Endomorphin-2: A Biased Agonist at the μ-Opioid Receptor

Previously we correlated the efficacy for G protein activation with that for arrestin recruitment for a number of agonists at the μ-opioid receptor (MOPr) stably expressed in HEK293 cells. We suggested that the endomorphins (endomorphin-1 and -2) might be biased toward arrestin recruitment. In the present study, we investigated this phenomenon in more detail for endomorphin-2, using endogenous MOPr in rat brain as well as MOPr stably expressed in HEK293 cells. For MOPr in neurons in brainstem locus ceruleus slices, the peptide agonists [d-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin (DAMGO) and endomorphin-2 activated inwardly rectifying K+ current in a concentration-dependent manner. Analysis of these responses with the operational model of pharmacological agonism confirmed that endomorphin-2 had a much lower operational efficacy for G protein-mediated responses than did DAMGO at native MOPr in mature neurons. However, endomorphin-2 induced faster desensitization of the K+ current than did DAMGO. In addition, in HEK293 cells stably expressing MOPr, the ability of endomorphin-2 to induce phosphorylation of Ser375 in the COOH terminus of the receptor, to induce association of arrestin with the receptor, and to induce cell surface loss of receptors was much more efficient than would be predicted from its efficacy for G protein-mediated signaling. Together, these results indicate that endomorphin-2 is an arrestin-biased agonist at MOPr and the reason for this is likely to be the ability of endomorphin-2 to induce greater phosphorylation of MOPr than would be expected from its ability to activate MOPr and to induce activation of G proteins.

[1]  R. Lefkowitz,et al.  Molecular mechanism of β-arrestin-biased agonism at seven-transmembrane receptors. , 2012, Annual review of pharmacology and toxicology.

[2]  Kunhong Xiao,et al.  Multiple ligand-specific conformations of the β2-adrenergic receptor. , 2011, Nature chemical biology.

[3]  M. Morgan,et al.  Analysis of opioid efficacy, tolerance, addiction and dependence from cell culture to human , 2011, British journal of pharmacology.

[4]  Sudarshan Rajagopal,et al.  Quantifying Ligand Bias at Seven-Transmembrane Receptors , 2011, Molecular Pharmacology.

[5]  S. Schulz,et al.  Agonist‐selective patterns of µ‐opioid receptor phosphorylation revealed by phosphosite‐specific antibodies , 2011, British journal of pharmacology.

[6]  R. Lefkowitz,et al.  β-Arrestin-mediated receptor trafficking and signal transduction. , 2011, Trends in pharmacological sciences.

[7]  A. Burlingame,et al.  Quantitative Encoding of the Effect of a Partial Agonist on Individual Opioid Receptors by Multisite Phosphorylation and Threshold Detection , 2011, Science Signaling.

[8]  L. Bohn,et al.  Agonist-directed Interactions with Specific β-Arrestins Determine μ-Opioid Receptor Trafficking, Ubiquitination, and Dephosphorylation* , 2011, The Journal of Biological Chemistry.

[9]  B. Chieng,et al.  Cellular Morphine Tolerance Produced by βArrestin-2-Dependent Impairment of μ-Opioid Receptor Resensitization , 2011, The Journal of Neuroscience.

[10]  M. von Zastrow,et al.  Recovery from μ-Opioid Receptor Desensitization after Chronic Treatment with Morphine and Methadone , 2011, The Journal of Neuroscience.

[11]  T. Kenakin Functional Selectivity and Biased Receptor Signaling , 2011, Journal of Pharmacology and Experimental Therapeutics.

[12]  S. Charlton,et al.  μ-Opioid Receptors: Correlation of Agonist Efficacy for Signalling with Ability to Activate Internalization , 2010, Molecular Pharmacology.

[13]  P. Molinari,et al.  Morphine-like Opiates Selectively Antagonize Receptor-Arrestin Interactions* , 2010, The Journal of Biological Chemistry.

[14]  S. Sirohi,et al.  Dosing protocol and analgesic efficacy determine opioid tolerance in the mouse , 2009, Psychopharmacology.

[15]  W. Dewey,et al.  Role of protein kinase C and μ-opioid receptor (MOPr) desensitization in tolerance to morphine in rat locus coeruleus neurons , 2009, The European Journal of Neuroscience.

[16]  H. Loh,et al.  Morphine-induced mu-opioid receptor rapid desensitization is independent of receptor phosphorylation and beta-arrestins. , 2008, Cellular signalling.

[17]  J. Woods,et al.  The Spinal Antinociceptive Effects of Endomorphins in Rats: Behavioral and G Protein Functional Studies , 2008, Anesthesia and analgesia.

[18]  John T. Williams,et al.  Agonist-Specific Regulation of μ-Opioid Receptor Desensitization and Recovery from Desensitization , 2008, Molecular Pharmacology.

[19]  B. Tian,et al.  Search of the human proteome for endomorphin-1 and endomorphin-2 precursor proteins. , 2007, Life sciences.

[20]  L. Kocsis,et al.  Partial and full agonism in endomorphin derivatives: Comparison by null and operational model , 2006, Peptides.

[21]  H. Okano,et al.  Separation of μ-Opioid Receptor Desensitization and Internalization: Endogenous Receptors in Primary Neuronal Cultures , 2006, The Journal of Neuroscience.

[22]  Graeme Henderson,et al.  75 years of opioid research: the exciting but vain quest for the Holy Grail , 2006, British journal of pharmacology.

[23]  S. Schulz,et al.  Morphine induces terminal μ‐opioid receptor desensitization by sustained phosphorylation of serine‐375 , 2004, The EMBO journal.

[24]  E. Kelly,et al.  μ-Opioid Receptor Desensitization in Mature Rat Neurons: Lack of Interaction between DAMGO and Morphine , 2003, The Journal of Neuroscience.

[25]  A. Magyar,et al.  Receptor constants for endomorphin-1 and endomorphin-1-ol indicate differences in efficacy and receptor occupancy. , 2001, European journal of pharmacology.

[26]  A. Basbaum,et al.  Postsynaptic Signaling via the μ-Opioid Receptor: Responses of Dorsal Horn Neurons to Exogenous Opioids and Noxious Stimulation , 2000, The Journal of Neuroscience.

[27]  C. Sternini,et al.  Activation and internalization of the μ-opioid receptor by the newly discovered endogenous agonists, endomorphin-1 and endomorphin-2 †† These authors contributed equally to this work. †† , 1999, Neuroscience.

[28]  J. Pintar,et al.  μ‐opioid receptor modulation of calcium channel current in periaqueductal grey neurons from C57B16/J mice and mutant mice lacking MOR‐1 , 1999, British journal of pharmacology.

[29]  M. Narita,et al.  Characterization of endomorphin-1 and -2 on [35S]GTPgammaS binding in the mouse spinal cord. , 1998, European journal of pharmacology.

[30]  V. Hruby,et al.  Endomorphin-1 and endomorphin-2 are partial agonists at the human mu-opioid receptor. , 1998, European journal of pharmacology.

[31]  D. Selley,et al.  Endomorphin‐Stimulated [35S]GTPγS Binding in Rat Brain: Evidence for Partial Agonist Activity at μ‐Opioid Receptors , 1998 .

[32]  W. Sadee,et al.  Specific G protein activation and mu-opioid receptor internalization caused by morphine, DAMGO and endomorphin I. , 1998, European journal of pharmacology.

[33]  G. Uhl,et al.  μ Opioid Receptor Phosphorylation, Desensitization, and Ligand Efficacy* , 1997, The Journal of Biological Chemistry.

[34]  John T. Williams,et al.  Characterization of acute homologous desensitization of μ‐opioid receptor‐induced currents in locus coeruleus neurones , 1995, British journal of pharmacology.

[35]  F. Ehlert The relationship between muscarinic receptor occupancy and adenylate cyclase inhibition in the rabbit myocardium. , 1985, Molecular pharmacology.

[36]  R. North,et al.  On the potassium conductance increased by opioids in rat locus coeruleus neurones. , 1985, The Journal of physiology.

[37]  J. Black,et al.  Operational models of pharmacological agonism , 1983, Proceedings of the Royal Society of London. Series B. Biological Sciences.

[38]  C. McArdle,et al.  Using high-content microscopy to study gonadotrophin-releasing hormone regulation of ERK. , 2010, Methods in molecular biology.

[39]  John T. Williams,et al.  Desensitization and Recovery from Desensitization , 2008 .

[40]  N. Abul-Husn,et al.  Mu Opioid Receptor , 2008 .

[41]  H. Okano,et al.  Separation of mu-opioid receptor desensitization and internalization: endogenous receptors in primary neuronal cultures. , 2006, The Journal of neuroscience : the official journal of the Society for Neuroscience.

[42]  A. Kastin,et al.  A potent and selective endogenous agonist for the mu-opiate receptor. , 1997, Nature.

[43]  G. Uhl,et al.  Mu opioid receptor phosphorylation, desensitization, and ligand efficacy. , 1997, The Journal of biological chemistry.