You are on page 1of 7

192 Current Pharmaceutical Design, 2012, 18, 192-198

Beta-arrestin Biased Agonism/Antagonism at Cardiovascular Seven Transmem-


brane-spanning Receptors

Anastasios Lymperopoulos*

Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL 33328, USA

Abstract: Heptahelical, G protein-coupled or seven transmembrane-spanning receptors, such as the -adrenergic and the angiotensin II
type 1 receptors, are the most diverse and therapeutically important family of receptors in the human genome, playing major roles in the
physiology of various organs/tissues including the heart and blood vessels. Ligand binding activates heterotrimeric G proteins that trans-
mit intracellular signals by regulating effector enzymes or ion channels. G protein signaling is terminated, in large part, by phosphoryla-
tion of the agonist-bound receptor by the G-protein coupled receptor kinases (GRKs), followed by arrestin binding, which uncouples the
phosphorylated receptor from the G protein and subsequently targets the receptor for internalization. As the receptor-arrestin complex
enters the cell, arrestin-1 and -2, the two mammalian arrestin isoforms, serve as ligand-regulated scaffolds that recruit a host of intra-
cellular proteins and signal transducers, thus promoting their own wave of signal transduction independently of G-proteins. A constantly
increasing number of studies over the past several years have begun to uncover specific roles played by these ubiquitously expressed re-
ceptor adapter proteins in signal transduction of several important heptahelical receptors regulating the physiology of various or-
gans/systems, including the cardiovascular (CV) system. Thus, arrestin-dependent signaling has increasingly been implicated in CV
physiology and pathology, presenting several exciting opportunities for therapeutic intervention in the treatment of CV disorders. Addi-
tionally, the discovery of this novel mode of heptahelical receptor signaling via arrestins has prompted a revision of classical pharma-
cological concepts such as receptor agonism/antagonism, as well as introduction of new terms such as biased signaling, which refers to
ligand-specific activation of selective signal transduction pathways by the very same receptor. The present review gives an overview of
the current knowledge in the field of arrestin-dependent signaling, with a specific focus on CV heptahelical receptor arrestin-mediated
signaling and on biased CV heptahelical receptor ligands that promote or inhibit it. Exciting new possibilities for cardiovascular thera-
peutics arising from the delineation of this arrestin-dependent signaling are also discussed.

Keywords: Heptahelical receptor (7TMR); cardiovascular; arrestin-dependent signalling; -adrenergic receptor; angiotensin II type 1 recep-
tor; arrestin biased agonist or antagonist.

INTRODUCTION with a well-conserved, central catalytic domain (~270 aa), similar


The heptahelical or seven-transmembrane spanning receptors to that of other serine-threonine kinases, flanked by an amino-
(7TMRs) or G protein-coupled receptors (GPCRs) are the largest terminal (NT) domain (~ 185 aa) and a variable-length carboxyl-
and most diverse superfamily of cell surface receptors. Approxi- terminal (CT) domain (~ 105-230 aa) that contains specific regula-
mately 600 distinct genes encoding non-olfactory 7TMRs make up tory sites dictating subcellular localization (4). GRK2 is the most
greater than 1% of the human genome [1,2]. Such evolutionary prominent member of this family, binding to and phosphorylating a
diversity enables 7TMRs to detect an extraordinary array of ex- vast majority of agonist-occupied 7TMRs, amongst them - and 2-
tracellular stimuli. 7TMRs function in neurotransmission, neuro- adrenergic receptors (ARs & 2ARs) and angiotensin II type 1
endocrine control of physiologic homeostasis and reproduction, and receptors (AT1Rs) [5]. It is also the most abundant GRK isoform in
regulation of hemodynamics and intermediary metabolism, and the heart and in the cardiovascular (CV) system in general [6].
they control the growth, proliferation, differentiation, and death of When the receptor is activated by agonist, agonist binding induces
multiple cell types. It is estimated that more than half of all drugs in conformational changes in the receptor that persist for a brief pe-
clinical use target 7TMRs, acting either to mimic endogenous riod, during which heterotrimeric G-proteins are activated and dis-
7TMR ligands, to block ligand access to the receptor, or to modu- sociate into G and G subunits. The CT of GRK2 (GRK2ct or
late ligand production [3]. Agonist binding promotes interaction of ARKct) interacts with the free G subunits resulting in transloca-
the receptor with heterotrimeric G proteins, which initiates the clas- tion of GRK2 (which is cytosolic at rest) to the plasma membrane
sical intracellular signaling of these receptors that ultimately leads where it can phosphorylate intracellular domains (i.e. the third in-
to a variety of cellular responses/physiological effects. tracellular loop or the CT tail) of agonist-occupied 7TMRs [4-7].
At the same time, agonist binding promotes the phenomenon of This phosphorylation enhances the affinity of the receptor for
homologous or agonist-dependent receptor desensitization, which is binding to the adapter proteins arrestins (arrs), which comprise
the molecular basis of the waning of the cellular responsiveness to two ubiquitously expressed isoforms in mammals, arrestin-1 and -
persistent receptor stimulation and constitutes a major classical 2 (arr1 and -2), also known as the two non-visual arrestins, ar-
homeostatic mechanism of cellular physiology [4]. This agonist- restin-2 and -3, respectively [4]. arrs are also abundantly ex-
dependent desensitization is conferred, at the molecular level, by pressed in the heart and in the vasculature, as well as in several
phosphorylation of the receptor by the family of kinases known as other tissues [4]. Similarly to the two other mammalian arrestins,
G-protein-coupled receptor kinases (GRKs). GRKs are serine- the visual arrestins (arrestin-1 and -4), arrs bind directly to GRK-
threonine protein kinases comprising a family of seven mem- phosphorylated 7TMRs, forming a stoichiometric complex that is
bers(GRK1-7), which all share a common structural architecture stereochemically incapable of further G protein coupling. arr acti-
vation occurs when the polar core located in the hinge region be-
tween the two globular domains of the arr molecule interacts with
*Address correspondence to this author at the Department of Pharma- GRK-phosphorylated residues on the receptor, displacing the arr
ceutical Sciences, Nova Southeastern University College of Pharmacy, 3200 CT and exposing the concave surface of the globular domains to
S. University Dr., HPD (Terry) Bldg/Room 1338, Fort Lauderdale, FL
33328, USA; Tel: 954-262-1338; FAX: 954-262-2278;
interact with the receptor [8]. Receptor binding produces significant
E-mail: al806@nova.edu conformational changes in the arr molecule [9], whereas, con-

1873-4286/12 $58.00+.00 2012 Bentham Science Publishers


Beta-arrestin Biased Agonism/Antagonism Current Pharmaceutical Design, 2012, Vol. 18, No. 2 193

versely, arr binding stabilizes a high-agonist affinity state of the ARR-DEPENDENT BIASED AGONISM/ANTAGONISM:
receptor, prompting some authors to characterize the receptor-arr GENERAL CONSIDERATIONS
complex as an alternative ternary complex analogous to the ter- Almost all agonists for 7TMRs have been characterized through
nary complex existing between agonist-receptor-G protein in the measurement of G protein signals mediating cell second messen-
absence of GTP [10]. arrs (but, interestingly enough, not the visual gers or calcium. However, as the measurement of arr signaling
arrestins) further dampen G protein signaling by linking receptors effects is becoming routine, agonists are now being classified as
to the clathrin-dependent endocytic machinery [4,11]. The arr CT actually being biased toward the arr system. For example, angio-
that is displaced upon engagement of the receptor directly binds tensin II (AngII) produces activation of G proteins and arrs
clathrin heavy chain and the 2 adaptin subunit of the AP-2 com- through binding to AT1Rs. However, the AngII agonist analog SII
plex, both important components of the endocytic machinery [12- (Sar1,Ile4,Ile8-AngII) (Fig. 1) [32] is perfectly biased toward pro-
15]. Clathrin/AP-2 binding causes arr-bound receptors to cluster in ducing activation of only the arr pathway through this receptor
clathrin-coated pits, which are pinched off the plasma membrane by [33,34] (Fig. 2). Other examples of perfect bias for arr activation
the motor protein dynamin. This arr-dependent endocytosis (re- can be found in standard -blockers. Thus, propranolol (Fig. 1), the
ceptor internalization or sequestration) removes receptors from the prototypic non-subtype selective -blocker and a known inverse
cell surface, rendering them less responsive to subsequent stimuli. agonist for G protein interaction with 2ARs [35], produces activa-
From that point on, most 7TMRs fall into one of two classes based tion of arrs [36,37]. Similar profiles are observed for carvedilol
on their affinity for the two arr isoforms and the longevity of the [38] and bucindolol [37] (Fig. 1). One of the major mechanisms of
receptor-arr interaction [16]. One class exhibits higher affinity for coding for functional selectivity of 7TMRs is the phosphorylation
arr2 than arr1 and forms transient receptor-arr complexes that of the ligand-bound receptor by the GRKs. This barcoding of the
dissociate soon after the receptor internalizes. These receptors (e.g. receptor by GRK phosphorylation forms the link between the ago-
the 2AR) rapidly recycle back to the plasma membrane ready to nist (as it stabilizes unique 7TMR conformations) and the rest of the
signal again upon the next encounter with agonist (receptor resensi- cytosolic machinery [39].
tization). The other class exhibits equivalent affinities for arr1 or -
2 and forms more stable receptor-arr complexes that remain intact The ultimate result of biased agonism/antagonism is a pheno-
as the receptor undergoes endosomal sorting. These receptors (e.g. typic response that can be unique to a given cell. The first step in
AT1R & vasopressin V2 receptor) are sequestered in endosomes and the prosecution of this mechanism for new drug discovery is to
tend to recycle slowly or undergo lysosomal targeting for degrada- have the ability to detect the effect. To this end, there has been vast
tion (receptor downregulation, i.e. total cellular receptor number technological progress in pharmacological assay systems over the
reduced). past few years. One of the most facile approaches has been whole-
system real-time response reading of pharmacologic response from
Unlike the catalytic 7TMR-G protein interaction, arr-bound human cells in culture rather than from isolated tissues. Recent
receptors form relatively stable complexes that persist on a time technological advances have expanded possible modes of detection
scale of minutes to hours [17]. It was the discovery that arrs serve of receptor function. A great deal of information is obtained from
as adapters not only in the context of 7TMR sequestration but also high-content assays based on imaging techniques that use fluores-
in linking activated receptors to other enzymatic effectors that ush- cent signals to yield information about receptor interaction with
ered in a new paradigm shift in 7TMR signal transduction [18-21]. arr and subsequent movement of the receptor/arr complex within
It is now clear that arrs bind a number of catalytically active pro- the cytoplasm. These responses can be monitored directly through
teins and recruit them to agonist-occupied 7TMRs, among them Src observation of receptor/arr green fluorescent protein (GFP) com-
family tyrosine kinases [22], components of the ERK1/2 and c-Jun plexes, with bioluminescence resonance energy transfer (BRET),
N-terminal kinase 3 (JNK3) mitogen-activated protein (MAP) with enzyme fragment complementation, with protease-activated
kinase cascades [23], the E3 ubiquitin ligase Mdm2 [24], the cAMP transcriptional reporter genes, or with whole-cell response meas-
phosphodiesterases (PDE) PDE4D3/5 [25], diacylglycerol kinase urements using optical biosensors that can measure dynamic mass
(DGK) [26], the inhibitor of nuclear factor (NF)-B IB [27], and redistribution signals from whole cells and measurement of the
the Ser/Thr protein phosphatase (PP) PP2A [28]. It is via these electrical impedance of layers of cells in culture caused by receptor-
interactions that arr binding to the receptor initiates secondary mediated changes in cell mass redistribution (see Ref. 30 for an
waves of 7TMR signal transduction independently of G proteins, excellent review of all the experimental approaches used in dissect-
which persist long after receptor signaling via the latter proteins has ing arr biased signaling, and references therein). Below, several
been terminated by the very same binding of arr. Of course, arr- specific examples of cardiovascular 7TMR arr biased signaling
mediated signaling displays several important qualitative differ- (and its agonism/antagonism) in specific organs/tissues of the car-
ences compared to G protein-mediated signal transduction; for in- diovascular system are discussed.
stance, arr signaling does not result in signal amplification, as it
usually proceeds through a 1:1 stoichiometry. Additionally, it ex- CARDIAC AR ARR-DEPENDENT BIASED SIGNALING
hibits specific subcellular localization, dictated by the location of arrs are abundantly expressed in cardiac muscle. As co-factors
the arr-based molecular signaling scaffold, which significantly of GRKs in AR desensitization/downregulation, they contribute to
affects the ultimate signaling events produced, i.e. arr signaling is the diminished inotropic and adrenergic reserve of the failing heart
more compartmentalized compared to the more diffuse, and their inhibition should theoretically be beneficial in acute heart
throughout the cell, G protein signaling [8,29]. failure, as it would enhance the G s-adenylyl cyclase-PKA-
Herein, we will review the current literature regarding mediated pro-contractile signaling of cardiac ARs which increases
arrestin-dependent signaling, with a specific focus on cardiovascu- cardiac contractility [6,40,41]. However, and exactly because arrs
lar 7TMRs and their biased ligands that selectively promote or do a lot more than merely ceasing G protein-mediated signaling
inhibit this signaling. Exciting new therapeutic possibilities emerg- (i.e. they actually promote signaling in their own right), a number
ing from uncovering the physiological roles in vivo in the cardio- of recent studies point to a beneficial role played by arr signaling
vascular system (and the pathological implications in various CV in the heart, especially when the cardiac 1AR is engaged. In heart
diseases) of this heptahelical receptor arrestin-dependent signaling failure, chronic catecholamine stimulation of the 1AR promotes
will also be discussed. For more extensive accounts of the physio- cardiac hypertrophy, decreased contractility, and increased myocyte
logical roles of arr signaling in vitro and in vivo in additional tis- apoptosis [42]. Especially this latter effect is, quite intriguingly,
sues and organ systems, the reader is referred to several excellent postulated to be opposed by cardiac 2AR signaling [42,43]. As a
recent reviews [29-31]. result, administration of -blockers is currently part of standard care
194 Current Pharmaceutical Design, 2012, Vol. 18, No. 2 Anastasios Lymperopoulos

Fig. (1). Structures of some important 7TMR ligands that display bias towards arr-dependent signaling. Structures of various AT1R arr biased ligands (ago-
nists & antagonists) are shown on the left, while the structures of some adrenergic receptor (and other 7TMR) arr biased ligands can be seen on the right.

in the clinical management of congestive heart failure [44,45]. arrs carbon within a series of phenylethylamines [47]. Stereoisomers
have been shown in vitro to mediate the mitogenic signaling of of fenoterol, a 2AR-selective agonist (Fig. 1), differentially acti-
EGF (Epidermal Growth Factor) receptor transactivation by the vate Gs and Gi proteins in rat cardiomyocytes [48]. It has been
1AR. As inhibition of EGF receptors contributes to dilated car- shown that, in addition to blockade of AR-induced G protein acti-
diomyopathy, 1AR signaling via arr2 appears to be protective vation, some -blockers produce activation of ERK through arr
rather than deleterious for the heart [46] and arr2-dependent EGF [36]. In view of the fact that a number of -blockers have been
receptor transactivation might exert a cardioprotective effect. tested in clinical trials for therapy of congestive heart failure and
Therefore, arr2-mediated 1AR signaling might be of therapeutic only a few have shown beneficial effect, this ERK-stimulating ef-
benefit in heart failure (Table 1). fect may be relevant. In particular, carvedilol has been identified as
With regards to arr biased agonism/antagonism at the AR, a producing beneficial effects in congestive heart failure [45], and it
considerable bias for association of 2ARs with arr was observed also has been shown to produce arr-mediated activation of ERK
for selective compounds containing an ethyl substitution of the [38].
Beta-arrestin Biased Agonism/Antagonism Current Pharmaceutical Design, 2012, Vol. 18, No. 2 195

Table 1. Pharmacological Interventions and Potential Therapeutic Effects of Targeting 7TMR arr Signaling in the Cardiovascu-
lar System

Tissue/Receptor Targeted Isoform Pharmacological Intervention Desired Therapeutic Effect (Disease Context)

Cardiac AT1R arr2 stimulation  contractility (HF)

Cardiac 1AR arr2 stimulation  survival-antiapoptosis (HF)

Adrenal AT1R arr1 inhibition  aldosterone (HF)

Adrenal 2AR arr1 (GRK2) inhibition  CA (HF)

VSM AT1 R arr1/arr2 stimulation/inhibition  VSM hyperplasia, hypertrophy (atherosclerosis)

VSM GPR109A arr1 inhibition vasodilation-flushing (niacin drug treatment)


Abbreviations: 7TMR, seven transmembrane-spanning receptor; arr, beta-arrestin; AT1R, angiotensin II type 1 receptor; 1AR, beta1-adrenergic receptor; 2AR, alpha2-adrenergic
receptor; arr1, beta-arrestin1; arr2, beta-arrestin2; CA, catecholamine; GPR109A, G protein-coupled receptor 109A (nicotinic acid receptor); GRK2, G protein-coupled receptor
kinase-2; HF, heart failure; VSM, vascular smooth muscle.

cardiomyocyte proliferation via its classical Gq/11 protein-PKC (pro-


tein kinase C) signaling on one hand, and on the other hand in-
SII creases cardiac contractility via arr2 signaling. Since arr2 is
bound to stop the G protein-mediated signaling of the receptor and
GRK2 also seems to oppose this pro-contractile signaling of arr2
[51], stimulation of arr2 activity at the cardiac AT1AR would ap-
pear therapeutically desirable for the treatment of heart failure and
cardiac hypertrophy (Table 1).
AT1R  2AR ARR-DEPENDENT BIASED SIGNALING & HY-
PERTENSION
The activation of 2ARs results in a beneficial hypotensive
response in hypertension but also in unwanted sedation [52]. This
latter effect can be gauged through observation of reduced coordi-
nation and balance in the rotorod test in mice. The resistance to
impairment of rotorod coordination to the 2AR agonist 5-bromo-
N-(4,5-dihydro-1Himidazol-2-yl)-6-quinoxalinamine (brimonidine
or UK14304) (Fig. 1) in arr2 knockout mice suggests that sedation
may be related to the arr signaling pathway [53]. Thus, a 2AR-
arr biased agonist devoid of arr2 stimulation properties might be of
therapeutic value in the pharmacotherapy of hypertension [54].
Fig. (2). The concept of arr-selective biased agonism exemplified by the ADRENAL AT1R &  2AR ARR-DEPENDENT SIGNALING
AngII analog SII (Sar1,Ile 4,Ile 8-AngII) at the AT1R. SII selectively activates
Aldosterone is one of a number of hormones whose levels are
arr signaling, while simultaneously prohibiting heterotrimeric G protein
elevated in heart failure and produces a multitude of negative ef-
activation from the receptor. See text for details. fects on the failing heart, including promotion of post-myocardial
CARDIAC AT1R ARR-DEPENDENT BIASED SIGNALING infarction adverse cardiac remodeling and heart failure progression
[55-57]. It is produced and secreted by the adrenocortical zona
The AT1R represents another very important heptahelical recep- glomerulosa cells in response to AT1R activation by AngII [58].
tor in CV physiology and pathology. An artificially constructed Until recently, the general consensus for AT1R signaling to aldos-
AT1AR mutant (AT1-i2m), which fails to activate G proteins but terone production was that it proceeded via activation of Gq/11 pro-
nonetheless interacts with arrs, when expressed in cardiomyocytes teins, to which the AT1R normally couples [59]. Recently however,
of transgenic mice in vivo, leads to greater cardiomyocyte hypertro- we described a crucial role for arr1 in mediating the signaling
phy, bradycardia, and fetal cardiac gene expression than compara- from AT1R to aldosterone synthesis and secretion in vitro and in
ble overexpression of the wild type receptor [49]. In primary car- vivo [60]. Specifically, arr1 was found to stimulate sustained
diomyocytes, the arr biased agonist at the AT1R SII stimulates ERK1/2 activation and subsequent upregulation of steroidogenic
cardiomyocyte proliferation independently of G proteins, but not acute regulatory protein (StAR), a steroid transport protein that
hypertrophy, which requires Gq/11 protein signaling [50]. In addi- catalyzes the rate-limting step in adrenal steroid biosynthesis.
tion, SII produces positive inotropic and lusitropic effects on iso- Moreover, arr1 was shown to promote aldosterone production
lated murine cardiomyocytes [51]. These effects require GRK6 and independently of G-proteins, since SII could recapitulate the effects
arr2, whereas GRK2 seems to oppose them, consistent with the of AngII on aldosterone turnover [60]. Importantly, in vivo, adrenal
specialized role of GRK isoforms described in a transfected system arr1 was shown to be a major regulator of circulating aldosterone
[52] and with the concept of GRK-induced receptor barcoding levels under normal healthy conditions, since its upregulation, spe-
discussed earlier [39]. On the other hand, SII does not produce cifically in the adrenal gland, caused hyperaldosteronism [60]. Sub-
inotropic or chronotropic effects in isolated Langendorff-perfused sequently, we investigated the effects of adrenal arr1 on aldoster-
cardiac preparations despite its ability to activate ERK1/2 [50]. one levels also in diseased animals, as they progressed to HF after a
Thus, it seems that the AT1AR promotes cardiac hypertrophy and vast experimental myocardial infarction [61]. We found that adrenal
196 Current Pharmaceutical Design, 2012, Vol. 18, No. 2 Anastasios Lymperopoulos

arr1 overexpression promoted aldosterone elevation post- mines (sympatholysis) as well as to lower aldosterone, both of
myocardial infarction, resulting in accelerated cardiac adverse re- which hormones contribute significantly to the morbidity and mor-
modeling and deterioration of left ventricular function. Importantly, tality of heart failure (Table 1).
these detrimental effects of aldosterone were prevented when adre-
nal arr1 was inhibited in vivo via gene therapy with a arr1 CT VASCULAR AT1R & NICOTINIC ACID RECEPTOR ARR-
domain-derived fragment-encoding mini-gene which we designed DEPENDENT SIGNALING
and developed. Adrenal arr1 inhibition with this mini-gene mark- In vascular smooth muscle (VSM), arr-dependent signaling
edly reduced circulating aldosterone levels and improved cardiac appears to promote development and progression of atherosclerotic
structure and function in post-myocardial infarction animals, vascular disease. Neointimal hyperplasia after carotid endothelial
largely preventing (and/or even reversing) the ensuing cardiac ad- injury is enhanced in arr1-knockout mice but diminished in arr2-
verse remodeling [61]. Thus, it appears that in the adrenal gland, knockout mice. Loss of arr2 appears to decrease 7TMR-stimulated
arr1 is a major driving force behind elevation of the cardiotoxic VSM cell proliferation and ERK1/2 activation/migration, consistent
aldosterone in HF, and its inhibition might be of therapeutic value with a role for arr2 signaling in the injury response. When the
in treatment of hyperaldosteronism of post-myocardial infarction low-density lipoprotein receptor-knockout mouse, a genetic model
heart failure (Table 1). of enhanced atherogenesis, is crossed onto a arr2-knockout back-
In addition, in that very same study, we found that losartan, the ground, atheromas are significantly reduced [65]. In vitro, both G
protoypic AT1R-selective antagonist (angiotensin receptor blocker, protein- and arr-dependent pathways elicited by the AT1AR con-
ARB or sartan), virtually fails to suppress adrenal arr1-dependent verge on the EGF receptor in primary VSM cells to stimulate pro-
post-myocardial infarction hyperaldosteronism [61]. This prompted liferation. Specifically, arr2 seems to enhance Src-dependent EGF
us to test various other sartans, structurally similar to losartan (i.e. receptor transactivation by the AT1AR [66]. Moreover, arr2-
biphenylmethyl derivatives, Fig. 1) and important drugs used in dependent ERK1/2 activation downregulates the pro-apoptotic
current clinical practice, in terms of their efficacy at inhibiting the phospho-BAD protein, thus inducing anti-apoptotic cytoprotective
AT1R-arr1 interaction and thereby suppressing AT1R-induced effects in rat VSM [67]. Thus, it appears that arr2 can promote
aldosterone production. Our latest data indicate that losartan, as AT1AR-dependent VSM proliferation and hypertrophy, thus con-
well as irbesartan (Fig. 1), are very weak biased antagonists to- tributing to the development and progression of atheromas, whereas
wards arr inhibition at the AT1R, displaying very little (if any) arr1 again (similarly to cardiac AT1AR pro-contractile signaling)
bias for arr inhibition vs. G protein inhibition and consequently opposes these actions of arr2. Therefore, in VSM and in athero-
are ineffective at curbing aldosterone production [Ref. 62 & Lym- sclerotic disease, arr2 inhibition and/or arr1 stimulation might be
peropoulos A et al., unpublished data]. In contrast, among the therapeutically desirable (Table 1).
AT1R antagonists tested, candesartan and valsartan (Fig. 1) were Another vascular-related physiological effect of arr-dependent
found to be the most potent arr inhibitors at the AT1R and the signaling appears to be facilitation of niacin (nicotinic acid)-
most biased arr antagonists, and this was reflected also on their induced flushing, a major adverse effect of pharmacotherapy with
effects on aldosterone production, which they dramatically sup- this very useful lipid-lowering drug that very often poses several
pressed in vitro and in vivo [Ref. 62 & Lymperopoulos A et al., limitations on its use in patients. Through its actions on GPR109A,
unpublished data]. Based on these data and on a crude comparison a 7TMR, nicotinic acid has been shown to decrease serum free fatty
of the structures of these biphenylmethyl derivatives (Fig. 1), which acids by activating Gi/o signaling and to increase cutaneous blood
are all very potent antagonists of the AT1R in terms of G protein flow/flushing in humans and in mice by stimulating cytosolic phos-
inhibition, some very interesting conclusions about the structural pholipase A2, leading to the production and secretion of prosta-
requirements for arr biased antagonism at the AT1R can be drawn. glandin D2 [68,69]. It was recently demonstrated that arr1, al-
Specifically, it appears that: a) the tetrazole ring substitution at the though dispensable for the beneficial effects of niacin on free fatty
R2 position of the biphenylmethyl backbone is necessary for arr acid levels, actually promotes the niacin-related flushing, as meas-
inhibition (Fig. 1), and b) the bulkier the substitution at the R 1 ured by perfusion of the ventral ear. The underlying mechanism
position, the more potent arr biased antagonist the compound is appears to be niacin-induced arr1-mediated activation of phos-
(Fig. 1), since candesartan and valsartan have the bulkiest R 1 pholipase A2, which stimulates arachidonic acid release, the pre-
substitutions, whereas losartan and irbesartan the smallest. We also cursor of vasodilatory prostaglandin D2 responsible for the flushing
tested a series of AngII analogs with various substitutions at amino- response [70]. Thus, arr1 inhibition at the GPR109A receptor or a
acid position 5 at their potency as arr biased agonists. These ana- biased GPR109A ligand capable of activating G protein signaling
logs had the sarcosine substitution at aminoacid position 1, so they without arr1 activation would be therapeutically desirable, as it
were all antagonists with regards to G protein activation. Based on would confer the same therapeutic effect of niacin on lipid metabo-
our data with these peptide analogs, we were able to conclude that, lism without the adverse effect of flushing (Table 1). Notably, a
when it comes to arr biased agonism at the AT1R, the bulkier series of pyrazole derivatives with GPR109A receptor agonist
the aminoacid substitution at position 5, the more potent arr biased properties have been synthesized that are devoid of the ability to
agonist the resultant compound is [Ref. 62 & Lymperopoulos A et internalize GPR109A and activate ERK, and thus they do not cause
al., unpublished data]. Of note, SII, the prototypic arr biased flushing [71]. One of these pyrazole derivatives, 3-(1H-tetrazol-5-
agonist at the AT1R, has no substitution at this position (it contains yl)-1,4,5,6-tetrahydrocyclopentapyrazole (MK-0354) (Fig. 1), de-
a substitution at aminoacid position 4 instead, Ile in the place of creases serum free fatty acids without producing cutaneous flush-
Tyr, Fig. 1). Of course, the substitutions at positions 1 and 8 are ing, probably by causing selective G protein activation and simulta-
always indispensable in order for the compound to be devoid of any neous arr1 inhibition at the GPR109A receptor, and appears thus
G protein activating properties [Refs. 32, 62 & Lymperopoulos A et superior to niacin for lipid lowering therapy [72].
al., unpublished data].
Finally, adrenal arr1, in conjunction with its co-factor GRK2, CONCLUDING REMARKS/FUTURE DIRECTIONS
also contributes to adrenal 2AR desensitization and downregula- The emerging and increasingly expanding field of 7TMR arr-
tion which results in chronically elevated catecholamine secretion, dependent signaling offers several exciting new opprtunities for
another enormous heurohormonal burden on the failing heart therapeutic intervention in cardiovascular disease. Despite being
[63,64]. Therefore, adrenal arr1 (and/or GRK2) inhibition poses as still in its infancy with respect to ascribing certain physiological
an attractive therapeutic strategy for lowering the neurohormonal and pathophysiological effects to specific arr isoforms and in spe-
burden of the failing heart, since it should lead to lower catechola- cific tissues/organs, the potential advantages of exploiting this area
Beta-arrestin Biased Agonism/Antagonism Current Pharmaceutical Design, 2012, Vol. 18, No. 2 197

for therapeutic purposes is enormous. Admittedly, the picture is still [10] Gurevich VV, Pals-Rylaarsdam R, Benovic JL, Hosey MM,
hazy regarding physiological actions of arrs, in particular with Onorato JJ. Agonist-receptor-arrestin, an alternative ternary com-
relation to the cardiovascular system. For instance, cardiac arr2 plex with high agonist affinity. J Biol Chem 1997; 272: 28849-52.
inhibits contractility by ARs but appears to enhance it by the [11] Stoffel RH 3rd, Pitcher JA, Lefkowitz RJ. Targeting G protein-
coupled receptor kinases to their receptor substrates. J Membr Biol
AT1R. It also appears to have beneficial effects in the heart as it 1997; 157: 1-8.
increases survival and cardiomyocyte proliferation, as well as [12] Goodman OB Jr, Krupnick JG, Santini F, et al. Beta-arrestin acts as
AngII-induced contractility, but it also promotes atherosclerosis in a clathrin adaptor in endocytosis of the beta2-adrenergic receptor.
the vasculature. On the other hand, arr1 seems to have negative Nature 1996; 383: 447-50.
effects in most cardiovascular tissues/organs whose physiology it is [13] Krupnick JG, Goodman OB Jr, Keen JH, Benovic JL. Ar-
involved in: it opposes cardiac AR (and AT1R) pro-contractile restin/clathrin interaction. Localization of the clathrin binding do-
signaling, promotes AngII-induced aldosterone produc- main of nonvisual arrestins to the carboxy terminus. J Biol Chem
tion/secretion by the adrenal glands and mediates niacin-induced 1997; 272: 15011-6.
flushing. Further, since it theoretically competes with arr2 for [14] Laporte SA, Oakley RH, Holt JA, Barak LS, Caron MG. The inter-
action of beta-arrestin with the AP-2 adaptor is required for the
receptor binding, inhibition of arr1 would indirectly enhance arr2 clustering of beta 2-adrenergic receptor into clathrin-coated pits. J
activity and its beneficial effects in the heart. Thus, it appears that Biol Chem 2000; 275: 23120-6.
these two arr isoforms by no means complement each other with [15] Laporte SA, Oakley RH, Zhang J, Holt JA, Ferguson SS, Caron
regard to their physiological actions, and, in fact, more often than MG, Barak LS. The beta2-adrenergic receptor/betaarrestin complex
not, their signaling pathways result in diametrically opposing ulti- recruits the clathrin adaptor AP-2 during endocytosis. Proc Natl
mate cellular effects. Therefore, the situation in terms of their Acad Sci USA 1999; 96: 3712-7.
therapeutic exploitation for cardiovascular disease treatment be- [16] Oakley RH, Laporte SA, Holt JA, Caron MG, Barak LS. Differen-
comes even more complicated and it is quite plausible that isoform- tial affinities of visual arrestin, beta arrestin1, and beta arrestin2 for
specific targeting might ultimately be warranted, if direct targeting G protein-coupled receptors delineate two major classes of recep-
tors. J Biol Chem 2000; 275: 17201-10.
of arr-dependent signaling (i.e. donwstream of the receptor) is [17] Charest PG, Terrillon S, Bouvier M. Monitoring agonist-promoted
ever going to be seriously considered as a therapeutic strategy for conformational changes of beta-arrestin in living cells by in-
the treatment of cardiovascular disease. For instance, inhibitors of tramolecular BRET. EMBO Rep 2005; 6: 334-40.
arr1 and enhancers of arr2 activity (or vice versa in the case of [18] Miller WE, Lefkowitz RJ. Expanding roles for beta-arrestins as
atherosclerosis) or inhibition of arr1 only, which would also indi- scaffolds and adapters in GPCR signaling and trafficking. Curr
rectly increase arr2 activity appear desirable for cardiovascular Opin Cell Biol 2001; 13: 139-45.
signal transduction therapy. Future studies will hopefully help clar- [19] Perry SJ, Lefkowitz RJ. Arresting developments in heptahelical
ify the picture with regards to the exact roles of arr-dependent receptor signaling and regulation. Trends Cell Biol 2002; 12: 130-
signaling in vivo in the cardiovascular system. For the time being, 8.
[20] Maudsley S, Martin B, Luttrell LM. The origins of diversity and
synthesis and development of new 7TMR biased ligands, which specificity in G protein-coupled receptor signaling. J Pharmacol
selectively activate one pathway over the other from the same re- Exp Ther 2005; 314: 485-94.
ceptor or even activate one pathway (e.g. G protein signaling) while [21] Shenoy SK, Lefkowitz RJ. Seven-transmembrane receptor signal-
inhibiting the other (e.g. arr signaling), depending each time on ing through beta-arrestin. Sci STKE 2005: cm10.
the specific 7TMR, cell type and desired cellular effect in question, [22] Luttrell LM, Ferguson SS, Daaka Y, et al. Beta- arrestin-dependent
seems the most reasonable and pragmatic approach of exploiting formation of beta2 adrenergic receptor-Src protein kinase com-
7TMR arr signaling therapeutically. In any case, this arr- plexes. Science 1999; 283: 655-61.
dependent 7TMR signal transduction represents a very exciting and [23] Luttrell LM, Roudabush FL, Choy EW, et al. Activation and target-
potentially very promising avenue of possibilities for novel thera- ing of extracellular signal-regulated kinases by beta-arrestin scaf-
folds. Proc Natl Acad Sci USA 2001; 98: 2449-54.
peutic approaches in cardiovascular disease, a disease area with an [24] Shenoy SK, McDonald PH, Kohout TA, Lefkowitz RJ. Regulation
overwhelmingly high mortality burden compared to the therapeutic of receptor fate by ubiquitination of activated beta 2-adrenergic re-
arsenal available to clinicians for combating it. ceptor and beta-arrestin. Science 2001; 294: 1307-13.
[25] Perry SJ, Baillie GS, Kohout TA, et al. Targeting of cyclic AMP
REFERENCES degradation to beta 2-adrenergic receptors by beta-arrestins. Sci-
[1] Lander E S, Linton LM, Birren B, et al. Initial sequencing and ence 2002; 298: 834-6.
analysis of the human genome. Nature 2001; 409: 860-921. [26] Nelson CD, Perry SJ, Regier DS, Prescott SM, Topham MK,
[2] Venter JC, Adams MD, Myers, et al. The sequence of the human Lefkowitz RJ. Targeting of diacylglycerol degradation to M1 mus-
genome. Science 2001; 291: 1304-51. carinic receptors by beta-arrestins. Science 2007; 315: 663-6.
[3] Flower DR. Modeling G-protein-coupled receptors for drug design. [27] Witherow DS, Garrison TR, Miller WE, Lefkowitz RJ. beta-
Biochem Biophys Acta 1999; 1422: 207-34. Arrestin inhibits NF-kappaB activity by means of its interaction
[4] Ferguson SS. Evolving concepts in G protein-coupled receptor with the NF-kappaB inhibitor IkappaBalpha. Proc Natl Acad Sci
endocytosis: the role in receptor desensitization and signaling. USA 2004; 101: 8603-7.
Pharmacol Rev 2001; 53: 1-24. [28] Beaulieu JM, Sotnikova TD, Marion S, Lefkowitz RJ, Gainetdinov
[5] Hata JA, Koch WJ. Phosphorylation of G protein-coupled recep- RR, Caron MG. An Akt/beta-arrestin 2/PP2A signaling complex
tors: GPCR kinases in heart disease. Mol Interv 2003; 3: 264-72. mediates dopaminergic neurotransmission and behavior. Cell 2005;
[6] Rockman HA, Koch WJ, Lefkowitz RJ. Seven-transmembrane- 122: 261-73.
spanning receptors and heart function. Nature 2002; 415: 206-12. [29] Luttrell LM, Gesty-Palmer D. Beyond desensitization: physiologi-
[7] Reiter E, Lefkowitz RJ. GRKs and beta-arrestins: roles in receptor cal relevance of arrestin-dependent signaling. Pharmacol Rev 2010;
silencing, trafficking and signaling. Tr Endocrinol Metab 2006; 17: 62: 305-30.
159-265. [30] Rajagopal S, Rajagopal K, Lefkowitz RJ. Teaching old receptors
[8] Gurevich VV, Gurevich EV. The structural basis of arrestin- new tricks: biasing seven-transmembrane receptors. Nat Rev Drug
mediated regulation of G-protein-coupled receptors. Pharmacol Discov 2010; 9: 373-86.
Ther 2006; 110: 465-502. [31] DeWire SM, Ahn S, Lefkowitz RJ, Shenoy SK. Beta-arrestins and
[9] Shukla AK, Violin JD, Whalen EJ, Gesty-Palmer D, Shenoy SK, cell signaling. Annu Rev Physiol 2007; 69: 483-510.
Lefkowitz RJ. Distinct conformational changes in beta-arrestin re- [32] Holloway AC, Qian H, Pipolo L, et al. Side-chain substitutions
port biased agonism at seven-transmembrane receptors. Proc Natl within angiotensin II reveal different requirements for signaling, in-
Acad Sci USA 2008; 105: 9988-93. ternalization, and phosphorylation of type 1A angiotensin recep-
tors. Mol Pharmacol 2002; 61: 768-77.
[33] Ahn S, Nelson CD, Garrison TR, Miller WE, Lefkowitz RJ. Desen-
sitization, internalization, and signaling functions of beta-arrestins
198 Current Pharmaceutical Design, 2012, Vol. 18, No. 2 Anastasios Lymperopoulos

demonstrated by RNA interference. Proc Natl Acad Sci USA 2003; [53] Wang Q, Zhao J, Brady AE, Feng J, et al. Spinophilin blocks ar-
100: 1740-4. restin actions in vitro and in vivo at G protein-coupled receptors.
[34] Wei H, Ahn S, Shenoy SK, et al. Independent beta-arrestin 2 and G Science 2004; 304: 1940-1944.
protein-mediated pathways for angiotensin II activation of extracel- [54] Schmid CL, Bohn LM. Physiological and pharmacological implica-
lular signal-regulated kinases 1 and 2. Proc Natl Acad Sci USA tions of beta-arrestin regulation. Pharmacol Ther 2009; 121: 285-
2003; 100: 10782-7. 93.
[35] Baker JG, Hall IP, Hill SJ. Agonist and inverse agonist actions of [55] Weber KT. Aldosterone in congestive heart failure. N Engl J Med
-blockers at the human 2-adrenoceptor provide evidence for 2001; 345: 1689-97.
agonist-directed signaling. Mol Pharmacol 2003; 64: 1357-69. [56] Connell JM, Davies E. The new biology of aldosterone. J Endocri-
[36] Azzi M, Charest PG, Angers S, Rousseau G, Kohout T, Bouvier M, nol 2005; 186: 1-20.
Pineyro G. - Arrestin-mediated activation of MAPK by inverse [57] Marney AM, Brown NJ. Aldosterone and end-organ damage. Clin
agonists reveals distinct active conformations for G protein- Sci (Lond) 2007; 113: 267-78.
coupled receptors. Proc Natl Acad Sci USA 2003; 100: 11406-11. [58] Ganguly A, Davis JS. Role of calcium and other mediators in al-
[37] Galandrin S, Oligny-Longpre G, Bonin H, Ogawa K, Gales C, dosterone secretion from the adrenal glomerulosa cells. Pharmacol
Bouvier M. Conformational rearrangements and signaling cascades Rev 1994; 46: 417-47.
involved in ligand-biased mitogen-activated protein kinase signal- [59] De Gasparo M, Catt KJ, Inagami T, Wright JW, Unger T. Interna-
ing through the 1-adrenergic receptor. Mol Pharmacol 2008; 74: tional union of pharmacology. XXIII. The angiotensin II receptors.
162-172. Pharmacol Rev 2000; 52: 415-72.
[38] Wisler JW, DeWire SM, Whalen EJ, et al. A unique mechanism of [60] Lymperopoulos A, Rengo G, Zincarelli C, Kim J, Soltys S, Koch
-blocker action: carvedilol stimulates -arrestin signaling. Proc WJ. An adrenal beta-arrestin 1-mediated signaling pathway under-
Natl Acad Sci USA 2007; 104: 16657-62. lies angiotensin II-induced aldosterone production in vitro and in
[39] Zidar DA, Violin JD, Whalen EJ, Lefkowitz RJ. Selective engage- vivo. Proc Natl Acad Sci USA 2009; 106: 5825-30.
ment of G protein coupled receptor kinases (GRKs) encodes dis- [61] Lymperopoulos A, Rengo G, Zincarelli C, Kim J, Koch WJ. Adre-
tinct functions of biased ligands. Proc Natl Acad Sci USA 2009; nal Beta-arrestin 1 inhibition in vivo attenuates post-myocardial in-
106: 9649-54. farction progression to heart failure and adverse remodeling via re-
[40] Rengo G, Lymperopoulos A, Koch WJ. Future g protein-coupled duction of circulating aldosterone levels. J Am Coll Cardiol 2011;
receptor targets for treatment of heart failure. Curr Treatment Op- 57: 356-65.
tions Cardiovasc Med 2009; 11: 328-38. [62] Lymperopoulos A, Rengo G, Magafa V, Cordopatis P, Koch WJ.
[41] Rengo G, Lymperopoulos A, Zincarelli C, et al. Myocardial adeno- Biased Agonism/Antagonism of -Arrestin activation by the An-
associated virus serotype 6-betaARKct gene therapy improves car- giotensin II Type 1 Receptor: A Study of Sartans and Angiotensin
diac function and normalizes the neurohormonal axis in chronic II Analogs Using Aldosterone Turnover as a Readout. J. Card. Fail.
heart failure. Circulation 2009; 119: 89-98. Suppl. 1 2010; 16: S30-1.
[42] Xiang Y, Kobilka BK. Myocyte adrenoceptor signaling pathways. [63] Lymperopoulos A, Rengo G, Funakoshi H, Eckhart AD, Koch WJ.
Science 2003; 300: 1530-2. Adrenal GRK2 upregulation mediates sympathetic overdrive in
[43] Communal, C, Singh K, Sawyer DB, Colucci WS. Opposing ef- heart failure. Nat Med 2007; 13: 315-23.
fects of beta(1)- and beta(2)-adrenergic receptors on cardiac myo- [64] Lymperopoulos A, Rengo G, Koch WJ. Adrenal adrenoceptors in
cyte apoptosis: role of a pertussis toxin-sensitive G protein. Circu- heart failure: fine-tuning cardiac stimulation. Tr Mol Med 2007;
lation 1999; 100: 2210-2. 13: 503-11.
[44] Bristow MR. -adrenergic receptor blockade in chronic heart fail- [65] Kim J, Zhang L, Peppel K, et al. Beta-arrestins regulate atheroscle-
ure. Circulation 2000; 101: 558-69. rosis and neointimal hyperplasia by controlling smooth muscle cell
[45] Packer M, Bristow MR, Cohn JN, Colucci WS, Fowler MB, proliferation and migration. Circ Res 2008; 103: 70-9.
Gilbert EM, Shusterman NH. The effect of carvedilol on morbidity [66] Kim J, Ahn S, Rajagopal K, Lefkowitz RJ. Independent beta-
and mortality in patients with chronic heart failure. N Engl J Med arrestin2 and Gq/protein kinase Czeta pathways for ERK stimu-
1996; 334: 1349-55. lated by angiotensin type 1A receptors in vascular smooth muscle
[46] Noma T, Lemaire A, Naga Prasad SV, et al. Beta-arrestin-mediated cells converge on transactivation of the epidermal growth factor re-
beta1-adrenergic receptor transactivation of the EGFR confers car- ceptor. J Biol Chem 2009; 284: 11953-62.
dioprotection. J Clin Invest 2007; 117: 2445-58. [67] Ahn S, Kim J, Hara MR, Ren XR, Lefkowitz RJ. Beta-arrestin-2
[47] Drake MT, Violin JD, Whalen EJ, Wisler JW, Shenoy SK, mediates anti-apoptotic signaling through regulation of BAD phos-
Lefkowitz RJ. -arrestin-biased agonism at the 2-adrenergic re- phorylation. J Biol Chem 2009; 284: 8855-65.
ceptor. J Biol Chem 2008; 283: 5669- 5676. [68] Kather H, Aktories K, Schulz G, Jakobs KH. Islet-activating pro-
[48] Woo AY, Wang TB, Zeng X, et al. Stereochemistry of an agonist tein discriminates the antilipolytic mechanism of insulin from that
determines coupling preference of beta2-adrenoceptor to different of other antilipolytic compounds. FEBS Lett 1983; 161: 149-52.
G proteins in cardiomyocytes. Mol Pharmacol 2009; 75: 158-65. [69] Pike NB. Flushing out the role of GPR109A (HM74A) in the clini-
[49] Zhai P, Yamamoto M, Galeotti J, et al. Cardiac-specific overex- cal efficacy of nicotinic acid. J Clin Invest 2005; 115: 3400-3.
pression of AT1 receptor mutant lacking G alpha q/G alpha i cou- [70] Walters RW, Shukla AK, et al. Beta-Arrestin1 mediates nicotinic
pling causes hypertrophy and bradycardia in transgenic mice. J Clin acid-induced flushing, but not its antilipolytic effect, in mice. J Clin
Invest 2005; 115: 3045-56. Invest 2009; 119: 1312-21.
[50] Aplin M, Christensen GL, Schneider M, et al. Differential extracel- [71] Richman JG, Kanemitsu-Parks M, Gaidarov I, et al. Nicotinic acid
lular signal-regulated kinases 1 and 2 activation by the angiotensin receptor agonists differentially activate downstream effectors. J
type 1 receptor supports distinct phenotypes of cardiac myocytes. Biol Chem 2007; 282: 18028-36.
Basic Clin Pharmacol Toxicol 2007; 100: 296-301. [72] Semple G, Skinner PJ, Gharbaoui T, et al. 3-(1H-tetrazol-5-yl)-
[51] Rajagopal K, Whalen EJ, Violin JD, et al. Beta-arrestin2-mediated 1,4,5,6-tetrahydro-cyclopentapyrazole (MK-0354): a partial agonist
inotropic effects of the angiotensin II type 1A receptor in isolated of the nicotinic acid receptor, G-protein coupled receptor 109a,
cardiac myocytes. Proc Natl Acad Sci USA 2006; 103: 16284-9. with antilipolytic but no vasodilatory activity in mice. J Med Chem
[52] Kukkonen JP. Regulation of receptor-coupling to (multiple) G 2008; 51: 5101-8.
proteins. A challenge for basic research and drug discovery. Recep-
tors and Channels 2004; 10: 167-83.

Received: September 4, 2011 Accepted: November 9, 2011

You might also like