPCRs) in both the purified, reconstituted state and in intact cells. These techniques span the entire spectrum from ligand binding to the receptors down to intracellular second messengers. They allow the determination and the visualization of signaling processes with high temporal and spatial resolution. With these techniques, it has been demonstrated that GPCR signals may show spatial and temporal patterning. In particular, evidence has been provided for spatial compartmentalization of GPCRs and their signals in intact cells and for distinct physiological consequences of such spatial patterning. We review here the FRET and BRET technologies that have been developed for G-protein-coupled receptors and their signaling proteins (G-proteins, effectors) and the concepts that result from such experiments.
Article
D(1) dopamine receptors are primary mediators of dopaminergic signaling in the CNS. These receptors internalize rapidly following agonist-induced activation, but the functional significance of this process is unknown. We investigated D(1) receptor endocytosis and signaling in HEK293 cells and cultured striatal neurons using real-time fluorescence imaging and cAMP biosensor technology. Agonist-induced activation of D(1) receptors promoted endocytosis of receptors with a time course overlapping that of acute cAMP accumulation. Inhibiting receptor endocytosis blunted acute D(1) receptor-mediated signaling in both dissociated cells and striatal slice preparations. Although endocytic inhibition markedly attenuated acute cAMP accumulation, inhibiting the subsequent recycling of receptors had no effect. Further, D(1) receptors localized in close proximity to endomembrane-associated trimeric G protein and adenylyl cyclase immediately after endocytosis. Together, these results suggest a previously unanticipated role of endocytosis, and the early endocytic pathway, in supporting rapid dopaminergic neurotransmission.
Article
Remarkable progress has been made in the field of G protein-coupled receptor (GPCR) structural biology during the past four years. Several obstacles to generating diffraction quality crystals of GPCRs have been overcome by combining innovative methods ranging from protein engineering to lipid-based screens and microdiffraction technology. The initial GPCR structures represent energetically stable inactive-state conformations. However, GPCRs signal through different G protein isoforms or G protein-independent effectors upon ligand binding suggesting the existence of multiple ligand-specific active states. These active-state conformations are unstable in the absence of specific cytosolic signaling partners representing new challenges for structural biology. Camelid single chain antibody fragments (nanobodies) show promise for stabilizing active GPCR conformations and as chaperones for crystallogenesis.