Activation profiles of opioid ligands in HEK cells expressing delta opioid receptors

Parham Gharagozlou, Hasan Demirci, J David Clark, Jelveh Lameh, Parham Gharagozlou, Hasan Demirci, J David Clark, Jelveh Lameh

Abstract

Background: The aim of the present study was to characterize the activation profiles of 15 opioid ligands in transfected human embryonic kidney cells expressing only delta opioid receptors. Activation profiles of most of these ligands at delta opioid receptors had not been previously characterized in vitro. Receptor activation was assessed by measuring the inhibition of forskolin-stimulated cAMP production.

Results: Naltrexone and nalorphine were classified as antagonists at delta opioid receptor. The other ligands studied were agonists at delta opioid receptors and demonstrated IC50 values of 0.1 nM to 2 microM, maximal inhibition of 39-77% and receptor binding affinities of 0.5 to 243 nM. The rank order of efficacy of the ligands tested was metazocine = xorphanol > or = fentanyl = SKF 10047 = etorphine = hydromorphone = butorphanol = lofentanil > WIN 44,441 = Nalbuphine = cyclazocine > or = met-enkephalin >> morphine = dezocine. For the first time these data describe and compare the function and relative efficacy of several ligands at delta opioid receptors.

Conclusions: The data produced from this study can lead to elucidation of the complete activation profiles of several opioid ligands, leading to clarification of the mechanisms involved in physiological effects of these ligands at delta opioid receptors. Furthermore, these data can be used as a basis for novel use of existing opioid ligands based on their pharmacology at delta opioid receptors.

Figures

Figure 1
Figure 1
Reproducibility of dose response curves for xorphanol at δ opioid receptors. This graph represents inter-experimental variability for dose response curves. Three independent experiments were carried out in duplicate. Varying concentrations of xorphanol were used to determine the potency and efficacy of xorphanol in inhibiting the effect of 5 μM forskolin in producing cAMP, as described under methods. Maximal cAMP levels were in the range of 400–1000 pmole/well. Data have been normalized as described under methods. Error bars represent standard error of the mean of duplicate measurements for each point.
Figure 2
Figure 2
Dose response curves of representative ligands on δ opioid receptors Varying concentrations of opioid ligands were used to determine the potency and efficacy of each ligand in inhibiting the effect of 5 μM forskolin in producing cAMP, as described under methods. Maximal cAMP levels were in the range of 400–1000 pmole/well. Data presented are the average data from 2 or more experiments carried out in duplicate. Data have been normalized as described under methods. Error bars represent standard error of the mean of the normalized data. (■) Etorphine, (□) Lofentanil, (▼) SKF 10047 and (●) Fentanyl.

References

    1. Reisine T, Bell GI. Molecular biology of opioid receptors. Trends Neurosci. 1993;16:506–510. doi: 10.1016/0166-2236(93)90194-Q.
    1. Dhawan BN, Cesselin F, Raghubir R, Reisine T, Bradley PB, Portoghese PS, Hamon M. International Union of Pharmacology. XII. Classification of opioid receptors. Pharmacol Rev. 1996;48:567–592.
    1. Abdelhamid EE, Sultana M, Portoghese PS, Takemori AE. Selective blockage of delta opioid receptors prevents the development of morphine tolerance and dependence in mice. J Pharmacol Exp Ther. 1991;258:299–303.
    1. Filizola M, Villar HO, Loew GH. Molecular determinants of non-specific recognition of delta, mu, and kappa opioid receptors. Bioorg Med Chem. 2001;9:69–76. doi: 10.1016/S0968-0896(00)00223-6.
    1. Filizola M, Villar HO, Loew GH. Differentiation of delta, mu, and kappa opioid receptor agonists based on pharmacophore development and computed physicochemical properties. J Comput Aided Mol Des. 2001;15:297–307. doi: 10.1023/A:1011187320095.
    1. Reisine T, Pasternak G. Opioid Analgesics and Antagonists. In: Hardman JG, Limbird LE, Molinoff PB, Ruddon RW, Gilman AG, editor. Goodman and Gilman's The Pharmacological Basis of Therapeutics. 9. New York: McGraw-Hill Companies; 1996. pp. 521–549.
    1. Prather PL, Song L, Piros ET, Law PY, Hales TG. delta-Opioid receptors are more efficiently coupled to adenylyl cyclase than to L-type Ca(2+) channels in transfected rat pituitary cells. J Pharmacol Exp Ther. 2000;295:552–562.
    1. Wang JF, Ren MF, Xue JC, Han JS. Cyclic AMP mediates mu and delta, but not kappa opioid analgesia in the spinal cord of the rat. Life Sci. 1993;52:1955–1960. doi: 10.1016/0024-3205(93)90636-H.
    1. Mantegazza P, Tammiso R, Zambotti F, Zecca L, Zonta N. Purine involvement in morphine antinociception. Br J Pharmacol. 1984;83:883–888.
    1. Pasternak GW. Pharmacological mechanisms of opioid analgesics. Clin Neuropharmacol. 1993;16:1–18.
    1. Brandt MR, Furness MS, Mello NK, Rice KC, Negus SS. Antinociceptive effects of delta-opioid agonists in Rhesus monkeys: effects on chemically induced thermal hypersensitivity. J Pharmacol Exp Ther. 2001;296:939–946.
    1. Kovelowski CJ, Bian D, Hruby VJ, Lai J, Ossipov MH, Porreca F. Selective opioid delta agonists elicit antinociceptive supraspinal/spinal synergy in the rat. Brain Res. 1999;843:12–17. doi: 10.1016/S0006-8993(99)01803-X.
    1. Takita K, Herlenius EA, Lindahl SG, Yamamoto Y. Actions of opioids on respiratory activity via activation of brainstem mu-, delta- and kappa-receptors; an in vitro study. Brain Res. 1997;778:233–241. doi: 10.1016/S0006-8993(97)01105-0.
    1. Negus SS, Picker MJ. Pharmacology of the nonpeptide delta opioid agonist BW373U86. CNS Drug Rev. 1996;2:68–90.
    1. Dykstra LA, Schoenbaum GM, Yarbrough J, McNutt R, Chang KJ. A novel delta opioid agonist, BW373U86, in squirrel monkeys responding under a schedule of shock titration. J Pharmacol Exp Ther. 1993;267:875–882.
    1. Wild KD, McCormick J, Bilsky EJ, Vanderah T, McNutt RW, Chang KJ, Porreca F. Antinociceptive actions of BW373U86 in the mouse. J Pharmacol Exp Ther. 1993;267:858–865.
    1. Schiller PW, Fundytus ME, Merovitz L, Weltrowska G, Nguyen TM, Lemieux C, Chung NN, Coderre TJ. The opioid mu agonist/delta antagonist DIPP-NH(2)[Psi] produces a potent analgesic effect, no physical dependence, and less tolerance than morphine in rats. J Med Chem. 1999;42:3520–3526. doi: 10.1021/jm980724+.
    1. Wells JL, Bartlett JL, Ananthan S, Bilsky EJ. In vivo pharmacological characterization of SoRI 9409, a nonpeptidic opioid mu-agonist/delta-antagonist that produces limited antinociceptive tolerance and attenuates morphine physical dependence. J Pharmacol Exp Ther. 2001;297:597–605.
    1. Gutstein HB, Akil H. Opioid Analgesics. In: Goodman LS, Hardman JG, Limbird LE, Gilman AG, editor. Goodman & Gilman's the pharmacological basis of therapeutics. 10. New York: McGraw-Hill, Medical Pub. Division; 2001.
    1. Davis PJ, Cook DR. Clinical pharmacokinetics of the newer intravenous anaesthetic agents. Clin Pharmacokinet. 1986;11:18–35.
    1. Clark MJ, Emmerson PJ, Mansour A, Akil H, Woods JH, Portoghese PS, Remmers AE, Medzihradsky F. Opioid efficacy in a C6 glioma cell line stably expressing the delta opioid receptor. J Pharmacol Exp Ther. 1997;283:501–510.
    1. Malatynska E, Wang Y, Knapp RJ, Waite S, Calderon S, Rice K, Hruby VJ, Yamamura HI, Roeske WR. Human delta opioid receptor: functional studies on stably transfected Chinese hamster ovary cells after acute and chronic treatment with the selective nonpeptidic agonist SNC-80. J Pharmacol Exp Ther. 1996;278:1083–1089.
    1. Raynor K, Kong H, Chen Y, Yasuda K, Yu L, Bell GI, Reisine T. Pharmacological characterization of the cloned kappa-, delta-, and mu-opioid receptors. Mol Pharmacol. 1994;45:330–334.
    1. Toll L, Berzetei-Gurske IP, Polgar WE, Brandt SR, Adapa ID, Rodriguez L, Schwartz RW, Haggart D, O'Brien A, White A, Kennedy JM, Craymer K, Farrington L, Auh JS. Standard binding and functional assays related to medications development division testing for potential cocaine and opiate narcotic treatment medications. NIDA Res Monogr. 1998;178:440–466.
    1. Fu MJ, Tsen LY, Lee TY, Lui PW, Chan SH. Involvement of cerulospinal glutamatergic neurotransmission in fentanyl-induced muscular rigidity in the rat. Anesthesiology. 1997;87:1450–1459. doi: 10.1097/00000542-199712000-00024.
    1. Jaros T, Kolasiewicz W. Attenuation of the fentanyl-induced muscle rigidity by the selective 5HT1A agonist 8-OH-DPAT. Pol J Pharmacol. 1995;47:19–24.
    1. Tsou MY, Lui PW, Lee TY, Pan JT, Chan SH. Differential effects of prazosin and yohimbine on fentanyl-induced muscular rigidity in rats. Neuropharmacology. 1989;28:1163–1168. doi: 10.1016/0028-3908(89)90206-2.
    1. Lui PW, Lee TY, Chan SH. Involvement of locus coeruleus and noradrenergic neurotransmission in fentanyl-induced muscular rigidity in the rat. Neurosci Lett. 1989;96:114–119. doi: 10.1016/0304-3940(89)90252-8.
    1. Viscomi CM, Bailey PL. Opioid-induced rigidity after intravenous fentanyl. Obstet Gynecol. 1997;89:822–824. doi: 10.1016/S0029-7844(97)81423-8.
    1. Howes JF, Villarreal JE, Harris LS, Essigmann EM, Cowan A. Xorphanol. Drug Alcohol Depend. 1985;14:373–380.
    1. McCarthy PS, Howlett GJ. Physical dependence induced by opiate partial agonists in the rat. Neuropeptides. 1984;5:11–14.
    1. Cruz SL, Villarreal JE, Volkow ND. Further evidence that naloxone acts as an inverse opiate agonist: implications for drug dependence and withdrawal. Life Sci. 1996;58:PL381–389. doi: 10.1016/0024-3205(96)00250-0.
    1. Arden JR, Segredo V, Wang Z, Lameh J, Sadée W. Phosphorylation and agonist-specific intracellular trafficking of an epitope-tagged μ-opioid receptor expressed in HEK 293 cells. Journal of Neurochemistry. 1995;65:1636–1645.
    1. Lameh J, Wang P, Elgart D, Meredith D, Shafer SL, Loew GH. Unraveling the Identity of Benzodiazepine Binding Sites in Rat Hippocampus and Olfactory Bulb. European Journal of Pharmacology. 2000;400:167–176. doi: 10.1016/S0014-2999(00)00398-8.
    1. Lameh J, Keohane A, Clark DJ, Loew GH. Characterization of Novel Benzodiazepine ligands in Sf-9 cells. Neurosci Lett. 2001;306:25–28. doi: 10.1016/S0304-3940(01)01856-0.

Source: PubMed

3
Sottoscrivi