Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Teaching old receptors new tricks: biasing seven-transmembrane receptors

Key Points

  • Seven-transmembrane receptors (7TMRs), also known as G protein-couple receptors, are the largest class of transmembrane receptors and a common target for therapeutics. Initially thought to signal only through heterotrimeric G proteins, it is now recognized that they also signal through the multifunctional adapter proteins β-arrestin 1 and β-arrestin 2.

  • β-arrestin-mediated signalling can have distinct functional consequences from G protein-mediated signalling. G protein-mediated signalling is usually accomplished via the generation of second messengers that signal to downstream partners. By contrast, β-arrestin-mediated signalling usually results in the formation of signalling complexes scaffolded by β-arrestins that lead to activation of kinases and other downstream targets.

  • Biased agonists are capable of signalling through only a restricted subset of all of the pathways usually available to the receptor. β-arrestin-biased agonists act as agonists for β-arrestin-mediated signalling and as weak partial agonists or antagonists of G protein-mediated signalling.

  • In many systems, receptor activation by β-arrestin-biased agonists results in functionally distinct responses from activation by non-biased agonists. For example, a β-arrestin-biased agonist of the parathyroid hormone receptor is capable of stimulating trabecular bone growth in mice without increasing bone resorption, whereas an unbiased full agonist stimulates both bone growth and resorption.

  • Cell-based assays used to facilitate the discovery of β-arrestin-biased agonists may be based on redistribution of labelled receptor or β-arrestin; proximity between receptor and β-arrestin; conformation of receptor or β-arrestin; or activation of downstream signalling pathways known to be regulated selectively by β-arrestins. Most of these assays have been adapted for high-throughput screening.

  • As we learn more about β-arrestin-mediated signalling at other 7TMRs, new targets for the development of β-arrestin-biased agonists will undoubtedly arise. Thus, it is important that we continue to focus on studying the physiological impact of β-arrestin-mediated signaling in cell-based and animal studies.

Abstract

Seven-transmembrane receptors (7TMRs; also known as G protein-coupled receptors) are the largest class of receptors in the human genome and are common targets for therapeutics. Originally identified as mediators of 7TMR desensitization, β-arrestins (arrestin 2 and arrestin 3) are now recognized as true adaptor proteins that transduce signals to multiple effector pathways. Signalling that is mediated by β-arrestins has distinct biochemical and functional consequences from those mediated by G proteins, and several biased ligands and receptors have been identified that preferentially signal through either G protein- or β-arrestin-mediated pathways. These ligands are not only useful tools for investigating the biochemistry of 7TMR signalling, they also have the potential to be developed into new classes of therapeutics.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: G protein and β-arrestin-mediated signalling.
Figure 2: Biased ligands and biased receptors.
Figure 3: Quantifying the pluridimensional efficacies of seven-transmembrane receptors.
Figure 4: Assays for β-arrestin recruitment and activation.

Similar content being viewed by others

References

  1. Lagerstrom, M. C. & Schioth, H. B. Structural diversity of G protein-coupled receptors and significance for drug discovery. Nature Rev. Drug Discov. 7, 339–357 (2008).

    Google Scholar 

  2. Ma, P. & Zemmel, R. Value of novelty? Nature Rev. Drug Discov. 1, 571–572 (2002).

    CAS  Google Scholar 

  3. Lefkowitz, R. J. Historical review: a brief history and personal retrospective of seven-transmembrane receptors. Trends Pharmacol. Sci. 25, 413–422 (2004).

    CAS  PubMed  Google Scholar 

  4. Lefkowitz, R. J. & Shenoy, S. K. Transduction of receptor signals by β-arrestins. Science 308, 512–517 (2005).

    CAS  PubMed  Google Scholar 

  5. DeWire, S. M., Ahn, S., Lefkowitz, R. J. & Shenoy, S. K. β-arrestins and cell signaling. Annu. Rev. Physiol. 69, 483–510 (2007).

    CAS  PubMed  Google Scholar 

  6. Benovic, J. L., Staniszewski, C., Mayor, F. Jr, Caron, M. G. & Lefkowitz, R. J. β-adrenergic receptor kinase. Activity of partial agonists for stimulation of adenylate cyclase correlates with ability to promote receptor phosphorylation. J. Biol. Chem. 263, 3893–3897 (1988).

    CAS  PubMed  Google Scholar 

  7. Violin, J. D. & Lefkowitz, R. J. β-arrestin-biased ligands at seven-transmembrane receptors. Trends Pharmacol. Sci. 28, 416–422 (2007).

    CAS  PubMed  Google Scholar 

  8. Kenakin, T. Collateral efficacy in drug discovery: taking advantage of the good (allosteric) nature of 7TM receptors. Trends Pharmacol. Sci. 28, 407–415 (2007).

    CAS  PubMed  Google Scholar 

  9. Lefkowitz, R. J. & Whalen, E. J. β-arrestins: traffic cops of cell signaling. Curr. Opin. Cell Biol. 16, 162–168 (2004).

    CAS  PubMed  Google Scholar 

  10. Wilden, U. & Kuhn, H. Light-dependent phosphorylation of rhodopsin: number of phosphorylation sites. Biochemistry 21, 3014–3022 (1982).

    CAS  PubMed  Google Scholar 

  11. Shichi, H. & Somers, R. L. Light-dependent phosphorylation of rhodopsin. Purification and properties of rhodopsin kinase. J. Biol. Chem. 253, 7040–7046 (1978).

    CAS  PubMed  Google Scholar 

  12. Benovic, J. L., DeBlasi, A., Stone, W. C., Caron, M. G. & Lefkowitz, R. J. β-adrenergic receptor kinase: primary structure delineates a multigene family. Science 246, 235–240 (1989).

    CAS  PubMed  Google Scholar 

  13. Wilden, U., Hall, S. W. & Kuhn, H. Phosphodiesterase activation by photoexcited rhodopsin is quenched when rhodopsin is phosphorylated and binds the intrinsic 48-kDa protein of rod outer segments. Proc. Natl Acad. Sci. USA 83, 1174–1178 (1986).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Stadel, J. M. et al. Catecholamine-induced desensitization of turkey erythrocyte adenylate cyclase is associated with phosphorylation of the β-adrenergic receptor. Proc. Natl Acad. Sci. USA 80, 3173–3177 (1983).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Benovic, J. L. et al. Functional desensitization of the isolated β-adrenergic receptor by the β-adrenergic receptor kinase: potential role of an analog of the retinal protein arrestin (48-kDa protein). Proc. Natl Acad. Sci. USA 84, 8879–8882 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Luttrell, L. M. et al. β-arrestin-dependent formation of β2 adrenergic receptor–Src protein kinase complexes. Science 283, 655–661 (1999).

    CAS  PubMed  Google Scholar 

  17. DeFea, K. A. et al. The proliferative and antiapoptotic effects of substance P are facilitated by formation of a β-arrestin-dependent scaffolding complex. Proc. Natl Acad. Sci. USA 97, 11086–11091 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. DeFea, K. A. et al. β-arrestin-dependent endocytosis of proteinase-activated receptor 2 is required for intracellular targeting of activated ERK1/2. J. Cell Biol. 148, 1267–1281 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Luttrell, L. M. et al. Activation and targeting of extracellular signal-regulated kinases by β-arrestin scaffolds. Proc. Natl Acad. Sci. USA 98, 2449–2454 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Tohgo, A., Pierce, K. L., Choy, E. W., Lefkowitz, R. J. & Luttrell, L. M. β-arrestin scaffolding of the ERK cascade enhances cytosolic ERK activity but inhibits ERK-mediated transcription following angiotensin AT1a receptor stimulation. J. Biol. Chem. 277, 9429–9436 (2002).

    CAS  PubMed  Google Scholar 

  21. Tohgo, A. et al. The stability of the G protein-coupled receptor–β-arrestin interaction determines the mechanism and functional consequence of ERK activation. J. Biol. Chem. 278, 6258–6267 (2003).

    CAS  PubMed  Google Scholar 

  22. Ahn, S., Shenoy, S. K., Wei, H. & Lefkowitz, R. J. Differential kinetic and spatial patterns of β-arrestin and G protein-mediated ERK activation by the angiotensin II receptor. J. Biol. Chem. 279, 35518–35525 (2004). This work demonstrates the different temporal and spatial patterns of β-arrestin- and G protein-mediated ERK. β-arrestin-mediated phosphorylated ERK peaks at late times in endosomes and G protein-mediated phosphorylated ERK peaks at early times with nuclear and cytoplasmic localization.

    CAS  PubMed  Google Scholar 

  23. Ge, L., Ly, Y., Hollenberg, M. & DeFea, K. A β-arrestin-dependent scaffold is associated with prolonged MAPK activation in pseudopodia during protease-activated receptor-2-induced chemotaxis. J. Biol. Chem. 278, 34418–34426 (2003).

    CAS  PubMed  Google Scholar 

  24. Hunton, D. L. et al. β-arrestin 2-dependent angiotensin II type 1A receptor-mediated pathway of chemotaxis. Mol. Pharmacol. 67, 1229–1236 (2005).

    CAS  PubMed  Google Scholar 

  25. Povsic, T. J., Kohout, T. A. & Lefkowitz, R. J. β-arrestin1 mediates insulin-like growth factor 1 (IGF-1) activation of phosphatidylinositol 3-kinase (PI3K) and anti-apoptosis. J. Biol. Chem. 278, 51334–51339 (2003).

    CAS  PubMed  Google Scholar 

  26. Goel, R., Phillips-Mason, P. J., Raben, D. M. & Baldassare, J. J. α-Thrombin induces rapid and sustained Akt phosphorylation by β-arrestin1-dependent and -independent mechanisms, and only the sustained Akt phosphorylation is essential for G1 phase progression. J. Biol. Chem. 277, 18640–18648 (2002).

    CAS  PubMed  Google Scholar 

  27. Ahn, S., Kim, J., Hara, M. R., Ren, X. R. & Lefkowitz, R. J. β-arrestin-2 mediates anti-apoptotic signaling through regulation of BAD phosphorylation. J. Biol. Chem. 284, 8855–8865 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Luan, B. et al. Deficiency of a β-arrestin-2 signal complex contributes to insulin resistance. Nature 457, 1146–1149 (2009).

    CAS  PubMed  Google Scholar 

  29. Beaulieu, J. M. et al. An Akt/β-arrestin 2/PP2A signaling complex mediates dopaminergic neurotransmission and behavior. Cell 122, 261–273 (2005).

    CAS  PubMed  Google Scholar 

  30. Xiao, K. et al. Functional specialization of β-arrestin interactions revealed by proteomic analysis. Proc. Natl Acad. Sci. USA 104, 12011–12016 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Ma, L. & Pei, G. β-arrestin signaling and regulation of transcription. J. Cell Sci. 120, 213–218 (2007).

    CAS  PubMed  Google Scholar 

  32. Neuhaus, E. M., Mashukova, A., Barbour, J., Wolters, D. & Hatt, H. Novel function of β-arrestin2 in the nucleus of mature spermatozoa. J. Cell Sci. 119, 3047–3056 (2006).

    CAS  PubMed  Google Scholar 

  33. Luan, B., Zhang, Z., Wu, Y., Kang, J. & Pei, G. β-arrestin2 functions as a phosphorylation-regulated suppressor of UV-induced NF-κB activation. EMBO J. 24, 4237–4246 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Witherow, D. S., Garrison, T. R., Miller, W. E. & Lefkowitz, R. J. β-arrestin inhibits NF-κB activity by means of its interaction with the NF-κB inhibitor IκBα. Proc. Natl Acad. Sci. USA 101, 8603–8607 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Piu, F., Gauthier, N. K. & Wang, F. β-arrestin 2 modulates the activity of nuclear receptor RAR β2 through activation of ERK2 kinase. Oncogene 25, 218–229 (2006).

    CAS  PubMed  Google Scholar 

  36. Bryja, V., Gradl, D., Schambony, A., Arenas, E. & Schulte, G. β-arrestin is a necessary component of Wnt/β-catenin signaling in vitro and in vivo. Proc. Natl Acad. Sci. USA 104, 6690–6695 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Rosano, L. et al. β-arrestin links endothelin A receptor to β-catenin signaling to induce ovarian cancer cell invasion and metastasis. Proc. Natl Acad. Sci. USA 106, 2806–2811 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Kang, J. et al. A nuclear function of β-arrestin1 in GPCR signaling: regulation of histone acetylation and gene transcription. Cell 123, 833–847 (2005).

    CAS  PubMed  Google Scholar 

  39. Mo, W. et al. Nuclear β-arrestin1 functions as a scaffold for the dephosphorylation of STAT1 and moderates the antiviral activity of IFN-γ. Mol. Cell 31, 695–707 (2008).

    CAS  PubMed  Google Scholar 

  40. Bhola, N. E. & Grandis, J. R. Crosstalk between G-protein-coupled receptors and epidermal growth factor receptor in cancer. Front. Biosci. 13, 1857–1865 (2008).

    CAS  PubMed  Google Scholar 

  41. Buchanan, F. G. et al. Role of β-arrestin 1 in the metastatic progression of colorectal cancer. Proc. Natl Acad. Sci. USA 103, 1492–1497 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Noma, T. et al. β-arrestin-mediated β1-adrenergic receptor transactivation of the EGFR confers cardioprotection. J. Clin. Invest. 117, 2445–2458 (2007). This paper suggests an important role for β-arrestin-mediated signalling in the heart. The β-arrestin-mediated pathway downstream of the β 1 -adrenergic receptor protects against cardiomyopathy induced by catecholamine infusion.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Kim, J., Ahn, S., Rajagopal, K. & Lefkowitz, R. J. Independent β-arrestin2 and Gq/protein kinase Cζ pathways for ERK stimulated by angiotensin type 1A receptors in vascular smooth muscle cells converge on transactivation of the epidermal growth factor receptor. J. Biol. Chem. 284, 11953–11962 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Kovacs, J. J. et al. β-arrestin-mediated localization of smoothened to the primary cilium. Science 320, 1777–1781 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Barnes, W. G. et al. β-arrestin 1 and Gαq/11 coordinately activate RhoA and stress fiber formation following receptor stimulation. J. Biol. Chem. 280, 8041–8050 (2005).

    CAS  PubMed  Google Scholar 

  46. Galandrin, S. & Bouvier, M. Distinct signaling profiles of β1 and β2 adrenergic receptor ligands toward adenylyl cyclase and mitogen-activated protein kinase reveals the pluridimensionality of efficacy. Mol. Pharmacol. 70, 1575–1584 (2006). In this work, the authors used a set of ligands (agonists, antagonists and inverse agonists) for the adrenergic receptors to identify ligand-specific differences in the balance between cAMP and ERK signalling.

    CAS  PubMed  Google Scholar 

  47. Roth, B. L. in Functional Selectivity of G Protein-Coupled Receptor Ligands (ed. Neve, K.) 3–7 (Springer, New York, 2009).

    Google Scholar 

  48. Eason, M. G., Kurose, H., Holt, B. D., Raymond, J. R. & Liggett, S. B. Simultaneous coupling of α2-adrenergic receptors to two G-proteins with opposing effects. Subtype-selective coupling of α2C10, α2C4, and α2C2 adrenergic receptors to Gi and Gs . J. Biol. Chem. 267, 15795–15801 (1992).

    CAS  PubMed  Google Scholar 

  49. Fisher, A. et al. Selective signaling via unique M1 muscarinic agonists. Ann. NY Acad. Sci. 695, 300–303 (1993).

    CAS  PubMed  Google Scholar 

  50. Gurwitz, D. et al. Discrete activation of transduction pathways associated with acetylcholine m1 receptor by several muscarinic ligands. Eur. J. Pharmacol. 267, 21–31 (1994).

    CAS  PubMed  Google Scholar 

  51. Kenakin, T. Agonist-receptor efficacy. II. Agonist trafficking of receptor signals. Trends Pharmacol. Sci. 16, 232–238 (1995). One of the first papers to approach the question of biased agonism from a theoretical perspective.

    CAS  PubMed  Google Scholar 

  52. Wei, H. et al. Independent β-arrestin 2 and G protein-mediated pathways for angiotensin II activation of extracellular signal-regulated kinases 1 and 2. Proc. Natl Acad. Sci. USA 100, 10782–10787 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Holloway, A. C. et al. Side-chain substitutions within angiotensin II reveal different requirements for signaling, internalization, and phosphorylation of type 1A angiotensin receptors. Mol. Pharmacol. 61, 768–777 (2002).

    CAS  PubMed  Google Scholar 

  54. Gesty-Palmer, D. et al. Distinct β-arrestin- and G protein-dependent pathways for parathyroid hormone receptor-stimulated ERK1/2 activation. J. Biol. Chem. 281, 10856–10864 (2006).

    CAS  PubMed  Google Scholar 

  55. Gaborik, Z. et al. The role of a conserved region of the second intracellular loop in AT1 angiotensin receptor activation and signaling. Endocrinology 144, 2220–2228 (2003).

    CAS  PubMed  Google Scholar 

  56. Shenoy, S. K. et al. β-arrestin-dependent, G protein-independent ERK1/2 activation by the β2 adrenergic receptor. J. Biol. Chem. 281, 1261–1273 (2006).

    CAS  PubMed  Google Scholar 

  57. Drake, M. T. et al. β-arrestin-biased agonism at the β2-adrenergic receptor. J. Biol. Chem. 283, 5669–5676 (2008).

    CAS  PubMed  Google Scholar 

  58. Leach, K., Sexton, P. M. & Christopoulos, A. Allosteric GPCR modulators: taking advantage of permissive receptor pharmacology. Trends Pharmacol. Sci. 28, 382–389 (2007).

    CAS  PubMed  Google Scholar 

  59. Whalen, E. J. et al. Regulation of β-adrenergic receptor signaling by S-nitrosylation of G-protein-coupled receptor kinase 2. Cell 129, 511–522 (2007).

    CAS  PubMed  Google Scholar 

  60. Kohout, T. A. et al. Differential desensitization, receptor phosphorylation, β-arrestin recruitment, and ERK1/2 activation by the two endogenous ligands for the CC chemokine receptor 7. J. Biol. Chem. 279, 23214–23222 (2004).

    CAS  PubMed  Google Scholar 

  61. Zidar, D. A., Violin, J. D., Whalen, E. J. & Lefkowitz, R. J. Selective engagement of G protein coupled receptor kinases (GRKs) encodes distinct functions of biased ligands. Proc. Natl Acad. Sci. USA 106, 9649–9654 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Wisler, J. W. et al. A unique mechanism of beta-blocker action: carvedilol stimulates β-arrestin signaling. Proc. Natl Acad. Sci. USA 104, 16657–16662 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Kim, I. M. et al. Beta-blockers alprenolol and carvedilol stimulate β-arrestin-mediated EGFR transactivation. Proc. Natl Acad. Sci. USA 105, 14555–14560 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Rajagopal, K. et al. β-arrestin2-mediated inotropic effects of the angiotensin II type 1A receptor in isolated cardiac myocytes. Proc. Natl Acad. Sci. USA 103, 16284–16289 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhai, P. et al. Cardiac-specific overexpression of AT1 receptor mutant lacking Gαq/Gαi coupling causes hypertrophy and bradycardia in transgenic mice. J. Clin. Invest. 115, 3045–3056 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Groer, C. E. et al. An opioid agonist that does not induce μ-opioid receptor–arrestin interactions or receptor internalization. Mol. Pharmacol. 71, 549–557 (2007).

    CAS  PubMed  Google Scholar 

  67. Bohn, L. M. et al. Enhanced morphine analgesia in mice lacking β-arrestin 2. Science 286, 2495–2498 (1999). One of the first examples of a physiological response that was regulated by β-arrestins in an animal model.

    CAS  PubMed  Google Scholar 

  68. Bohn, L. M., Dykstra, L. A., Lefkowitz, R. J., Caron, M. G. & Barak, L. S. Relative opioid efficacy is determined by the complements of the G protein-coupled receptor desensitization machinery. Mol. Pharmacol. 66, 106–112 (2004).

    CAS  PubMed  Google Scholar 

  69. Raehal, K. M., Walker, J. K. & Bohn, L. M. Morphine side effects in β-arrestin 2 knockout mice. J. Pharmacol. Exp. Ther. 314, 1195–1201 (2005).

    CAS  PubMed  Google Scholar 

  70. Enjalbert, A. & Bockaert, J. Pharmacological characterization of the D2 dopamine receptor negatively coupled with adenylate cyclase in rat anterior pituitary. Mol. Pharmacol. 23, 576–584 (1983).

    CAS  PubMed  Google Scholar 

  71. Beaulieu, J. M., Gainetdinov, R. R. & Caron, M. G. Akt/GSK3 signaling in the action of psychotropic drugs. Annu. Rev. Pharmacol. Toxicol. 49, 327–347 (2009).

    CAS  PubMed  Google Scholar 

  72. Beaulieu, J. M. et al. A β-arrestin 2 signaling complex mediates lithium action on behavior. Cell 132, 125–136 (2008).

    CAS  PubMed  Google Scholar 

  73. Bohn, L. M. et al. Enhanced rewarding properties of morphine, but not cocaine, in β(arrestin)-2 knock-out mice. J. Neurosci. 23, 10265–10273 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Gainetdinov, R. R., Premont, R. T., Bohn, L. M., Lefkowitz, R. J. & Caron, M. G. Desensitization of G protein-coupled receptors and neuronal functions. Annu. Rev. Neurosci. 27, 107–144 (2004).

    CAS  PubMed  Google Scholar 

  75. Berg, K. A. & Clarke, W. P. in Functional Selectivity of G Protein-Coupled Receptor Ligands (ed. Neve, K.) 155–176 (Springer, New York, 2009).

    Google Scholar 

  76. Berg, K. A. et al. Effector pathway-dependent relative efficacy at serotonin type 2A and 2C receptors: evidence for agonist-directed trafficking of receptor stimulus. Mol. Pharmacol. 54, 94–104 (1998).

    CAS  PubMed  Google Scholar 

  77. Moya, P. R. et al. Functional selectivity of hallucinogenic phenethylamine and phenylisopropylamine derivatives at human 5-hydroxytryptamine (5-HT)2A and 5-HT2C receptors. J. Pharmacol. Exp. Ther. 321, 1054–1061 (2007).

    CAS  PubMed  Google Scholar 

  78. Gelber, E. I. et al. Structure and function of the third intracellular loop of the 5-hydroxytryptamine2A receptor: the third intracellular loop is α-helical and binds purified arrestins. J. Neurochem. 72, 2206–2214 (1999).

    CAS  PubMed  Google Scholar 

  79. Schmid, C. L., Raehal, K. M. & Bohn, L. M. Agonist-directed signaling of the serotonin 2A receptor depends on β-arrestin-2 interactions in vivo. Proc. Natl Acad. Sci. USA 105, 1079–1084 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Balabanian, K. et al. WHIM syndromes with different genetic anomalies are accounted for by impaired CXCR4 desensitization to CXCL12. Blood 105, 2449–2457 (2005).

    CAS  PubMed  Google Scholar 

  81. Gesty-Palmer, D. et al. A β-arrestin biased agonist of a parathyroid hormone receptor (PTH1R) promotes bone formation independent of G protein activation. Sci. Transl. Med. 1, 1ra1 (2009). One of the first studies to demonstrate the effects of a biased agonist in an animal model. Compared with the unbiased agonist PTH(1–34), which stimulates both bone formation and resorption, the biased agonist PTH-βarr stimulates bone formation only.

    PubMed  PubMed Central  Google Scholar 

  82. Bodor, E. T. & Offermanns, S. Nicotinic acid: an old drug with a promising future. Br. J. Pharmacol. 153 (Suppl. 1), 68–75 (2008).

    Google Scholar 

  83. Walters, R. W. et al. β-arrestin1 mediates nicotinic acid-induced flushing, but not its antilipolytic effect, in mice. J. Clin. Invest. 119, 1312–1321 (2009). An example in which development of a biased agonist could lead to a therapeutic with fewer side effects.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Semple, G. et al. 3-(1H-tetrazol-5-yl)-1,4,5,6-tetrahydro-cyclopentapyrazole (MK-0354): a partial agonist of the nicotinic acid receptor, G-protein coupled receptor 109a, with antilipolytic but no vasodilatory activity in mice. J. Med. Chem. 51, 5101–5108 (2008).

    CAS  PubMed  Google Scholar 

  85. Eglen, R. M., Bosse, R. & Reisine, T. Emerging concepts of guanine nucleotide-binding protein-coupled receptor (GPCR) function and implications for high throughput screening. Assay Drug Dev. Technol. 5, 425–451 (2007).

    CAS  PubMed  Google Scholar 

  86. Siehler, S. Cell-based assays in GPCR drug discovery. Biotechnol. J. 3, 471–483 (2008).

    CAS  PubMed  Google Scholar 

  87. Verkaar, F. et al. G protein-independent cell-based assays for drug discovery on seven-transmembrane receptors. Biotechnol. Annu. Rev. 14, 253–274 (2008). A review on current technologies used for high-throughput screening of 7TMRs with a focus on assays of β-arrestin activity.

    CAS  PubMed  Google Scholar 

  88. Hudson, C. C., Oakley, R. H., Sjaastad, M. D. & Loomis, C. R. High-content screening of known G protein-coupled receptors by arrestin translocation. Methods Enzymol. 414, 63–78 (2006).

    CAS  PubMed  Google Scholar 

  89. Henriksen, U., Fog, J., Loechel, F. & Praestegaard, M. Profiling of multiple signal pathway activities by multiplexing antibody and GFP-based translocation assays. Comb. Chem. High Throughput Screen. 11, 537–544 (2008).

    CAS  PubMed  Google Scholar 

  90. Hanyaloglu, A. C. & von Zastrow, M. Regulation of GPCRs by endocytic membrane trafficking and its potential implications. Annu. Rev. Pharmacol. Toxicol. 48, 537–568 (2008).

    CAS  PubMed  Google Scholar 

  91. Krasel, C., Bunemann, M., Lorenz, K. & Lohse, M. J. β-arrestin binding to the β2-adrenergic receptor requires both receptor phosphorylation and receptor activation. J. Biol. Chem. 280, 9528–9535 (2005).

    CAS  PubMed  Google Scholar 

  92. Bertrand, L. et al. The BRET2/arrestin assay in stable recombinant cells: a platform to screen for compounds that interact with G protein-coupled receptors (GPCRs). J. Recept. Signal. Transduct. Res. 22, 533–541 (2002).

    CAS  PubMed  Google Scholar 

  93. Olson, K. R. & Eglen, R. M. Beta galactosidase complementation: a cell-based luminescent assay platform for drug discovery. Assay Drug Dev. Technol. 5, 137–144 (2007).

    CAS  PubMed  Google Scholar 

  94. Luker, K. E., Gupta, M. & Luker, G. D. Imaging CXCR4 signaling with firefly luciferase complementation. Anal. Chem. 80, 5565–5573 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Barnea, G. et al. The genetic design of signaling cascades to record receptor activation. Proc. Natl Acad. Sci. USA 105, 64–69 (2008).

    CAS  PubMed  Google Scholar 

  96. Vilardaga, J. P., Bunemann, M., Krasel, C., Castro, M. & Lohse, M. J. Measurement of the millisecond activation switch of G protein-coupled receptors in living cells. Nature Biotech. 21, 807–812 (2003).

    CAS  Google Scholar 

  97. Rochais, F. et al. Real-time optical recording of β1-adrenergic receptor activation reveals supersensitivity of the Arg389 variant to carvedilol. J. Clin. Invest. 117, 229–235 (2007). A study of the β 1 -adrenergic receptor that monitored distinct structural responses of polymorphic receptor variants in response to treatment with a clinically used beta blocker.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Charest, P. G., Terrillon, S. & Bouvier, M. Monitoring agonist-promoted conformational changes of β-arrestin in living cells by intramolecular BRET. EMBO Rep. 6, 334–340 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Shukla, A. K. et al. Distinct conformational changes in β-arrestin report biased agonism at seven-transmembrane receptors. Proc. Natl Acad. Sci. USA 105, 9988–9993 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Mailman, R. B. GPCR functional selectivity has therapeutic impact. Trends Pharmacol. Sci. 28, 390–396 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Lohse, M. J., Benovic, J. L., Codina, J., Caron, M. G. & Lefkowitz, R. J. β-arrestin: a protein that regulates β-adrenergic receptor function. Science 248, 1547–1550 (1990).

    CAS  PubMed  Google Scholar 

  102. Perry, S. J. et al. Targeting of cyclic AMP degradation to β2-adrenergic receptors by β-arrestins. Science 298, 834–836 (2002).

    CAS  PubMed  Google Scholar 

  103. Ferguson, S. S. et al. Role of β-arrestin in mediating agonist-promoted G protein-coupled receptor internalization. Science 271, 363–366 (1996).

    CAS  PubMed  Google Scholar 

  104. Goodman, O. B. Jr et al. β-arrestin acts as a clathrin adaptor in endocytosis of the β2-adrenergic receptor. Nature 383, 447–450 (1996). One of the first papers to describe a role for β-arrestins as adapter proteins, in this case for clathrin, leading to receptor endocytosis.

    CAS  PubMed  Google Scholar 

  105. Barlic, J. et al. Regulation of tyrosine kinase activation and granule release through β-arrestin by CXCRI. Nature Immunol. 1, 227–233 (2000).

    CAS  Google Scholar 

  106. Imamura, T. et al. β-arrestin-mediated recruitment of the Src family kinase Yes mediates endothelin-1-stimulated glucose transport. J. Biol. Chem. 276, 43663–43667 (2001).

    CAS  PubMed  Google Scholar 

  107. Gao, H. et al. Identification of β-arrestin2 as a G protein-coupled receptor-stimulated regulator of NF-κB pathways. Mol. Cell 14, 303–317 (2004).

    CAS  PubMed  Google Scholar 

  108. Lagane, B. et al. CXCR4 dimerization and β-arrestin-mediated signaling account for the enhanced chemotaxis to CXCL12 in WHIM syndrome. Blood 112, 34–44 (2008).

    CAS  PubMed  Google Scholar 

  109. Alloway, P. G., Howard, L. & Dolph, P. J. The formation of stable rhodopsin–arrestin complexes induces apoptosis and photoreceptor cell degeneration. Neuron 28, 129–138 (2000).

    CAS  PubMed  Google Scholar 

  110. Kiselev, A. et al. A molecular pathway for light-dependent photoreceptor apoptosis in Drosophila. Neuron 28, 139–152 (2000).

    CAS  PubMed  Google Scholar 

  111. Revankar, C. M., Vines, C. M., Cimino, D. F. & Prossnitz, E. R. Arrestins block G protein-coupled receptor-mediated apoptosis. J. Biol. Chem. 279, 24578–24584 (2004).

    CAS  PubMed  Google Scholar 

  112. DeWire, S. M. et al. β-arrestin-mediated signaling regulates protein synthesis. J. Biol. Chem. 283, 10611–10620 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. De Lean, A., Stadel, J. M. & Lefkowitz, R. J. A ternary complex model explains the agonist-specific binding properties of the adenylate cyclase-coupled β-adrenergic receptor. J. Biol. Chem. 255, 7108–7117 (1980).

    CAS  PubMed  Google Scholar 

  114. Samama, P., Cotecchia, S., Costa, T. & Lefkowitz, R. J. A mutation-induced activated state of the β2-adrenergic receptor. Extending the ternary complex model. J. Biol. Chem. 268, 4625–4636 (1993).

    CAS  PubMed  Google Scholar 

  115. Weiss, J. M., Morgan, P. H., Lutz, M. W. & Kenakin, T. P. The cubic ternary complex receptor-occupancy model. III. resurrecting efficacy. J. Theor. Biol. 181, 381–397 (1996).

    CAS  PubMed  Google Scholar 

  116. Audet, N. et al. Bioluminescence resonance energy transfer assays reveal ligand-specific conformational changes within preformed signaling complexes containing δ-opioid receptors and heterotrimeric G proteins. J. Biol. Chem. 283, 15078–15088 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Vilardaga, J. P., Steinmeyer, R., Harms, G. S. & Lohse, M. J. Molecular basis of inverse agonism in a G protein-coupled receptor. Nature Chem. Biol. 1, 25–28 (2005).

    CAS  Google Scholar 

  118. Gurevich, V. V., Pals-Rylaarsdam, R., Benovic, J. L., Hosey, M. M. & Onorato, J. J. Agonist–receptor–arrestin, an alternative ternary complex with high agonist affinity. J. Biol. Chem. 272, 28849–28852 (1997).

    CAS  PubMed  Google Scholar 

  119. Schwartz, T. W. & Holst, B. Allosteric enhancers, allosteric agonists and ago-allosteric modulators: where do they bind and how do they act? Trends Pharmacol. Sci. 28, 366–373 (2007).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank E. Whalen, J. Violin, S. Ahn and A. Shukla for critical review of the manuscript. We thank D. Addison and E. Hall for secretarial assistance. This work was supported in part by National Institutes of Health (NIH) Grants HL16037 and HL70631 to R.J.L. R.J.L. is an Investigator with the Howard Hughes Medical Institute. S.R. is supported by NIH T32 training grant HL07101-34.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Robert J. Lefkowitz.

Ethics declarations

Competing interests

Robert J. Lefkowitz is a co-founder of Trevena, a company that is developing drugs to target seven transmembrane receptors.

Related links

Related links

DATABASES

IUPHAR Databse of Receptors and Ion Channels 

5-HT2A

5HT-2C

AT1A

β1-adrenergic

β2-adrenergic

CXCR4

D2

δ-opioid

EP4

GPR109A

κ-opioid

NK1

PAR2

PTH1

FURTHER INFORMATION

Laboaratory homepage of Robert J. Lefkowitz

Glossary

G protein

A heterotrimeric protein that exchanges GDP for GTP in its α-subunit on agonist binding to a seven-transmembrane receptor, resulting in dissociation of the complex into Gα and Gβγ subunits. These subunits lead to the activation of second messenger systems through the regulation of enzymes such as adenylyl cyclase or phospholipase C.

Arrestins

Multifunctional adaptor proteins that are important in regulating desensitization and signalling by seven-transmembrane receptors and other transmembrane receptors.

β-catenin

A multifunctional adaptor protein. One of its roles includes the regulation of TCF/LEF transcription factors in response to signalling by Wnts through the frizzled seven-transmembrane receptors.

Transactivation

The activation of a transmembrane receptor that results from signalling caused by activation of another receptor. In the case of epidermal growth factor receptor (EGFR) transactivation, agonist binding at a number of seven-transmembrane receptors activates a pathway that releases a membrane-bound EGF ligand by proteolytic cleavage, which then activates the EGFR.

Biased agonist

A ligand that results in the activation of select, but not all, available signalling pathways that are known to be activated by the receptor.

Positive inotropes

An agent that increases the force of the heart's contraction. Negative inotropes decrease the force of contraction.

Antagonist

A term that is commonly used to refer broadly to neutral antagonists, weak partial agonists and inverse agonists.

Partial agonist

A ligand that when bound to a receptor results in a submaximal response. Partial agonists can antagonize full agonists.

Inverse agonist

A ligand that decreases the signalling activity of the receptor on binding compared with the ligand-unbound state.

Neutral antagonist

A ligand that results in no change in activity of the receptor on binding compared with the ligand-unbound state.

Allosteric site

A binding site on a seven-transmembrane receptor that is different than the orthosteric site.

Orthosteric site

The binding site on a seven-transmembrane receptor to which the endogenous agonist binds.

Allosteric modulator

A ligand that binds to the allosteric site of the receptor and affects receptor responses to orthosteric ligands. Some allosteric modulators are capable of generating biased responses.

Lusitropic

Relates to the relaxation and filling of the heart. Positive lusitropic agents improve the heart's relaxation and filling.

Full agonist

A ligand that completely activates the receptor on binding.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Rajagopal, S., Rajagopal, K. & Lefkowitz, R. Teaching old receptors new tricks: biasing seven-transmembrane receptors. Nat Rev Drug Discov 9, 373–386 (2010). https://doi.org/10.1038/nrd3024

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd3024

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing