SPECIAL ARTICLE Circ J 2008; 72: 1725–1729

Beta--Mediated Signaling in the Heart

Priyesh A. Patel, BS; Douglas G. Tilley, PhD*; Howard A. Rockman, MD*,**

Beta-arrestin is a multifunctional adapter well known for its role in G-protein-coupled receptor (GPCR) desensitization. Exciting new evidence indicates thatβ-arrestin is also a signaling molecule capable of initiating its own G-protein-independent signaling at GPCRs. One of the best-studiedβ-arrestin signaling pathways is the one involvingβ-arrestin-dependent activation of a mitogen-activated protein kinase cascade, the extracellular regulated kinase (ERK). ERK signaling, which is classically activated by agonist stimulation of the epidermal growth factor receptor (EGFR), can be activated by a number of GPCRs in aβ-arrestin-dependent manner. Recent work in animal models of heart failure suggests thatβ-arrestin-dependent activation of EGFR/ERK signaling by theβ-1-, and possibly the angiotensin II Type 1A receptor, are cardioprotective. Hence, a new model of signaling at cardiac GPCRs has emerged and implicates classical G-protein-mediated signaling with promoting harmful remodeling in heart failure, while concurrently linkingβ-arrestin-dependent, G-protein-inde- pendent signaling with cardioprotective effects. Based on this paradigm, a new class of drugs could be identified, termed “biased ligands”, which simultaneously block harmful G-protein signaling, while also promoting cardio- protectiveβ-arrestin-dependent signaling, leading to a potential breakthrough in the treatment of chronic cardiac disease. (Circ J 2008; 72: 1725–1729) Key Words: Beta-arrestin; Beta-1-adrenergic receptor; Extracellular regulated kinase (ERK); G protein- coupled receptor

-protein-coupled receptors (GPCR) are ubiquitously Beta-Arrestin as a Desensitizing expressed, 7-transmembrane spanning receptors and Signaling Molecule G that regulate a vast multitude of physiologic proc- esses.1 The classic paradigm is that agonist stimulation of a Desensitization GPCR activates a receptor-associated G protein to promote Beta-arrestin exists as 2 isoforms, β-arrestin1 and β- downstream signaling via generation of second messengers.1 arrestin2, both of which are important in the regulation of Termination of GPCR signaling occurs by a process known GPCR signaling. It has long been known that β- as desensitization,2 which involves phosphorylation of the prevent G protein signaling by physically uncoupling the activated receptor by a GPCR kinase and subsequent recruit- interaction between a GPCR and its associated G protein.1,3 ment of the multifunctional adapter proteinβ-arrestin. The Moreover, recent studies of the β-2-adrenergic receptor β-arrestin in turn physically uncouples the G protein from (β2AR) and the muscarinic M1 receptor show thatβ-arres- the receptor, effectively terminating signaling.1,3 Although tins directly promote degradation of second messengers via the role ofβ-arrestin in desensitization is well established, recruitment of phosphodiesterase and diacylglycerol kinase recent evidence indicates thatβ-arrestin is far more func- respectively, hinting at a more elegant and versatile role for tionally versatile than previously appreciated.1,3 β-arrestins in desensitization than previously understood.5,6 Perhaps the most important discovery in the past decade In addition to their role in desensitization, bothβ-arrestin1 is the ability ofβ-arrestin to facilitate trafficking of the re- andβ-arrestin2 are intricately involved in GPCR trafficking, ceptor and initiate cell signaling events in its own right, recycling, and degradation following agonist stimulation independent of G protein activation. This new paradigm (reviewed by Moore et al3). establishes that, while agonist stimulation of a GPCR leads to immediate G-protein-mediated signaling,β-arrestin itself Signaling promotes a second wave of β-arrestin-dependent, G-pro- Although β-arrestins indirectly alter signaling through tein-independent signaling.1,3,4 Here we present evidence their roles in receptor desensitization and trafficking, new highlighting the importance of distinguishing the discrete data show that they can initiate signaling independent of G physiologic outcomes ofβ-arrestin-dependent vs G-protein- protein activation. The best studied example of aβ-arrestin- dependent signaling at GPCRs. activated signaling pathway is the extracellular regulated kinase (ERK) cascade. ERK signaling is prototypically activated by growth factor stimulation of receptor tyrosine (Received August 1, 2008; accepted August 1, 2008; released online 7 October 7, 2008) kinases (RTKs). However, recent studies have shown that School of Medicine, *Department of Medicine and **Department of several GPCRs can themselves initiate ERK signaling by Cell Biology and Genetics, Duke University, Durham, NC, USA both G-protein- andβ-arrestin-dependent processes. Exam- Mailing address: Howard A. Rockman, MD, Department of Medicine, ples of such receptors include the angiotensin II Type 1A Duke University Medical Center, DUMC 3104, 226 CARL Building, receptor (AT1AR),8β-1-adrenergic receptor (β1AR),9β2AR,10 Research Drive, Durham, NC 27710, USA. E-mail: h.rockman@ vasopressin V2 receptor11 and parathyroid hormone recep- duke.edu Supported by grants from the National Institutes of Health 12 to HAR (HL56687 and HL75443) tor. Interestingly, the time course and molecular conse- All rights are reserved to the Japanese Circulation Society. For per- quences of activating ERK signaling through G-protein- missions, please e-mail: [email protected] mediated pathways vs β-arrestin-mediated pathways are

Circulation Journal Vol.72, November 2008 1726 PATEL PA et al.

Fig1. Theβ-arrestin-dependent and G-protein-de- pendent pathways differentially activate extracellular regulated kinase (ERK) signaling. The angiotensin II Type 1A receptor (AT1AR) is a Gq coupled receptor that can activate ERK signaling by both G-protein- andβ-arrestin-dependent processes. Upon angioten- sin II stimulation, inhibition ofβ-arrestin signaling by the AT1AR usingβ-arrestin2 siRNA reveals that Gq promotes rapid, short-lived activation of ERK (blue line). In contrast,β-arrestin mediates a late, extended phase of ERK activation as seen by PKC inhibition of Gq signaling (orange line). Inhibiting both G protein and β-arrestin-signaling prevents ERK activation (green line), suggesting that the AT1AR cannot acti- vate ERK in a manner independent of G protein orβ- arrestin. PKC, protein kinase C; CTL, control siRNA. Printed with permission from Science 2005; 308: 512–517 and J Biol Chem 2004; 279: 35518–35525.

Fig2. Theβ-1-adrenergic receptor (β1AR)-mediated transactivation of epidermal growth factor receptor (EGFR) isβ- arrestin dependent. HEK293 cells stably overexpressing wild-typeβ1AR were transiently transfected with EGFR-GFP and eitherβ-arrestin si-RNA or control si-RNA. After treatment with theβ1AR agonist dobutamine (Dob), EGFR redis- tributed into cellular aggregates, thereby indicating activation of the EGFR (panel 2, arrowheads). Redistribution of the EGFR into aggregates was prevented in the presence ofβ-arrestin siRNA, in which case EGFR remained localized to the plasma membrane (panel 5, arrow). Taken together, these findings indicate thatβ-arrestin is necessary forβ1AR-mediated transactivation of the EGFR. ICI, ICI118551, a selectiveβ-2-adrenergic receptor blocker; IB, immunoblot; EGF, epidermal growth factor; GFP, green fluorescent protein. Adapted with permission from J Clin Invest 2007; 117: 2445–2458. quite different. For example, in HEK293 cells overexpress- creasingly been investigated in order to uncover the cross- ing the AT1AR, G protein activation leads to peak activity talk between the GPCR and RTK signaling pathways. ERK within 2min and promotes both nuclear and cytoplasmic signaling is classically activated by agonist stimulation of localization of activated ERK. In contrast,β-arrestin-medi- RTKs, an example being the epidermal growth factor recep- ated signaling following angiotensin II stimulation of tor (EGFR). EGFR initiates a signaling cascade consisting AT1AR has a slower and more prolonged pattern and of Raf, MEK, and ERK, as well as recruitment of several promotes only cytoplasmic ERK localization (Fig1).13 In adapter and scaffolding , including Src, Grb2 and addition, G-protein-mediated nuclear localization of ERK Ras, to promote mitogenic and anti-apoptotic effects.7,15 promotes increased transcription of early growth response 1, Anti-apoptotic signaling can be initiated through ERK phos- indicating that only G-protein-dependent signaling promotes phorylation of Bad, thereby allowing homodimerization of transcriptional events.8,13,14 These results suggest that G- Bcl-2 to promote pro-survival pathways.16,17 ERK has also protein- vsβ-arrestin-dependent signaling promote distinct been shown to phosphorylate and inactivate pro-apoptotic ERK signaling events downstream of the AT1AR. proteins such as caspase 918 and Bim,19 while also enhancing cellular proliferation by activation of proteins involved in Beta-Arrestin-Mediated Activation of nucleic acid synthesis,20 transcription,21,22 and translation.23 The β1AR can activate the EGFR to signal through EGFR Initiates ERK Signaling ERK pathways in aβ-arrestin-dependent manner through a Mitogenic ERK signaling activated by GPCRs has in- process known as “transactivation” (Fig2).9 Transactivation

Circulation Journal Vol.72, November 2008 Beta-Arrestin Signaling 1727

Fig3. Theβ-arrestin-mediated transactivation of the epidermal growth factor receptor is cardioprotective. Comparisons of (A) left ventricular function by echocardiography, (B) fractional shortening, and (C) TUNEL staining in non-transgenic (NTG) vs transgenic mice stably expressing the wild-typeβ-1-adrenergic receptor (WTβ1AR TG) or G-protein coupled receptor kinase-β-1-adrenergic receptor (GRK-β1AR TG, aβ1AR mutant that is unable to signal throughβ-arrestin-medi- ated pathways). GRK-β1AR mice have (A) greatly enlarged ventricle (enlarged black space), (B) decreased fractional shortening, and (C) increased apoptosis (arrowheads) vs NTG and WTβ1AR TG mice after chronic exposure to isopro- terenol (ISO). Adapted with permission from Clin Invest 2007; 117: 2445–2458.

Fig4. Theβ-1-adrenergic receptor (β1AR) mediated transactivation of the epidermal growth factor receptor (EGFR). Stimulation of theβ1AR activates signaling through 2 independent pathways, Gs-mediated pathways and newly discovered β-arrestin-mediated pathways. Gs activates adenylyl cyclase (AC) to generate the second messenger cAMP to promote downstream signaling. Gs-signaling promotes inotropy in the short term, but with chronic catecholamine stimulation pro- motes harmful cardiac remodeling including myocyte apoptosis, increased ventricular dilatation, and decreased fractional shortening. Following recruitment ofβ-arrestin to GRK5/6-phosphorylated receptors, theβ1AR initiates a second wave of cardioprotectiveβ-arrestin- mediated signaling via “transactivation” of the EGFR. Transactivation involves Src-dependent matrix-metalloproteinase (MMP)-mediated shedding of Heparin-Binding epidermal growth factor (HB-EGF) into the extracellular space, where HB-EGF acts as a ligand for the EGFR. PKA, cAMP-dependent protein kinase A; GRK, G-pro- tein coupled receptor kinase; ERK, extracellular regulated kinase; MAPK, mitogen-activated protein kinase; P, phospho- rylation site. Adapted with permission from J Clin Invest 2007; 117: 2396–2398.

of the EGFR following catecholamine stimulation of the by activation and internalization of EGFR and phosphoryla- β1AR involves Src-dependent matrix-metalloproteinase- tion of ERK.9 Although molecular signaling events in the mediated shedding of heparin-binding epidermal growth ERK signaling pathway have been extensively investigated, factor (HB-EGF). HB-EGF in turn acts as a ligand for the exciting new studies reveal the in vivo importance ofβ-ar- EGFR to initiate mitogenic signaling pathways, as evidenced restin-dependent ERK signaling in the heart.9

Circulation Journal Vol.72, November 2008 1728 PATEL PA et al.

Beta-Arrestin Dependent Signaling gest that identification of β-arrestin-biased ARBs and β- Confers Cardioprotection blockers may provide new therapeutic opportunities in the treatment of cardiac disease. Recent data indicates that β-arrestin-dependent, G-pro- tein-independent activation of the EGFR by the β1AR confers cardioprotection in mice chronically stimulated To the Future with catecholamine.9 Theβ1AR is a Gs-coupled receptor that Although the discovery of distinctβ-arrestin-dependent, regulates cardiac inotropy and chronotropy via activation G-protein-independent signaling is indeed exciting, all the of cAMP-mediated protein kinase A (PKA).24,25 Long-term mechanisms underlyingβ-arrestin-mediated signaling have chronic catecholamine stimulation of theβ1AR, as seen in yet to be fully appreciated. For instance, the 2 isoforms of heart failure, promotes receptor downregulation and harm- β-arrestin, β-arrestin1 and β-arrestin2, appear to have di- ful cardiac remodeling.26,27 vergent and sometimes opposing roles in signaling at a Interestingly, transgenic mice overexpressing mutant number of receptors, including the protease-activated β1ARs that are unable to recruitβ-arrestins show marked receptor (PAR) 1,34 PAR 2,35 parathyroid hormone receptor myocyte apoptosis and left ventricular dilatation under con- type 1,36 and the RTK toll-like receptor 4.37 Also, studies ditions of chronic catecholamine stress (Fig3).9 Moreover, with the β2AR indicate that β-arrestin dimerization is wild-type mice chronically stimulated with catecholamines needed forβ-arrestin-dependent ERK signaling, suggesting show the same deterioration in cardiac function when pre- that interactions between individualβ-arrestins themselves treated with the selective EGFR inhibitor, erlotinib.9 Those are necessary to initiateβ-arrestin-dependent signaling.38,39 results indicate that β-arrestin-mediated signaling by the Interestingly, proteomic analysis of β-arrestin1 and β- β1AR is cardioprotective under conditions of chronic cate- arrestin2 show that they may directly interact with as many cholamine stress, and further, that the mechanism of this as 337 proteins from various protein families,40 which sug- cardioprotection involvesβ-arrestin-dependent transactiva- gests thatβ-arrestins are likely players in a complex web of tion of the EGFR by theβ1AR (Fig4). protein–protein interactions and cellular signaling events. Similarβ-arrestin-mediated cardioprotection may exist at Studies revealing the cardioprotective function of β- the AT1AR, which also activates ERK in aβ-arrestin-depen- arrestin provide the most clinically applicable information dent manner. Indeed, transgenic mice with cardiac-specific about it to date. Understanding the mechanism of cardiopro- overexpression of AT1AR that are unable to couple with G tection conferred by β-arrestin-dependent, G-protein-inde- protein show less myocardial apoptosis and fibrosis, and pendent signaling will help to enhance our understanding enhanced cardiac hypertrophy, after chronic stimulation of chronic cardiac disease and aid in the development of with angiotensin II,28 suggesting that β-arrestin-dependent novel therapies for cardiac disease. ERK signaling by the AT1AR may promote these cardio- protective effects. References 1. Lefkowitz RJ, Shenoy SK. Transduction of receptor signals by beta- Super Receptor Blockers: Developing arrestins. Science 2005; 308: 512–517. a Novel Class of Cardiac Drugs 2. Freedman NJ, Liggett SB, Drachmann DE, Pei G, Caron MG, Lefkowitz RJ. Phosphorylation and desensitization of the human Studies with the AT1AR and theβ1AR suggest that classi- beta 1-adrenergic receptor: Involvement of G protein-coupled recep- cal G protein signaling is detrimental in heart failure, where- tor kinases and cAMP-dependent protein kinase. J Biol Chem 1995; as β-arrestin-dependent, G-protein-independent signaling 270: 17953–17961. by these receptors is cardioprotective.9,28 Because chronic 3. Moore CA, Milano SK, Benovic JL. Regulation of receptor traffick- ing by GRKs and arrestins. Annu Rev Physiol 2007; 69: 451–482. stimulation of theβ1AR and AT1AR, as seen in heart failure, 4. Rajagopal K, Lefkowitz RJ, Rockman HA. When 7 transmembrane has been shown to be harmful, many of the best therapies for receptors are not G protein-coupled receptors. J Clin Invest 2005; heart disease block signaling at these receptors. Although 115: 2971–2974. most of the angiotensin-receptor blockers (ARB) and β- 5. Nelson CD, Perry SJ, Regier DS, Prescott SM, Topham MK, Lefkowitz RJ. Targeting of diacylglycerol degradation to M1 adrenergic receptor blockers (β-blockers) in use today block muscarinic receptors by beta-arrestins. Science 2007; 315: 663–666. both G-protein- andβ-arrestin-mediated signaling at their 6. Perry SJ, Baillie GS, Kohout TA, McPhee I, Magiera MM, Ang KL, respective receptors, these new data suggest that a novel et al. Targeting of cyclic AMP degradation to beta 2-adrenergic re- class of receptor blockers can be identified, termed “biased ceptors by beta-arrestins. Science 2002; 298: 834–836. 7. McKay MM, Morrison DK. Integrating signals from RTKs to ligands”, which preferentially exploit cardioprotective β- ERK/MAPK. Oncogene 2007; 26: 3113–3121. arrestin signaling while simultaneously inhibiting harmful 8. Wei H, Ahn S, Shenoy SK, Karnik SS, Hunyady L, Luttrell LM, et G protein signaling. Such a ligand would be expected to al. Independent beta-arrestin 2 and G protein-mediated pathways for prevent or even reverse the harmful cardiac remodeling angiotensin II activation of extracellular signal-regulated kinases 1 caused by excess circulating levels of catecholamines and and 2. Proc Natl Acad Sci USA 2003; 100: 10782–10787. 1 9. Noma T, Lemaire A, Naga Prasad SV, Barki-Harrington L, Tilley angiotensin II in heart failure. DG, Chen J, et al. Beta-arrestin-mediated beta(1)-adrenergic receptor In fact, 2 such biased ligands have been described in the transactivation of the EGFR confers cardioprotection. J Clin Invest literature. The AT1AR ligand [Sar1, Ile4, Ile8]-ANG (SII) 2007; 117: 2445–2458. selectively activates β-arrestin mediated signaling while 10. Shenoy SK, Drake MT, Nelson CD, Houtz DA, Xiao K, Madabushi S, et al. Beta-arrestin-dependent, G protein-independent ERK1/2 simultaneously inhibiting G protein-mediated signaling at activation by the beta2 adrenergic receptor. J Biol Chem 2006; 281: the AT1AR to promote the beneficial effects of positive 1261–1273. lusitropy and inotropy in cardiac myocytes.29 Even more re- 11. Charest PG, Oligny-Longpre G, Bonin H, Azzi M, Bouvier M. The cently, it was shown that carvedilol, 1 of only 3β-blockers V2 vasopressin receptor stimulates ERK1/2 activity independently 30–32 of heterotrimeric G protein signalling. Cell Signal 2007; 19: 32–41. known to improve mortality in heart failure, weakly 12. Gesty-Palmer D, Chen M, Reiter E, Ahn S, Nelson CD, Wang S, et activates β-arrestin signaling pathways while blocking G al. Distinct beta-arrestin- and G protein-dependent pathways for protein signaling at theβ2AR.33 Collectively, these data sug- parathyroid hormone receptor-stimulated ERK1/2 activation. J Biol

Circulation Journal Vol.72, November 2008 Beta-Arrestin Signaling 1729

Chem 2006; 281: 10856–10864. alpha q/G alpha i coupling causes hypertrophy and bradycardia in 13. Ahn S, Shenoy SK, Wei H, Lefkowitz RJ. Differential kinetic and transgenic mice. J Clin Invest 2005; 115: 3045–3056. spatial patterns of beta-arrestin and G protein-mediated ERK activa- 29. Rajagopal K, Whalen EJ, Violin JD, Stiber JA, Rosenberg PB, tion by the angiotensin II receptor. J Biol Chem 2004; 279: 35518– Premont RT, et al. Beta-arrestin2-mediated inotropic effects of the 35525. angiotensin II type 1A receptor in isolated cardiac myocytes. Proc 14. Tohgo A, Choy EW, Gesty-Palmer D, Pierce KL, Laporte S, Oakley Natl Acad Sci USA 2006; 103: 16284–16289. RH, et al. The stability of the G protein-coupled receptor-beta-arrestin 30. Hjalmarson A, Goldstein S, Fagerberg B, Wedel H, Waagstein F, interaction determines the mechanism and functional consequence of Kjekshus J, et al. Effects of controlled-release metoprolol on total ERK activation. J Biol Chem 2003; 278: 6258–6267. mortality, hospitalizations, and well-being in patients with heart fail- 15. Jarpe MB, Widmann C, Knall C, Schlesinger TK, Gibson S, Yujiri T, ure: The Metoprolol CR/XL Randomized Intervention Trial in con- et al. Anti-apoptotic versus pro-apoptotic signal transduction: Check- gestive heart failure (MERIT-HF). MERIT-HF Study Group. JAMA points and stop signs along the road to death. Oncogene 1998; 17(11 2000; 283: 1295–1302. Reviews): 1475–1482. 31. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II). A ran- 16. Zha J, Harada H, Yang E, Jockel J, Korsmeyer SJ. Serine phosphory- domised trial. Lancet 1999; 353: 9–13. lation of death agonist BAD in response to survival factor results in 32. Packer M, Fowler MB, Roecker EB, Coats AJ, Catus HA, Krum H, et binding to 14-3-3 not BCL-X(L). Cell 1996; 87: 619–628. al. Effect of carvedilol on the morbidity of patients with severe 17. McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, chronic heart failure: Results of the carvedilol prospective randomized Chang F, et al. Roles of the Raf/MEK/ERK pathway in cell growth, cumulative survival (COPERNICUS) study. Circulation 2002; 106: malignant transformation and drug resistance. Biochim Biophys Acta 2194–2199. 2007; 1773: 1263–1284. 33. Wisler JW, DeWire SM, Whalen EJ, Violin JD, Drake MT, Ahn S, et 18. Allan LA, Morrice N, Brady S, Magee G, Pathak S, Clarke PR. In- al. A unique mechanism of beta-blocker action: Carvedilol stimulates hibition of caspase-9 through phosphorylation at Thr 125 by ERK beta-arrestin signaling. Proc Natl Acad Sci USA 2007; 104: 16657– MAPK. Nat Cell Biol 2003; 5: 647–654. 16662. 19. Luciano F, Jacquel A, Colosetti P, Herrant M, Cagnol S, Pages G, et 34. Kuo FT, Lu TL, Fu HW. Opposing effects of beta-arrestin1 and beta- al. Phosphorylation of Bim-EL by Erk1/2 on serine 69 promotes its arrestin2 on activation and degradation of Src induced by protease- degradation via the proteasome pathway and regulates its proapoptotic activated receptor 1. Cell Signal 2006; 18: 1914–1923. function. Oncogene 2003; 22: 6785–6793. 35. Kumar P, Lau CS, Mathur M, Wang P, DeFea KA. Differential effects 20. Graves LM, Guy HI, Kozlowski P, Huang M, Lazarowski E, Pope of beta-arrestins on the internalization, desensitization and ERK1/2 RM, et al. Regulation of carbamoyl phosphate synthetase by MAP activation downstream of protease activated receptor-2. Am J Physiol kinase. Nature 2000; 403: 328–332. Cell Physiol 2007; 293: C346–C357. 21. Stefanovsky V, Langlois F, Gagnon-Kubler T, Rothblum LI, Moss T. 36. Sneddon WB, Friedman PA. Beta-arrestin-dependent parathyroid hor- Growth factor signaling regulates elongation of RNA polymerase I mone-stimulated extracellular signal-regulated kinase activation and transcription in mammals via UBF phosphorylation and r-chromatin parathyroid hormone type 1 receptor internalization. Endocrinology remodeling. Mol Cell 2006; 21: 629–639. 2007; 148: 4073–4079. 22. Stefanovsky VY, Langlois F, Bazzett-Jones D, Pelletier G, Moss T. 37. Fan H, Luttrell LM, Tempel GE, Senn JJ, Halushka PV, Cook JA, et ERK modulates DNA bending and enhancesome structure by phos- al. Beta-arrestins 1 and 2 differentially regulate LPS-induced signal- phorylating HMG1-boxes 1 and 2 of the RNA polymerase I transcrip- ing and pro-inflammatory expression. Mol Immunol 2007; 44: tion factor UBF. Biochemistry 2006; 45: 3626–3634. 3092–3099. 23. Waskiewicz AJ, Flynn A, Proud CG, Cooper JA. Mitogen-activated 38. Xu TR, Baillie GS, Bhari N, Houslay TM, Pitt AM, Adams DR, et al. protein kinases activate the serine/threonine kinases Mnk1 and Mnk2. Mutations of beta-arrestin 2 that limit self-association also interfere EMBO J 1997; 16: 1909–1920. with interactions with the beta2-adrenoceptor and the ERK1/2 24. Brodde OE, Michel MC. Adrenergic and muscarinic receptors in the MAPKs: Implications for beta2-adrenoceptor signalling via the human heart. Pharmacol Rev 1999; 51: 651–690. ERK1/2 MAPKs. Biochem J 2008; 413: 51–60. 25. Rockman HA, Koch WJ, Lefkowitz RJ. Seven-transmembrane-span- 39. DeFea KA. Beta-arrestin multimers: Does a crowd help or hinder ning receptors and heart function. Nature 2002; 415: 206–212. function? Biochem J 2008; 413: e1–e3. 26. Ungerer M, Bohm M, Elce JS, Erdmann E, Lohse MJ. Altered expres- 40. Xiao K, McClatchy DB, Shukla AK, Zhao Y, Chen M, Shenoy SK, sion of beta-adrenergic receptor kinase and beta 1-adrenergic recep- et al. Functional specialization of beta-arrestin interactions revealed tors in the failing human heart. Circulation 1993; 87: 454–463. by proteomic analysis. Proc Natl Acad Sci USA 2007; 104: 12011– 27. Xiang Y, Kobilka BK. Myocyte adrenoceptor signaling pathways. 12016. Science 2003; 300: 1530–1532. 41. Engelhardt S. Alternative signaling: Cardiomyocyte beta1-adrenergic 28. Zhai P, Yamamoto M, Galeotti J, Liu J, Masurekar M, Thaisz J, et al. receptors signal through EGFRs. J Clin Invest 2007; 117: 2396– Cardiac-specific overexpression of AT1 receptor mutant lacking G 2398.

Circulation Journal Vol.72, November 2008