Skip to main content
Log in

Effector antagonism by the regulators of G protein signalling (RGS) proteins causes desensitization of mu-opioid receptors in the CNS

  • Review
  • Published:
Psychopharmacology Aims and scope Submit manuscript

Abstract

Rationale

In cell culture systems, agonists can promote the phosphorylation and internalization of receptors coupled to G proteins (GPCR), leading to their desensitization. However, in the CNS opioid agonists promote a profound desensitization of their analgesic effects without diminishing the presence of their receptors in the neuronal membrane. Recent studies have indicated that CNS proteins of the RGS family, specific regulators of G protein signalling, may be involved in mu-opioid receptor desensitization in vivo.

Objective

In this work we review the role played by RGS proteins in the intensity and duration of the effects of mu-opioid receptor agonists, and how they influence the delayed tolerance that develops in response to specific doses of opioids.

Results

RGS proteins are GTPase-activating proteins (GAP) that accelerate the hydrolysis of GαGTP to terminate signalling at effectors. The GAP activity of RGS-R4 and RGS-Rz proteins restricts the amplitude of opioid analgesia, and the efficient deactivation of GαzGTP subunits by RGS-Rz proteins prevents mu receptor desensitization. However, RGS-R7 proteins antagonize effectors by binding to and sequestering mu receptor-activated Gαi/o/z subunits. Thus, they reduce the pool of receptor-regulated G proteins and hence, the effects of agonists. The delayed tolerance observed following morphine administration correlates with the transfer of Gα subunits from mu receptors to RGS-R7 proteins and the subsequent stabilization of this association.

Conclusion

In the CNS, the RGS proteins control the activity of mu opioid receptors through GAP-dependent (RGS-R4 and RGS-Rz) as well as by GAP-independent mechanisms (RGS-R7). As a result, they can both antagonize effectors and desensitize receptors under certain circumstances.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  • Anger T, Zhang W, Mende U (2004) Differential contribution of GTPase activation and effector antagonism to the inhibitory effect of RGS proteins on Gq-mediated signaling in vivo. J Biol Chem 279:3906–3915

    Google Scholar 

  • Arden JR, Segredo V, Wang Z, Lameh J, Sadee W (1995) Phosphorylation and agonist-specific intracellular trafficking of an epitope-tagged mu-opioid receptor expressed in HEK 293 cells. J Neurochem 65:1636–1645

    Google Scholar 

  • Axelrod JD, Miller JR, Shulman JM, Moon RT, Rerrimon N (1998) Differential recruitment of Dishevelled provides signaling specificity in the planar cell polarity and Wingless signaling pathways. Genes Dev 12:2610–2622

    Google Scholar 

  • Barker SA, Wang J, Sierra DA, Ross EM (2001) RGSZ1 and Ret RGS: two of several splice variants from the gene RGS20. Genomics 78:223–229

    Google Scholar 

  • Belcheva MM, Wong YH, Coscia CJ (2000) Evidence for transduction of mu but not kappa opioid modulation of extracellular signal-regulated kinase activity by Gz and G12 proteins. Cell Signal 12:481–489

    Google Scholar 

  • Berman DM, Gilman AG (1998) Mammalian RGS proteins: barbarians at the gate. J Biol Chem 273:1269–1272

    Google Scholar 

  • Berman DM, Wilkie TM, Gilman AG (1996a) GAIP and RGS4 are GTPase-activating proteins for the Gi subfamily of G protein alpha subunits. Cell 86:445–452

    Google Scholar 

  • Berman DM, Kozasa T, Gilman AG (1996b) The GTPase-activating protein RGS4 stabilizes the transition state for nucleotide hydrolysis. J Biol Sci 271:27209–27212

    Google Scholar 

  • Bernstein LS, Ramineni S, Hague C, Cladman W, Chidiac P, Levey AI, Hepler JR (2004) RGS2 binds directly and selectively to the M1 muscarinic acetylcholine receptor third intracellular loop to modulate Gq/11α signaling. J Biol Chem 279:21248–21256

    Google Scholar 

  • Bohn LM, Gainetdinov RR, Lin FT, Leftkowitz RJ, Caron MG (2000) Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature 408:720–723

    Google Scholar 

  • Burchett SA (2003) In through the out door: nuclear localization of the regulators of G protein signaling. J Neurochem 87:551–559

    Google Scholar 

  • Buzas B, Rosenberger J, Cox BM (1996) Mu and delta opioid receptor gene expression after chronic treatment with opioid agonist. NeuroReport 7:1505–1508

    Google Scholar 

  • Carman CV, Parent JL, Day PW, Pronin AN, Sternweis PM, Wedegaertner PB, Gilman AG, Benovic JL, Kozasa T (1999) Selective regulation of Galpha(q/11) by an RGS domain in the G protein-coupled receptor kinase, GRK2. J Biol Chem 274:34483–34492

    Google Scholar 

  • Chalecka-Franaszek E, Weems HB, Crowder AT, Cox BM, Côte TE (2000) Immunoprecipitation of high-affinity, guanine nucleotide-sensitive, solubilized μ-opioid receptors from rat brain: coimmunoprecipitation of the G proteins Gαo, Gαi1, and Gαi3. J Neurochem 74:1068–1078

    Google Scholar 

  • Chatterjee TK, Eapen A, Fisher RA (1997) A truncated form of RGS3 negatively regulates G protein-coupled receptor stimulation of adenylyl cyclase and phosphoinositide phospholipase C. J Biol Chem 272:15481–15487

    Google Scholar 

  • Chavkin C, McLaughlin JP, Celver JP (2001) Regulation of opioid receptor function by chronic agonist exposure: constitutive activity and desensitization. Mol Pharmacol 60:20–25

    Google Scholar 

  • De Vries L, Elenko E, Hubler L, Jones TL, Faquhar MG (1996) GAIP is membrane-anchored by palmitoylation and interacts with the activated (GTP-bound) form of Gαi subunits. Proc Natl Acad Sci U S A 93:15203–15208

    Google Scholar 

  • De Vries L, Lou X, Zhao G, Zheng B, Farquhar MG (1998) GIPC, a PDZ domain containing protein, interacts specifically with the C terminus of RGS-GAIP. Proc Natl Acad Sci U S A 95:12340–12345

    Google Scholar 

  • Doupnik CA, Davidson N, Lester HA, Kofuji P (1997) RGS proteins reconstitute the rapid gating kinetics of Gβγ-activated inwardly rectifying K+ channels. Proc Natl Acad Sci U S A 94:10461–10466

    Google Scholar 

  • Doupnik CA, Xu T, Shinaman JM (2001) Profile of RGS expression in single rat atrial myocytes. Biochim Biophys Acta 1522:97–107

    Google Scholar 

  • Doupnik CA, Jaen C, Zhang Q (2004) Measuring the modulatory effects of RGS proteins on GIRK channels. Meth Enzymol 389:131–154

    Google Scholar 

  • Dowal L, Elliot J, Popov S, Wilkie TM, Scarlate S (2001) Determination of the contact energies between a regulator of G protein signaling and G protein subunits and phospholipase Cβ1. Biochemistry 40:414–421

    Google Scholar 

  • Finn AK, Whistler JL (2001) Endocytosis of the mu opioid receptor reduces tolerance and a cellular hallmark of opiate withdrawal. Neuron 32:829–839

    Google Scholar 

  • Fischer T, De Vries L, Meerloo T, Farquhar MG (2003) Promotion of G alpha i3 subunit down-regulation by GIPN, a putative E3 ubiquitin ligase that interacts with RGS-GAIP. Proc Natl Acad Sci U S A 100:8270–8275

    Google Scholar 

  • Garzón J, Sánchez-Blázquez P (2001) Administration of myr+-Gi2α subunits prevents acute tolerance (tachyphylaxis) to mu-opioid effects in mice. Neuropharmacology 40:560–569

    Google Scholar 

  • Garzón J, Juarros JL, Castro MA, Sánchez-Blázquez P (1995) Antibodies to the cloned mu-opioid receptor detect various molecular weight forms in areas of mouse brain. Mol Pharmacol 47:738–744

    Google Scholar 

  • Garzón J, García-España A, Sánchez-Blázquez P (1997a) Opioids binding mu and delta receptors exhibit diverse efficacy in the activation of Gi2 and Gx/z transducer proteins in mouse periaqueductal gray matter. J Pharmacol Exp Ther 281:549–557

    Google Scholar 

  • Garzón J, Martínez-Peña Y, Sánchez-Blázquez P (1997b) Gx/z is regulated by μ but not δ opioid receptors in the stimulation of the low Km GTPase activity in mouse periaqueductal grey matter. Eur J Neurosci 9:1194–1200

    Google Scholar 

  • Garzón J, Castro M, Sánchez-Blázquez P (1998) Influence of Gz and Gi2 transducer proteins in the affinity of opioid agonists to μ receptors. Eur J Neurosci 10:2557–2564

    Google Scholar 

  • Garzón J, De Antonio I, Sánchez-Blázquez P (2000) In vivo modulation of G proteins and opioid receptor function by antisense oligodeoxynucleotides. Methods Enzymol 314:3–20

    Google Scholar 

  • Garzón J, Rodríguez-Díaz M, López-Fando A, Sánchez-Blázquez P (2001) RGS9 proteins facilitate acute tolerance to mu-opioid effects. Eur J Neurosci 13:801–811

    Google Scholar 

  • Garzón J, Rodríguez-Díaz M, López-Fando A, García-España A, Sánchez-Blázquez P (2002) Glycosylated phosducin-like protein long regulates opioid receptor function in mouse brain. Neuropharmacology 42:813–828

    Google Scholar 

  • Garzón J, López-Fando A, Sánchez-Blázquez P (2003) The R7 subfamily of RGS proteins assists tachyphylaxis and acute tolerance at μ-opioid receptors. Neuropsychopharmacology 28:1983–1990

    Google Scholar 

  • Garzón J, Rodríguez-Muñoz M, López-Fando A, García-España A, Sánchez-Blázquez P (2004) RGSZ1 and GAIP regulate μ- but not δ-opioid receptors in mouse CNS: role in tachyphylaxis and acute tolerance. Neuropsychopharmacology 29:1091–1104

    Google Scholar 

  • Garzón J, Rodríguez-Muñoz M, López-Fando A, Sánchez-Blázquez P (2005a) Activation of Mu-opioid receptors transfers control of Gα subunits to the regulator of G-protein signaling RGS9-2. Role in receptor desensitization. J Biol Chem 280:8951–8960

    Google Scholar 

  • Garzón J, Rodríguez-Muñoz M, Sánchez-Blázquez P (2005b) In mouse periaqueductal grey matter, morphine alters the selective association between Mu-opioid receptors and specific RGS proteins. Neuropharmacology 48:853–868

    Google Scholar 

  • Georgoussi Z, Milligan G, Zioudrou C (1995) Immunoprecipitation of opioid receptor-Go-protein complexes using specific GTP-binding-proteins antisera. Biochem J 306:71–75

    Google Scholar 

  • Giudice A, Gould JA, Freeman KB, Rastan S, Hertzog P, Kola I, Iannello RC (2001) Identification and characterization of alternatively spliced murine Rgs11 isoforms: genomic structure and gene analysis. Cytogenet Cell Genet 94:216–224

    Google Scholar 

  • Gold SJ, Ni YG, Dohlmam HG, Nestler EJ (1997) Regulators of G-protein signaling (RGS) proteins; region-specific expression of nine subtypes in rat brain. J Neurosci 17:8024–8037

    Google Scholar 

  • Hepler JR (1999) Emerging roles for RGS proteins in cell signalling. Trends Pharmacol Sci 20:376–382

    Google Scholar 

  • Hepler JR, Berman DM, Gilman AG, Kozasa T (1997) RGS4 and GAIP are GTPase-activating proteins for Gqα and block activation of phospholipase Cβ by γ-thio-GTP-Gqα. Proc Natl Acad Sci U S A 94:428–432

    Google Scholar 

  • Hooks SB, Waldo GL, Corbitt J, Bodor ET, Krumins AM, Harden TK (2003) RGS6, RGS7, RGS9, and RGS11 stimulate GTPase activity of Gi family G-proteins with differential selectivity and maximal activity. J Biol Chem 278:10087–10093

    Google Scholar 

  • Hollinger S, Hepler JR (2002) Cellular regulation of RGS proteins: modulators and integrators of G protein signaling. Pharmacol Rev 54:527–559

    Google Scholar 

  • Hollinger S, Taylor JB, Goldman EH, Hepler JR (2001) RGS14 is a bifunctional regulator of Gαi/o activity that exists in multiple populations in brain. J Neurochem 79:941–949

    Google Scholar 

  • Huidobro F, Huidobro-Toro JP, Way EL (1976) Studies on tolerance development to single doses of morphine in mice. J Pharmacol Exp Ther 198:318–326

    Google Scholar 

  • Hunt RA, Edris W, Chanda PK, Nieuwenhuijsen B, Young KH (2003) Snapin interacts with the N-terminus of regulator of G protein signaling 7. Biochem Biophys Res Commun 303:594–599

    Google Scholar 

  • Keith DE, Murray SR, Zaki PA, Chu PC, Lissin DV, Kang L, Evans CJ, von Zastrow M (1996) Morphine activates opioid receptors without causing their rapid internalization. J Biol Chem 271:19021–19024

    Google Scholar 

  • Kimple RJ, De Vries L, Tronchère H, Behe CI, Morris RA, Farquhar MG, Siderovski DP (2001) RGS12 and RGS14 GoLoco motifs are Gαi interaction sites with guanine nucleotide dissociation inhibitor activity. J Biol Chem 276:29275–29281

    Google Scholar 

  • Lefkowitz RJ (1998) G protein-coupled receptors. III. New roles for receptor kinases and beta-arrestins in receptor signaling and desensitization. J Biol Chem 273:18677–18680

    Article  CAS  PubMed  Google Scholar 

  • Levis MJ, Bourne HR (1992) Activation of the alpha subunit of Gs in intact cells alters its abundance, rate of degradation, and membrane avidity. J Cell Biol 119:1297–1307

    Google Scholar 

  • Longenecker KL, Lewis ME, Chikumi H, Gutkind JS, Derewenda ZS (2001) Structure of the RGS-like domain from PDZ-RhoGEF: linking heterotrimeric G protein-coupled signaling to Rho GTPases. Structure 9:559–569

    Google Scholar 

  • López-Fando A, Rodríguez-Muñoz M, Sánchez-Blázquez P, Garzón J (2005) Expression of neural RGS-R7 and Gβ5 proteins in response to acute and chronic morphine. Neuropsychopharmacology 30:99–110

    Google Scholar 

  • Lou X, Yano H, Lee F, Chao MV, Farquhar MG (2001) GIPC and GAIP form a complex with TrkA: a putative link between G protein and receptor tyrosine kinase pathways. Mol Biol Cell 12:615–627

    Google Scholar 

  • Lui-Chen LY, Chongguang C, Phillips CA (1993) β-[3H]Funaltrexamine-labeled μ-opioid receptors: species variations in molecular mass and glycosylation by complex-type, N-linked oligosacharides. Mol Pharmacol 44:749–756

    Google Scholar 

  • Mao H, Zhao Q, Daigle M, Ghahremani MH, Chidiac P, Albert PR (2004) RGS17/RGSZ2, a novel regulator of Gi/o, Gz and Gq signaling. J Biol Chem 279:26314–26322

    Google Scholar 

  • Massotte D, Brillet K, Kieffer BL, Milligan G (2002) Agonists activate Giα or Gi2α fused to the human mu opioid receptor differentially. J Neurochem 81:1372–1382

    Google Scholar 

  • McLaughlin JP, Myers LC, Zarek PE, Caron MG, Leftkowitz TJ, Czyzyk TA, Pintar JE, Chavkin C (2004) Prolonged kappa opioid receptor phosphorylation mediated by G-protein receptor kinase underlies sustained analgesic tolerance. J Biol Chem 279:1810–1818

    Google Scholar 

  • Posner BA, Gilman AG, Harris BA (1999) Regulators of G protein signaling 6 and 7. Purification of complexes with Gβ5 and assessment of their effects on G protein-mediated signaling pathways. J Biol Chem 274:31087–31093

    Google Scholar 

  • Rahman Z, Gold SJ, Potenza MN, Cowan CW, Ni YG, He W, Wensel TG, Nestler EJ (1999) Cloning and characterization of RGS9-2: a striatal-enriched alternatively spliced product of the RGS9 gene. J Neurosci 19:2016–2026

    Google Scholar 

  • Rose JJ, Taylor JB, Cockett MI, Jones PG, Hepler JR (2000) RGS7 is palmitoylated and exists as biochemically distinct forms. J Neurochem 75:2103–2112

    Google Scholar 

  • Ross EM, Wilkie TM (2000) GTPase-activating proteins for heterotrimeric G proteins: regulators of G protein signaling (RGS) and RGS-like proteins. Annu Rev Biochem 69:795–827

    Google Scholar 

  • Saitoh O, Kubo Y, Odagiri M, Ichikawa M, Yamagata K, Sekine T (1999) RGS7 and RGS8 differentially accelerate G protein-mediated modulation of K+ currents. J Biol Chem 14:9899–9904

    Google Scholar 

  • Sánchez-Blázquez P, García-España A, Garzón J (1995) In vivo injection of antisense oligodeoxynucleotides to Gα subunits and supraspinal analgesia evoked by mu and delta opioid agonists. J Pharmacol Exp Ther 275:1590–1596

    Google Scholar 

  • Sánchez-Blázquez P, Gómez-Serranillos P, Garzón J (2001) Agonist determine the pattern of G-protein activation in μ-opioid receptor-mediated supraspinal analgesia. Brain Res Bull 54:229–235

    Google Scholar 

  • Sánchez-Blázquez P, Rodríguez-Díaz M, López-Fando A, Rodríguez-Muñoz M, Garzón J (2003) The GBeta5 subunit that associates with the R7 subfamily of RGS proteins regulates mu-opioid effects. Neuropharmacology 45:85–98

    Google Scholar 

  • Sánchez-Blázquez P, Rodríguez-Muñoz M, Montero C, Garzón J (2005) RGS-Rz and RGS9-2 proteins control mu-opioid receptor desensitization in CNS: the role of activated Gαz subunits. Neuropharmacology 48:123–139

    Google Scholar 

  • Schmidt H, Schulz S, Klutzny M, Koch T, Handel M, Hollt V (2000) Involvement of mitogen-activated protein kinase in agonist-induced phosphorylation of the mu-opioid receptor in HEK 293 cells. J Neurochem 74:414–422

    Google Scholar 

  • Shuey DJ, Betty M, Jones PJ, Khawaja XZ, Cockett MI (1998) RGS7 attenuates signal transduction through the Gαq family of heterotrimeric G proteins in mammalian cells. J Neurochem 70:1964–1972

    Google Scholar 

  • Sierra DA, Gilbert DJ, Householder D, Grishin NV, Yu K, Ukidwe P, Barker SA, He W, Wensel TG, Otero G, Brown G, Copeland NG, Jenkins NA, Wilkie TM (2002) Evolution of the regulators of G-protein signaling multigene family in mouse and human. Genomics 79:177–185

    Google Scholar 

  • Sinnarajah S, Dessauer CW, Srikumar D, Chen J, Yuen J, Yilma S, Dennis JC, Morrison EE, Vodyanoy V, Kehrl JH (2001) RGS2 regulates signal transduction in olfactory neurons by attenuating activation of adenylyl cyclase III. Nature 409:1051–1055

    Article  CAS  PubMed  Google Scholar 

  • Skiba NP, Hopp JA, Arshavsky VY (2000) The effector enzyme regulates the duration of G protein signaling in vertebrate photoreceptors by increasing the affinity between transducin and RGS protein. J Biol Chem 275:32716–322720

    Google Scholar 

  • Snow BE, Krumins AM, Brothers GM, Lee S-F, Wall MA, Chung S, Mangion J, Arya S, Gilman AG, Siderovski DP (1998) A G protein γ subunit-like domain shared between RGS11 and other RGS proteins specifies binding to Gβ5 subunits. Proc Natl Acad Sci U S A 95:13307–13312

    Google Scholar 

  • Sondek J, Siderovski DP (2001) Gγ-like (GGL) domains: new frontiers in G-protein signaling and beta-propeller scaffolding. Biochem Pharmacol 61:1329–1337

    Google Scholar 

  • Stanasila L, Lim WK, Neubig RR, Pattus F (2000) Coupling efficacy and selectivity of the human μ-opioid receptor expressed as receptor-Gα fusion proteins in Escherichia coli. J Neurochem 75:1190–1199

    Google Scholar 

  • Standifer KM, Rossi GC, Pasternak GW (1996) Differential blockade of opioid analgesia by antisense oligodeoxynucleotides directed against various G protein α subunits. Mol Pharmacol 50:293–298

    Google Scholar 

  • Sternini C, Spann M, Anton B, Keith DE Jr, Bunnett NW, von Zastrow M, Evans C, Brecha NC (1996) Agonist-selective endocytosis of mu opioid receptor by neurons in vivo. Proc Natl Acad Sci U S A 93:9241–9246

    Article  Google Scholar 

  • Takida S, Fischer CC, Wedegaertner PB (2004) Palmitoylation and plasma membrane targeting of RGS7 are promoted by αo. Mol Pharmacol 67:132–139

    Google Scholar 

  • Tesmer JJ, Berman DM, Gilman AG, Sprang SR (1997) Structure of RGS4 bound to AlF4-activated Giα1: stabilization of the transition state for GTP hydrolysis. Cell 89:251–261

    Google Scholar 

  • Thomas EA, Danielson PE, Sutcliffe JG (1998) RGS9: a regulator of G-protein signalling with specific expression in rat and mouse striatum. J Neurosci Res 52:118–124

    Google Scholar 

  • Watson AJ, Aragay AM, Slepak VZ, Simon MI (1996) A novel form of the G protein β subunit Gβ5 is specifically expressed in the vertebrate retina. J Biol Chem 271:28154–28160

    Google Scholar 

  • Werling LL, McMahon PN, Cox BM (1989) Selective changes in mu opioid receptor properties induced by chronic morphine exposure. Proc Natl Acad Sci U S A 86:6393–6397

    Google Scholar 

  • Wise A, Lee TW, McEwan DJ, Milligan G (1995) Degradation of G11 alpha/Gq alpha is accelerated by agonist occupancy of alpha 1A/D, alpha 1B, and alpha 1C adrenergic receptors. J Biol Chem 270:17196–17203

    Article  CAS  PubMed  Google Scholar 

  • Yaksh TL, Yeung JC, Rudy TA (1976) Systematic examination in the rat of brain sites sensitive to the direct application of morphine: observation of differential effects within the periaqueductal grey. Brain Res 114:83–103

    Google Scholar 

  • Zachariou V, Georgescu D, Sanchez N, Rahman Z, DiLeone R, Berton O, Neve RL, Sim-Selley LJ, Selley DE, Gold SJ, Nestler EJ (2003) Essential role for RGS9 in opiate action. Proc Natl Acad Sci U S A 100:13656–13661

    Google Scholar 

  • Zeng W, Xu X, Popov S, Mukhopadhyay S, Chidiac P, Swistok J, Danho W, Yagaloff KA, Fisher SL, Ross EM, Muallem S, Wilkie TM (1998) The N-terminal domain of RGS4 confers receptor-selective inhibition of G protein signaling. J Biol Chem 273:34687–34690

    Google Scholar 

  • Zhang JH, Simonds WF (2000) Copurification of brain G-protein β5 with RGS6 and RGS7. J Neurosci 20:(RC59)1–5

    CAS  PubMed  Google Scholar 

  • Zhang J, Ferguson SS, Barak LS, Bodduluri SR, Laporte SA, Law PY, Caron MG (1998) Role for G protein-coupled receptor kinase in agonist-specific regulation of mu-opioid receptor responsiveness. Proc Natl Acad Sci U S A 95:7157–7162

    Google Scholar 

  • Zhang JH, Barr VA, Mo Y, Rojkova AM, Liu S, Simonds WF (2001) Nuclear localization of G protein beta 5 and regulator of G protein signaling 7 in neurons and brain. J Biol Chem 276:10284–10289

    Google Scholar 

  • Zheng B, De Vries L, Gist FM (1999) Divergence of RGS proteins: evidence for the existence of six mammalian RGS subfamilies. Trends Biochem Sci 24:411–414

    Google Scholar 

  • Zheng B, Ma YC, Ostrom RS, Lavoie C, Gill GN, Insel PA, Huang XY, Farquhar MG (2001) RGS-PX1, a GAP for Gαs and sorting nexin in vesicular trafficking. Science 294:1939–1942

    Google Scholar 

Download references

Acknowledgements

This work was supported by the SAF2003-01121, ISCIII G03/005 and MCYT BMC2002-03228.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Javier Garzón.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Garzón, J., Rodríguez-Muñoz, M., de la Torre-Madrid, E. et al. Effector antagonism by the regulators of G protein signalling (RGS) proteins causes desensitization of mu-opioid receptors in the CNS. Psychopharmacology 180, 1–11 (2005). https://doi.org/10.1007/s00213-005-2248-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00213-005-2248-9

Keywords

Navigation