B-Arrestin Mediated Receptor Trafficking and Signal Traduction - Lefkowitz 2011

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Review

b-arrestin-mediated receptor
trafficking and signal transduction
Sudha K. Shenoy1,2 and Robert J. Lefkowitz1,3,4
1
Department of Medicine, Duke University Medical Center, Box 3821, Durham, NC 27710, USA
2
Department of Cell Biology, Duke University Medical Center, Box 3821, Durham, NC 27710, USA
3
Department of Biochemistry, Duke University Medical Center, Box 3821, Durham, NC 27710, USA
4
Howard Hughes Medical Institute, Duke University Medical Center, Box 3821, Durham, NC 27710, USA

b-Arrestins function as endocytic adaptors and mediate established that b-arrestins and GRKs serve as universal
trafficking of a variety of cell-surface receptors, including regulators of the 7TMR family.
seven-transmembrane receptors (7TMRs). In the case of 7TMRs respond to a wide array of extracellular stimuli
7TMRs, b-arrestins carry out these tasks while simulta- that includes hormones, neurotransmitters, lipids, pep-
neously inhibiting upstream G-protein-dependent sig- tides, ions and sensory stimuli and elicit a wide variety
naling and promoting alternate downstream signaling of physiological responses, making them important mole-
pathways. The mechanisms by which b-arrestins inter- cules for drug discovery. It is estimated that at least 40% of
act with a continuously expanding ensemble of protein marketed prescription drugs are either agonists or antago-
partners and perform their multiple functions including nists acting either directly or indirectly at the 7TMRs [9]. It
trafficking and signaling are currently being uncovered. is noteworthy that the two nonvisual arrestins and four
Molecular changes at the level of protein conformation ubiquitously expressed GRKs regulate most of the large
as well as post-translational modifications of b-arrestins number of members of the7TMR superfamily. Although
probably form the basis for their dynamic interactions the various 7TMRs show very different patterns of expres-
during receptor trafficking and signaling. It is becoming sion and functions, they are all related by virtue of their
increasingly evident that b-arrestins, originally discov- highly conserved heptahelical structure.
ered as 7TMR adaptor proteins, indeed have much Upon agonist stimulation, 7TMRs undergo conforma-
broader and more versatile roles in maintaining cellular tional changes, thus exposing binding sites for heterotri-
homeostasis. In this review paper, we assess the tradi- meric G proteins in their intracellular domains. This leads
tional and novel functions of b-arrestins and discuss the to exchange of GDP for GTP on the Ga subunit initiating
molecular attributes that might facilitate multiple inter- the dissociation of G protein a and bg dimers that act as
actions in regulating cell signaling and receptor traffick- signaling units and activate various effectors (e.g. adenylyl
ing. cyclase by the a subunit and K+ channels by bg dimers)
[10]. Thus, traditionally, most 7TMR signaling has been
Regulation of 7-transmembrane receptors (7TMRs) considered to be mediated by G-protein-receptor coupling.
b-Arrestins 1 and 2 (also known as arrestins 2 and 3) along Agonist-occupied 7TMRs become immediate substrates for
with the visual rod (arrestin 1) and cone (arrestin 4) GRK-mediated phosphorylation and, in turn, the phos-
arrestins constitute a small family of cytosolic adaptor phorylated receptors recruit the cytosolic adaptors b-
proteins [1–5]. The initial discovery and cloning of b-arrest- arrestins 1 and 2. b-arrestin-binding blocks further G
ins 1 and 2 immediately followed the characterization of protein activation by sterically hindering access to
the visual rod arrestin [6]. The three proteins were identi- the receptor binding domains causing desensitization
fied as crucial mediators of desensitization of their cognate of 7TMR–G protein signaling (Figure 1). In addition,
7TMRs, namely the b2 adrenergic receptor (b2AR, for b- b-arrestins also scaffold enzymes, such as phosphodies-
arrestins) and Rhodopsin (for rod arrestin). A series of in terases and diacylglycerol kinases, which degrade second
vitro reconstitution experiments involving b2AR com- messengers generated by G protein coupling, thus provid-
plexes and the quenching of the activity of the effector ing additional mechanisms for efficient dampening of sig-
adenylyl cyclase led to the conclusion that 7TMR desensi- naling [11,12]. This scaffolding function of b-arrestins has
tization minimally involves two steps, namely seryl/threo- only been recently appreciated, raising the possibility that
nyl phosphorylation of agonist-stimulated receptors by the it might be a general mechanism by which b-arrestins
G-protein-coupled receptor kinases (GRKs) followed by engage different enzymatic machineries to deplete second
high-affinity interactions of b2AR and nonvisual arrestins messenger pools.
[7]. The GRK family consists of seven homologs, of which
GRKs 1 and 7 phosphorylate the visual opsins, whereas the b-Arrestins and 7TMR endocytosis
GRKs 2–6 regulate various members of the huge family of An almost universal property of cell-surface receptors
7TMRs as well as other cell-surface receptors [8]. It is now (including 7TMRs) is their ability to internalize upon
Corresponding authors: Shenoy, S.K. (sudha@receptor-biol.duke.edu); Lefkowitz,
exposure to a ligand [13–15]. Many 7TMRs internalize
R.J. (lefko001@receptor-biol.duke.edu) via clathrin-coated vesicles, but other mechanisms of
0165-6147/$ – see front matter ! 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.tips.2011.05.002 Trends in Pharmacological Sciences, September 2011, Vol. 32, No. 9 521
(Figure_1)TD$IG][ Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

MULTIFACETED FUNCTIONS OF β-ARRESTINS


DESENSITIZATION ENDOCYTOSIS SIGNALING
Agonist

7TMR

PP

PP
n
PP

sti
ti n arreClathrin β-arrestin
β rr es β-
α

MAPKKK
a GRK
β-

MAPK
MAPKK
γ AP2 NSF

PDE4D Arf6

DAG-K Mdm2

USP33

G protein uncoupling Internalization Chemotaxis


2ndmessenger Intracellular trafficking: Apoptosis
degradation Recycling or lysosomal Metastasis
degradation Protein translation
TRENDS in Pharmacological Sciences

Figure 1. Multifaceted functions of b-arrestins. Desensitization: agonist stimulation of 7TMRs leads to G protein coupling and activation, following which the receptors are
rapidly phosphorylated by G-protein-coupled receptor kinases (GRKs). Phosphorylated receptors present high-affinity binding surfaces to recruit the cytosolic adaptors, b-
arrestins. Steric binding by b-arrestin interferes with further G protein coupling leading to the desentization of G protein-dependent signaling. b-Arrestins also scaffold the
second messenger degrading enzymes (phosphodiesterase 4D, PDE4D, that degrades cAMP and diacylglycerol kinase, DAG-K, that converts diacylglycerol to phosphatidic
acid). Endocytosis: agonist stimulation promotes rapid internalization of cell-surface 7TMRs into clathrin-coated vesicles. This internalization is facilitated by b-arrestin
binding, which has specific binding domains for clathrin and AP2 interactions. b-Arrestin binding to other endocytic proteins is also required for efficient receptor
internalization: N-ethylmaleimide-sensitive fusion (NSF) protein and ADP ribosylation factor 6 (Arf6). The interaction between b-arrestin and the E3 ubiquitin ligase Mdm2
(mouse double minute 2) promotes ubiquitination of b-arrestin, which facilitates robust binding of b-arrestin with both cargo (7TMR) as well as endocytic machinery
(clathrin and AP2). Receptor internalization is followed by postendocytic sorting of internalized receptors for recycling or lysosomal degradation. Signaling: b-arrestin
acquires an active conformation upon forming a complex with agonist-stimulated 7TMRs and scaffolds MAP kinase, MAP kinase kinase and MAP kinase kinase kinase,
leading to the robust activation of MAP kinase, and subsequently targets the activated kinase to distinct subcellular compartments. Such b-arrestin-dependent MAP kinase
activity has been shown to regulate cellular chemotaxis, apoptosis, cancer metastasis and protein translation.

internalization also exist. Receptor trafficking is an intri- of amino acids towards the C terminal region of b-arrestins
cate process that involves dynamic protein–protein inter- 1 and 2 [17]. A recent X-ray crystal structure of b-arrestin 1
actions as well as activation or deactivation of associated with clathrin suggests that additional interacting regions
proteins. Many well-characterized 7TMRs utilize b-arrest- in b-arrestin 1 (splice variant, L) might regulate dynamic
ins as endocytic adaptors, but mechanisms involving al- b-arrestin–clathrin interactions [21,22]. Studies in the
ternate adaptors also exist [15]. The idea that b-arrestins past decade have identified additional binding partners
are not just signal terminators was fueled by several of b-arrestin that facilitate receptor internalization. These
studies that demonstrated novel roles for b-arrestins in include interactions with the clathrin adaptor AP2, the N-
mediating 7TMR endocytosis via clathrin-coated pits ethylmaleimide-sensitive fusion protein (NSF), the small
(CCPs) [16,17], which are invaginations occurring at the G protein ARF6 and PI4P kinase, all of which exploit the
plasma membrane that are enveloped with clathrin and adaptor functions of b-arrestin in mediating 7TMR endo-
adaptor–protein2 complexes. The CCPs, aided by the ac- cytosis (Figure 1) [23,24]. Mass spectrometry-based analy-
tivity of the GTPase Dynamin, pinch off as clathrin-coated ses of b-arrestin immunoprecipitates also suggest that a
vesicles (CCVs) resulting in the internalization of cell- complex network of b-arrestin interacting proteins regu-
surface receptors. Overexpression of b-arrestin was shown late endocytosis [25]. The interaction of b-arrestins with
to enhance agonist-stimulated b2AR internalization and the clathrin adaptor AP2 involves a peptide motif in the
rescue the endocytosis of defective mutant receptors [18]. carboxyl terminal region of b-arrestin [26,27]. This region
That b-arrestins are essential for endocytosis of 7TMRs is homologous to the AP2 binding domain in autosomal
has been demonstrated by studies performed in mouse recessive hypercholesterolemia (ARH) protein, an adaptor
embryonic fibroblast cells isolated from b-arrestins 1 that links the low-density lipoprotein (LDL) receptor to
and 2 double knockout embryos [19]. More recently, this AP2 and the clathrin machinery [27]. Upon receptor acti-
has been corroborated by small interfering RNA (siRNA)- vation of b-arrestin, this homologous peptide assumes an
mediated downregulation of b-arrestin isoforms in differ- a-helical conformation triggering b-arrestin-binding with
ent cell types [20]. the b2-appendage platform in AP2, thus promoting recruit-
Studies that characterized a direct interaction between ment of 7TMR–b-arrestin complexes to CCVs [28].
b-arrestin 1 and clathrin have shed light on the mecha- Upon agonist stimulation, many 7TMRs induce translo-
nisms by which b-arrestins promote 7TMR endocytosis cation of cytosolic b-arrestins to the plasma membrane,
[15]. Clathrin-binding involves a highly conserved stretch which can occur even in the absence of G protein activation

522
Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

and can be visualized by monitoring green fluorescent pro- platforms, which facilitate multiple interactions [48]. In-
tein (GFP)–b-arrestin [29,30]. The translocated b-arrestin deed, Ub has been suggested to play such a role in other
forms a complex with the 7TMR, and the stability of these protein–protein interactions and cargo recognition along
complexes governs the subsequent intracellular trafficking the endocytic pathway, because many endocytic proteins
itinerary of the activated receptors [31,32]. Specific molecu- contain Ub binding and Ub recognition domains [49].
lar determinants, namely the occurrence of a serine/threo- Interestingly, PalF, an Aspergillus nidulans protein that
nine motif in the receptor C-tail, and the pattern of agonist- is distantly related to the arrestins, is also ubiquitinated
induced ubiquitination (covalent modification with Ubiqui- upon activation of the 7TMR PalH by alkaline pH, suggest-
tin or Ub) of b-arrestin determine the stability of 7TMR–b- ing that b-arrestin ubiquitination could be a modification
arrestin complexes [32–34]. 7TMRs, such as the angiotensin that has been conserved during the evolutionary process
II 1a (AT1a) and vasopressin 2 (V2) receptors that contain [50].
these clustered phosphorylation sites, induce sustained In addition to ubiquitination, b-arrestin 2 is SUMOy-
ubiquitination of b-arrestin 2 and form tight complexes that lated (SUMO: small ubiquitin like modifier, which is cova-
cointernalize into endocytic vesicles. These complexes are lently attached to lysines just as in ubiquitination [51]).
also associated with sustained mitogen-activated protein The conserved SUMO motif c-K-x-E that serves as the
kinase (MAPK) activity and function as ‘signalosomes’ recognition domain for the single SUMO-conjugating en-
(denoting a signaling receptor complex or scaffold associated zyme UBC-9 makes it possible to predict SUMOylation of
with endosomes). Receptors that show this pattern of b- substrate proteins in contrast with ubiquitination that
arrestin recruitment are referred to as ‘class B’ receptors does not involve a consensus motif and involves an array
[32]. By contrast, 7TMRs, such as the b2AR that contain of E3 and E2 enzymes [52].
different phosphorylation codes in their cytoplasmic The lysine within the motif LKDE (c-K-x-E) in the
domains than the V2 or AT1a receptors, induce transient carboxyl terminal region of bovine b-arrestin 2 is suggested
ubiquitination of b-arrestin 2 and form only transient sig- to be the major site of modification and, interestingly, this
naling complexes at the plasma membrane. Such receptors consensus SUMO motif is absent in human and murine b-
are referred to as ‘class A’ receptors [32]. Notably, replacing arrestin [53]. Unlike ubiquitination, the reported SUMOy-
the b2AR carboxyl tail with that of either the AT1aR or the lation of b-arrestin 2 does not influence the affinity of
V2R, or alternatively fusing Ub to the C terminus of b- 7TMR interaction, but is important for AP-2 interaction
arrestin 2 converts b2AR–b-arrestin complexes to stable and receptor internalization via CCVs [53]. It is notewor-
signalosomes [33,35]. The serine/threonine clusters gener- thy that consensus SUMO motifs are also present in b-
ally occur 9–19 residues downstream of the palmitoylation arrestin 1 and GRKs as well as b1AR, b2AR and a2AR, but
site found in the Rhodopsin family of 7TMRs and mutation absent in AT1aR and V2R (Table 1). The roles of SUMOyla-
of these clusters to alanines diminishes receptor phosphor- tion in b-arrestin 1, GRKs and 7TMRs remain to be deter-
ylation and stability of b-arrestin recruitment for the neu- mined. It is intriguing that two of the well-characterized
rotensin-1, oxytocin, AT1a and V2 receptors, but not for class B receptors lack these sites, whereas the established
urotensin II receptor [34,36]. In some 7TMRs, where there class A receptors contain them, which could be yet another
are more than one phosphorylation cluster, the sites associ- factor differentiating trafficking properties of these two
ated with b-arrestin binding are not necessarily the sites of classes of 7TMRs.
predominant phosphorylation [37–39]. The stability of b-
arrestin–7TMR interaction is also influenced by additional b-Arrestins facilitate trafficking of unconventional
binding determinants present in other intracellular 7TMRs
domains of 7TMRs [40–44]. The role of receptor carboxyl Frizzleds (FZDs) are 7TMRs that are activated by the
tail phosphorylation on b-arrestin recruitment and subse- Wingless/Int-1 (WNT) family of secreted lipoglycoproteins.
quent b-arrestin functions (desensitization, internalization, WNT/FZD signaling is important for embryonic develop-
signaling) is also dependent upon the particular GRK iso- ment, generation of cell polarity, maintenance of stem cell
form involved (see section on ‘Phosphorylation codes set by pluripotency and its dysregulation leads to degenerative
GRKs’ below and [45] for more details). diseases and cancer. FZDs bind to phosphorylated adaptor
The interaction of b-arrestin 2 with the E3 ubiquitin proteins called disheveled (DVL), and activation of FZDs
ligase Mdm2 has been shown to be crucial for mediating leads to stabilization of the transcriptional factor b-cate-
7TMR endocytosis. Mdm2 specifically and transiently ubi- nin, its subsequent nuclear translocation and transcription
quitinates b-arrestin2 upon agonist stimulation of the of specific genes [54]. b-Arrestin 2 is recruited to the DVL–
b2AR [46,47]. The functional effects of Mdm2-mediated FZD complex, and b-arrestin–DVL binding is required for
b-arrestin 2 ubiquitination have been addressed by multi- the clathrin-dependent internalization of FZD4 receptor
ple experimental approaches. Reduction of Mdm2 activity [55]. Additionally, WNT-11 induces the formation of a
by using dominant negative forms, siRNA mediated knock- FZD7–DVL–AP-2 complex that translocates to Ras related
down or Mdm2 null cells all ablate b-arrestin ubiquitina- gene from brain 5 or RAB5-positive endosomes in the
tion and b2AR internalization [46,47]. Moreover, Xenopus dorsal marginal zone in a b-arrestin-dependent
ubiquitinated b-arrestin shows enhanced clathrin binding, manner, and this process is crucial for planar cell polarity
7TMR interaction as well as scaffolding of MAPKs, where- signaling [56].
as nonubiquitinated b-arrestins are impaired in each of Another atypical 7TMR that is regulated by b-arrestin
these functional interactions. This suggests that the Ub is the Smoothened (Smo) receptor that transduces hedge-
moieties on b-arrestin might serve as efficient binding hog (Hh) signaling, which is important for vertebrate

523
Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

Table 1. Sumoylation sites in b-arrestins, GRKs and 7TMRs


Protein name Species Accession number Residue number Sequence SUMO motif
b-Arrestin 1 Homo sapiens NP_004032 294 GKLKHED Type I: C-K-X-E
357 PKPKEEP Type I: C-K-X-E
400 KGMKDDK Type II: non-consensus
403 KDDKEEE Type II: non-consensus
b-Arrestin 2 Homo sapiens NP_004304 295 GKLKHED Type I: C-K-X-E
b-Arrestin 2 Bos taurus NP_001192206 295 GKLKHED Type I: C-K-X-E
411 KGLKDED Type I: C-K-X-E
GRK2 Homo sapiens NP_001610 90 EEIKKYE Type II: non-consensus
94 KYEKLET Type II: non-consensus
644 VQWKKEL Type I: C-K-X-E
GRK3 Homo sapiens NP_005151 90 EEIKEYE Type II: non-consensus
644 VQWKKEL Type I: C-K-X-E
GRK4 Homo sapiens NP_892027 134 LGLKEEN Type I: C-K-X-E
227 KKRKGEA Type II: non-consensus
314 RDLKPEN Type I: C-K-X-E
394 EKVKWEE Type I: C-K-X-E
403 QRIKNDT Type II: non-consensus
527 DINKSES Type II: non-consensus
GRK5 Homo sapiens CAI15804 226 KKRKGES Type II: non-consensus
313 RDLKPEN Type I: C-K-X-E
393 EKVKREE Type I: C-K-X-E
GRK6 Homo sapiens NP_001004106 226 KKRKGEA Type II: non-consensus
313 RDLKPEN Type I: C-K-X-E
358 EVVKNER Type I: C-K-X-E
393 KKIKREE Type I: C-K-X-E
466 PPFKPDP Type II: non-consensus
b2AR Homo sapiens P07550 60 AIAKFER Type I: C-K-X-E
227 QEAKRQL Type II: non-consensus
232 QLQKIDK Type II: non-consensus
235 KIDKSEG Type II: non-consensus
b1AR Homo sapiens P08588 85 AIAKTPR Type II: non-consensus
253 EAQKQVK Type II: non-consensus
476 SESKV Type II: non-consensus
a2AR Homo sapiens NP_000672 80 RALKAPQ Type II: non-consensus
AT1aR Homo sapiens ABY87526 No sites predicted
V2R Homo sapiens ACR39021 No sites predicted
Sites were identified by sequence analysis with the software SUMOsp 2.0 [135].

development. b-Arrestin 2 is recruited to Smo after GRK2 b-Arrestins regulate endocytosis of non7TMRs and ion
phosphorylation of Smo and leads to its clathrin-depen- channels
dent internalization [57]. At cilia, b-arrestin 2 interacts Although b-arrestins were identified in the context of
with Kif3a, a subunit of the kinesin-2 motor complex, 7TMR regulation, there is increasing evidence that they
which is an anterograde molecular motor that transports function as adaptors for diverse cell surface receptor mole-
protein complexes to and within cilia. This interaction cules. The first example was the demonstration that b-
between b-arrestin and Kif3a is required for the localiza- arrestin 1 facilitates clathrin-dependent endocytosis of the
tion of Smo in primary cilia and for efficient Gli-dependent insulin-like growth factor receptor (IGF-1R) [63]. b-
transcription as well as proper functioning of the centro- Arrestin 2 can also bind to and enhance endocytosis of
some [58,59]. Knockdown of b-arrestin 2 in zebrafish the LDL receptor via clathrin-coated vesicles and further
results in a phenotype with multiple defects, including influence lipoprotein metabolism [64]. Generally, b-arrest-
defective somite patterning and craniofacial development, ins function as adaptors for phosphorylated forms of re-
which are regulated by Hh [60]. The novel role of b- ceptor and nonreceptor proteins. However, their binding to
arrestin 2 in Smo signaling is associated with GRK2- nephrin, a single transmembrane receptor is prevented
mediated phosphorylation of the Smo receptor cyto- upon tyrosine phosphorylation of the receptor by Src family
plasmic domains as demonstrated in HEK-293 cells and member Yes [65]. Nephrin proteins are essential for proper
zebrafish embryos [61,62]. Knockdown of zebrafish GRK2/ renal filtration. Nephrin molecules in the glomerular slit
3 results in attenuation of Hh transcriptional activity, diaphragm form a porous structure that results from the
impaired muscle development and neural patterning. interaction between the extracellular domains (which are
These findings underscore a central role for the GRK-b– IgG-like and 35 nm in length) of neighboring nephrin
arrestin system in Smo signaling in vertebrates. molecules [66]. Internalization of nephrin–b-arrestin 2

524
Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

complexes is preceded by a loss of these homophilic inter- androgen receptor [81,82]. Interestingly Mdm2 ubiquiti-
actions at the extracellular domains, decreased phosphor- nates b-arrestin 2 itself and when scaffolded by b-arrestin
ylation by Yes and decreased binding to podocin. Loss of 2, it acts as an E3 ligase for GRK2 and phosphodiesterase
cell-surface nephrin molecules disrupts the porous struc- (PDE) 4D [46,83,84].
ture of the glomerular slit diaphragm [65]. In contrast to The adaptor role of b-arrestin 1 has recently been shown
the scenario with nephrins, b-arrestin 2 binds to trans- for TRPV4 and NHE1 ubiquitination, in which the E3
forming growth factor (TGF)-b type III receptor upon ligases AIP4 and Nedd4-1 are involved, respectively
receptor phosphorylation by the TGF-b type II receptor. [68,72]. In both cases, ubiquitination leads to internaliza-
This leads to endocytosis and downregulation of TGF-b tion and subsequent downregulation of the ubiquitinated
signaling [67]. Akin to the TGF-b III receptor, the Na+/H+ species. TRPV4 ubiquitination mediated by b-arrestin 1–
exchanger NHE1 and NHE5 isoforms also internalize and AIP4 is induced upon ligand activation of the AT1aR recep-
are downregulated in a b-arrestin-dependent manner tor, thus linking a 7TMR and an ion channel and providing
[68,69]. In addition, b-arrestins are key regulators of the a mechanism for ubiquitin-dependent Ca2+ regulation. The
ligand-gated ion channel nicotinic cholinergic receptor E3 ligase adaptor role of b-arrestins is also observed
[70], cardiac Ca(v)1 voltage-gated channels [71] and the beyond the mammalian system: Kurtz, the Drosophila b-
transient receptor potential (TRP) ion channel, TRPV4 [72] arrestin homolog binds and escorts the E3 ligase deltex to
(see below). mediate ubiquitination of the Notch receptor [85]. Ub-
dependent postendocytic sorting of internalized receptors
b-Arrestins act as adaptors for ubiquitin ligases and involves recognition of ubiquitinated cargo by the protein
deubiquitinases and control postendocytic sorting complexes termed ESCRTs, whose role has been exten-
Ubiquitination of cell-surface receptors has emerged as an sively characterized in yeast [86]. An interaction between
important post-translational modification that defines the b-arrestin 1 and STAM-1, a protein subunit of ESCRT-0,
trafficking itinerary of internalized receptors [73,74]. Ubi- has recently been suggested to inhibit lysosomal degrada-
quitination is a highly regulated process in which the C tion of CXCR4 [87].
terminal glycine of Ub is appended covalently to the epsi- b-Arrestin 2 also acts as an adaptor for two homologous
lon amino group of a lysine residue in the substrate [75]. deubiquitinases (USP20 and USP33) that deubiquitinate
Transfer of ubiquitin to substrate requires an enzymatic the b2AR [88]. An increase in USP20/33 expression pro-
cascade involving three distinct enzyme activities: first Ub motes receptor deubiquitination. This leads to an increase
is activated by the enzyme E1, activated Ub is bound and in recycling and resensitization of the b2AR, whereas
carried by the E2 enzyme and finally substrate modifica- inhibition of their enzymatic activity stabilizes receptor
tion proceeds with the aid of the third enzyme called E3 ubiquitination and promotes lysosomal degradation [88].
ubiquitin ligase (for a detailed review on this, see [75]). It is Deubiquitination of Protease Activated Receptor 2 (PAR2)
estimated that !600 E3 ligases ubiquitinate thousands of involves other USP enzymes, but whether b-arrestins play
protein substrates with remarkable specificity and timing. a role in this process is not known [89].
Recognition of each substrate is a tightly regulated process,
and it appears that E3 ligases have tailored distinct mech- b-Arrestin-related proteins: the a-arrestins
anisms for different substrates. For example, they can bind b-Arrestin homologs are not expressed in yeast and lower
to phosphorylated domains or utilize adaptor proteins. As metazoans, but structurally similar b-arrestin-related pro-
discussed below, b-arrestins act as indispensable E3 ligase teins, called arrestin-related trafficking adaptors (ARTs)
adaptors for 7TMRs and non7TMRs to mediate ubiquiti- with the characteristic ‘arrestin domain’ fold, have been
nation, and they can play an equally important role in shown to function as E3 ligase adaptors in yeast [90–92].
escorting deubiquitinases (Figure 2). b-Arrestins can also The yeast ARTs are localized at the late Golgi and trans-
be involved in determining the dynamics of ubiquitination/ locate to the plasma membrane in response to chemical
deubiquitination of endocytic proteins that are important stress or changes in nutrients. At the membrane, they
for proper steering of internalizing cargo across various recognize and mediate ubiquitination of specific plasma
vesicular pathways. membrane nutrient transporters, leading to their intracel-
For mammalian 7TMRs, ubiquitination was first dem- lular trafficking and degradation in the yeast vacuole [92].
onstrated for the b2AR and the chemokine CXCR4 recep- The interesting feature that sets these ARTs apart from
tors [46,76]. Although prior studies had indicated that the mammalian b-arrestins is the presence of characteris-
endocytosis of yeast 7TMRs Ste2 and Ste3 depends on tic polyproline motifs that are known to bind to WW
ubiquitination, in the case of most mammalian 7TMRs, domains contained in the HECT E3 ligase Rsp5 (yeast
ubiquitination is not required for receptors to internalize, homolog of mammalian Nedd4). Whether the yeast ARTs
but is required for their targeting to lysosomal compart- regulate ubiquitination and trafficking of the 7TMRs, Ste2
ments for degradation [77–80]. b2AR ubiquitination is and Ste3 remains to be determined.
detected in b-arrestin-null mouse embryonic fibroblasts Mammalian cells express five ART homologs, termed
only after re-expression of b-arrestin 2, which acts as arrestin-domain containing proteins (ARRDC 1–5). In addi-
the adaptor for recruiting the HECT domain containing tion, vacuole sorting protein 26 VPS26, which has no se-
E3 ligase Nedd4 to the activated b2AR complex [46,80]. b- quence identity with b-arrestins, was also shown to share a
Arrestin 1 acts as an essential adaptor for the ubiquitina- similar structural fold with mammalian arrestins [93,94].
tion of the IGF-1R by the RING domain containing E3 Vps26 forms part of a retromer subcomplex involved
ligase Mdm2, whereas b-arrestin 2 serves the role for the in recognition of cargo at the endosome and facilitates

525
(Figure_2)TD$IG][ Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

TRPV4 AT1 aR
(a) 7TMR
(c)

UU UU

PP
UU PP Deubiquitination U U
Ubiquitination
B
DU

E3 Ligase
Ubiquitination
Internalization

β-arrest
E3 Ligase
in
C
la
AP

th
2

r
in

(b) (d)

Na+
NHE1
IGF-1R

H+
U
U U
Ubiquitination
Ubiquitination
Internalization
E3 Ligase
E3 Ligase
β-arrest
in
C
la
AP

th
2

rin

TRENDS in Pharmacological Sciences

Figure 2. b-Arrestins function as versatile adaptors for different cell-surface receptors. (a) For 7TMRs, b-arrestins act as clathrin–AP2 adaptors to facilitate receptor
internalization, E3 ubiquitin ligase and deubiquitinase adaptors to facilitate receptor ubiquitination and deubiquitination. (b) For the insulin-like growth factor receptor (IGF-
1R), b-arrestin 1 functions as a clathrin adaptor as well as an E3 ligase adaptor, facilitating internalization and ubiquitination of IGF-activated receptors. (c) The cell surface
transient receptor potential vanilloid 4 (TRPV4) channel remains complexed with the angiotensin 1a receptor (AT1aR), and upon AT1aR activation becomes ubiquitinated in a
b-arrestin 1-dependent manner. In this process, b-arrestin 1 functions as an E3 ubiquitin ligase adaptor. (d) The Na+/H+ exchanger channel NHE1 is internalized and
ubiquitinated in a b-arrestin-dependent manner.

retrograde transport to the Golgi complex. b-Arrestins and the b-arrestins and a-arrestins, b-arrestins have unique
visual arrestins contain a ‘polar core’ region in their struc- and crucial roles in mediating cellular homeostasis and
ture, disruption of which is thought to precede conforma- embryonic development.
tional changes and activation of arrestins [95]. VPS26 also
has a similar polar core, but the structural contacts involve b-Arrestins balance 7TMR trafficking and signal
different side chains [93]. transduction and lead to physiological outcomes
Future studies should reveal the exact role of these Receptor internalization was originally considered a
arrestin-related proteins in conjunction with those of b- means for turning off signaling because activated receptors
arrestins during receptor trafficking and signaling. It is are sequestered away from the cell surface and cannot bind
probable that b-arrestins utilize these extra adaptors to extracellular ligands. However, it is now evident that, in
drive internalized receptor complexes through distinct many cases, signal transduction persists after the receptor
vesicle populations. The fact that double knockout of b- has internalized, especially in the initiation of b-arrestin-
arrestins 1 and 2 is associated with prenatal or perinatal dependent signaling, which coincides with the early stages
mortality [19] indicates that the arrestin-related proteins of receptor endocytosis. b-Arrestins 1 and 2 bind various
do not complement b-arrestin functions in vivo. Thus, kinases and regulatory proteins, and in many cases b-
although there could be some commonalities between arrestins function as receptor-activated scaffolds to coor-

526
Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

dinate upstream and downstream components of particu- 7TMR GPR120 and recruiting b-arrestins [107]. The
lar signal transduction pathways [8,96]. For example, b- GPR120–b-arrestin complex internalizes and sequesters
arrestin binds cRAF-1 (upstream MAPK kinase kinase) the TGF-b activated kinase 1 binding protein, TAB1, and
and ERK2 (downstream MAPK) and recruits MEK1 blocks the downstream activation of nuclear factor-kB and
(MAPK kinase), thus assembling the core components Janus kinase, thus inhibiting the inflammatory responses
for activating ERK2 (Figure 1) [97,98]. Furthermore, such mediated by toll-like receptor 4 and tumor necrosis factor a.
scaffold assembly is often potentiated by 7TMR activation, This blockade of inflammatory cytokine receptor signaling
thus providing a stimulus-dependent signal transduction promotes the return of anti-inflammatory macrophages to
event to direct the biological response of the cell. An addi- adipose tissue and restores insulin sensitivity in obese mice
tional feature of such signaling scaffolds is that upon high [107]. b-Arrestin-biased signaling and trafficking that are
affinity interaction of b-arrestin and 7TMR, they become prevalent for the b2AR, b1AR, AT1aR and parathyroid hor-
localized on endosomes (i.e. signalosomes), which bestows mone1 receptor (PTH1R) in model cells are likely to be linked
compartmentalization on 7TMR signaling [99,100]. Thus, b- to distinct physiological effects observed in these cases,
arrestin-dependent signals are not only temporally distinct namely mitigation of catecholamine-induced cardiomyocyte
from the initial second-messenger responses generated by G apoptosis for the bARs and mediation of b-arrestin-depen-
protein signaling, they are also spatially segregated from dent anabolic bone formation for the PTH1R [105]. These
them. Although a direct connection has not been demon- studies suggest the exciting possibility that b-arrestin-de-
strated between endocytosis and all b-arrestin-dependent pendent signaling via these receptors can be exploited for
signaling pathways characterized thus far, it is probable developing new drugs to prevent heart failure and treat
that the two processes are intimately linked. osteoporosis and that b-arrestin–receptor signalosomes
In general, a 7TMR agonist is capable of activation of G could also be targets for therapeutic intervention.
protein-dependent pathways and subsequent regulation
and signaling by GRKs and b-arrestins. In the presence Molecular mechanisms regulating b-arrestin functions
of bound b-arrestin 2, phosphorylated b2AR and M2 mus- b-Arrestins undergo post-translational modifications,
carinic receptor (M2R) display increased affinity to ago- most often in response to 7TMR stimulation (Figure 3)
nists, suggesting that similar to G protein–receptor and, as discussed below, these molecular changes in b-
complexes, b-arrestin–phosphoreceptor complexes can ex- arrestin could represent its activated conformation(s) and
ist as ‘active’ ternary complexes [101]. It is now evident form the basis of its various functional effects in mediating
that certain ligands can preferentially activate b-arrestins receptor trafficking and signaling.
while blocking or minimally activating G proteins or vice
versa [102,103]. This ligand-directed signaling is defined Phosphorylation of b-arrestin
as biased agonism, in which receptor-activated signal b-Arrestins 1 and 2 are cytosolic phosphoproteins that,
transduction (and perhaps physiological effects) are chan- upon agonist-stimulated recruitment to the b2AR, become
neled through only one form of active coupling (either G rapidly dephosphorylated (Figure 3a) [108–110]. In b-
protein or b-arrestin) [104,105]. The existence of distinct b- arrestin 1, serine 412 is phosphorylated by ERK1/2 when
arrestin-biased signaling suggests the prevalence of dis- b2ARs are activated and by GRK5 when 5-HT4 receptors
tinct activated conformation(s) of the receptor for signaling are activated [108,111]. Threonine 383 in rat b-arrestin 2
through G proteins or b-arrestins. This necessitates fur- (which is threonine 382 of bovine b-arrestin 2) is phosphor-
ther modification of receptor active-state models to include ylated by casein kinase II [110,112]. Studies conducted
additional conformation(s) of the receptor and formation of with both phosphomimetic (serine/threonine mutated to
additional ternary complexes [103,104]. Examples of bi- aspartate) and phosphorylation-impaired (serine/threo-
ased-signaling and related physiological effects where a nine mutated to alanine) mutants have indicated that
direct relationship to b-arrestin-dependent trafficking was dephosphorylation of b-arrestins 1 and 2 is important
demonstrated are discussed below (for a more extensive for clathrin interaction and clathrin-dependent–receptor
discussion and examples, see [105]). internalization. Such mutations of serine 412 or threonine
Serotonin-stimulated ERK1/2 activation and internali- 383, respectively, in b-arrestins 1 or 2 do not alter their
zation of 5-HT2ARs requires b-arrestin 2 and leads to a properties with regard to receptor binding and desensiti-
head-twitch response in mice (this is an indication that the zation. However, when b-arrestin 1 is phosphorylated by
7TMR, serotonin receptor is activated [106]). In b-arrestin GRK5, it triggers a unique sequence of events in which the
2 knockout mice, these effects are not initiated by seroto- G protein-independent signaling of 5-HT4 receptor to c-
nin, but another hallucinogenic ligand 2,5-dimethoxy-4- Src–ERK1/2 is inhibited [111]. A tyrosine (Y54) residue
iodoamphetamine induces ERK1/2, receptor trafficking found only in the b-arrestin 1 isoform is phosphorylated by
and head-twitch response in the absence of b-arrestin 2. c-Src and this inhibits interaction with the m subunit of
This provides an example of an interesting scenario in adaptin 2, thus lowering its ability to augment b2AR
which a downstream pathway of receptor activity and its internalization [113]. b-Arrestin 2 has a natural substitu-
conformation is ligand-specific. tion at this residue (phenylalanine 54) and has a more
A recent study reports a completely novel b-arrestin- productive interaction with AP2 than b-arrestin 1.
dependent trafficking and signaling effect in which docosa- The exact mechanism of dephosphorylation of b-arrest-
hexaenoic acid and eicosapentaenoic acid, the major natural ins remains to be determined; however, the protein phos-
v-3 fatty acid constituents of dietary supplements such as phatase PP2A has been shown to interact with b-arrestin 2
fish oil, cause anti-inflammatory effects by stimulating the upon dopamine D2R as well as AT1aR activation

527
(Figure_3)TD$IG][ Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

PP2A
(a) N C
P ERK1/2, GRK5
CKII

E3 Ligase(s) U U
U U U

U
U
(b) N C

U
U U U
U
DUB(s)

eNOS
N C S-NO
(c)
De-Nitrosylation

extended conformation
(N and C domains move apart)
(d) Conformational
N C changes
closed conformation
(N and C domains move together)

AT1 aR
V2R

Endocytic adaptor
PPP PP
P
(e) N C
C
la
AP

th
2

rin

GRK2
GRK 3

AT1 aR
V2R Signaling adaptor
MAPK

N C
MAPKK
MAPKKK

P PP P

GRK5
GRK6
TRENDS in Pharmacological Sciences

Figure 3. Schematics showing molecular changes in b-arrestin. (a) b-Arrestins are phosphorylated at seryl or threonyl residues in the C terminal domain (see text for
details) and become dephosphorylated upon 7TMR activation. Dephosphorylated b-arrestins interact more efficiently with clathrin than phosphorylated forms. Upon
receptor internalization, b-arrestins become rephosphorylated. ERK1/2: extracellular signal regulated kinase, GRK: G-protein-coupled receptor kinase, CKII: casein kinase II,
PP2A: protein phosphatase 2A. (b) 7TMR activation leads to ubiquitination of b-arrestin 2, the site of ubiquitination is unique to a particular 7TMR–b-arrestin pair, thus
leading to differential modifications or differential display of ubiquitinated b-arrestins. (c) The C terminal cysteine residue in b-arrestin 2 is nitrosylated and this modification
promotes its interaction with clathrin. (d) Upon binding to 7TMRs, the b-arrestin molecule undergoes conformational rearrangement, which could either push the amino (N)
and carboxyl (C) domains apart or pull them together. The conformational changes have been assessed by various biochemical methods (limited proteolysis of purified b-
arrestins, resonance energy transfer between fluorophores attached to the two domains, etc.). (e) When b-arrestin 2 is recruited to the AT1aR or V2R that are phosphorylated
by GRK2 or GRK3 isoforms, it is able to function as an endocytic adaptor, but not as a signaling adaptor. By contrast, when b-arrestin 2 binds to the same receptors that are
phosphorylated by GRK5 or GRK6 isoforms, it functions as a signaling adaptor leading to robust activation of MAP kinases ERK1 and ERK2. It is suggested that the different
GRK isoforms set respective phosphorylation ‘barcodes’ to dictate the downstream activity of b-arrestin.

[25,114,115], and the PP2A inhibitor okadaic acid thus promoting their cointernalization and colocalization
increases b-arrestin serine 412 phosphorylation resulting on endosomes [47]. Comparison of two modified b-arrest-
from insulin treatment [116]. Phosphorylation of b-arrest- ins, one which lacks all ubiquitin acceptor sites and the
ins appears to be a mechanism of feedback regulation for other in which stable ubiquitination is conferred by fusing
specific protein interactions during endocytosis in general ubiquitin at the carboxyl terminus, have provided insights
and can regulate signaling at 7TMRs. as to how b-arrestin ubiquitin might regulate both b2AR
endocytosis and ERK1/2 signaling [48]. The stably ubiqui-
Ubiquitination of b-arrestins tinated b-arrestin 2–Ub fusion protein bound the activated
As discussed above, Mdm2-dependent ubiquitination of b- b2AR, clathrin and RFP–ERK2 with higher affinity than
arrestin 2 is crucial for rapid agonist-promoted internali- wild-type b-arrestin and formed stable signalosomes,
zation of the b2AR. b-Arrestin ubiquitination is also im- whereas the nonubiquitinated b-arrestin showed only
portant for forming a high-affinity complex with the b2AR weak or very transient binding with these partners [48].
[33,48]. Overexpression of Mdm2 augments b-arrestin Ubiquitination is also targeted to different domains of b-
ubiquitination and stabilizes b-arrestin–b2AR binding, arrestin 2 depending on the 7TMR that is being activated

528
Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

[35], thus providing a means of generating an ensemble of Consistent with this structural information, conforma-
ubiquitinated b-arrestins, differentially adorned with Ub tional changes have been demonstrated for b-arrestins 1
moieties at distinct sites (Figure 3b). Differential ubiqui- and 2 in the presence of a phosphopeptide that mimics the
tination could be a consequence of distinct conformational GRK-phosphorylated carboxyl tail of the V2R [122,123].
changes induced in b-arrestins when they bind to specific Addition of the phosphopeptide leads to the exposure of a
7TMRs. It might also form the molecular basis by which b- buried tryptic cleavage site (arginine 393 in b-arrestin 1
arrestins bind a variety of protein partners in distinct and arginine 394 in b-arrestin 2) as well as to the release of
subcellular compartments, leading to distinct trafficking buried carboxyl terminus of b-arrestin, which contains the
and signaling pathways. previously mapped sites for clathrin interaction [124]. In
The roles of b-arrestin ubiquitination in receptor bind- this ‘activated’ conformation induced by the phosphopep-
ing, endocytosis and signaling are further illuminated by tide, b-arrestin binding to clathrin is much more robust
the discovery that it can be reversed by a specific deubi- than in the presence of a nonphosphorylated peptide.
quitinating enzyme, USP33 [47]. Upon b2AR activation, Recently, conformational changes in b-arrestins have
the recruited b-arrestins are rapidly ubiquitinated and been demonstrated by monitoring intramolecular biolumi-
deubiquitinated and this is facilitated by a high-affinity nescence resonance energy transfer (BRET) between a
interaction between b-arrestin 2 and USP33. This interac- donor enzyme, luciferase, which is fused at the amino
tion leads to deubiquitination of b-arrestin and its dissoci- terminus of b-arrestin 2 and an acceptor fluorophore,
ation from internalizing b2AR, thus promoting only yellow fluorescent protein appended to the carboxyl termi-
transient binding with activated ERK1/2. Indeed, deple- nus of the same b-arrestin molecule. Any intramolecular
tion of USP33 not only stabilizes b-arrestin ubiquitination reorientation of b-arrestin causes a change in the amount
but also promotes its stable interaction with the internal- of BRET, reflective of a conformational change in b-
ized b2AR on endosomes; it also increases the magnitude arrestin [125]. Recent studies with the above BRET bio-
and duration of b-arrestin-dependent ERK1/2 activity [47]. sensor have indicated a net decrease in BRET elicited by b-
By contrast, depletion of the E3 ubiquitin ligase Mdm2 arrestin-biased mutant receptors or b-arrestin-biased
ablates both b-arrestin ubiquitination and b-arrestin-de- ligands and an increase in BRET by the unbiased ligands
pendent ERK1/2 activity [47]. Thus, the kinetics of b- or receptors [126]. These conformational changes probably
arrestin ubiquitination and deubiquitination are tightly stabilize b-arrestins, in particular orientations that expose
regulated by these cellular enzymes, which ensures the or bury domains important in partner protein interactions.
appropriate duration and magnitude of b-arrestin-biased Because each receptor–b-arrestin pair can be bound in a
signaling. particular manner, the conformational changes induced
could be unique for a particular 7TMR–b-arrestin pair.
S-Nitrosylation of b-arrestin 2 Specific conformations of b-arrestins induced by different
b-Arrestin 2 interacts with endothelial NO synthase 7TMR binding can also determine the kinetics and stabili-
(eNOS), an enzyme that releases NO in blood vessels. b- ty of interaction of b-arrestin with the deubiquitinating
Arrestin 2 is S-nitrosylated at a single cysteine (C terminal enzyme USP33 [47].
residue 410) by eNOS and S-nitrosylation of b-arrestin 2 is
promoted by endogenous S-nitrosoglutathione [117]. Inter- Phosphorylation codes set by GRKs
estingly, S-nitrosylation of cysteine 410 enhances the in- GRK-mediated phosphorylation has been considered as a
teraction of b-arrestin 2 with clathrin and b-adaptin, in generic and global process that affects all available phos-
striking contrast to the inhibitory effect of S-nitrosylation phorylation sites on the receptors, leading to b-arrestin
on the interaction between b-arrestin 2 and eNOS [117]. binding. However, recent analyses of GRK functions utiliz-
Although S-nitrosylation of b-arrestin 2 was reported to ing siRNA-mediated knockdown of individual GRKs have
increase b2AR internalization, its effect on b-arrestin-de- suggested that receptor phosphorylation is a more specific
pendent signaling is not currently known (Figure 3c). and targeted process. Moreover, the downstream effects of
receptor phosphorylation by a particular GRK isoform are
Conformational changes in b-arrestin specialized and distinct. For example, if the AT1aR or V2R is
b-Arrestins 1 and 2 interact with numerous receptors and phosphorylated by GRKs 2 or 3, b-arrestin-dependent
nonreceptor proteins and many of these interactions are ERK1/2 signaling is diminished, whereas receptor endocy-
regulated by 7TMR stimulation. It is probable that the tosis and the magnitude of b-arrestin recruitment remain
basal conformation of free form of b-arrestins is changed to the same [127,128]. Furthermore, phosphorylation by GRKs
an ‘active’ conformation upon recruitment to agonist-occu- 5 and 6 does not promote robust internalization and b-
pied 7TMRs (Figure 3d) [8,95,118]. The X-ray structures of arrestin-binding, but is required for b-arrestin-dependent
bovine visual arrestin and b-arrestin 1 in the basal inactive ERK1/2 activation [127,128]. This reciprocal regulation of b-
state indicate that arrestin is an elongated molecule with arrestin-dependent processes by GRK-mediated receptor
two domains (N and C domains), connected through a 12- phosphorylation suggests that specific GRKs target specific
residue linker region [119,120]. A notable feature is that of phosphorylation sites on 7TMRs and that the b-arrestin
a hydrogen-bonded polar core, embedded between the N conformation recruited to such differentially modified recep-
and C domains, which is disrupted by the phosphate tors could be different leading to distinct functional outputs.
moieties on the activated receptors, resulting in a confor- In this framework, the differential patterns of receptor
mational rearrangement within the b-arrestin molecule phosphorylation establish a ‘barcode’ for b-arrestin recruit-
[121]. ment and activity (Figure 3e) [127,128].

529
Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

Although the exact sites of phosphorylation targeted by arrestin is a cause or consequence of the conformational
each GRK are yet to be determined, several other studies changes of b-arrestin is a debatable issue. Perhaps the
support the barcode hypothesis. GRKs 2 and 3 target conformational changes in b-arrestin lead to distinct ubi-
phosphosites within the third intracellular loop of the quitin modifications of b-arrestins by specific E3 ligases,
D2 dopamine receptor, and this is important for postendo- which could provide unique protein interaction surfaces
cytic recycling of the receptor [129]. Phosphorylation of a based on the nature and length of polyubiquitin chain
subset of serines in the carboxyl tail of the follicle-stimu- modifications on b-arrestin. By contrast, the covalent at-
lating hormone receptor is required for receptor desensiti- tachment of ubiquitin or ubiquitin chains could further
zation, but not for b-arrestin-dependent ERK1/2 signaling shift domains within b-arrestins causing changes in over-
[130]. Mutagenesis studies of distinct sets of serines and all conformation. Nonetheless, these molecular features
threonines on the b2AR carboxyl tail showed striking confer utmost specificity in space and time to b-arrestin-
differences with regard to b-arrestin recruitment and re- dependent trafficking and signaling mechanisms.
ceptor internalization [131]. In the somatin subtype 2A The discovery of b-arrestin-biased ligands has opened
receptor, internalization and desensitization involves dif- up a new approach for treating various diseases, which
ferent phosphorylation sites [132]. In the thyrotropin-re- might lead to the formulation of drugs with higher
leasing hormone receptor, the proximal (but not distal) efficacy, specificity and less side effects than the 7TMR-
phosphosites on the C tail evoke b-arrestin-dependent targeted medications currently available. Another devel-
internalization and desensitization after its recruitment oping idea is that the ‘active’ conformational states of b-
[39]. For the chemokine receptor CCR7, two endogenous arrestins are favored by distinct patterns of phosphory-
agonists lead to phosphorylation of the receptor by differ- lation in 7TMRs (the barcode hypothesis), which enables
ent GRK isoforms: CCL19 leads to GRK3- and 6-mediated transformation of recruited b-arrestins into different
phosphorylation. By contrast, CCL21 engages only GRK6, forms of active b-arrestins. This facilitates regulation
leading to differential consequences, namely robust b- of a wide variety of cellular responses, including endocytic
arrestin binding and desensitization only by CCL19, but trafficking, signaling in various cellular compartments,
b-arrestin-dependent ERK1/2 activation by both agonists gene transcription, protein translation and processes
[133]. Using tandem mass spectrometry, phosphosite-spe- such as chemotaxis and apoptosis. The analogy between
cific antibodies and selective knockdown of GRK isoforms, b-arrestins and related arrestin domain-containing adap-
distinct phosphorylation sites, were mapped on the CXCR4 tors in intracellular trafficking suggests that the endocy-
[134]. This study also revealed that distinct sites of phos- tic adaptor role of b-arrestins might be evolutionarily
phorylation differentially regulated b-arrestin recruitment conserved. However, the mammalian b-arrestins have
and ERK1/2 activation. Thus, the downstream effects of b- acquired additional specialization with regard to the
arrestin binding to the activated receptor could involve regulation of the superfamily of 7TMRs that make them
reading a barcode preset by distinct GRK-mediated phos- particularly well suited for targeting specific pathways in
phorylation events. Furthermore, GRK-mediated regula- drug development.
tion could be differentially effective based on the tissue-
specific expression of each GRK isoform, thus providing yet References
another level of specificity for b-arrestin-dependent inter- 1 Lefkowitz, R.J. (2004) Historical review: a brief history and personal
retrospective of seven-transmembrane receptors. Trends Pharmacol.
nalization and signaling.
Sci. 25, 413–422
2 Yamaki, K. et al. (1987) Molecular cloning of the S-antigen cDNA from
Concluding remarks bovine retina. Biochem. Biophys. Res. Commun. 142, 904–910
Over the past two decades our understanding about and 3 Lohse, M.J. et al. (1990) beta-Arrestin: a protein that regulates beta-
appreciation of the multifaceted functions of b-arrestins adrenergic receptor function. Science 248, 1547–1550
4 Attramadal, H. et al. (1992) beta-Arrestin2, a novel member of the
have greatly evolved. Nonetheless, many aspects of the arrestin/beta-arrestin gene family. J. Biol. Chem. 267, 17882–17890
functional roles of b-arrestins remain poorly understood. 5 Smith, W.C. et al. (2000) Cloning and functional characterization of
How does b-arrestin bind the growing ensemble of inter- salamander rod and cone arrestins. Invest. Ophthalmol. Vis. Sci. 41,
actors? How are these novel interactions choreographed 2445–2455
during trafficking? How are the additional interactions/ 6 Kuhn, H. and Wilden, U. (1987) Deactivation of photoactivated
rhodopsin by rhodopsin-kinase and arrestin. J. Recept. Res. 7, 283–298
functions favored over its traditional roles in receptor 7 Benovic, J.L. et al. (1987) Functional desensitization of the isolated
binding? These are some outstanding questions that need beta-adrenergic receptor by the beta-adrenergic receptor kinase:
to be addressed from a molecular and structural stand- potential role of an analog of the retinal protein arrestin (48-kDa
point in the future. protein). Proc. Natl. Acad. Sci. U.S.A. 84, 8879–8882
8 Lefkowitz, R.J. and Shenoy, S.K. (2005) Transduction of receptor
Even though mapping of binding domains of several
signals by beta-arrestins. Science 308, 512–517
partners has been shown to involve distinct structural 9 Wise, A. et al. (2002) Target validation of G-protein coupled receptors.
regions in b-arrestin, simultaneous binding of multiple Drug Discov. Today 7, 235–246
partners is not likely. It is more probable that different 10 Neves, S.R. et al. (2002) G protein pathways. Science 296, 1636–1639
b-arrestin molecules engage in different interactions and 11 Perry, S.J. et al. (2002) Targeting of cyclic AMP degradation to beta 2-
adrenergic receptors by beta-arrestins. Science 298, 834–836
these can occur simultaneously in different subcellular
12 Nelson, C.D. et al. (2007) Targeting of diacylglycerol degradation to
compartments. Furthermore, many b-arrestin–partner M1 muscarinic receptors by beta-arrestins. Science 315, 663–666
interactions could be dynamic in nature, precisely timed 13 Brown, M.S. et al. (1979) Receptor-mediated uptake of lipoprotein-
and regulated by various molecular changes in b-arrestins cholesterol and its utilization for steroid synthesis in the adrenal
such as ubiquitination. Whether ubiquitination of b- cortex. Recent Prog. Horm. Res. 35, 215–257

530
Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

14 Drake, M.T. et al. (2006) Trafficking of G protein-coupled receptors. 38 Milasta, S. et al. (2005) The sustainability of interactions between the
Circ. Res. 99, 570–582 orexin-1 receptor and beta-arrestin-2 is defined by a single C-terminal
15 Moore, C.A. et al. (2007) Regulation of receptor trafficking by GRKs cluster of hydroxy amino acids and modulates the kinetics of ERK
and arrestins. Annu. Rev. Physiol. 69, 451–482 MAPK regulation. Biochem. J. 387, 573–584
16 Zhang, J. et al. (1996) Dynamin and beta-arrestin reveal distinct 39 Jones, B.W. and Hinkle, P.M. (2008) Arrestin binds to different
mechanisms for G protein-coupled receptor internalization. J. Biol. phosphorylated regions of the thyrotropin-releasing hormone
Chem. 271, 18302–18305 receptor with distinct functional consequences. Mol. Pharmacol. 74,
17 Goodman, O.B., Jr et al. (1996) beta-Arrestin acts as a clathrin adaptor 195–202
in endocytosis of the beta2-adrenergic receptor. Nature 383, 447–450 40 Marion, S. et al. (2006) A beta-arrestin binding determinant common
18 Ferguson, S.S. et al. (1996) Role of beta-arrestin in mediating agonist- to the second intracellular loops of rhodopsin family G protein-coupled
promoted G protein-coupled receptor internalization. Science 271, receptors. J. Biol. Chem. 281, 2932–2938
363–366 41 Nakamura, K. et al. (2000) Seven non-contiguous intracellular
19 Kohout, T.A. et al. (2001) beta-Arrestin 1 and 2 differentially regulate residues of the lutropin/choriogonadotropin receptor dictate the
heptahelical receptor signaling and trafficking. Proc. Natl. Acad. Sci. rate of agonist-induced internalization and its sensitivity to non-
U.S.A. 98, 1601–1606 visual arrestins. J. Biol. Chem. 275, 241–247
20 Ahn, S. et al. (2003) Desensitization, internalization, and signaling 42 Kim, O.J. et al. (2004) The role of phosphorylation in D1 dopamine
functions of beta-arrestins demonstrated by RNA interference. Proc. receptor desensitization: evidence for a novel mechanism of arrestin
Natl. Acad. Sci. U.S.A. 100, 1740–1744 association. J. Biol. Chem. 279, 7999–8010
21 Kern, R.C. et al. (2009) Arrestin2/clathrin interaction is regulated by 43 Ouedraogo, M. et al. (2008) Distinct motifs of neuropeptide Y receptors
key N- and C-terminal regions in arrestin2. Biochemistry 48, 7190–7200 differentially regulate trafficking and desensitization. Traffic 9,
22 Kang, D.S. et al. (2009) Structure of an arrestin2-clathrin complex 305–324
reveals a novel clathrin binding domain that modulates receptor 44 Lan, H. et al. (2009) An intracellular loop 2 amino acid residue
trafficking. J. Biol. Chem. 284, 29860–29872 determines differential binding of arrestin to the dopamine D2 and
23 Lefkowitz, R.J. et al. (2006) New roles for beta-arrestins in cell D3 receptors. Mol. Pharmacol. 75, 19–26
signaling: not just for seven-transmembrane receptors. Mol. Cell 45 Tobin, A.B. et al. (2008) Location, location, location. . .site-specific
24, 643–652 GPCR phosphorylation offers a mechanism for cell-type-specific
24 Nelson, C.D. et al. (2008) Beta-arrestin scaffolding of signalling. Trends Pharmacol. Sci. 29, 413–420
phosphatidylinositol 4-phosphate 5-kinase Ialpha promotes agonist- 46 Shenoy, S.K. et al. (2001) Regulation of receptor fate by ubiquitination
stimulated sequestration of the beta2-adrenergic receptor. J. Biol. of activated beta 2-adrenergic receptor and beta-arrestin. Science 294,
Chem. 283, 21093–21101 1307–1313
25 Xiao, K. et al. (2007) Functional specialization of beta-arrestin 47 Shenoy, S.K. et al. (2009) Beta-arrestin-dependent signaling and
interactions revealed by proteomic analysis. Proc. Natl. Acad. Sci. trafficking of 7-transmembrane receptors is reciprocally regulated
U.S.A. 104, 12011–12016 by the deubiquitinase USP33 and the E3 ligase Mdm2. Proc. Natl.
26 Laporte, S.A. et al. (2000) The interaction of beta-arrestin with the Acad. Sci. U.S.A. 106, 6650–6655
AP-2 adaptor is required for the clustering of beta 2-adrenergic 48 Shenoy, S.K. et al. (2007) Ubiquitination of beta-arrestin links seven-
receptor into clathrin-coated pits. J. Biol. Chem. 275, 23120–23126 transmembrane receptor endocytosis and ERK activation. J. Biol.
27 Mishra, S.K. et al. (2005) Functional dissection of an AP-2 beta2 Chem. 282, 29549–29562
appendage-binding sequence within the autosomal recessive 49 Hicke, L. et al. (2005) Ubiquitin-binding domains. Nat. Rev. Mol. Cell
hypercholesterolemia protein. J. Biol. Chem. 280, 19270–19280 Biol. 6, 610–621
28 Edeling, M.A. et al. (2006) Molecular switches involving the AP-2 50 Penalva, M.A. et al. (2008) Ambient pH gene regulation in fungi:
beta2 appendage regulate endocytic cargo selection and clathrin coat making connections. Trends Microbiol. 16, 291–300
assembly. Dev. Cell 10, 329–342 51 Johnson, E.S. (2004) Protein modification by SUMO. Annu. Rev.
29 Oakley, R.H. et al. (2002) The cellular distribution of fluorescently Biochem. 73, 355–382
labeled arrestins provides a robust, sensitive, and universal assay for 52 Muller, S. et al. (2001) SUMO, ubiquitin’s mysterious cousin. Nat. Rev.
screening G protein-coupled receptors. Assay Drug Dev. Technol. 1, Mol. Cell Biol. 2, 202–210
21–30 53 Wyatt, D. et al. (2011) Small ubiquitin-like modifier modification
30 DeWire, S.M. et al. (2007) beta-Arrestins and cell signaling. Annu. of arrestin-3 regulates receptor trafficking. J. Biol. Chem. 286,
Rev. Physiol. 69, 483–510 3884–3893
31 Oakley, R.H. et al. (1999) Association of beta-arrestin with G protein- 54 Schulte, G. et al. (2010) beta-Arrestins – scaffolds and signalling
coupled receptors during clathrin-mediated endocytosis dictates the elements essential for WNT/Frizzled signalling pathways? Br. J.
profile of receptor resensitization. J. Biol. Chem. 274, 32248–32257 Pharmacol. 159, 1051–1058
32 Oakley, R.H. et al. (2000) Differential affinities of visual arrestin, beta 55 Chen, W. et al. (2003) Dishevelled 2 recruits beta-arrestin 2 to
arrestin1, and beta arrestin2 for G protein-coupled receptors delineate mediate Wnt5A-stimulated endocytosis of Frizzled 4. Science 301,
two major classes of receptors. J. Biol. Chem. 275, 17201–17210 1391–1394
33 Shenoy, S.K. and Lefkowitz, R.J. (2003) Trafficking patterns of beta- 56 Kim, G.H. and Han, J.K. (2007) Essential role for beta-arrestin 2 in
arrestin and G protein-coupled receptors determined by the kinetics of the regulation of Xenopus convergent extension movements. EMBO J.
beta-arrestin deubiquitination. J. Biol. Chem. 278, 14498–14506 26, 2513–2526
34 Oakley, R.H. et al. (2001) Molecular determinants underlying the 57 Chen, W. et al. (2004) Activity-dependent internalization of
formation of stable intracellular G protein-coupled receptor-beta- smoothened mediated by beta-arrestin 2 and GRK2. Science 306,
arrestin complexes after receptor endocytosis. J. Biol. Chem. 276, 2257–2260
19452–19460 58 Kovacs, J.J. et al. (2008) beta-Arrestin-mediated localization of
35 Shenoy, S.K. and Lefkowitz, R.J. (2005) Receptor-specific smoothened to the primary cilium. Science 320, 1777–1781
ubiquitination of beta-arrestin directs assembly and targeting of 59 Molla-Herman, A. et al. (2008) Targeting of beta-arrestin2 to the
seven-transmembrane receptor signalosomes. J. Biol. Chem. 280, centrosome and primary cilium: role in cell proliferation control.
15315–15324 PLoS ONE 3, e3728
36 Proulx, C.D. et al. (2005) Involvement of a cytoplasmic-tail serine 60 Wilbanks, A.M. et al. (2004) beta-Arrestin 2 regulates zebrafish
cluster in urotensin II receptor internalization. Biochem. J. 385, development through the hedgehog signaling pathway. Science 306,
115–123 2264–2267
37 Neuschafer-Rube, F. et al. (2004) Identification of a Ser/Thr cluster in 61 Meloni, A.R. et al. (2006) Smoothened signal transduction is promoted
the C-terminal domain of the human prostaglandin receptor EP4 that by G protein-coupled receptor kinase 2. Mol. Cell. Biol. 26, 7550–7560
is essential for agonist-induced beta-arrestin1 recruitment but differs 62 Philipp, M. et al. (2008) Smoothened signaling in vertebrates is
from the apparent principal phosphorylation site. Biochem. J. 379, facilitated by a G protein-coupled receptor kinase. Mol. Biol. Cell
573–585 19, 5478–5489

531
Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

63 Lin, F.T. et al. (1998) beta-Arrestins regulate mitogenic signaling and 88 Berthouze, M. et al. (2009) The deubiquitinases USP33 and USP20
clathrin-mediated endocytosis of the insulin-like growth factor I coordinate beta2 adrenergic receptor recycling and resensitization.
receptor. J. Biol. Chem. 273, 31640–31643 EMBO J. 28, 1684–1696
64 Wu, J.H. et al. (2003) The adaptor protein beta-arrestin2 enhances 89 Murphy, J.E. et al. (2009) Endosomes: a legitimate platform for the
endocytosis of the low density lipoprotein receptor. J. Biol. Chem. 278, signaling train. Proc. Natl. Acad. Sci. U.S.A. 106, 17615–17622
44238–44245 90 Aubry, L. et al. (2009) The arrestin fold: variations on a theme. Curr.
65 Quack, I. et al. (2006) beta-Arrestin2 mediates nephrin endocytosis Genomics 10, 133–142
and impairs slit diaphragm integrity. Proc. Natl. Acad. Sci. U.S.A. 91 Nikko, E. et al. (2008) Arrestin-like proteins mediate ubiquitination
103, 14110–14115 and endocytosis of the yeast metal transporter Smf1. EMBO Rep. 9,
66 Patrakka, J. and Tryggvason, K. (2007) Nephrin – a unique structural 1216–1221
and signaling protein of the kidney filter. Trends Mol. Med. 13, 92 Lin, C.H. et al. (2008) Arrestin-related ubiquitin-ligase adaptors
396–403 regulate endocytosis and protein turnover at the cell surface. Cell
67 Chen, W. et al. (2003) beta-Arrestin 2 mediates endocytosis of type III 135, 714–725
TGF-beta receptor and down-regulation of its signaling. Science 301, 93 Collins, B.M. et al. (2008) Structure of Vps26B and mapping of its
1394–1397 interaction with the retromer protein complex. Traffic 9, 366–379
68 Simonin, A. and Fuster, D. (2010) Nedd4-1 and beta-arrestin-1 are 94 Shi, H. et al. (2006) The retromer subunit Vps26 has an arrestin fold
key regulators of Na+/H+ exchanger 1 ubiquitylation, endocytosis, and binds Vps35 through its C-terminal domain. Nat. Struct. Mol.
and function. J. Biol. Chem. 285, 38293–38303 Biol. 13, 540–548
69 Szabo, E.Z. et al. (2005) beta-Arrestins bind and decrease cell-surface 95 Gurevich, V.V. and Gurevich, E.V. (2004) The molecular acrobatics of
abundance of the Na+/H+ exchanger NHE5 isoform. Proc. Natl. Acad. arrestin activation. Trends Pharmacol. Sci. 25, 105–111
Sci. U.S.A. 102, 2790–2795 96 Miller, W.E. and Lefkowitz, R.J. (2001) Expanding roles for beta-
70 Dasgupta, P. et al. (2006) Nicotine induces cell proliferation by beta- arrestins as scaffolds and adapters in GPCR signaling and trafficking.
arrestin-mediated activation of Src and Rb-Raf-1 pathways. J. Clin. Curr. Opin. Cell Biol. 13, 139–145
Invest. 116, 2208–2217 97 DeFea, K.A. et al. (2000) beta-Arrestin-dependent endocytosis of
71 Lipsky, R. et al. (2008) beta-Adrenergic receptor activation induces proteinase-activated receptor 2 is required for intracellular
internalization of cardiac Cav1.2 channel complexes through a beta- targeting of activated ERK1/2. J. Cell Biol. 148, 1267–1281
Arrestin 1-mediated pathway. J. Biol. Chem. 283, 17221–17226 98 Luttrell, L.M. et al. (2001) Activation and targeting of extracellular
72 Shukla, A.K. et al. (2010) Arresting a transient receptor potential signal-regulated kinases by beta-arrestin scaffolds. Proc. Natl. Acad.
(TRP) channel: beta-arrestin 1 mediates ubiquitination and Sci. U.S.A. 98, 2449–2454
functional down-regulation of TRPV4. J. Biol. Chem. 285, 30115– 99 DeWire, S.M. et al. (2008) beta-Arrestin-mediated signaling regulates
30125 protein synthesis. J. Biol. Chem. 283, 10611–10620
73 Mukhopadhyay, D. and Riezman, H. (2007) Proteasome-independent 100 DeFea, K.A. (2011) beta-Arrestins as regulators of signal termination
functions of ubiquitin in endocytosis and signaling. Science 315, and transduction: how do they determine what to scaffold? Cell
201–205 Signal. 23, 621–629
74 Shenoy, S.K. (2007) Seven-transmembrane receptors and 101 Gurevich, V.V. et al. (1997) Agonist-receptor-arrestin, an alternative
ubiquitination. Circ. Res. 100, 1142–1154 ternary complex with high agonist affinity. J. Biol. Chem. 272,
75 Hershko, A. and Ciechanover, A. (1998) The ubiquitin system. Annu. 28849–28852
Rev. Biochem. 67, 425–479 102 Violin, J.D. and Lefkowitz, R.J. (2007) beta-Arrestin-biased ligands at
76 Marchese, A. and Benovic, J.L. (2001) Agonist-promoted seven-transmembrane receptors. Trends Pharmacol. Sci. 28, 416–422
ubiquitination of the G protein-coupled receptor CXCR4 mediates 103 Kenakin, T. (2007) Collateral efficacy in drug discovery: taking
lysosomal sorting. J. Biol. Chem. 276, 45509–45512 advantage of the good (allosteric) nature of 7TM receptors. Trends
77 Hicke, L. and Dunn, R. (2003) Regulation of membrane protein Pharmacol. Sci. 28, 407–415
transport by ubiquitin and ubiquitin-binding proteins. Annu. Rev. 104 Rajagopal, S. et al. (2010) Teaching old receptors new tricks: biasing
Cell Dev. Biol. 19, 141–172 seven-transmembrane receptors. Nat. Rev. Drug Discov. 9, 373–386
78 Marchese, A. et al. (2008) G protein-coupled receptor sorting to 105 Whalen, E.J. et al. (2010) Therapeutic potential of beta-arrestin- and
endosomes and lysosomes. Annu. Rev. Pharmacol. Toxicol. 48, G protein-biased agonists. Trends Mol. Med. 17, 126–139
601–629 106 Schmid, C.L. et al. (2008) Agonist-directed signaling of the serotonin
79 Hislop, J.N. and von Zastrow, M. (2010) Role of ubiquitination in 2A receptor depends on beta-arrestin-2 interactions in vivo.
endocytic trafficking of G-protein-coupled receptors. Traffic 12, Proc. Natl. Acad. Sci. U.S.A. 105, 1079–1084
137–148 107 Oh, D.Y. et al. (2010) GPR120 is an omega-3 fatty acid receptor
80 Shenoy, S.K. et al. (2008) Nedd4 mediates agonist-dependent mediating potent anti-inflammatory and insulin-sensitizing effects.
ubiquitination, lysosomal targeting, and degradation of the beta2- Cell 142, 687–698
adrenergic receptor. J. Biol. Chem. 283, 22166–22176 108 Lin, F.T. et al. (1997) Clathrin-mediated endocytosis of the beta-
81 Girnita, L. et al. (2005) {beta}-Arrestin is crucial for ubiquitination adrenergic receptor is regulated by phosphorylation/
and down-regulation of the insulin-like growth factor-1 receptor by dephosphorylation of beta-arrestin1. J. Biol. Chem. 272,
acting as adaptor for the MDM2 E3 ligase. J. Biol. Chem. 280, 31051–31057
24412–24419 109 Lin, F.T. et al. (1999) Feedback regulation of beta-arrestin1 function
82 Lakshmikanthan, V. et al. (2009) Identification of betaArrestin2 as a by extracellular signal-regulated kinases. J. Biol. Chem. 274, 15971–
corepressor of androgen receptor signaling in prostate cancer. Proc. 15974
Natl. Acad. Sci. U.S.A. 106, 9379–9384 110 Lin, F.T. et al. (2002) Phosphorylation of beta-arrestin2 regulates its
83 Li, X. et al. (2009) Mdm2 directs the ubiquitination of beta-arrestin- function in internalization of beta(2)-adrenergic receptors.
sequestered cAMP phosphodiesterase-4D5. J. Biol. Chem. 284, Biochemistry 41, 10692–10699
16170–16182 111 Barthet, G. et al. (2009) Beta-arrestin1 phosphorylation by GRK5
84 Salcedo, A. et al. (2006) Mdm2 is involved in the ubiquitination and regulates G protein-independent 5-HT4 receptor signalling. EMBO J.
degradation of G-protein-coupled receptor kinase 2. EMBO J. 25, 28, 2706–2718
4752–4762 112 Kim, Y.M. et al. (2002) Regulation of arrestin-3 phosphorylation by
85 Mukherjee, A. et al. (2005) Regulation of Notch signalling by non- casein kinase II. J. Biol. Chem. 277, 16837–16846
visual beta-arrestin. Nat. Cell Biol. 7, 1191–1201 113 Marion, S. et al. (2007) N-terminal tyrosine modulation of the
86 Saksena, S. et al. (2007) ESCRTing proteins in the endocytic pathway. endocytic adaptor function of the beta-arrestins. J. Biol. Chem.
Trends Biochem. Sci. 32, 561–573 282, 18937–18944
87 Malik, R. and Marchese, A. (2010) Arrestin-2 interacts with the 114 Beaulieu, J.M. et al. (2005) An Akt/beta-arrestin 2/PP2A signaling
endosomal sorting complex required for transport machinery to complex mediates dopaminergic neurotransmission and behavior.
modulate endosomal sorting of CXCR4. Mol. Biol. Cell 21, 2529–2541 Cell 122, 261–273

532
Review Trends in Pharmacological Sciences September 2011, Vol. 32, No. 9

115 Xiao, K. et al. (2010) Global phosphorylation analysis of beta-arrestin- 126 Shukla, A.K. et al. (2008) Distinct conformational changes in beta-
mediated signaling downstream of a seven transmembrane receptor arrestin report biased agonism at seven-transmembrane receptors.
(7TMR). Proc. Natl. Acad. Sci. U.S.A. 107, 15299–15304 Proc. Natl. Acad. Sci. U.S.A. 105, 9988–9993
116 Hupfeld, C.J. et al. (2005) Insulin-induced beta-arrestin1 Ser-412 127 Kim, J. et al. (2005) Functional antagonism of different G protein-
phosphorylation is a mechanism for desensitization of ERK coupled receptor kinases for beta-arrestin-mediated angiotensin II
activation by Gai-coupled receptors. J. Biol. Chem. 280, 1016–1023 receptor signaling. Proc. Natl. Acad. Sci. U.S.A. 102, 1442–1447
117 Ozawa, K. et al. (2008) S-Nitrosylation of beta-arrestin regulates beta- 128 Ren, X.R. et al. (2005) Different G protein-coupled receptor kinases
adrenergic receptor trafficking. Mol. Cell 31, 395–405 govern G protein and beta-arrestin-mediated signaling of V2
118 Palczewski, K. et al. (1991) Phosphorylated rhodopsin and heparin vasopressin receptor. Proc. Natl. Acad. Sci. U.S.A. 102, 1448–1453
induce similar conformational changes in arrestin. J. Biol. Chem. 266, 129 Namkung, Y. et al. (2009) G protein-coupled receptor kinase-mediated
18649–18654 phosphorylation regulates post-endocytic trafficking of the D2
119 Hirsch, J.A. et al. (1999) The 2.8 A crystal structure of visual arrestin: dopamine receptor. J. Biol. Chem. 284, 15038–15051
a model for arrestin’s regulation. Cell 97, 257–269 130 Kara, E. et al. (2006) A phosphorylation cluster of five serine and
120 Han, M. et al. (2001) Crystal structure of beta-arrestin at 1.9 A: threonine residues in the C-terminus of the follicle-stimulating
possible mechanism of receptor binding and membrane hormone receptor is important for desensitization but not for beta-
translocation. Structure 9, 869–880 arrestin-mediated ERK activation. Mol. Endocrinol. 20, 3014–3026
121 Vishnivetskiy, S.A. et al. (2000) An additional phosphate-binding 131 Krasel, C. et al. (2008) Dual role of the beta2-adrenergic receptor C
element in arrestin molecule. Implications for the mechanism of terminus for the binding of beta-arrestin and receptor internalization.
arrestin activation. J. Biol. Chem. 275, 41049–41057 J. Biol. Chem. 283, 31840–31848
122 Xiao, K. et al. (2004) Activation-dependent conformational changes in 132 Liu, Q. et al. (2008) Distinct phosphorylation sites in the SST2A
{beta}-arrestin 2. J. Biol. Chem. 279, 55744–55753 somatostatin receptor control internalization, desensitization, and
123 Nobles, K.N. et al. (2007) The active conformation of beta-arrestin1: arrestin binding. Mol. Pharmacol. 73, 292–304
direct evidence for the phosphate sensor in the N-domain and 133 Zidar, D.A. et al. (2009) Selective engagement of G protein coupled
conformational differences in the active states of beta-arrestins1 receptor kinases (GRKs) encodes distinct functions of biased ligands.
and -2. J. Biol. Chem. 282, 21370–21381 Proc. Natl. Acad. Sci. U.S.A. 106, 9649–9654
124 Krupnick, J.G. et al. (1997) Arrestin/clathrin interaction. Localization 134 Busillo, J.M. et al. (2010) Site-specific phosphorylation of CXCR4 is
of the clathrin binding domain of nonvisual arrestins to the carboxy dynamically regulated by multiple kinases and results in differential
terminus. J. Biol. Chem. 272, 15011–15016 modulation of CXCR4 signaling. J. Biol. Chem. 285, 7805–7817
125 Charest, P.G. et al. (2005) Monitoring agonist-promoted 135 Ren, J. et al. (2009) Systematic study of protein sumoylation:
conformational changes of beta-arrestin in living cells by development of a site-specific predictor of SUMOsp 2.0. Proteomics
intramolecular BRET. EMBO Rep. 6, 334–340 9, 3409–3412

533

You might also like