메뉴 건너뛰기




Volumn 5, Issue 3, 2012, Pages 199-226

Direct association of Mu-Opioid and NMDA glutamate receptors supports their cross-regulation: Molecular implications for opioid tolerance

Author keywords

Analgesia; Antinociception; Calciumcalmodulin dependent kinase II; Morphine; Mu opioid receptor; Nervous tissue; Nitric oxide synthase; NMDA receptor; Opioid; Protein kinase A; Protein kinase C; Receptor desensitization; Receptor tolerance; Redox signaling; Regulators of G protein signaling proteins; Zinc ions

Indexed keywords

AGMATINE; BROMOCRIPTINE; CALCITONIN GENE RELATED PEPTIDE; CALCIUM; CALMODULIN; CHOLECYSTOKININ; CYCLIC AMP DEPENDENT PROTEIN KINASE; CYCLIN DEPENDENT KINASE 2; CYCLOOXYGENASE 2 INHIBITOR; DELTA OPIATE RECEPTOR; G PROTEIN COUPLED RECEPTOR; GLUTAMATE RECEPTOR; GLUTAMIC ACID; GLUTAMINE; MORPHINE; MU OPIATE RECEPTOR; N METHYL DEXTRO ASPARTIC ACID; N METHYL DEXTRO ASPARTIC ACID RECEPTOR; NIFEDIPINE; NIMESULIDE; NITRIC OXIDE; NITRIC OXIDE SYNTHASE; NOLPITANTIUM; OPIATE; OXYTOCIN; PROTEIN KINASE C; RITANSERIN; SUBSTANCE P; UNINDEXED DRUG; VERAPAMIL;

EID: 84869138744     PISSN: 18744737     EISSN: 18744745     Source Type: Journal    
DOI: 10.2174/1874473711205030199     Document Type: Article
Times cited : (75)

References (304)
  • 1
    • 0017118916 scopus 로고
    • Studies on tolerance development to single doses of morphine in mice
    • Huidobro F, Huidobro-Toro JP, Way EL. Studies on tolerance development to single doses of morphine in mice. J Pharmacol Exp Ther 1976; 198: 318-29.
    • (1976) J Pharmacol Exp Ther , vol.198 , pp. 318-329
    • Huidobro, F.1    Huidobro-Toro, J.P.2    Way, E.L.3
  • 2
    • 0014434474 scopus 로고
    • Morphine: Single-dose tolerance
    • Kornetsky C, Bain G. Morphine: single-dose tolerance. Science 1968; 162: 1011-2.
    • (1968) Science , vol.162 , pp. 1011-1012
    • Kornetsky, C.1    Bain, G.2
  • 3
    • 0030967429 scopus 로고    scopus 로고
    • Acute tolerance to spinally administered morphine compares mechanistically with chronically induced morphine tolerance
    • Fairbanks CA, Wilcox GL. Acute tolerance to spinally administered morphine compares mechanistically with chronically induced morphine tolerance. J Pharmacol Exp Ther 1997; 282: 1408-17.
    • (1997) J Pharmacol Exp Ther , vol.282 , pp. 1408-1417
    • Fairbanks, C.A.1    Wilcox, G.L.2
  • 4
    • 0030439407 scopus 로고    scopus 로고
    • Effects of naloxone and D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 and the protein kinase inhibitors H7 and H8 on acute morphine dependence and antinociceptive tolerance in mice
    • Bilsky EJ, Bernstein RN, Wang Z, Sadee W, Porreca F. Effects of naloxone and D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 and the protein kinase inhibitors H7 and H8 on acute morphine dependence and antinociceptive tolerance in mice. J Pharmacol Exp Ther 1996; 277: 484-90.
    • (1996) J Pharmacol Exp Ther , vol.277 , pp. 484-490
    • Bilsky, E.J.1    Bernstein, R.N.2    Wang, Z.3    Sadee, W.4    Porreca, F.5
  • 5
    • 0018226370 scopus 로고
    • Single-dose tolerance to antinociception, and physical dependence on beta-endorphin in mice
    • Huidobro-Toro JP, Way EL. Single-dose tolerance to antinociception, and physical dependence on beta-endorphin in mice. Eur J Pharmacol 1978; 52: 179-89.
    • (1978) Eur J Pharmacol , vol.52 , pp. 179-189
    • Huidobro-Toro, J.P.1    Way, E.L.2
  • 6
    • 34548040700 scopus 로고    scopus 로고
    • Morphine induces endocytosis of neuronal m-opioid receptors through the sustained transfer of Ga subunits to RGSZ2 proteins
    • Rodríguez-Muñoz M, de la Torre-Madrid E, Sánchez-Blázquez P, Garzón J. Morphine induces endocytosis of neuronal m-opioid receptors through the sustained transfer of Ga subunits to RGSZ2 proteins. Mol Pain 2007; 3: 19.
    • (2007) Mol Pain , vol.3 , pp. 19
    • Rodríguez-Muñoz, M.1    de la Torre-Madrid, E.2    Sánchez-Blázquez, P.3    Garzón, J.4
  • 8
    • 0037169345 scopus 로고    scopus 로고
    • Regulation of opioid receptor trafficking and morphine tolerance by receptor oligomerization
    • He L, Fong J, von Zastrow M, Whistler JL. Regulation of opioid receptor trafficking and morphine tolerance by receptor oligomerization. Cell 2002; 108: 271-82.
    • (2002) Cell , vol.108 , pp. 271-282
    • He, L.1    Fong, J.2    von Zastrow, M.3    Whistler, J.L.4
  • 9
    • 33749239627 scopus 로고    scopus 로고
    • Tracking the opioid receptors on the way of desensitization
    • Marie N, Aguila B, Allouche S. Tracking the opioid receptors on the way of desensitization. Cell Signal 2006; 18: 1815-33.
    • (2006) Cell Signal , vol.18 , pp. 1815-1833
    • Marie, N.1    Aguila, B.2    Allouche, S.3
  • 10
    • 33748571086 scopus 로고    scopus 로고
    • Phosphorylation: A molecular switch in opioid tolerance
    • Wang ZJ, Wang LX. Phosphorylation: a molecular switch in opioid tolerance. Life Sci 2006; 79: 1681-91.
    • (2006) Life Sci , vol.79 , pp. 1681-1691
    • Wang, Z.J.1    Wang, L.X.2
  • 11
    • 0035819078 scopus 로고    scopus 로고
    • Endocytosis of the mu opioid receptor reduces tolerance and a cellular hallmark of opiate withdrawal
    • Finn AK, Whistler JL. Endocytosis of the mu opioid receptor reduces tolerance and a cellular hallmark of opiate withdrawal. Neuron 2001; 32: 829-39.
    • (2001) Neuron , vol.32 , pp. 829-839
    • Finn, A.K.1    Whistler, J.L.2
  • 12
    • 0034084767 scopus 로고    scopus 로고
    • Downregulation of G protein-coupled receptors
    • Tsao P, von Zastrow M. Downregulation of G protein-coupled receptors. Curr Opin Neurobiol 2000; 10: 365-9.
    • (2000) Curr Opin Neurobiol , vol.10 , pp. 365-369
    • Tsao, P.1    von Zastrow, M.2
  • 13
    • 79953003987 scopus 로고    scopus 로고
    • Recovery from mu-opioid receptor desensitization after chronic treatment with morphine and methadone
    • Quillinan N, Lau EK, Virk M, von ZM, Williams JT. Recovery from mu-opioid receptor desensitization after chronic treatment with morphine and methadone. J Neurosci 2011; 31: 4434-43.
    • (2011) J Neurosci , vol.31 , pp. 4434-4443
    • Quillinan, N.1    Lau, E.K.2    Virk, M.3    von, Z.M.4    Williams, J.T.5
  • 14
    • 79956302829 scopus 로고    scopus 로고
    • Cellular morphine tolerance produced by barrestin-2-dependent impairment of muopioid receptor resensitization
    • Dang VC, Chieng B, Azriel Y, Christie MJ. Cellular morphine tolerance produced by barrestin-2-dependent impairment of muopioid receptor resensitization. J Neurosci 2011; 31: 7122-30.
    • (2011) J Neurosci , vol.31 , pp. 7122-7130
    • Dang, V.C.1    Chieng, B.2    Azriel, Y.3    Christie, M.J.4
  • 15
    • 0032585754 scopus 로고    scopus 로고
    • oa subunits restore the efficacy of opioids, clonidine and neurotensin giving rise to antinociception in G-protein knock-down mice
    • oa subunits restore the efficacy of opioids, clonidine and neurotensin giving rise to antinociception in G-protein knock-down mice. Neuropharmacology 1999; 38: 1861-73.
    • (1999) Neuropharmacology , vol.38 , pp. 1861-1873
    • Garzón, J.1    Rodríguez-Díaz, M.2    de Antonio, I.3
  • 16
    • 0035089407 scopus 로고    scopus 로고
    • i2a subunits prevents acute tolerance (tachyphylaxis) to mu-opioid effects in mice
    • i2a subunits prevents acute tolerance (tachyphylaxis) to mu-opioid effects in mice. Neuropharmacology 2001; 40: 560-9.
    • (2001) Neuropharmacology , vol.40 , pp. 560-569
    • Garzón, J.1    Sánchez-Blázquez, P.2
  • 17
    • 20644443702 scopus 로고    scopus 로고
    • Mechanisms of mu opioid receptor/G-protein desensitization in brain by chronic heroin administration
    • Maher CE, Martin TJ, Childers SR. Mechanisms of mu opioid receptor/G-protein desensitization in brain by chronic heroin administration. Life Sci 2005; 77: 1140-54.
    • (2005) Life Sci , vol.77 , pp. 1140-1154
    • Maher, C.E.1    Martin, T.J.2    Childers, S.R.3
  • 18
    • 0034660289 scopus 로고    scopus 로고
    • Chronic heroin self-administration desensitizes m opioid receptoractivated G-proteins in specific regions of rat brain
    • Sim-Selley LJ, Selley DE, Vogt LJ, Childers SR, Martin TJ. Chronic heroin self-administration desensitizes m opioid receptoractivated G-proteins in specific regions of rat brain. J Neurosci 2000; 20: 4555-62.
    • (2000) J Neurosci , vol.20 , pp. 4555-4562
    • Sim-Selley, L.J.1    Selley, D.E.2    Vogt, L.J.3    Childers, S.R.4    Martin, T.J.5
  • 22
    • 21244499733 scopus 로고    scopus 로고
    • Effector antagonism by the regulators of G protein signalling (RGS) proteins causes desensitization of mu-opioid receptors in the CNS
    • Garzón J, Rodriguez-Muñoz M, de la Torre-Madrid E, Sánchez-Blázquez P. Effector antagonism by the regulators of G protein signalling (RGS) proteins causes desensitization of mu-opioid receptors in the CNS. Psychopharmacology (Berl) 2005; 180: 1-11.
    • (2005) Psychopharmacology (Berl) , vol.180 , pp. 1-11
    • Garzón, J.1    Rodriguez-Muñoz, M.2    de la Torre-Madrid, E.3    Sánchez-Blázquez, P.4
  • 23
    • 11144258839 scopus 로고    scopus 로고
    • RGS-Rz and RGS9-2 proteins control mu-opioid receptor desensitisation in CNS: The role of activated Gaz subunits
    • Sánchez-Blázquez P, Rodríguez-Muñoz M, Montero C, Garzón J. RGS-Rz and RGS9-2 proteins control mu-opioid receptor desensitisation in CNS: the role of activated Gaz subunits. Neuropharmacology 2005; 48: 134-50.
    • (2005) Neuropharmacology , vol.48 , pp. 134-150
    • Sánchez-Blázquez, P.1    Rodríguez-Muñoz, M.2    Montero, C.3    Garzón, J.4
  • 24
    • 15744387869 scopus 로고    scopus 로고
    • Activation of m-opioid receptors transfers control of Ga subunits to the regulator of G-protein signaling RGS9-2: Role in receptor desensitization
    • Garzón J, Rodríguez-Muñoz M, López-Fando A, Sánchez-Blázquez P. Activation of m-opioid receptors transfers control of Ga subunits to the regulator of G-protein signaling RGS9-2: role in receptor desensitization. J Biol Chem 2005; 280: 8951-60.
    • (2005) J Biol Chem , vol.280 , pp. 8951-8960
    • Garzón, J.1    Rodríguez-Muñoz, M.2    López-Fando, A.3    Sánchez-Blázquez, P.4
  • 25
    • 79955787551 scopus 로고    scopus 로고
    • A unique role of RGS9-2 in the striatum as a positive or negative regulator of opiate analgesia
    • Psifogeorgou K, Terzi D, Papachatzaki MM, et al. A unique role of RGS9-2 in the striatum as a positive or negative regulator of opiate analgesia. J Neurosci 2011; 31: 5617-24.
    • (2011) J Neurosci , vol.31 , pp. 5617-5624
    • Psifogeorgou, K.1    Terzi, D.2    Papachatzaki, M.M.3
  • 26
    • 83055160848 scopus 로고    scopus 로고
    • Involvement of NOX1/NADPH Oxidase in Morphine-Induced Analgesia and Tolerance
    • Ibi M, Matsuno K, Matsumoto M, et al. Involvement of NOX1/NADPH Oxidase in Morphine-Induced Analgesia and Tolerance. J Neurosci 2011; 31: 18094-103.
    • (2011) J Neurosci , vol.31 , pp. 18094-18103
    • Ibi, M.1    Matsuno, K.2    Matsumoto, M.3
  • 27
    • 0032930258 scopus 로고    scopus 로고
    • Cholecystokinin receptor mechanism(s) and morphine tolerance in mice
    • Zarrindast MR, Nikfar S, Rezayat M. Cholecystokinin receptor mechanism(s) and morphine tolerance in mice. Pharmacol Toxicol 1999; 84: 46-50.
    • (1999) Pharmacol Toxicol , vol.84 , pp. 46-50
    • Zarrindast, M.R.1    Nikfar, S.2    Rezayat, M.3
  • 28
    • 0028217236 scopus 로고
    • CCK receptor activation may prevent tolerance to morphine in mice
    • Rezayat M, Nikfar S, Zarrindast MR. CCK receptor activation may prevent tolerance to morphine in mice. Eur J Pharmacol 1994; 254: 21-6.
    • (1994) Eur J Pharmacol , vol.254 , pp. 21-26
    • Rezayat, M.1    Nikfar, S.2    Zarrindast, M.R.3
  • 29
    • 0030665019 scopus 로고    scopus 로고
    • Neuroanatomical localization, pharmacological characterization and functions of CGRP, related peptides and their receptors
    • van Rossum D, Hanisch UK, Quirion R. Neuroanatomical localization, pharmacological characterization and functions of CGRP, related peptides and their receptors. Neurosci Biobehav Rev 1997; 21: 649-78.
    • (1997) Neurosci Biobehav Rev , vol.21 , pp. 649-678
    • van Rossum, D.1    Hanisch, U.K.2    Quirion, R.3
  • 30
    • 0025441936 scopus 로고
    • Tachykinins and calcitonin gene-related peptide enhance release of endogenous glutamate and aspartate from the rat spinal dorsal horn slice
    • Kangrga I, Randic M. Tachykinins and calcitonin gene-related peptide enhance release of endogenous glutamate and aspartate from the rat spinal dorsal horn slice. J Neurosci 1990; 10: 2026-38.
    • (1990) J Neurosci , vol.10 , pp. 2026-2038
    • Kangrga, I.1    Randic, M.2
  • 31
    • 0029945840 scopus 로고    scopus 로고
    • A calcitonin gene-related peptide receptor antagonist prevents the development of tolerance to spinal morphine analgesia
    • Menard DP, van RD, Kar S, et al. A calcitonin gene-related peptide receptor antagonist prevents the development of tolerance to spinal morphine analgesia. J Neurosci 1996; 16: 2342-51.
    • (1996) J Neurosci , vol.16 , pp. 2342-2351
    • Menard, D.P.1    van, R.D.2    Kar, S.3
  • 32
    • 79952990379 scopus 로고    scopus 로고
    • Blockade of adrenomedullin receptors reverses morphine tolerance and its neurochemical mechanisms
    • Wang D, Chen P, Li Q, Quirion R, Hong Y. Blockade of adrenomedullin receptors reverses morphine tolerance and its neurochemical mechanisms. Behav Brain Res 2011; 221: 83-90.
    • (2011) Behav Brain Res , vol.221 , pp. 83-90
    • Wang, D.1    Chen, P.2    Li, Q.3    Quirion, R.4    Hong, Y.5
  • 33
    • 0033707952 scopus 로고    scopus 로고
    • Blockade and reversal of spinal morphine tolerance by peptide and non-peptide calcitonin generelated peptide receptor antagonists
    • Powell KJ, Ma W, Sutak M, et al. Blockade and reversal of spinal morphine tolerance by peptide and non-peptide calcitonin generelated peptide receptor antagonists. Br J Pharmacol 2000; 131: 875-84.
    • (2000) Br J Pharmacol , vol.131 , pp. 875-884
    • Powell, K.J.1    Ma, W.2    Sutak, M.3
  • 35
    • 33645095476 scopus 로고    scopus 로고
    • Paradigm shift in neuroprotection by NMDA receptor blockade: Memantine and beyond
    • Lipton SA. Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond. Nat Rev Drug Discov 2006; 5: 160-70.
    • (2006) Nat Rev Drug Discov , vol.5 , pp. 160-170
    • Lipton, S.A.1
  • 36
    • 37349049304 scopus 로고    scopus 로고
    • The role of glutamate in mood disorders: Results from the ketamine in major depression study and the presumed cellular mechanism underlying its antidepressant effects
    • Maeng S, Zarate Jr CA. The role of glutamate in mood disorders: results from the ketamine in major depression study and the presumed cellular mechanism underlying its antidepressant effects. Curr Psychiatry Rep 2007; 9: 467-74.
    • (2007) Curr Psychiatry Rep , vol.9 , pp. 467-474
    • Maeng, S.1    Zarate Jr, C.A.2
  • 37
    • 1142302573 scopus 로고    scopus 로고
    • Glutaminergic hypothesis of schizophrenia: Clinical research studies with ketamine
    • Mechri A, Saoud M, Khiari G, et al. Glutaminergic hypothesis of schizophrenia: clinical research studies with ketamine. Encephale 2001; 27: 53-9.
    • (2001) Encephale , vol.27 , pp. 53-59
    • Mechri, A.1    Saoud, M.2    Khiari, G.3
  • 38
    • 0028063771 scopus 로고
    • Electrophysiologic analysis of preemptive effects of spinal opioids on N-methyl-D-aspartate receptor-mediated events
    • Chapman V, Haley JE, Dickenson AH. Electrophysiologic analysis of preemptive effects of spinal opioids on N-methyl-D-aspartate receptor-mediated events. Anesthesiology 1994; 81: 1429-35.
    • (1994) Anesthesiology , vol.81 , pp. 1429-1435
    • Chapman, V.1    Haley, J.E.2    Dickenson, A.H.3
  • 39
    • 69649087572 scopus 로고    scopus 로고
    • Ketamine produces effective and long-term pain relief in patients with Complex Regional Pain Syndrome Type 1
    • Sigtermans MJ, van Hilten JJ, Bauer MC, et al. Ketamine produces effective and long-term pain relief in patients with Complex Regional Pain Syndrome Type 1. Pain 2009; 145: 304-11.
    • (2009) Pain , vol.145 , pp. 304-311
    • Sigtermans, M.J.1    van Hilten, J.J.2    Bauer, M.C.3
  • 40
    • 0028810883 scopus 로고
    • Structure and function of the NMDA receptor channel
    • Mori H, Mishina M. Structure and function of the NMDA receptor channel. Neuropharmacology 1995; 34: 1219-37.
    • (1995) Neuropharmacology , vol.34 , pp. 1219-1237
    • Mori, H.1    Mishina, M.2
  • 42
    • 0032954744 scopus 로고    scopus 로고
    • In vivo studies of the cerebral glutamate receptor/NO/cGMP pathway
    • Fedele E, Raiteri M. In vivo studies of the cerebral glutamate receptor/NO/cGMP pathway. Prog Neurobiol 1999; 58: 89-120.
    • (1999) Prog Neurobiol , vol.58 , pp. 89-120
    • Fedele, E.1    Raiteri, M.2
  • 43
    • 0042844731 scopus 로고    scopus 로고
    • Locus-specific rescue of GluRe1 NMDA receptors in mutant mice identifies the brain regions important for morphine tolerance and dependence
    • Inoue M, Mishina M, Ueda H. Locus-specific rescue of GluRe1 NMDA receptors in mutant mice identifies the brain regions important for morphine tolerance and dependence. J Neurosci 2003; 23: 6529-36.
    • (2003) J Neurosci , vol.23 , pp. 6529-6536
    • Inoue, M.1    Mishina, M.2    Ueda, H.3
  • 44
    • 0029618055 scopus 로고
    • Perspectives on the Nmethyl-D-aspartate/nitric oxide cascade and opioid tolerance
    • Pasternak GW, Kolesnikov YA, Babey AM. Perspectives on the Nmethyl-D-aspartate/nitric oxide cascade and opioid tolerance. Neuropsychopharmacology 1995; 13: 309-13.
    • (1995) Neuropsychopharmacology , vol.13 , pp. 309-313
    • Pasternak, G.W.1    Kolesnikov, Y.A.2    Babey, A.M.3
  • 45
    • 0025971938 scopus 로고
    • Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801
    • Trujillo KA, Akil H. Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science 1991; 251: 85-7.
    • (1991) Science , vol.251 , pp. 85-87
    • Trujillo, K.A.1    Akil, H.2
  • 46
    • 0036618951 scopus 로고    scopus 로고
    • The neurobiology of opiate tolerance, dependence and sensitization: Mechanisms of NMDA receptor-dependent synaptic plasticity
    • Trujillo KA. The neurobiology of opiate tolerance, dependence and sensitization: mechanisms of NMDA receptor-dependent synaptic plasticity. Neurotox Res 2002; 4: 373-91.
    • (2002) Neurotox Res , vol.4 , pp. 373-391
    • Trujillo, K.A.1
  • 47
    • 37349105318 scopus 로고    scopus 로고
    • Post-synaptic action of morphine on glutamatergic neuronal transmission related to the descending antinociceptive pathway in the rat thalamus
    • Narita M, Hashimoto K, Amano T, et al. Post-synaptic action of morphine on glutamatergic neuronal transmission related to the descending antinociceptive pathway in the rat thalamus. J Neurochem 2008; 104: 469-78.
    • (2008) J Neurochem , vol.104 , pp. 469-478
    • Narita, M.1    Hashimoto, K.2    Amano, T.3
  • 48
    • 37549050119 scopus 로고    scopus 로고
    • Activation of mu-opioid receptor selectively potentiates NMDA-induced outward currents in rat locus coeruleus neurons
    • Koyama S, Akaike N. Activation of mu-opioid receptor selectively potentiates NMDA-induced outward currents in rat locus coeruleus neurons. Neurosci Res 2008; 60: 22-8.
    • (2008) Neurosci Res , vol.60 , pp. 22-28
    • Koyama, S.1    Akaike, N.2
  • 49
    • 0025895334 scopus 로고
    • Sustained potentiation of NMDA receptormediated glutamate responses through activation of protein kinase C by a m opioid
    • Chen L, Huang LY. Sustained potentiation of NMDA receptormediated glutamate responses through activation of protein kinase C by a m opioid. Neuron 1991; 7: 319-26.
    • (1991) Neuron , vol.7 , pp. 319-326
    • Chen, L.1    Huang, L.Y.2
  • 50
    • 0037052624 scopus 로고    scopus 로고
    • Potentiation of the excitatory action of NMDA in ventrolateral periaqueductal gray by the mu-opioid receptor agonist, DAMGO
    • Kow LM, Commons KG, Ogawa S, Pfaff DW. Potentiation of the excitatory action of NMDA in ventrolateral periaqueductal gray by the mu-opioid receptor agonist, DAMGO. Brain Res 2002; 935: 87-102.
    • (2002) Brain Res , vol.935 , pp. 87-102
    • Kow, L.M.1    Commons, K.G.2    Ogawa, S.3    Pfaff, D.W.4
  • 51
    • 0025897304 scopus 로고
    • Modulation of NMDA-induced currents by mu-opioid receptor agonist DAGO in acutely isolated rat spinal dorsal horn neurons
    • Rusin KI, Randic M. Modulation of NMDA-induced currents by mu-opioid receptor agonist DAGO in acutely isolated rat spinal dorsal horn neurons. Neurosci Lett 1991; 124: 208-12.
    • (1991) Neurosci Lett , vol.124 , pp. 208-212
    • Rusin, K.I.1    Randic, M.2
  • 52
    • 0031032086 scopus 로고    scopus 로고
    • mu-Opioid receptors modulate NMDA receptor-mediated responses in nucleus accumbens neurons
    • Martin G, Nie Z, Siggins GR. mu-Opioid receptors modulate NMDA receptor-mediated responses in nucleus accumbens neurons. J Neurosci 1997; 17: 11-22.
    • (1997) J Neurosci , vol.17 , pp. 11-22
    • Martin, G.1    Nie, Z.2    Siggins, G.R.3
  • 53
    • 0033571936 scopus 로고    scopus 로고
    • Opioid enhancement of calcium oscillations and burst events involving NMDA receptors and L-type calcium channels in cultured hippocampal neurons
    • Przewlocki R, Parsons KL, Sweeney DD, et al. Opioid enhancement of calcium oscillations and burst events involving NMDA receptors and L-type calcium channels in cultured hippocampal neurons. J Neurosci 1999; 19: 9705-15.
    • (1999) J Neurosci , vol.19 , pp. 9705-9715
    • Przewlocki, R.1    Parsons, K.L.2    Sweeney, D.D.3
  • 54
    • 0028177985 scopus 로고
    • The NMDA receptor antagonists, LY274614 and MK-801, and the nitric oxide synthase inhibitor, NG-nitro-L-arginine, attenuate analgesic tolerance to the mu-opioid morphine but not to kappa opioids
    • Elliott K, Minami N, Kolesnikov YA, Pasternak GW, Inturrisi CE. The NMDA receptor antagonists, LY274614 and MK-801, and the nitric oxide synthase inhibitor, NG-nitro-L-arginine, attenuate analgesic tolerance to the mu-opioid morphine but not to kappa opioids. Pain 1994; 56: 69-75.
    • (1994) Pain , vol.56 , pp. 69-75
    • Elliott, K.1    Minami, N.2    Kolesnikov, Y.A.3    Pasternak, G.W.4    Inturrisi, C.E.5
  • 55
    • 0029562938 scopus 로고
    • The effects of NMDA receptor antagonists and nitric oxide synthase inhibitors on opioid tolerance and withdrawal. Medication development issues for opiate addiction
    • Herman BH, Vocci F, Bridge P. The effects of NMDA receptor antagonists and nitric oxide synthase inhibitors on opioid tolerance and withdrawal. Medication development issues for opiate addiction. Neuropsychopharmacology 1995; 13: 269-93.
    • (1995) Neuropsychopharmacology , vol.13 , pp. 269-293
    • Herman, B.H.1    Vocci, F.2    Bridge, P.3
  • 56
    • 0026589139 scopus 로고
    • The NMDA receptor antagonist MK-801 prevents long-lasting non-associative morphine tolerance in the rat
    • Ben-Eliyahu S, Marek P, Vaccarino AL, et al. The NMDA receptor antagonist MK-801 prevents long-lasting non-associative morphine tolerance in the rat. Brain Res 1992; 575: 304-8.
    • (1992) Brain Res , vol.575 , pp. 304-308
    • Ben-Eliyahu, S.1    Marek, P.2    Vaccarino, A.L.3
  • 57
    • 0025883699 scopus 로고
    • Delayed application of MK-801 attenuates development of morphine tolerance in rats
    • Marek P, Ben Eliyahu S, Vaccarino AL, Liebeskind JC. Delayed application of MK-801 attenuates development of morphine tolerance in rats. Brain Res 1991; 558: 163-5.
    • (1991) Brain Res , vol.558 , pp. 163-165
    • Marek, P.1    Ben Eliyahu, S.2    Vaccarino, A.L.3    Liebeskind, J.C.4
  • 58
    • 0026036359 scopus 로고
    • Glutamate antagonists prevent morphine withdrawal in mice and guinea pigs
    • Tanganelli S, Antonelli T, Morari M, Bianchi C, Beani L. Glutamate antagonists prevent morphine withdrawal in mice and guinea pigs. Neurosci Lett 1991; 122: 270-2.
    • (1991) Neurosci Lett , vol.122 , pp. 270-272
    • Tanganelli, S.1    Antonelli, T.2    Morari, M.3    Bianchi, C.4    Beani, L.5
  • 59
    • 0030444685 scopus 로고    scopus 로고
    • Competitive and non-competitive NMDA antagonists block the development of antinociceptive tolerance to morphine, but not to selective m or d opioid agonists in mice
    • Bilsky EJ, Inturrisi CE, Sadee W, Hruby VJ, Porreca F. Competitive and non-competitive NMDA antagonists block the development of antinociceptive tolerance to morphine, but not to selective m or d opioid agonists in mice. Pain 1996; 68: 229-37.
    • (1996) Pain , vol.68 , pp. 229-237
    • Bilsky, E.J.1    Inturrisi, C.E.2    Sadee, W.3    Hruby, V.J.4    Porreca, F.5
  • 60
    • 49549108443 scopus 로고    scopus 로고
    • NMDAR-nNOS generated zinc recruits PKCgamma to the HINT1-RGS17 complex bound to the C terminus of Muopioid receptors
    • Rodríguez-Muñoz M, Torre-Madrid E, Sánchez-Blázquez P, Wang JB, Garzón J. NMDAR-nNOS generated zinc recruits PKCgamma to the HINT1-RGS17 complex bound to the C terminus of Muopioid receptors. Cell Signal 2008; 20: 1855-64.
    • (2008) Cell Signal , vol.20 , pp. 1855-1864
    • Rodríguez-Muñoz, M.1    Torre-Madrid, E.2    Sánchez-Blázquez, P.3    Wang, J.B.4    Garzón, J.5
  • 61
    • 0030272102 scopus 로고    scopus 로고
    • Immunocytochemical distribution of ionotropic glutamate receptor subunits in the spinal cord of the rabbit
    • Bonnot A, Corio M, Tramu G, Viala D. Immunocytochemical distribution of ionotropic glutamate receptor subunits in the spinal cord of the rabbit. J Chem Neuroanat 1996; 11: 267-78.
    • (1996) J Chem Neuroanat , vol.11 , pp. 267-278
    • Bonnot, A.1    Corio, M.2    Tramu, G.3    Viala, D.4
  • 62
    • 0028022125 scopus 로고
    • Differential NMDA NR1 mRNA expression among spinal trigeminal neurons that project to different targets
    • Dohrn CS, Beitz AJ. Differential NMDA NR1 mRNA expression among spinal trigeminal neurons that project to different targets. Neuroreport 1994; 5: 1857-60.
    • (1994) Neuroreport , vol.5 , pp. 1857-1860
    • Dohrn, C.S.1    Beitz, A.J.2
  • 63
    • 0028087773 scopus 로고
    • The NMDA receptor subunits NR2A and NR2B show histological and ultrastructural localization patterns similar to those of NR1
    • Petralia RS, Wang YX, Wenthold RJ. The NMDA receptor subunits NR2A and NR2B show histological and ultrastructural localization patterns similar to those of NR1. J Neurosci 1994; 14: 6102-20.
    • (1994) J Neurosci , vol.14 , pp. 6102-6120
    • Petralia, R.S.1    Wang, Y.X.2    Wenthold, R.J.3
  • 64
    • 0028157362 scopus 로고
    • Light and electron microscope distribution of the NMDA receptor subunit NMDAR1 in the rat nervous system using a selective anti-peptide antibody
    • Petralia RS, Yokotani N, Wenthold RJ. Light and electron microscope distribution of the NMDA receptor subunit NMDAR1 in the rat nervous system using a selective anti-peptide antibody. J Neurosci 1994; 14: 667-96.
    • (1994) J Neurosci , vol.14 , pp. 667-696
    • Petralia, R.S.1    Yokotani, N.2    Wenthold, R.J.3
  • 65
    • 0029014593 scopus 로고
    • Cellular and subcellular distribution of NMDAR1 splice variant mRNA in the rat lumbar spinal cord
    • Tolle TR, Berthele A, Laurie DJ, Seeburg PH, Zieglgansberger W. Cellular and subcellular distribution of NMDAR1 splice variant mRNA in the rat lumbar spinal cord. Eur J Neurosci 1995; 7: 1235-44.
    • (1995) Eur J Neurosci , vol.7 , pp. 1235-1244
    • Tolle, T.R.1    Berthele, A.2    Laurie, D.J.3    Seeburg, P.H.4    Zieglgansberger, W.5
  • 66
    • 0030953255 scopus 로고    scopus 로고
    • Synchronized overproduction of AMPA, kainate, and NMDA glutamate receptors during human spinal cord development
    • Kalb RG, Fox AJ. Synchronized overproduction of AMPA, kainate, and NMDA glutamate receptors during human spinal cord development. J Comp Neurol 1997; 384: 200-10.
    • (1997) J Comp Neurol , vol.384 , pp. 200-210
    • Kalb, R.G.1    Fox, A.J.2
  • 67
    • 0033552067 scopus 로고    scopus 로고
    • Mu-opioid and NMDA-type glutamate receptors are often colocalized in spiny neurons within patches of the caudate-putamen nucleus
    • Wang H, Gracy KN, Pickel VM. Mu-opioid and NMDA-type glutamate receptors are often colocalized in spiny neurons within patches of the caudate-putamen nucleus. J Comp Neurol 1999; 412: 132-46.
    • (1999) J Comp Neurol , vol.412 , pp. 132-146
    • Wang, H.1    Gracy, K.N.2    Pickel, V.M.3
  • 68
    • 0030974668 scopus 로고    scopus 로고
    • Dual ultrastructural localization of m-opioid receptors and NMDA-type glutamate receptors in the shell of the rat nucleus accumbens
    • Gracy KN, Svingos AL, Pickel VM. Dual ultrastructural localization of m-opioid receptors and NMDA-type glutamate receptors in the shell of the rat nucleus accumbens. J Neurosci 1997; 17: 4839-48.
    • (1997) J Neurosci , vol.17 , pp. 4839-4848
    • Gracy, K.N.1    Svingos, A.L.2    Pickel, V.M.3
  • 69
    • 0034608272 scopus 로고    scopus 로고
    • Rostrocaudal variation in targeting of N-methyl-D-aspartate and mu-opioid receptors in the rat medial nucleus of the solitary tract
    • Huang J, Wang H, Pickel VM. Rostrocaudal variation in targeting of N-methyl-D-aspartate and mu-opioid receptors in the rat medial nucleus of the solitary tract. J Comp Neurol 2000; 421: 400-11.
    • (2000) J Comp Neurol , vol.421 , pp. 400-411
    • Huang, J.1    Wang, H.2    Pickel, V.M.3
  • 70
    • 70149120493 scopus 로고    scopus 로고
    • Ultrastructural relationship between N-methyl-D-aspartate-NR1 receptor subunit and mu-opioid receptor in the mouse central nucleus of the amygdala
    • Glass MJ, Vanyo L, Quimson L, Pickel VM. Ultrastructural relationship between N-methyl-D-aspartate-NR1 receptor subunit and mu-opioid receptor in the mouse central nucleus of the amygdala. Neuroscience 2009; 163: 857-67.
    • (2009) Neuroscience , vol.163 , pp. 857-867
    • Glass, M.J.1    Vanyo, L.2    Quimson, L.3    Pickel, V.M.4
  • 71
    • 0033553834 scopus 로고    scopus 로고
    • Frequent colocalization of mu opioid and NMDA-type glutamate receptors at postsynaptic sites in periaqueductal gray neurons
    • Commons KG, van Bockstaele EJ, Pfaff DW. Frequent colocalization of mu opioid and NMDA-type glutamate receptors at postsynaptic sites in periaqueductal gray neurons. J Comp Neurol 1999; 408: 549-59.
    • (1999) J Comp Neurol , vol.408 , pp. 549-559
    • Commons, K.G.1    van Bockstaele, E.J.2    Pfaff, D.W.3
  • 72
    • 5044224260 scopus 로고    scopus 로고
    • PDZ domain proteins of synapses
    • Kim E, Sheng M. PDZ domain proteins of synapses. Nat Rev Neurosci 2004; 5: 771-81.
    • (2004) Nat Rev Neurosci , vol.5 , pp. 771-781
    • Kim, E.1    Sheng, M.2
  • 73
    • 28544447721 scopus 로고    scopus 로고
    • mPins modulates PSD-95 and SAP102 trafficking and influences NMDA receptor surface expression
    • Sans N, Wang PY, Du Q, et al. mPins modulates PSD-95 and SAP102 trafficking and influences NMDA receptor surface expression. Nat Cell Biol 2005; 7: 1179-90.
    • (2005) Nat Cell Biol , vol.7 , pp. 1179-1190
    • Sans, N.1    Wang, P.Y.2    Du, Q.3
  • 74
    • 0000927212 scopus 로고
    • The major tyrosinephosphorylated protein in the postsynaptic density fraction is Nmethyl-D-aspartate receptor subunit 2B
    • Moon IS, Apperson ML, Kennedy MB. The major tyrosinephosphorylated protein in the postsynaptic density fraction is Nmethyl-D-aspartate receptor subunit 2B. Proc Natl Acad Sci U S A 1994; 91: 3954-8.
    • (1994) Proc Natl Acad Sci U S A , vol.91 , pp. 3954-3958
    • Moon, I.S.1    Apperson, M.L.2    Kennedy, M.B.3
  • 75
    • 0029093990 scopus 로고
    • Differential tyrosine phosphorylation of Nmethyl-D-aspartate receptor subunits
    • Lau LF, Huganir RL. Differential tyrosine phosphorylation of Nmethyl-D-aspartate receptor subunits. J Biol Chem 1995; 270: 20036-41.
    • (1995) J Biol Chem , vol.270 , pp. 20036-20041
    • Lau, L.F.1    Huganir, R.L.2
  • 76
    • 4143110134 scopus 로고    scopus 로고
    • Effect of protein kinase C blockade on phosphorylation of NR1 in dorsal horn and spinothalamic tract cells caused by intradermal capsaicin injection in rats
    • Zou X, Lin Q, Willis WD. Effect of protein kinase C blockade on phosphorylation of NR1 in dorsal horn and spinothalamic tract cells caused by intradermal capsaicin injection in rats. Brain Res 2004; 1020: 95-105.
    • (2004) Brain Res , vol.1020 , pp. 95-105
    • Zou, X.1    Lin, Q.2    Willis, W.D.3
  • 77
    • 23744503691 scopus 로고    scopus 로고
    • Protein phosphatase modulates the phosphorylation of spinal cord NMDA receptors in rats following intradermal injection of capsaicin
    • Zhang X, Wu J, Lei Y, Fang L, Willis WD. Protein phosphatase modulates the phosphorylation of spinal cord NMDA receptors in rats following intradermal injection of capsaicin. Brain Res Mol Brain Res 2005; 138: 264-72.
    • (2005) Brain Res Mol Brain Res , vol.138 , pp. 264-272
    • Zhang, X.1    Wu, J.2    Lei, Y.3    Fang, L.4    Willis, W.D.5
  • 78
    • 1842731881 scopus 로고    scopus 로고
    • Src kinases: A hub for NMDA receptor regulation
    • Salter MW, Kalia LV. Src kinases: a hub for NMDA receptor regulation. Nat Rev Neurosci 2004; 5: 317-28.
    • (2004) Nat Rev Neurosci , vol.5 , pp. 317-328
    • Salter, M.W.1    Kalia, L.V.2
  • 79
    • 1542614147 scopus 로고    scopus 로고
    • Interactions between Src family protein tyrosine kinases and PSD-95
    • Kalia LV, Salter MW. Interactions between Src family protein tyrosine kinases and PSD-95. Neuropharmacology 2003; 45: 720-8.
    • (2003) Neuropharmacology , vol.45 , pp. 720-728
    • Kalia, L.V.1    Salter, M.W.2
  • 80
    • 0033366541 scopus 로고    scopus 로고
    • G-protein-coupled receptors act via protein kinase C and Src to regulate NMDA receptors
    • Lu WY, Xiong ZG, Lei S, et al. G-protein-coupled receptors act via protein kinase C and Src to regulate NMDA receptors. Nat Neurosci 1999; 2: 331-8.
    • (1999) Nat Neurosci , vol.2 , pp. 331-338
    • Lu, W.Y.1    Xiong, Z.G.2    Lei, S.3
  • 81
    • 67349153367 scopus 로고    scopus 로고
    • Brain-specific Gaz interacts with Src tyrosine kinase to regulate Mu-opioid receptor-NMDAR signaling pathway
    • Sánchez-Blázquez P, Rodríguez-Muñoz M, de la Torre-Madrid E, Garzón J. Brain-specific Gaz interacts with Src tyrosine kinase to regulate Mu-opioid receptor-NMDAR signaling pathway. Cell Signal 2009; 21: 1444-54.
    • (2009) Cell Signal , vol.21 , pp. 1444-1454
    • Sánchez-Blázquez, P.1    Rodríguez-Muñoz, M.2    de la Torre-Madrid, E.3    Garzón, J.4
  • 82
    • 0032422059 scopus 로고    scopus 로고
    • Regulation of N-methyl-Daspartate receptor function by constitutively active protein kinase C
    • Xiong ZG, Raouf R, Lu WY, et al. Regulation of N-methyl-Daspartate receptor function by constitutively active protein kinase C. Mol Pharmacol 1998; 54: 1055-63.
    • (1998) Mol Pharmacol , vol.54 , pp. 1055-1063
    • Xiong, Z.G.1    Raouf, R.2    Lu, W.Y.3
  • 83
    • 5644256896 scopus 로고    scopus 로고
    • Group I metabotropic glutamate receptor NMDA receptor coupling and signaling cascade mediate spinal dorsal horn NMDA receptor 2B tyrosine phosphorylation associated with inflammatory hyperalgesia
    • Guo W, Wei F, Zou S, et al. Group I metabotropic glutamate receptor NMDA receptor coupling and signaling cascade mediate spinal dorsal horn NMDA receptor 2B tyrosine phosphorylation associated with inflammatory hyperalgesia. J Neurosci 2004; 24: 9161-73.
    • (2004) J Neurosci , vol.24 , pp. 9161-9173
    • Guo, W.1    Wei, F.2    Zou, S.3
  • 84
    • 0037101627 scopus 로고    scopus 로고
    • Tyrosine phosphorylation of the NR2B subunit of the NMDA receptor in the spinal cord during the development and maintenance of inflammatory hyperalgesia
    • Guo W, Zou S, Guan Y, et al. Tyrosine phosphorylation of the NR2B subunit of the NMDA receptor in the spinal cord during the development and maintenance of inflammatory hyperalgesia. J Neurosci 2002; 22: 6208-17.
    • (2002) J Neurosci , vol.22 , pp. 6208-6217
    • Guo, W.1    Zou, S.2    Guan, Y.3
  • 85
    • 0026419327 scopus 로고
    • Molecular cloning and characterization of the rat NMDA receptor
    • Moriyoshi K, Masu M, Ishii T, et al. Molecular cloning and characterization of the rat NMDA receptor. Nature 1991; 354: 31-7.
    • (1991) Nature , vol.354 , pp. 31-37
    • Moriyoshi, K.1    Masu, M.2    Ishii, T.3
  • 86
    • 0028343648 scopus 로고
    • Developmental and regional expression in the rat brain and functional properties of four NMDA receptors
    • Monyer H, Burnashev N, Laurie DJ, Sakmann B, Seeburg PH. Developmental and regional expression in the rat brain and functional properties of four NMDA receptors. Neuron 1994; 12: 529-40.
    • (1994) Neuron , vol.12 , pp. 529-540
    • Monyer, H.1    Burnashev, N.2    Laurie, D.J.3    Sakmann, B.4    Seeburg, P.H.5
  • 87
    • 0028855530 scopus 로고
    • Down-regulation of Nmethyl-D-aspartate (NMDA) receptors of brain regions and spinal cord of rats treated chronically with morphine
    • Bhargava HN, Reddy PL, Gudehithlu KP. Down-regulation of Nmethyl-D-aspartate (NMDA) receptors of brain regions and spinal cord of rats treated chronically with morphine. Gen Pharmacol 1995; 26: 131-6.
    • (1995) Gen Pharmacol , vol.26 , pp. 131-136
    • Bhargava, H.N.1    Reddy, P.L.2    Gudehithlu, K.P.3
  • 88
    • 4644261905 scopus 로고    scopus 로고
    • Chronic morphine treatment alters N-methyl-D-aspartate receptors in freshly isolated neurons from nucleus accumbens
    • Martin G, Guadano-Ferraz A, Morte B, et al. Chronic morphine treatment alters N-methyl-D-aspartate receptors in freshly isolated neurons from nucleus accumbens. J Pharmacol Exp Ther 2004; 311: 265-73.
    • (2004) J Pharmacol Exp Ther , vol.311 , pp. 265-273
    • Martin, G.1    Guadano-Ferraz, A.2    Morte, B.3
  • 89
    • 33644905540 scopus 로고    scopus 로고
    • Chronic morphine treatment alters expression of N-methyl-Daspartate receptor subunits in the extended amygdala
    • Bajo M, Crawford EF, Roberto M, Madamba SG, Siggins GR. Chronic morphine treatment alters expression of N-methyl-Daspartate receptor subunits in the extended amygdala. J Neurosci Res 2006; 83: 532-7.
    • (2006) J Neurosci Res , vol.83 , pp. 532-537
    • Bajo, M.1    Crawford, E.F.2    Roberto, M.3    Madamba, S.G.4    Siggins, G.R.5
  • 90
    • 34247170735 scopus 로고    scopus 로고
    • Nucleus accumbens NMDA receptor subunit expression and function is enhanced in morphine-dependent rats
    • Murray F, Harrison NJ, Grimwood S, Bristow LJ, Hutson PH. Nucleus accumbens NMDA receptor subunit expression and function is enhanced in morphine-dependent rats. Eur J Pharmacol 2007; 562: 191-7.
    • (2007) Eur J Pharmacol , vol.562 , pp. 191-197
    • Murray, F.1    Harrison, N.J.2    Grimwood, S.3    Bristow, L.J.4    Hutson, P.H.5
  • 91
    • 0038124239 scopus 로고    scopus 로고
    • Region-specific changes in NMDA receptor mRNA induced by chronic morphine treatment are prevented by the co-administration of the competitive NMDA receptor antagonist LY274614
    • Zhu H, Brodsky M, Gorman AL, Inturrisi CE. Region-specific changes in NMDA receptor mRNA induced by chronic morphine treatment are prevented by the co-administration of the competitive NMDA receptor antagonist LY274614. Brain Res Mol Brain Res 2003; 114: 154-62.
    • (2003) Brain Res Mol Brain Res , vol.114 , pp. 154-162
    • Zhu, H.1    Brodsky, M.2    Gorman, A.L.3    Inturrisi, C.E.4
  • 94
    • 0035895932 scopus 로고    scopus 로고
    • Threonine 180 is required for G-protein-coupled receptor kinase 3-and barrestin 2-mediated desensitization of the m-opioid receptor in Xenopus oocytes
    • Celver JP, Lowe J, Kovoor A, Gurevich VV, Chavkin C. Threonine 180 is required for G-protein-coupled receptor kinase 3-and barrestin 2-mediated desensitization of the m-opioid receptor in Xenopus oocytes. J Biol Chem 2001; 276: 4894-900.
    • (2001) J Biol Chem , vol.276 , pp. 4894-4900
    • Celver, J.P.1    Lowe, J.2    Kovoor, A.3    Gurevich, V.V.4    Chavkin, C.5
  • 95
    • 0040735779 scopus 로고    scopus 로고
    • Role for the C-terminus in agonistinduced m opioid receptor phosphorylation and desensitization
    • Deng HB, Yu Y, Pak Y, et al. Role for the C-terminus in agonistinduced m opioid receptor phosphorylation and desensitization. Biochemistry 2000; 39: 5492-9.
    • (2000) Biochemistry , vol.39 , pp. 5492-5499
    • Deng, H.B.1    Yu, Y.2    Pak, Y.3
  • 96
    • 0035918284 scopus 로고    scopus 로고
    • 375 residues within the carboxyl tail differentially regulates m-opioid receptor internalization
    • 375 residues within the carboxyl tail differentially regulates m-opioid receptor internalization. J Biol Chem 2001; 276: 12774-80.
    • (2001) J Biol Chem , vol.276 , pp. 12774-12780
    • El Kouhen, R.1    Burd, A.L.2    Erickson-Herbrandson, L.J.3
  • 97
    • 0033944519 scopus 로고    scopus 로고
    • Allelic variation S268P of the human m-opioid receptor affects both desensitization and G protein coupling
    • Koch T, Kroslak T, Averbeck M, et al. Allelic variation S268P of the human m-opioid receptor affects both desensitization and G protein coupling. Mol Pharmacol 2000; 58: 328-34.
    • (2000) Mol Pharmacol , vol.58 , pp. 328-334
    • Koch, T.1    Kroslak, T.2    Averbeck, M.3
  • 98
    • 0030875721 scopus 로고    scopus 로고
    • Agonist-induced desensitization of the m opioid receptor is determined by threonine 394 preceded by acidic amino acids in the COOH-terminal tail
    • Pak Y, O'Dowd BF, George SR. Agonist-induced desensitization of the m opioid receptor is determined by threonine 394 preceded by acidic amino acids in the COOH-terminal tail. J Biol Chem 1997; 272: 24961-5.
    • (1997) J Biol Chem , vol.272 , pp. 24961-24965
    • Pak, Y.1    O'Dowd, B.F.2    George, S.R.3
  • 99
    • 23944437411 scopus 로고    scopus 로고
    • Morphine promotes rapid, arrestin-dependent endocytosis of m-opioid receptors in striatal neurons
    • Haberstock-Debic H, Kim KA, Yu YJ, von Zastrow M. Morphine promotes rapid, arrestin-dependent endocytosis of m-opioid receptors in striatal neurons. J Neurosci 2005; 25: 7847-57.
    • (2005) J Neurosci , vol.25 , pp. 7847-7857
    • Haberstock-Debic, H.1    Kim, K.A.2    Yu, Y.J.3    von Zastrow, M.4
  • 100
    • 18544411827 scopus 로고    scopus 로고
    • m-Opioid receptor internalization: Opiate drugs have differential effects on a conserved endocytic mechanism in vitro and in the mammalian brain
    • Keith DE, Anton B, Murray SR, et al. m-Opioid receptor internalization: opiate drugs have differential effects on a conserved endocytic mechanism in vitro and in the mammalian brain. Mol Pharmacol 1998; 53: 377-84.
    • (1998) Mol Pharmacol , vol.53 , pp. 377-384
    • Keith, D.E.1    Anton, B.2    Murray, S.R.3
  • 101
    • 0029856581 scopus 로고    scopus 로고
    • Functional analysis of cloned opioid receptors in transfected cell lines
    • Piros ET, Hales TG, Evans CJ. Functional analysis of cloned opioid receptors in transfected cell lines. Neurochem Res 1996; 21: 1277-85.
    • (1996) Neurochem Res , vol.21 , pp. 1277-1285
    • Piros, E.T.1    Hales, T.G.2    Evans, C.J.3
  • 102
    • 0034681348 scopus 로고    scopus 로고
    • Tolerance to morphine at the m-opioid receptor differentially induced by cAMP-dependent protein kinase activation and morphine
    • Wang Z, Sadee W. Tolerance to morphine at the m-opioid receptor differentially induced by cAMP-dependent protein kinase activation and morphine. Eur J Pharmacol 2000; 389: 165-71.
    • (2000) Eur J Pharmacol , vol.389 , pp. 165-171
    • Wang, Z.1    Sadee, W.2
  • 103
    • 0034176522 scopus 로고    scopus 로고
    • Spinal PKC activity and expression: Role in tolerance produced by continuous spinal morphine infusion
    • Granados-Soto V, Kalcheva I, Hua X, Newton A, Yaksh TL. Spinal PKC activity and expression: role in tolerance produced by continuous spinal morphine infusion. Pain 2000; 85: 395-404.
    • (2000) Pain , vol.85 , pp. 395-404
    • Granados-Soto, V.1    Kalcheva, I.2    Hua, X.3    Newton, A.4    Yaksh, T.L.5
  • 104
    • 0029042975 scopus 로고
    • Inhibition of protein kinase C, but not of protein kinase A, blocks the development of acute antinociceptive tolerance to an intrathecally administered mopioid receptor agonist in the mouse
    • Narita M, Narita M, Mizoguchi H, Tseng LF. Inhibition of protein kinase C, but not of protein kinase A, blocks the development of acute antinociceptive tolerance to an intrathecally administered mopioid receptor agonist in the mouse. Eur J Pharmacol 1995; 280: R1-R3.
    • (1995) Eur J Pharmacol , vol.280
    • Narita, M.1    Narita, M.2    Mizoguchi, H.3    Tseng, L.F.4
  • 105
    • 2642511324 scopus 로고    scopus 로고
    • PKC and PKA inhibitors reverse tolerance to morphine-induced hypothermia and supraspinal analgesia in mice
    • Javed RR, Dewey WL, Smith PA, Smith FL. PKC and PKA inhibitors reverse tolerance to morphine-induced hypothermia and supraspinal analgesia in mice. Eur J Pharmacol 2004; 492: 149-57.
    • (2004) Eur J Pharmacol , vol.492 , pp. 149-157
    • Javed, R.R.1    Dewey, W.L.2    Smith, P.A.3    Smith, F.L.4
  • 106
    • 0142026927 scopus 로고    scopus 로고
    • The expression of a high level of morphine antinociceptive tolerance in mice involves both PKC and PKA
    • Smith FL, Javed RR, Elzey MJ, Dewey WL. The expression of a high level of morphine antinociceptive tolerance in mice involves both PKC and PKA. Brain Res 2003; 985: 78-88.
    • (2003) Brain Res , vol.985 , pp. 78-88
    • Smith, F.L.1    Javed, R.R.2    Elzey, M.J.3    Dewey, W.L.4
  • 107
    • 33845966383 scopus 로고    scopus 로고
    • PKC and PKA inhibitors reinstate morphine-induced behaviors in morphine tolerant mice
    • Smith FL, Javed RR, Smith PA, Dewey WL, Gabra BH. PKC and PKA inhibitors reinstate morphine-induced behaviors in morphine tolerant mice. Pharmacol Res 2006; 54: 474-80.
    • (2006) Pharmacol Res , vol.54 , pp. 474-480
    • Smith, F.L.1    Javed, R.R.2    Smith, P.A.3    Dewey, W.L.4    Gabra, B.H.5
  • 108
    • 0037146929 scopus 로고    scopus 로고
    • Prolonged reversal of morphine tolerance with no reversal of dependence by protein kinase C inhibitors
    • Smith FL, Javed R, Elzey MJ, et al. Prolonged reversal of morphine tolerance with no reversal of dependence by protein kinase C inhibitors. Brain Res 2002; 958: 28-35.
    • (2002) Brain Res , vol.958 , pp. 28-35
    • Smith, F.L.1    Javed, R.2    Elzey, M.J.3
  • 109
    • 0034011235 scopus 로고    scopus 로고
    • Protein kinase C-mediated acute tolerance to peripheral m-opioid analgesia in the bradykinin-nociception test in mice
    • Inoue M, Ueda H. Protein kinase C-mediated acute tolerance to peripheral m-opioid analgesia in the bradykinin-nociception test in mice. J Pharmacol Exp Ther 2000; 293: 662-9.
    • (2000) J Pharmacol Exp Ther , vol.293 , pp. 662-669
    • Inoue, M.1    Ueda, H.2
  • 110
    • 0028567776 scopus 로고
    • A protein kinase inhibitor, H-7, inhibits the development of tolerance to opioid antinociception
    • Narita M, Feng Y, Makimura M, Hoskins B, Ho IK. A protein kinase inhibitor, H-7, inhibits the development of tolerance to opioid antinociception. Eur J Pharmacol 1994; 271: 543-5.
    • (1994) Eur J Pharmacol , vol.271 , pp. 543-545
    • Narita, M.1    Feng, Y.2    Makimura, M.3    Hoskins, B.4    Ho, I.K.5
  • 111
    • 2642520288 scopus 로고    scopus 로고
    • Effects of mGlu1 and mGlu5 metabotropic glutamate antagonists to reverse morphine tolerance in mice
    • Smith FL, Smith PA, Dewey WL, Javed RR. Effects of mGlu1 and mGlu5 metabotropic glutamate antagonists to reverse morphine tolerance in mice. Eur J Pharmacol 2004; 492: 137-42.
    • (2004) Eur J Pharmacol , vol.492 , pp. 137-142
    • Smith, F.L.1    Smith, P.A.2    Dewey, W.L.3    Javed, R.R.4
  • 112
    • 0029060391 scopus 로고
    • Protein kinase C and lipid signaling for sustained cellular responses
    • Nishizuka Y. Protein kinase C and lipid signaling for sustained cellular responses. FASEB J 1995; 9: 484-96.
    • (1995) FASEB J , vol.9 , pp. 484-496
    • Nishizuka, Y.1
  • 113
    • 79957530585 scopus 로고    scopus 로고
    • NO-released zinc supports the simultaneous binding of Raf-1 and PKCgamma cysteine-rich domains to HINT1 protein at the mu-opioid receptor
    • Rodríguez-Muñoz M, Torre-Madrid E, Sánchez-Blázquez P, Garzón J. NO-released zinc supports the simultaneous binding of Raf-1 and PKCgamma cysteine-rich domains to HINT1 protein at the mu-opioid receptor. Antioxid Redox Signal 2011; 14: 2413-25.
    • (2011) Antioxid Redox Signal , vol.14 , pp. 2413-2425
    • Rodríguez-Muñoz, M.1    Torre-Madrid, E.2    Sánchez-Blázquez, P.3    Garzón, J.4
  • 114
    • 0037113925 scopus 로고    scopus 로고
    • Zinc release from protein kinase C as the common event during activation by lipid second messenger or reactive oxygen
    • Korichneva I, Hoyos B, Chua R, Levi E, Hammerling U. Zinc release from protein kinase C as the common event during activation by lipid second messenger or reactive oxygen. J Biol Chem 2002; 277: 44327-31.
    • (2002) J Biol Chem , vol.277 , pp. 44327-44331
    • Korichneva, I.1    Hoyos, B.2    Chua, R.3    Levi, E.4    Hammerling, U.5
  • 115
    • 79551674547 scopus 로고    scopus 로고
    • Are zinc-finger domains of protein kinase C dynamic structures that unfold by lipid or redox activation?
    • Zhao F, Ilbert M, Varadan R, et al. Are zinc-finger domains of protein kinase C dynamic structures that unfold by lipid or redox activation? Antioxid Redox Signal 2011; 14: 757-66.
    • (2011) Antioxid Redox Signal , vol.14 , pp. 757-766
    • Zhao, F.1    Ilbert, M.2    Varadan, R.3
  • 116
    • 0037129803 scopus 로고    scopus 로고
    • Phosphorylation of p47phox sites by PKC alpha, beta II, delta, and zeta: Effect on binding to p22phox and on NADPH oxidase activation
    • Fontayne A, Dang PM, Gougerot-Pocidalo MA, El-Benna J. Phosphorylation of p47phox sites by PKC alpha, beta II, delta, and zeta: effect on binding to p22phox and on NADPH oxidase activation. Biochemistry 2002; 41: 7743-50.
    • (2002) Biochemistry , vol.41 , pp. 7743-7750
    • Fontayne, A.1    Dang, P.M.2    Gougerot-Pocidalo, M.A.3    El-Benna, J.4
  • 117
    • 70349356001 scopus 로고    scopus 로고
    • Emerging evidence for the importance of phosphorylation in the regulation of NADPH oxidases
    • Bokoch GM, Diebold B, Kim JS, Gianni D. Emerging evidence for the importance of phosphorylation in the regulation of NADPH oxidases. Antioxid Redox Signal 2009; 11: 2429-41.
    • (2009) Antioxid Redox Signal , vol.11 , pp. 2429-2441
    • Bokoch, G.M.1    Diebold, B.2    Kim, J.S.3    Gianni, D.4
  • 118
    • 0034604723 scopus 로고    scopus 로고
    • Superoxide-induced stimulation of protein kinase C via thiol modification and modulation of zinc content
    • Knapp LT, Klann E. Superoxide-induced stimulation of protein kinase C via thiol modification and modulation of zinc content. J Biol Chem 2000; 275: 24136-45.
    • (2000) J Biol Chem , vol.275 , pp. 24136-24145
    • Knapp, L.T.1    Klann, E.2
  • 119
    • 77956188649 scopus 로고    scopus 로고
    • Spinal NADPH oxidase is a source of superoxide in the development of morphine-induced hyperalgesia and antinociceptive tolerance
    • Doyle T, Bryant L, Muscoli C, et al. Spinal NADPH oxidase is a source of superoxide in the development of morphine-induced hyperalgesia and antinociceptive tolerance. Neurosci Lett 2010; 483: 85-9.
    • (2010) Neurosci Lett , vol.483 , pp. 85-89
    • Doyle, T.1    Bryant, L.2    Muscoli, C.3
  • 120
    • 0028948646 scopus 로고
    • Increases in protein kinase C gamma immunoreactivity in the spinal cord of rats associated with tolerance to the analgesic effects of morphine
    • Mao J, Price DD, Phillips LL, Lu J, Mayer DJ. Increases in protein kinase C gamma immunoreactivity in the spinal cord of rats associated with tolerance to the analgesic effects of morphine. Brain Res 1995; 677: 257-67.
    • (1995) Brain Res , vol.677 , pp. 257-267
    • Mao, J.1    Price, D.D.2    Phillips, L.L.3    Lu, J.4    Mayer, D.J.5
  • 121
    • 0030881680 scopus 로고    scopus 로고
    • Preserved acute pain and reduced neuropathic pain in mice lacking PKCg
    • Malmberg AB, Chen C, Tonegawa S, Basbaum AI. Preserved acute pain and reduced neuropathic pain in mice lacking PKCg. Science 1997; 278: 279-83.
    • (1997) Science , vol.278 , pp. 279-283
    • Malmberg, A.B.1    Chen, C.2    Tonegawa, S.3    Basbaum, A.I.4
  • 122
    • 0035805632 scopus 로고    scopus 로고
    • Enhanced m-opioid responses in the spinal cord of mice lacking protein kinase Cg isoform
    • Narita M, Mizoguchi H, Suzuki T, et al. Enhanced m-opioid responses in the spinal cord of mice lacking protein kinase Cg isoform. J Biol Chem 2001; 276: 15409-14.
    • (2001) J Biol Chem , vol.276 , pp. 15409-15414
    • Narita, M.1    Mizoguchi, H.2    Suzuki, T.3
  • 123
    • 0035214481 scopus 로고    scopus 로고
    • Reduced development of tolerance to the analgesic effects of morphine and clonidine in PKCg mutant mice
    • Zeitz KP, Malmberg AB, Gilbert H, Basbaum AI. Reduced development of tolerance to the analgesic effects of morphine and clonidine in PKCg mutant mice. Pain 2001; 94: 245-53.
    • (2001) Pain , vol.94 , pp. 245-253
    • Zeitz, K.P.1    Malmberg, A.B.2    Gilbert, H.3    Basbaum, A.I.4
  • 124
    • 0037008344 scopus 로고    scopus 로고
    • Inhibition of spinal protein kinase Ca expression by an antisense oligonucleotide attenuates morphine infusion-induced tolerance
    • Hua XY, Moore A, Malkmus S, et al. Inhibition of spinal protein kinase Ca expression by an antisense oligonucleotide attenuates morphine infusion-induced tolerance. Neuroscience 2002; 113: 99-107.
    • (2002) Neuroscience , vol.113 , pp. 99-107
    • Hua, X.Y.1    Moore, A.2    Malkmus, S.3
  • 125
    • 0030935925 scopus 로고    scopus 로고
    • Modulation of immunoreactive protein kinase C-a and b isoforms and G proteins by acute and chronic treatments with morphine and other opiate drugs in rat brain
    • Ventayol P, Busquets X, Garcia-Sevilla JA. Modulation of immunoreactive protein kinase C-a and b isoforms and G proteins by acute and chronic treatments with morphine and other opiate drugs in rat brain. Naunyn Schmiedebergs Arch Pharmacol 1997; 355: 491-500.
    • (1997) Naunyn Schmiedebergs Arch Pharmacol , vol.355 , pp. 491-500
    • Ventayol, P.1    Busquets, X.2    Garcia-Sevilla, J.A.3
  • 126
    • 33845681648 scopus 로고    scopus 로고
    • Determination of the role of conventional, novel and atypical PKC isoforms in the expression of morphine tolerance in mice
    • Smith FL, Gabra BH, Smith PA, Redwood MC, Dewey WL. Determination of the role of conventional, novel and atypical PKC isoforms in the expression of morphine tolerance in mice. Pain 2007; 127: 129-39.
    • (2007) Pain , vol.127 , pp. 129-139
    • Smith, F.L.1    Gabra, B.H.2    Smith, P.A.3    Redwood, M.C.4    Dewey, W.L.5
  • 127
    • 0023198109 scopus 로고
    • Binding of a nuclear protein to the cyclic-AMP response element of the somatostatin gene
    • Montminy MR, Bilezikjian LM. Binding of a nuclear protein to the cyclic-AMP response element of the somatostatin gene. Nature 1987; 328: 175-8.
    • (1987) Nature , vol.328 , pp. 175-178
    • Montminy, M.R.1    Bilezikjian, L.M.2
  • 128
    • 0027439945 scopus 로고
    • NMDA receptor activation increases cyclic AMP in area CA1 of the hippocampus via calcium/calmodulin stimulation of adenylyl cyclase
    • Chetkovich DM, Sweatt JD. NMDA receptor activation increases cyclic AMP in area CA1 of the hippocampus via calcium/calmodulin stimulation of adenylyl cyclase. J Neurochem 1993; 61: 1933-42.
    • (1993) J Neurochem , vol.61 , pp. 1933-1942
    • Chetkovich, D.M.1    Sweatt, J.D.2
  • 129
    • 0035797491 scopus 로고    scopus 로고
    • A model of synaptic memory: A CaMKII/PP1 switch that potentiates transmission by organizing an AMPA receptor anchoring assembly
    • Lisman JE, Zhabotinsky AM. A model of synaptic memory: a CaMKII/PP1 switch that potentiates transmission by organizing an AMPA receptor anchoring assembly. Neuron 2001; 31: 191-201.
    • (2001) Neuron , vol.31 , pp. 191-201
    • Lisman, J.E.1    Zhabotinsky, A.M.2
  • 130
    • 0041836230 scopus 로고    scopus 로고
    • An ultrasensitive Ca2+/calmodulin-dependent protein kinase II-protein phosphatase 1 switch facilitates specificity in postsynaptic calcium signaling
    • Bradshaw JM, Kubota Y, Meyer T, Schulman H. An ultrasensitive Ca2+/calmodulin-dependent protein kinase II-protein phosphatase 1 switch facilitates specificity in postsynaptic calcium signaling. Proc Natl Acad Sci U S A 2003; 100: 10512-7.
    • (2003) Proc Natl Acad Sci U S A , vol.100 , pp. 10512-10517
    • Bradshaw, J.M.1    Kubota, Y.2    Meyer, T.3    Schulman, H.4
  • 131
    • 0032546994 scopus 로고    scopus 로고
    • Gating of CaMKII by cAMP-regulated protein phosphatase activity during LTP
    • Blitzer RD, Connor JH, Brown GP, et al. Gating of CaMKII by cAMP-regulated protein phosphatase activity during LTP. Science 1998; 280: 1940-2.
    • (1998) Science , vol.280 , pp. 1940-1942
    • Blitzer, R.D.1    Connor, J.H.2    Brown, G.P.3
  • 132
    • 0033118508 scopus 로고    scopus 로고
    • Adenylyl cyclase activation modulates activity-dependent changes in synaptic strength and Ca2+/calmodulin-dependent kinase II autophosphorylation
    • Makhinson M, Chotiner JK, Watson JB, O'Dell TJ. Adenylyl cyclase activation modulates activity-dependent changes in synaptic strength and Ca2+/calmodulin-dependent kinase II autophosphorylation. J Neurosci 1999; 19: 2500-10.
    • (1999) J Neurosci , vol.19 , pp. 2500-2510
    • Makhinson, M.1    Chotiner, J.K.2    Watson, J.B.3    O'Dell, T.J.4
  • 133
    • 0034567867 scopus 로고    scopus 로고
    • Role of cAMP-dependent protein kinase (PKA) in opioid agonist-induced m-opioid receptor downregulation and tolerance in mice
    • Shen J, Benedict GA, Gallagher A, Stafford K, Yoburn BC. Role of cAMP-dependent protein kinase (PKA) in opioid agonist-induced m-opioid receptor downregulation and tolerance in mice. Synapse 2000; 38: 322-7.
    • (2000) Synapse , vol.38 , pp. 322-327
    • Shen, J.1    Benedict, G.A.2    Gallagher, A.3    Stafford, K.4    Yoburn, B.C.5
  • 134
    • 0031561764 scopus 로고    scopus 로고
    • Effects of spinal versus supraspinal administration of cyclic nucleotide-dependent protein kinase inhibitors on morphine tolerance in mice
    • Bernstein MA, Welch SP. Effects of spinal versus supraspinal administration of cyclic nucleotide-dependent protein kinase inhibitors on morphine tolerance in mice. Drug Alcohol Depend 1997; 44: 41-6.
    • (1997) Drug Alcohol Depend , vol.44 , pp. 41-46
    • Bernstein, M.A.1    Welch, S.P.2
  • 135
    • 0034727888 scopus 로고    scopus 로고
    • Colocalization of the m-opioid receptor and calcium/calmodulin-dependent kinase II in distinct pain-processing brain regions
    • Brüggemann I, Schulz S, Wiborny D, Höllt V. Colocalization of the m-opioid receptor and calcium/calmodulin-dependent kinase II in distinct pain-processing brain regions. Brain Res Mol Brain Res 2000; 85: 239-50.
    • (2000) Brain Res Mol Brain Res , vol.85 , pp. 239-250
    • Brüggemann, I.1    Schulz, S.2    Wiborny, D.3    Höllt, V.4
  • 136
    • 0026613087 scopus 로고
    • Activation of Ca2+/calmodulin-dependent protein kinase II and protein kinase C by glutamate in cultured rat hippocampal neurons
    • Fukunaga K, Soderling TR, Miyamoto E. Activation of Ca2+/calmodulin-dependent protein kinase II and protein kinase C by glutamate in cultured rat hippocampal neurons. J Biol Chem 1992; 267: 22527-33.
    • (1992) J Biol Chem , vol.267 , pp. 22527-22533
    • Fukunaga, K.1    Soderling, T.R.2    Miyamoto, E.3
  • 137
    • 0347990407 scopus 로고    scopus 로고
    • Increased expression of Ca2+/calmodulin-dependent protein kinase IIa during chronic morphine exposure
    • Liang D, Li X, Clark JD. Increased expression of Ca2+/calmodulin-dependent protein kinase IIa during chronic morphine exposure. Neuroscience 2004; 123: 769-75.
    • (2004) Neuroscience , vol.123 , pp. 769-775
    • Liang, D.1    Li, X.2    Clark, J.D.3
  • 138
    • 0032586887 scopus 로고    scopus 로고
    • Modulation of Ca2+/calmodulindependent protein kinase II activity by acute and chronic morphine administration in rat hippocampus: Differential regulation of a and b isoforms
    • Lou L, Zhou T, Wang P, Pei G. Modulation of Ca2+/calmodulindependent protein kinase II activity by acute and chronic morphine administration in rat hippocampus: differential regulation of a and b isoforms. Mol Pharmacol 1999; 55: 557-63.
    • (1999) Mol Pharmacol , vol.55 , pp. 557-563
    • Lou, L.1    Zhou, T.2    Wang, P.3    Pei, G.4
  • 139
    • 0033039909 scopus 로고    scopus 로고
    • Inhibition of calcium/calmodulin-dependent protein kinase II in rat hippocampus attenuates morphine tolerance and dependence
    • Fan GH, Wang LZ, Qiu HC, Ma L, Pei G. Inhibition of calcium/calmodulin-dependent protein kinase II in rat hippocampus attenuates morphine tolerance and dependence. Mol Pharmacol 1999; 56: 39-45.
    • (1999) Mol Pharmacol , vol.56 , pp. 39-45
    • Fan, G.H.1    Wang, L.Z.2    Qiu, H.C.3    Ma, L.4    Pei, G.5
  • 140
    • 0034703585 scopus 로고    scopus 로고
    • Inhibition of the amygdala and hippocampal calcium/calmodulin-dependent protein kinase II attenuates the dependence and relapse to morphine differently in rats
    • Lu L, Zeng S, Liu D, Ceng X. Inhibition of the amygdala and hippocampal calcium/calmodulin-dependent protein kinase II attenuates the dependence and relapse to morphine differently in rats. Neurosci Lett 2000; 291: 191-5.
    • (2000) Neurosci Lett , vol.291 , pp. 191-195
    • Lu, L.1    Zeng, S.2    Liu, D.3    Ceng, X.4
  • 141
    • 0037470848 scopus 로고    scopus 로고
    • 2+/calmodulin-dependent protein kinase II
    • 2+/calmodulin-dependent protein kinase II. Eur J Pharmacol 2003; 465: 199-200.
    • (2003) Eur J Pharmacol , vol.465 , pp. 199-200
    • Wang, Z.J.1    Tang, L.2    Xin, L.3
  • 142
    • 0027981633 scopus 로고
    • Calcium/calmodulindependent protein kinase types II and IV differentially regulate CREB-dependent gene expression
    • Matthews RP, Guthrie CR, Wailes LM, et al. Calcium/calmodulindependent protein kinase types II and IV differentially regulate CREB-dependent gene expression. Mol Cell Biol 1994; 14: 6107-16.
    • (1994) Mol Cell Biol , vol.14 , pp. 6107-6116
    • Matthews, R.P.1    Guthrie, C.R.2    Wailes, L.M.3
  • 143
    • 0038813716 scopus 로고    scopus 로고
    • Regulation of phosducin-like protein by casein kinase 2 and N-terminal splicing
    • Humrich J, Bermel C, Grubel T, Quitterer U, Lohse MJ. Regulation of phosducin-like protein by casein kinase 2 and N-terminal splicing. J Biol Chem 2003; 278: 4474-81.
    • (2003) J Biol Chem , vol.278 , pp. 4474-4481
    • Humrich, J.1    Bermel, C.2    Grubel, T.3    Quitterer, U.4    Lohse, M.J.5
  • 144
    • 0027449386 scopus 로고
    • Phosducin-like protein: An ethanol-responsive potential modulator of guanine nucleotidebinding protein function
    • Miles MF, Barhite S, Sganga M, Elliott M. Phosducin-like protein: an ethanol-responsive potential modulator of guanine nucleotidebinding protein function. Proc Natl Acad Sci U S A 1993; 90: 10831-5.
    • (1993) Proc Natl Acad Sci U S A , vol.90 , pp. 10831-10835
    • Miles, M.F.1    Barhite, S.2    Sganga, M.3    Elliott, M.4
  • 145
    • 38149077089 scopus 로고    scopus 로고
    • Calcium/calmodulin-dependent protein kinase II supports morphine antinociceptive tolerance by phosphorylation of glycosylated phosducin-like protein
    • Sánchez-Blázquez P, Rodríguez-Muñoz M, Montero C, de la Torre-Madrid E, Garzón J. Calcium/calmodulin-dependent protein kinase II supports morphine antinociceptive tolerance by phosphorylation of glycosylated phosducin-like protein. Neuropharmacology 2008; 54: 319-30.
    • (2008) Neuropharmacology , vol.54 , pp. 319-330
    • Sánchez-Blázquez, P.1    Rodríguez-Muñoz, M.2    Montero, C.3    de la Torre-Madrid, E.4    Garzón, J.5
  • 146
    • 73049103800 scopus 로고    scopus 로고
    • The role of mitogen-activated protein kinase (MAPK) in morphine tolerance and dependence
    • Chen Y, Sommer C. The role of mitogen-activated protein kinase (MAPK) in morphine tolerance and dependence. Mol Neurobiol 2009; 40: 101-7.
    • (2009) Mol Neurobiol , vol.40 , pp. 101-107
    • Chen, Y.1    Sommer, C.2
  • 147
    • 0345687446 scopus 로고    scopus 로고
    • Phosphoinositide 3-kinase cascade facilitates mu-opioid desensitization in sensory neurons by altering G-protein-effector interactions
    • Tan M, Groszer M, Tan AM, et al. Phosphoinositide 3-kinase cascade facilitates mu-opioid desensitization in sensory neurons by altering G-protein-effector interactions. J Neurosci 2003; 23: 10292-301.
    • (2003) J Neurosci , vol.23 , pp. 10292-10301
    • Tan, M.1    Groszer, M.2    Tan, A.M.3
  • 148
    • 0035783434 scopus 로고    scopus 로고
    • Chronic morphine exposure increases the phosphorylation of MAP kinases and the transcription factor CREB in dorsal root ganglion neurons: An in vitro and in vivo study
    • Ma W, Zheng WH, Powell K, Jhamandas K, Quirion R. Chronic morphine exposure increases the phosphorylation of MAP kinases and the transcription factor CREB in dorsal root ganglion neurons: an in vitro and in vivo study. Eur J Neurosci 2001; 14: 1091-104.
    • (2001) Eur J Neurosci , vol.14 , pp. 1091-1104
    • Ma, W.1    Zheng, W.H.2    Powell, K.3    Jhamandas, K.4    Quirion, R.5
  • 149
    • 31344448048 scopus 로고    scopus 로고
    • Activation of p38 mitogen-activated protein kinase in spinal microglia mediates morphine antinociceptive tolerance
    • Cui Y, Chen Y, Zhi JL, et al. Activation of p38 mitogen-activated protein kinase in spinal microglia mediates morphine antinociceptive tolerance. Brain Res 2006; 1069: 235-43.
    • (2006) Brain Res , vol.1069 , pp. 235-243
    • Cui, Y.1    Chen, Y.2    Zhi, J.L.3
  • 150
    • 28644433404 scopus 로고    scopus 로고
    • Activation of the spinal ERK signaling pathway contributes naloxone-precipitated withdrawal in morphine-dependent rats
    • Cao JL, He JH, Ding HL, Zeng YM. Activation of the spinal ERK signaling pathway contributes naloxone-precipitated withdrawal in morphine-dependent rats. Pain 2005; 118: 336-49.
    • (2005) Pain , vol.118 , pp. 336-349
    • Cao, J.L.1    He, J.H.2    Ding, H.L.3    Zeng, Y.M.4
  • 151
    • 0032509718 scopus 로고    scopus 로고
    • Inhibition of protein phosphatases alters the expression of morphine tolerance in mice
    • Bernstein MA, Welch SP. Inhibition of protein phosphatases alters the expression of morphine tolerance in mice. Eur J Pharmacol 1998; 341: 173-7.
    • (1998) Eur J Pharmacol , vol.341 , pp. 173-177
    • Bernstein, M.A.1    Welch, S.P.2
  • 152
    • 0037466354 scopus 로고    scopus 로고
    • Inhibition by immunophilin ligands of morphine-induced tolerance and dependence in guinea pig ileum
    • Mehr SE, Samini M, Namiranian K, et al. Inhibition by immunophilin ligands of morphine-induced tolerance and dependence in guinea pig ileum. Eur J Pharmacol 2003; 467: 205-10.
    • (2003) Eur J Pharmacol , vol.467 , pp. 205-210
    • Mehr, S.E.1    Samini, M.2    Namiranian, K.3
  • 153
    • 0037368572 scopus 로고    scopus 로고
    • The effects of FK506 on the development and expression of morphine tolerance and dependence in mice
    • Homayoun H, Khavandgar S, Mehr SE, Namiranian K, Dehpour AR. The effects of FK506 on the development and expression of morphine tolerance and dependence in mice. Behav Pharmacol 2003; 14: 121-7.
    • (2003) Behav Pharmacol , vol.14 , pp. 121-127
    • Homayoun, H.1    Khavandgar, S.2    Mehr, S.E.3    Namiranian, K.4    Dehpour, A.R.5
  • 154
    • 0034623146 scopus 로고    scopus 로고
    • Inhibition of neuronal nitric-oxide synthase by calcium/calmodulin-dependent protein kinase IIalpha through Ser847 phosphorylation in NG108-15 neuronal cells
    • Komeima K, Hayashi Y, Naito Y, Watanabe Y. Inhibition of neuronal nitric-oxide synthase by calcium/calmodulin-dependent protein kinase IIalpha through Ser847 phosphorylation in NG108-15 neuronal cells. J Biol Chem 2000; 275: 28139-43.
    • (2000) J Biol Chem , vol.275 , pp. 28139-28143
    • Komeima, K.1    Hayashi, Y.2    Naito, Y.3    Watanabe, Y.4
  • 155
    • 0033783132 scopus 로고    scopus 로고
    • GTPase-activating proteins for heterotrimeric G proteins: Regulators of G protein signaling (RGS) and RGS-like proteins
    • Ross EM, Wilkie TM. GTPase-activating proteins for heterotrimeric G proteins: regulators of G protein signaling (RGS) and RGS-like proteins. Annu Rev Biochem 2000; 69: 795-827.
    • (2000) Annu Rev Biochem , vol.69 , pp. 795-827
    • Ross, E.M.1    Wilkie, T.M.2
  • 156
    • 50849086372 scopus 로고    scopus 로고
    • Coordinate regulation of G protein signaling via dynamic interactions of receptor and GAP
    • Turcotte M, Tang W, Ross EM. Coordinate regulation of G protein signaling via dynamic interactions of receptor and GAP. PLoS Comput Biol 2008; 4: e1000148.
    • (2008) PLoS Comput Biol , vol.4
    • Turcotte, M.1    Tang, W.2    Ross, E.M.3
  • 157
    • 77957068247 scopus 로고    scopus 로고
    • Structure and function of regulator of G protein signaling homology domains
    • Tesmer JJ. Structure and function of regulator of G protein signaling homology domains. Prog Mol Biol Transl Sci 2009; 86: 75-113.
    • (2009) Prog Mol Biol Transl Sci , vol.86 , pp. 75-113
    • Tesmer, J.J.1
  • 158
    • 16844384197 scopus 로고    scopus 로고
    • Morphine alters the selective association between mu-opioid receptors and specific RGS proteins in mouse periaqueductal gray matter
    • Garzón J, Rodriguez-Muñoz M, Sánchez-Blázquez P. Morphine alters the selective association between mu-opioid receptors and specific RGS proteins in mouse periaqueductal gray matter. Neuropharmacology 2005; 48: 853-68.
    • (2005) Neuropharmacology , vol.48 , pp. 853-868
    • Garzón, J.1    Rodriguez-Muñoz, M.2    Sánchez-Blázquez, P.3
  • 159
    • 32244436712 scopus 로고    scopus 로고
    • Selective interactions between G protein subunits and RGS4 with the Cterminal domains of the m-and d-opioid receptors regulate opioid receptor signaling
    • Georgoussi Z, Leontiadis L, Mazarakou G, et al. Selective interactions between G protein subunits and RGS4 with the Cterminal domains of the m-and d-opioid receptors regulate opioid receptor signaling. Cell Signal 2006; 18: 771-82.
    • (2006) Cell Signal , vol.18 , pp. 771-782
    • Georgoussi, Z.1    Leontiadis, L.2    Mazarakou, G.3
  • 160
    • 34249045098 scopus 로고    scopus 로고
    • Regulator of G protein signaling proteins differentially modulate signaling of m and d opioid receptors
    • Xie Z, Li Z, Guo L, et al. Regulator of G protein signaling proteins differentially modulate signaling of m and d opioid receptors. Eur J Pharmacol 2007; 565: 45-53.
    • (2007) Eur J Pharmacol , vol.565 , pp. 45-53
    • Xie, Z.1    Li, Z.2    Guo, L.3
  • 161
    • 33748590520 scopus 로고    scopus 로고
    • DEP-domain-mediated regulation of GPCR signaling responses
    • Ballon DR, Flanary PL, Gladue DP, et al. DEP-domain-mediated regulation of GPCR signaling responses. Cell 2006; 126: 1079-93.
    • (2006) Cell , vol.126 , pp. 1079-1093
    • Ballon, D.R.1    Flanary, P.L.2    Gladue, D.P.3
  • 162
    • 6944233607 scopus 로고    scopus 로고
    • Role of mPKCI, a novel mu-opioid receptor interactive protein, in receptor desensitization, phosphorylation, and morphine-induced analgesia
    • Guang W, Wang H, Su T, Weinstein IB, Wang JB. Role of mPKCI, a novel mu-opioid receptor interactive protein, in receptor desensitization, phosphorylation, and morphine-induced analgesia. Mol Pharmacol 2004; 66: 1285-92.
    • (2004) Mol Pharmacol , vol.66 , pp. 1285-1292
    • Guang, W.1    Wang, H.2    Su, T.3    Weinstein, I.B.4    Wang, J.B.5
  • 163
    • 33847021440 scopus 로고    scopus 로고
    • RGSZ1 interacts with protein kinase C interacting protein PKCI-1 and modulates mu opioid receptor signaling
    • Ajit SK, Ramineni S, Edris W, et al. RGSZ1 interacts with protein kinase C interacting protein PKCI-1 and modulates mu opioid receptor signaling. Cell Signal 2007; 19: 723-30.
    • (2007) Cell Signal , vol.19 , pp. 723-730
    • Ajit, S.K.1    Ramineni, S.2    Edris, W.3
  • 164
    • 80051799852 scopus 로고    scopus 로고
    • The histidine triad nucleotide-binding protein 1 supports muopioid receptor-glutamate NMDA receptor cross-regulation
    • Rodríguez-Muñoz M, Sánchez-Blázquez P, Vicente-Sánchez A, et al. The histidine triad nucleotide-binding protein 1 supports muopioid receptor-glutamate NMDA receptor cross-regulation. Cell Mol Life Sci 2011; 68: 2933-49.
    • (2011) Cell Mol Life Sci , vol.68 , pp. 2933-2949
    • Rodríguez-Muñoz, M.1    Sánchez-Blázquez, P.2    Vicente-Sánchez, A.3
  • 165
    • 49549084688 scopus 로고    scopus 로고
    • Do pharmacological approaches that prevent opioid tolerance target different elements in the same regulatory machinery?
    • Garzón J, Rodriguez-Muñoz M, Sánchez-Blázquez P. Do pharmacological approaches that prevent opioid tolerance target different elements in the same regulatory machinery? Curr Drug Abuse Rev 2008; 1: 222-38.
    • (2008) Curr Drug Abuse Rev , vol.1 , pp. 222-238
    • Garzón, J.1    Rodriguez-Muñoz, M.2    Sánchez-Blázquez, P.3
  • 166
    • 84857140534 scopus 로고    scopus 로고
    • mu-Opioid receptors and regulators of G protein signaling (RGS) proteins: From a symposium on new concepts in mu-opioid pharmacology
    • Traynor J. mu-Opioid receptors and regulators of G protein signaling (RGS) proteins: From a symposium on new concepts in mu-opioid pharmacology. Drug Alcohol Depend 2012; 121: 173-80.
    • (2012) Drug Alcohol Depend , vol.121 , pp. 173-180
    • Traynor, J.1
  • 167
    • 0037371746 scopus 로고    scopus 로고
    • Up-regulation of regulator of G protein signaling 4 expression in a model of neuropathic pain and insensitivity to morphine
    • Garnier M, Zaratin PF, Ficalora G, et al. Up-regulation of regulator of G protein signaling 4 expression in a model of neuropathic pain and insensitivity to morphine. J Pharmacol Exp Ther 2003; 304: 1299-306.
    • (2003) J Pharmacol Exp Ther , vol.304 , pp. 1299-1306
    • Garnier, M.1    Zaratin, P.F.2    Ficalora, G.3
  • 168
    • 19244367478 scopus 로고    scopus 로고
    • The R7 subfamily of RGS proteins assists tachyphylaxis and acute tolerance at mopioid receptors
    • Garzón J, López-Fando A, Sánchez-Blázquez P. The R7 subfamily of RGS proteins assists tachyphylaxis and acute tolerance at mopioid receptors. Neuropsychopharmacology 2003; 28: 1983-90.
    • (2003) Neuropsychopharmacology , vol.28 , pp. 1983-1990
    • Garzón, J.1    López-Fando, A.2    Sánchez-Blázquez, P.3
  • 170
    • 2342587417 scopus 로고    scopus 로고
    • Opioid peptide receptor studies. 17. Attenuation of chronic morphine effects after antisense oligodeoxynucleotide knock-down of RGS9 protein in cells expressing the cloned Mu opioid receptor
    • Xu H, Wang X, Wang J, Rothman RB. Opioid peptide receptor studies. 17. Attenuation of chronic morphine effects after antisense oligodeoxynucleotide knock-down of RGS9 protein in cells expressing the cloned Mu opioid receptor. Synapse 2004; 52: 209-17.
    • (2004) Synapse , vol.52 , pp. 209-217
    • Xu, H.1    Wang, X.2    Wang, J.3    Rothman, R.B.4
  • 173
    • 33645642111 scopus 로고    scopus 로고
    • Brainspecific regulator of G-protein signaling 9-2 selectively interacts with alpha-actinin-2 to regulate calcium-dependent inactivation of NMDA receptors
    • Bouhamdan M, Yan HD, Yan XH, Bannon MJ, Andrade R. Brainspecific regulator of G-protein signaling 9-2 selectively interacts with alpha-actinin-2 to regulate calcium-dependent inactivation of NMDA receptors. J Neurosci 2006; 26: 2522-30.
    • (2006) J Neurosci , vol.26 , pp. 2522-2530
    • Bouhamdan, M.1    Yan, H.D.2    Yan, X.H.3    Bannon, M.J.4    Andrade, R.5
  • 174
    • 0033557999 scopus 로고    scopus 로고
    • Interactions of calmodulin and alpha-actinin with the NR1 subunit modulate Ca2+-dependent inactivation of NMDA receptors
    • Krupp JJ, Vissel B, Thomas CG, Heinemann SF, Westbrook GL. Interactions of calmodulin and alpha-actinin with the NR1 subunit modulate Ca2+-dependent inactivation of NMDA receptors. J Neurosci 1999; 19: 1165-78.
    • (1999) J Neurosci , vol.19 , pp. 1165-1178
    • Krupp, J.J.1    Vissel, B.2    Thomas, C.G.3    Heinemann, S.F.4    Westbrook, G.L.5
  • 175
    • 27744598288 scopus 로고    scopus 로고
    • The RGSZ2 protein exists in a complex with m-opioid receptors and regulates the desensitizing capacity of Gz proteins
    • Garzón J, Rodríguez-Muñoz M, López-Fando A, Sánchez-Blázquez P. The RGSZ2 protein exists in a complex with m-opioid receptors and regulates the desensitizing capacity of Gz proteins. Neuropsychopharmacology 2005; 30: 1632-48.
    • (2005) Neuropsychopharmacology , vol.30 , pp. 1632-1648
    • Garzón, J.1    Rodríguez-Muñoz, M.2    López-Fando, A.3    Sánchez-Blázquez, P.4
  • 177
    • 79960393240 scopus 로고    scopus 로고
    • RGSZ2 binds to the neural nitric oxide synthase PDZ domain to regulate mu-opioid receptor-mediated potentiation of the N-methyl-Daspartate receptor-calmodulin-dependent protein kinase II pathway
    • Garzón J, Rodríguez-Muñoz M, Vicente-Sánchez A, et al. RGSZ2 binds to the neural nitric oxide synthase PDZ domain to regulate mu-opioid receptor-mediated potentiation of the N-methyl-Daspartate receptor-calmodulin-dependent protein kinase II pathway. Antioxid Redox Signal 2011; 15: 873-87.
    • (2011) Antioxid Redox Signal , vol.15 , pp. 873-887
    • Garzón, J.1    Rodríguez-Muñoz, M.2    Vicente-Sánchez, A.3
  • 178
    • 0031058739 scopus 로고    scopus 로고
    • A GTPaseactivating protein for the G protein Galphaz. Identification, purification, and mechanism of action
    • Wang J, Tu Y, Woodson J, Song X, Ross EM. A GTPaseactivating protein for the G protein Galphaz. Identification, purification, and mechanism of action. J Biol Chem 1997; 272: 5732-40.
    • (1997) J Biol Chem , vol.272 , pp. 5732-5740
    • Wang, J.1    Tu, Y.2    Woodson, J.3    Song, X.4    Ross, E.M.5
  • 179
    • 33947275782 scopus 로고    scopus 로고
    • Sumoylated RGS-Rz proteins act as scaffolds for Mu-opioid receptors and G-protein complexes in mouse brain
    • Rodríguez-Muñoz M, Bermúdez D, Sánchez-Blázquez P, Garzón J. Sumoylated RGS-Rz proteins act as scaffolds for Mu-opioid receptors and G-protein complexes in mouse brain. Neuropsychopharmacology 2007; 32: 842-50.
    • (2007) Neuropsychopharmacology , vol.32 , pp. 842-850
    • Rodríguez-Muñoz, M.1    Bermúdez, D.2    Sánchez-Blázquez, P.3    Garzón, J.4
  • 181
    • 4444311166 scopus 로고    scopus 로고
    • Morphine induces terminal m-opioid receptor desensitization by sustained phosphorylation of serine-375
    • Schulz S, Mayer D, Pfeiffer M, et al. Morphine induces terminal m-opioid receptor desensitization by sustained phosphorylation of serine-375. EMBO J 2004; 23: 3282-9.
    • (2004) EMBO J , vol.23 , pp. 3282-3289
    • Schulz, S.1    Mayer, D.2    Pfeiffer, M.3
  • 183
    • 77955356174 scopus 로고    scopus 로고
    • Mu-opioid receptors transiently activate the Akt-nNOS pathway to produce sustained potentiation of PKC-mediated NMDAR-CaMKII signaling
    • Sánchez-Blázquez P, Rodríguez-Muñoz M, Garzón J. Mu-opioid receptors transiently activate the Akt-nNOS pathway to produce sustained potentiation of PKC-mediated NMDAR-CaMKII signaling. PLoS One 2010; 5: e11278.
    • (2010) PLoS One , vol.5
    • Sánchez-Blázquez, P.1    Rodríguez-Muñoz, M.2    Garzón, J.3
  • 184
    • 63449102832 scopus 로고    scopus 로고
    • Regulator of G-protein signaling 14 (RGS14) is a selective H-Ras effector
    • Willard FS, Willard MD, Kimple AJ, et al. Regulator of G-protein signaling 14 (RGS14) is a selective H-Ras effector. PLoS One 2009; 4: e4884.
    • (2009) PLoS One , vol.4
    • Willard, F.S.1    Willard, M.D.2    Kimple, A.J.3
  • 185
    • 71349084088 scopus 로고    scopus 로고
    • RGS14 is a multifunctional scaffold that integrates G protein and Ras/Raf MAPkinase signalling pathways
    • Shu FJ, Ramineni S, Hepler JR. RGS14 is a multifunctional scaffold that integrates G protein and Ras/Raf MAPkinase signalling pathways. Cell Signal 2010; 22: 366-76.
    • (2010) Cell Signal , vol.22 , pp. 366-376
    • Shu, F.J.1    Ramineni, S.2    Hepler, J.R.3
  • 186
    • 0037162696 scopus 로고    scopus 로고
    • Ras and Rap control AMPA receptor trafficking during synaptic plasticity
    • Zhu JJ, Qin Y, Zhao M, Van AL, Malinow R. Ras and Rap control AMPA receptor trafficking during synaptic plasticity. Cell 2002; 110: 443-55.
    • (2002) Cell , vol.110 , pp. 443-455
    • Zhu, J.J.1    Qin, Y.2    Zhao, M.3    Van, A.L.4    Malinow, R.5
  • 187
    • 0029552394 scopus 로고
    • In vivo injection of antisense oligodeoxynucleotides to G alpha subunits and supraspinal analgesia evoked by mu and delta opioid agonists
    • Sánchez-Blázquez P, García-España A, Garzón J. In vivo injection of antisense oligodeoxynucleotides to G alpha subunits and supraspinal analgesia evoked by mu and delta opioid agonists. J Pharmacol Exp Ther 1995; 275: 1590-6.
    • (1995) J Pharmacol Exp Ther , vol.275 , pp. 1590-1596
    • Sánchez-Blázquez, P.1    García-España, A.2    Garzón, J.3
  • 188
    • 0035862538 scopus 로고    scopus 로고
    • Agonists determine the pattern of G-protein activation in m-opioid receptormediated supraspinal analgesia
    • Sánchez-Blázquez P, Gómez-Serranillos P, Garzón J. Agonists determine the pattern of G-protein activation in m-opioid receptormediated supraspinal analgesia. Brain Research Bulletin 2001; 54: 229-35.
    • (2001) Brain Research Bulletin , vol.54 , pp. 229-235
    • Sánchez-Blázquez, P.1    Gómez-Serranillos, P.2    Garzón, J.3
  • 189
    • 0029980110 scopus 로고    scopus 로고
    • Threedimensional structure of human protein kinase C interacting protein 1, a member of the HIT family of proteins
    • Lima CD, Klein MG, Weinstein IB, Hendrickson WA. Threedimensional structure of human protein kinase C interacting protein 1, a member of the HIT family of proteins. Proc Natl Acad Sci U S A 1996; 93: 5357-62.
    • (1996) Proc Natl Acad Sci U S A , vol.93 , pp. 5357-5362
    • Lima, C.D.1    Klein, M.G.2    Weinstein, I.B.3    Hendrickson, W.A.4
  • 190
    • 0025248445 scopus 로고
    • Amino acid sequence and characterization of a protein inhibitor of protein kinase C
    • Pearson JD, DeWald DB, Mathews WR, et al. Amino acid sequence and characterization of a protein inhibitor of protein kinase C. J Biol Chem 1990; 265: 4583-91.
    • (1990) J Biol Chem , vol.265 , pp. 4583-4591
    • Pearson, J.D.1    DeWald, D.B.2    Mathews, W.R.3
  • 191
    • 0023112569 scopus 로고
    • Properties and distribution of the protein inhibitor (Mr 17,000) of protein kinase C
    • McDonald JR, Groschel-Stewart U, Walsh MP. Properties and distribution of the protein inhibitor (Mr 17,000) of protein kinase C. Biochem J 1987; 242: 695-705.
    • (1987) Biochem J , vol.242 , pp. 695-705
    • McDonald, J.R.1    Groschel-Stewart, U.2    Walsh, M.P.3
  • 192
    • 0035413601 scopus 로고    scopus 로고
    • Protein kinase C: Structural and spatial regulation by phosphorylation, cofactors, and macromolecular interactions
    • Newton AC. Protein kinase C: structural and spatial regulation by phosphorylation, cofactors, and macromolecular interactions. Chem Rev 2001; 101: 2353-64.
    • (2001) Chem Rev , vol.101 , pp. 2353-2364
    • Newton, A.C.1
  • 193
    • 64549163306 scopus 로고    scopus 로고
    • Zinc fingers as biologic redox switches?
    • Kroncke KD, Klotz LO. Zinc fingers as biologic redox switches? Antioxid Redox Signal 2009; 11: 1015-27.
    • (2009) Antioxid Redox Signal , vol.11 , pp. 1015-1027
    • Kroncke, K.D.1    Klotz, L.O.2
  • 194
    • 15744401979 scopus 로고    scopus 로고
    • Calcium/calmodulin-dependent protein kinase II (CaMKII) localization acts in concert with substrate targeting to create spatial restriction for phosphorylation
    • Tsui J, Inagaki M, Schulman H. Calcium/calmodulin-dependent protein kinase II (CaMKII) localization acts in concert with substrate targeting to create spatial restriction for phosphorylation. J Biol Chem 2005; 280: 9210-6.
    • (2005) J Biol Chem , vol.280 , pp. 9210-9216
    • Tsui, J.1    Inagaki, M.2    Schulman, H.3
  • 195
    • 33750927850 scopus 로고    scopus 로고
    • Zinc dynamics in the myocardial redox signaling network
    • Korichneva I. Zinc dynamics in the myocardial redox signaling network. Antioxid Redox Signal 2006; 8: 1707-21.
    • (2006) Antioxid Redox Signal , vol.8 , pp. 1707-1721
    • Korichneva, I.1
  • 197
    • 33646200560 scopus 로고    scopus 로고
    • Gbg inhibits Ga GTPaseactivating proteins by inhibition of Ga-GTP binding during stimulation by receptor
    • Tang W, Tu Y, Nayak SK, et al. Gbg inhibits Ga GTPaseactivating proteins by inhibition of Ga-GTP binding during stimulation by receptor. J Biol Chem 2006; 281: 4746-53.
    • (2006) J Biol Chem , vol.281 , pp. 4746-4753
    • Tang, W.1    Tu, Y.2    Nayak, S.K.3
  • 198
    • 0034693139 scopus 로고    scopus 로고
    • The effector enzyme regulates the duration of G protein signaling in vertebrate photoreceptors by increasing the affinity between transducin and RGS protein
    • Skiba NP, Hopp JA, Arshavsky VY. 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 2000; 275: 32716-20.
    • (2000) J Biol Chem , vol.275 , pp. 32716-32720
    • Skiba, N.P.1    Hopp, J.A.2    Arshavsky, V.Y.3
  • 199
    • 0035813162 scopus 로고    scopus 로고
    • RGS9-G beta 5 substrate selectivity in photoreceptors. Opposing effects of constituent domains yield high affinity of RGS interaction with the G protein-effector complex
    • Skiba NP, Martemyanov KA, Elfenbein A, et al. RGS9-G beta 5 substrate selectivity in photoreceptors. Opposing effects of constituent domains yield high affinity of RGS interaction with the G protein-effector complex. J Biol Chem 2001; 276: 37365-72.
    • (2001) J Biol Chem , vol.276 , pp. 37365-37372
    • Skiba, N.P.1    Martemyanov, K.A.2    Elfenbein, A.3
  • 200
    • 0038198731 scopus 로고    scopus 로고
    • Specificity of G protein-RGS protein recognition is regulated by affinity adapters
    • Martemyanov KA, Hopp JA, Arshavsky VY. Specificity of G protein-RGS protein recognition is regulated by affinity adapters. Neuron 2003; 38: 857-62.
    • (2003) Neuron , vol.38 , pp. 857-862
    • Martemyanov, K.A.1    Hopp, J.A.2    Arshavsky, V.Y.3
  • 201
    • 33750901634 scopus 로고    scopus 로고
    • Zinc coordination environments in proteins as redox sensors and signal transducers
    • Maret W. Zinc coordination environments in proteins as redox sensors and signal transducers. Antioxid Redox Signal 2006; 8: 1419-41.
    • (2006) Antioxid Redox Signal , vol.8 , pp. 1419-1441
    • Maret, W.1
  • 202
    • 79952833794 scopus 로고    scopus 로고
    • Nitric oxide signaling and nitrosative stress in neurons: Role for s-nitrosylation
    • Shahani N, Sawa A. Nitric oxide signaling and nitrosative stress in neurons: role for s-nitrosylation. Antioxid Redox Signal 2011; 14: 1493-504.
    • (2011) Antioxid Redox Signal , vol.14 , pp. 1493-1504
    • Shahani, N.1    Sawa, A.2
  • 203
    • 0034058549 scopus 로고    scopus 로고
    • The function of zinc metallothionein: A link between cellular zinc and redox state
    • Maret W. The function of zinc metallothionein: a link between cellular zinc and redox state. J Nutr 2000; 130: 1455S-8S.
    • (2000) J Nutr , vol.130
    • Maret, W.1
  • 205
    • 33745865202 scopus 로고    scopus 로고
    • Zinc center as redox switch-new function for an old motif
    • Ilbert M, Graf PC, Jakob U. Zinc center as redox switch-new function for an old motif. Antioxid Redox Signal 2006; 8: 835-46.
    • (2006) Antioxid Redox Signal , vol.8 , pp. 835-846
    • Ilbert, M.1    Graf, P.C.2    Jakob, U.3
  • 206
    • 0032584166 scopus 로고    scopus 로고
    • Thiolate ligands in metallothionein confer redox activity on zinc clusters
    • Maret W, Vallee BL. Thiolate ligands in metallothionein confer redox activity on zinc clusters. Proc Natl Acad Sci U S A 1998; 95: 3478-82.
    • (1998) Proc Natl Acad Sci U S A , vol.95 , pp. 3478-3482
    • Maret, W.1    Vallee, B.L.2
  • 207
    • 0035369919 scopus 로고    scopus 로고
    • NMDA receptor regulation by Src kinase signalling in excitatory synaptic transmission and plasticity
    • Ali DW, Salter MW. NMDA receptor regulation by Src kinase signalling in excitatory synaptic transmission and plasticity. Curr Opin Neurobiol 2001; 11: 336-42.
    • (2001) Curr Opin Neurobiol , vol.11 , pp. 336-342
    • Ali, D.W.1    Salter, M.W.2
  • 208
    • 29144452123 scopus 로고    scopus 로고
    • Endogenous and synthetic inhibitors of the Src-family protein tyrosine kinases
    • Chong YP, Ia KK, Mulhern TD, Cheng HC. Endogenous and synthetic inhibitors of the Src-family protein tyrosine kinases. Biochim Biophys Acta 2005; 1754: 210-20.
    • (2005) Biochim Biophys Acta , vol.1754 , pp. 210-220
    • Chong, Y.P.1    Ia, K.K.2    Mulhern, T.D.3    Cheng, H.C.4
  • 209
    • 0034268796 scopus 로고    scopus 로고
    • Src tyrosine kinase is a novel direct effector of G proteins
    • Ma YC, Huang J, Ali S, Lowry W, Huang XY. Src tyrosine kinase is a novel direct effector of G proteins. Cell 2000; 102: 635-46.
    • (2000) Cell , vol.102 , pp. 635-646
    • Ma, Y.C.1    Huang, J.2    Ali, S.3    Lowry, W.4    Huang, X.Y.5
  • 210
    • 0038267569 scopus 로고    scopus 로고
    • H-Ras modulates N-methyl-D-aspartate receptor function via inhibition of Src tyrosine kinase activity
    • Thornton C, Yaka R, Dinh S, Ron D. H-Ras modulates N-methyl-D-aspartate receptor function via inhibition of Src tyrosine kinase activity. J Biol Chem 2003; 278: 23823-9.
    • (2003) J Biol Chem , vol.278 , pp. 23823-23829
    • Thornton, C.1    Yaka, R.2    Dinh, S.3    Ron, D.4
  • 211
    • 0344392135 scopus 로고    scopus 로고
    • Reciprocal modulation of phospholipase Cb isoforms: Adaptation to chronic morphine
    • Chakrabarti S, Liu NJ, Gintzler AR. Reciprocal modulation of phospholipase Cb isoforms: adaptation to chronic morphine. Proc Natl Acad Sci U S A 2003; 100: 13686-91.
    • (2003) Proc Natl Acad Sci U S A , vol.100 , pp. 13686-13691
    • Chakrabarti, S.1    Liu, N.J.2    Gintzler, A.R.3
  • 212
    • 0032106760 scopus 로고    scopus 로고
    • Tyrosine kinase potentiates NMDA receptor currents by reducing tonic zinc inhibition
    • Zheng F, Gingrich MB, Traynelis SF, Conn PJ. Tyrosine kinase potentiates NMDA receptor currents by reducing tonic zinc inhibition. Nat Neurosci 1998; 1: 185-91.
    • (1998) Nat Neurosci , vol.1 , pp. 185-191
    • Zheng, F.1    Gingrich, M.B.2    Traynelis, S.F.3    Conn, P.J.4
  • 213
    • 0023043778 scopus 로고
    • Translocation of protein kinase C activity may mediate hippocampal long-term potentiation
    • Akers RF, Lovinger DM, Colley PA, Linden DJ, Routtenberg A. Translocation of protein kinase C activity may mediate hippocampal long-term potentiation. Science 1986; 231: 587-9.
    • (1986) Science , vol.231 , pp. 587-589
    • Akers, R.F.1    Lovinger, D.M.2    Colley, P.A.3    Linden, D.J.4    Routtenberg, A.5
  • 214
    • 0030611666 scopus 로고    scopus 로고
    • Ca2+ influx amplifies protein kinase C potentiation of recombinant NMDA receptors
    • Zheng X, Zhang L, Wang AP, Bennett MV, Zukin RS. Ca2+ influx amplifies protein kinase C potentiation of recombinant NMDA receptors. J Neurosci 1997; 17: 8676-86.
    • (1997) J Neurosci , vol.17 , pp. 8676-8686
    • Zheng, X.1    Zhang, L.2    Wang, A.P.3    Bennett, M.V.4    Zukin, R.S.5
  • 215
    • 0030764618 scopus 로고    scopus 로고
    • Regulation of the AKAP79-protein kinase C interaction by Ca2+/Calmodulin
    • Faux MC, Scott JD. Regulation of the AKAP79-protein kinase C interaction by Ca2+/Calmodulin. J Biol Chem 1997; 272: 17038-44.
    • (1997) J Biol Chem , vol.272 , pp. 17038-17044
    • Faux, M.C.1    Scott, J.D.2
  • 217
    • 33845213288 scopus 로고    scopus 로고
    • Calmodulin potentiates Gbg activation of phospholipase C-b3
    • McCullar JS, Malencik DA, Vogel WK, et al. Calmodulin potentiates Gbg activation of phospholipase C-b3. Biochem Pharmacol 2007; 73: 270-8.
    • (2007) Biochem Pharmacol , vol.73 , pp. 270-278
    • McCullar, J.S.1    Malencik, D.A.2    Vogel, W.K.3
  • 218
    • 0031243489 scopus 로고    scopus 로고
    • RC3/neurogranin, a postsynaptic calpacitin for setting the response threshold to calcium influxes
    • Gerendasy DD, Sutcliffe JG. RC3/neurogranin, a postsynaptic calpacitin for setting the response threshold to calcium influxes. Mol Neurobiol 1997; 15: 131-63.
    • (1997) Mol Neurobiol , vol.15 , pp. 131-163
    • Gerendasy, D.D.1    Sutcliffe, J.G.2
  • 219
    • 0033618426 scopus 로고    scopus 로고
    • Calmodulin binding to G proteincoupling domain of opioid receptors
    • Wang D, Sadee W, Quillan JM. Calmodulin binding to G proteincoupling domain of opioid receptors. J Biol Chem 1999; 274: 22081-8.
    • (1999) J Biol Chem , vol.274 , pp. 22081-22088
    • Wang, D.1    Sadee, W.2    Quillan, J.M.3
  • 221
    • 0030611335 scopus 로고    scopus 로고
    • The Ca2+-dependent binding of calmodulin to an N-terminal motif of the heterotrimeric G protein b subunit
    • Liu M, Yu B, Nakanishi O, Wieland T, Simon M. The Ca2+-dependent binding of calmodulin to an N-terminal motif of the heterotrimeric G protein b subunit. J Biol Chem 1997; 272: 18801-7.
    • (1997) J Biol Chem , vol.272 , pp. 18801-18807
    • Liu, M.1    Yu, B.2    Nakanishi, O.3    Wieland, T.4    Simon, M.5
  • 222
    • 0242468744 scopus 로고    scopus 로고
    • Regulation and organization of adenylyl cyclases and cAMP
    • Cooper DM. Regulation and organization of adenylyl cyclases and cAMP. Biochem J 2003; 375: 517-29.
    • (2003) Biochem J , vol.375 , pp. 517-529
    • Cooper, D.M.1
  • 223
    • 0033516701 scopus 로고    scopus 로고
    • Regulation of NMDA receptors by an associated phosphatase-kinase signaling complex
    • Westphal RS, Tavalin SJ, Lin JW, et al. Regulation of NMDA receptors by an associated phosphatase-kinase signaling complex. Science 1999; 285: 93-6.
    • (1999) Science , vol.285 , pp. 93-96
    • Westphal, R.S.1    Tavalin, S.J.2    Lin, J.W.3
  • 224
    • 0031814749 scopus 로고    scopus 로고
    • Distinct differences between morphine-and [D-Ala2, N-MePhe4, Gly-ol5]-enkephalin-mu-opioid receptor complexes demonstrated by cyclic AMP-dependent protein kinase phosphorylation
    • Chakrabarti S, Law PY, Loh HH. Distinct differences between morphine-and [D-Ala2, N-MePhe4, Gly-ol5]-enkephalin-mu-opioid receptor complexes demonstrated by cyclic AMP-dependent protein kinase phosphorylation. J Neurochem 1998; 71: 231-9.
    • (1998) J Neurochem , vol.71 , pp. 231-239
    • Chakrabarti, S.1    Law, P.Y.2    Loh, H.H.3
  • 226
    • 0027326410 scopus 로고
    • Protein kinase C alpha activates RAF-1 by direct phosphorylation
    • Kolch W, Heidecker G, Kochs G, et al. Protein kinase C alpha activates RAF-1 by direct phosphorylation. Nature 1993; 364: 249-52.
    • (1993) Nature , vol.364 , pp. 249-252
    • Kolch, W.1    Heidecker, G.2    Kochs, G.3
  • 227
    • 34547469939 scopus 로고    scopus 로고
    • Raf kinases: Function, regulation and role in human cancer
    • Leicht DT, Balan V, Kaplun A, et al. Raf kinases: function, regulation and role in human cancer. Biochim Biophys Acta 2007; 1773: 1196-212.
    • (2007) Biochim Biophys Acta , vol.1773 , pp. 1196-1212
    • Leicht, D.T.1    Balan, V.2    Kaplun, A.3
  • 228
    • 0029050573 scopus 로고
    • Distribution and targeting of a m-opioid receptor (MOR1) in brain and spinal cord
    • Arvidsson U, Riedl M, Chakrabarti S, et al. Distribution and targeting of a m-opioid receptor (MOR1) in brain and spinal cord. J Neurosci 1995; 15: 3328-41.
    • (1995) J Neurosci , vol.15 , pp. 3328-3341
    • Arvidsson, U.1    Riedl, M.2    Chakrabarti, S.3
  • 229
    • 0031578951 scopus 로고    scopus 로고
    • Dual ultrastructural immunocytochemical labeling of m and d opioid receptors in the superficial layers of the rat cervical spinal cord
    • Cheng PY, Liu-Chen LY, Pickel VM. Dual ultrastructural immunocytochemical labeling of m and d opioid receptors in the superficial layers of the rat cervical spinal cord. Brain Res 1997; 778: 367-80.
    • (1997) Brain Res , vol.778 , pp. 367-380
    • Cheng, P.Y.1    Liu-Chen, L.Y.2    Pickel, V.M.3
  • 230
    • 77955606622 scopus 로고    scopus 로고
    • Coexpression of delta-and mu-opioid receptors in nociceptive sensory neurons
    • Wang HB, Zhao B, Zhong YQ, et al. Coexpression of delta-and mu-opioid receptors in nociceptive sensory neurons. Proc Natl Acad Sci U S A 2010; 107: 13117-22.
    • (2010) Proc Natl Acad Sci U S A , vol.107 , pp. 13117-13122
    • Wang, H.B.1    Zhao, B.2    Zhong, Y.Q.3
  • 231
    • 1842687121 scopus 로고    scopus 로고
    • A role for heterodimerization of m and d opiate receptors in enhancing morphine analgesia
    • Gomes I, Gupta A, Filipovska J, et al. A role for heterodimerization of m and d opiate receptors in enhancing morphine analgesia. Proc Natl Acad Sci U S A 2004; 101: 5135-9.
    • (2004) Proc Natl Acad Sci U S A , vol.101 , pp. 5135-5139
    • Gomes, I.1    Gupta, A.2    Filipovska, J.3
  • 232
    • 30044442319 scopus 로고    scopus 로고
    • Opioid-induced tolerance and dependence in mice is modulated by the distance between pharmacophores in a bivalent ligand series
    • Daniels DJ, Lenard NR, Etienne CL, et al. Opioid-induced tolerance and dependence in mice is modulated by the distance between pharmacophores in a bivalent ligand series. Proc Natl Acad Sci U S A 2005; 102: 19208-13.
    • (2005) Proc Natl Acad Sci U S A , vol.102 , pp. 19208-19213
    • Daniels, D.J.1    Lenard, N.R.2    Etienne, C.L.3
  • 233
    • 0037220638 scopus 로고    scopus 로고
    • 3 cells changes the functional response to m agonists from inhibitory to excitatory
    • 3 cells changes the functional response to m agonists from inhibitory to excitatory. Mol Pharmacol 2003; 63: 89-95.
    • (2003) Mol Pharmacol , vol.63 , pp. 89-95
    • Charles, A.C.1    Mostovskaya, N.2    Asas, K.3
  • 234
    • 0034714335 scopus 로고    scopus 로고
    • Oligomerization of m-and d-opioid receptors. Generation of novel functional properties
    • George SR, Fan T, Xie Z, et al. Oligomerization of m-and d-opioid receptors. Generation of novel functional properties. J Biol Chem 2000; 275: 26128-35.
    • (2000) J Biol Chem , vol.275 , pp. 26128-26135
    • George, S.R.1    Fan, T.2    Xie, Z.3
  • 235
    • 0031808936 scopus 로고    scopus 로고
    • Delta opioid receptor subtypes activate inositol-signaling pathways in the production of antinociception
    • Sánchez-Blázquez P, Garzón J. Delta opioid receptor subtypes activate inositol-signaling pathways in the production of antinociception. J Pharmacol Exp Ther 1998; 285: 820-7.
    • (1998) J Pharmacol Exp Ther , vol.285 , pp. 820-827
    • Sánchez-Blázquez, P.1    Garzón, J.2
  • 236
    • 0035862538 scopus 로고    scopus 로고
    • Agonists determine the pattern of G-protein activation in mu-opioid receptor-mediated supraspinal analgesia
    • Sánchez-Blázquez P, Gómez-Serranillos P, Garzón J. Agonists determine the pattern of G-protein activation in mu-opioid receptor-mediated supraspinal analgesia. Brain Res Bull 2001; 54: 229-35.
    • (2001) Brain Res Bull , vol.54 , pp. 229-235
    • Sánchez-Blázquez, P.1    Gómez-Serranillos, P.2    Garzón, J.3
  • 237
    • 0027474649 scopus 로고
    • d-Opioid receptor subtypes and cross-talk with m-receptors
    • Traynor JR, Elliott J. d-Opioid receptor subtypes and cross-talk with m-receptors. Trends Pharmacol Sci 1993; 14: 84-6.
    • (1993) Trends Pharmacol Sci , vol.14 , pp. 84-86
    • Traynor, J.R.1    Elliott, J.2
  • 238
    • 0025827095 scopus 로고
    • Selective blockage of delta opioid receptors prevents the development of morphine tolerance and dependence in mice
    • 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.
    • (1991) J Pharmacol Exp Ther , vol.258 , pp. 299-303
    • Abdelhamid, E.E.1    Sultana, M.2    Portoghese, P.S.3    Takemori, A.E.4
  • 239
    • 0028788399 scopus 로고
    • Attenuation of morphine tolerance and dependence with the highly selective d-opioid receptor antagonist TIPP[y]
    • Fundytus ME, Schiller PW, Shapiro M, Weltrowska G, Coderre TJ. Attenuation of morphine tolerance and dependence with the highly selective d-opioid receptor antagonist TIPP[y]. Eur J Pharmacol 1995; 286: 105-8.
    • (1995) Eur J Pharmacol , vol.286 , pp. 105-108
    • Fundytus, M.E.1    Schiller, P.W.2    Shapiro, M.3    Weltrowska, G.4    Coderre, T.J.5
  • 240
    • 0030947808 scopus 로고    scopus 로고
    • Antisense oligodeoxynucleotides to opioid mu and delta receptors reduced morphine dependence in mice: Role of delta-2 opioid receptors
    • Sánchez-Blázquez P, García-España A, Garzón J. Antisense oligodeoxynucleotides to opioid mu and delta receptors reduced morphine dependence in mice: role of delta-2 opioid receptors. J Pharmacol Exp Ther 1997; 280: 1423-31.
    • (1997) J Pharmacol Exp Ther , vol.280 , pp. 1423-1431
    • Sánchez-Blázquez, P.1    García-España, A.2    Garzón, J.3
  • 241
    • 0033198831 scopus 로고    scopus 로고
    • Retention of supraspinal delta-like analgesia and loss of morphine tolerance in d opioid receptor knockout mice
    • Zhu Y, King MA, Schuller AG, et al. Retention of supraspinal delta-like analgesia and loss of morphine tolerance in d opioid receptor knockout mice. Neuron 1999; 24: 243-52.
    • (1999) Neuron , vol.24 , pp. 243-252
    • Zhu, Y.1    King, M.A.2    Schuller, A.G.3
  • 242
    • 0033539134 scopus 로고    scopus 로고
    • 2 [y] produces a potent analgesic effect, no physical dependence, and less tolerance than morphine in rats
    • 2 [y] produces a potent analgesic effect, no physical dependence, and less tolerance than morphine in rats. J Med Chem 1999; 42: 3520-6.
    • (1999) J Med Chem , vol.42 , pp. 3520-3526
    • Schiller, P.W.1    Fundytus, M.E.2    Merovitz, L.3
  • 243
    • 0035040627 scopus 로고    scopus 로고
    • In vivo pharmacological characterization of SoRI 9409, a nonpeptidic opioid m-agonist/d-antagonist that produces limited antinociceptive tolerance and attenuates morphine physical dependence
    • Wells JL, Bartlett JL, Ananthan S, Bilsky EJ. In vivo pharmacological characterization of SoRI 9409, a nonpeptidic opioid m-agonist/d-antagonist that produces limited antinociceptive tolerance and attenuates morphine physical dependence. J Pharmacol Exp Ther 2001; 297: 597-605.
    • (2001) J Pharmacol Exp Ther , vol.297 , pp. 597-605
    • Wells, J.L.1    Bartlett, J.L.2    Ananthan, S.3    Bilsky, E.J.4
  • 244
    • 33846029187 scopus 로고    scopus 로고
    • Trafficking of d-opioid receptors and other G-protein-coupled receptors: Implications for pain and analgesia
    • Cahill CM, Holdridge SV, Morinville A. Trafficking of d-opioid receptors and other G-protein-coupled receptors: implications for pain and analgesia. Trends Pharmacol Sci 2007; 28: 23-31.
    • (2007) Trends Pharmacol Sci , vol.28 , pp. 23-31
    • Cahill, C.M.1    Holdridge, S.V.2    Morinville, A.3
  • 245
    • 78650958254 scopus 로고    scopus 로고
    • Facilitation of mu-opioid receptor activity by preventing delta-opioid receptor-mediated codegradation
    • He SQ, Zhang ZN, Guan JS, et al. Facilitation of mu-opioid receptor activity by preventing delta-opioid receptor-mediated codegradation. Neuron 2011; 69: 120-31.
    • (2011) Neuron , vol.69 , pp. 120-131
    • He, S.Q.1    Zhang, Z.N.2    Guan, J.S.3
  • 246
    • 79956189726 scopus 로고    scopus 로고
    • Heteromerization of the mu-and deltaopioid receptors produces ligand-biased antagonism and alters mureceptor trafficking
    • Milan-Lobo L, Whistler JL. Heteromerization of the mu-and deltaopioid receptors produces ligand-biased antagonism and alters mureceptor trafficking. J Pharmacol Exp Ther 2011; 337: 868-75.
    • (2011) J Pharmacol Exp Ther , vol.337 , pp. 868-875
    • Milan-Lobo, L.1    Whistler, J.L.2
  • 247
    • 63849206531 scopus 로고    scopus 로고
    • Disruption of Cdk5-associated phosphorylation of residue threonine-161 of the delta-opioid receptor: Impaired receptor function and attenuated morphine antinociceptive tolerance
    • Xie WY, He Y, Yang YR, et al. Disruption of Cdk5-associated phosphorylation of residue threonine-161 of the delta-opioid receptor: impaired receptor function and attenuated morphine antinociceptive tolerance. J Neurosci 2009; 29: 3551-64.
    • (2009) J Neurosci , vol.29 , pp. 3551-3564
    • Xie, W.Y.1    He, Y.2    Yang, Y.R.3
  • 248
    • 33746932103 scopus 로고    scopus 로고
    • Role of delivery and trafficking of dopioid peptide receptors in opioid analgesia and tolerance
    • Zhang X, Bao L, Guan JS. Role of delivery and trafficking of dopioid peptide receptors in opioid analgesia and tolerance. Trends Pharmacol Sci 2006; 27: 324-9.
    • (2006) Trends Pharmacol Sci , vol.27 , pp. 324-329
    • Zhang, X.1    Bao, L.2    Guan, J.S.3
  • 249
    • 0037066747 scopus 로고    scopus 로고
    • KEPI, a PKC-dependent protein phosphatase 1 inhibitor regulated by morphine
    • Liu QR, Zhang PW, Zhen Q, et al. KEPI, a PKC-dependent protein phosphatase 1 inhibitor regulated by morphine. J Biol Chem 2002; 277: 13312-20.
    • (2002) J Biol Chem , vol.277 , pp. 13312-13320
    • Liu, Q.R.1    Zhang, P.W.2    Zhen, Q.3
  • 250
    • 70349205440 scopus 로고    scopus 로고
    • Supraspinal Gbetagamma-dependent stimulation of PLCbeta originating from G inhibitory protein-mu opioid receptor-coupling is necessary for morphine induced acute hyperalgesia
    • Bianchi E, Norcini M, Smrcka A, Ghelardini C. Supraspinal Gbetagamma-dependent stimulation of PLCbeta originating from G inhibitory protein-mu opioid receptor-coupling is necessary for morphine induced acute hyperalgesia. J Neurochem 2009; 111: 171-80.
    • (2009) J Neurochem , vol.111 , pp. 171-180
    • Bianchi, E.1    Norcini, M.2    Smrcka, A.3    Ghelardini, C.4
  • 251
    • 58149400019 scopus 로고    scopus 로고
    • A novel Gbg-subunit inhibitor selectively modulates mu-opioid-dependent antinociception and attenuates acute morphine-induced antinociceptive tolerance and dependence
    • Mathews JL, Smrcka AV, Bidlack JM. A novel Gbg-subunit inhibitor selectively modulates mu-opioid-dependent antinociception and attenuates acute morphine-induced antinociceptive tolerance and dependence. J Neurosci 2008; 28: 12183-9.
    • (2008) J Neurosci , vol.28 , pp. 12183-12189
    • Mathews, J.L.1    Smrcka, A.V.2    Bidlack, J.M.3
  • 252
    • 0030743849 scopus 로고    scopus 로고
    • Functionally differentiating two neuronal nitric oxide synthase isoforms through antisense mapping: Evidence for opposing NO actions on morphine analgesia and tolerance
    • Kolesnikov YA, Pan YX, Babey AM, et al. Functionally differentiating two neuronal nitric oxide synthase isoforms through antisense mapping: evidence for opposing NO actions on morphine analgesia and tolerance. Proc Natl Acad Sci U S A 1997; 94: 8220-5.
    • (1997) Proc Natl Acad Sci U S A , vol.94 , pp. 8220-8225
    • Kolesnikov, Y.A.1    Pan, Y.X.2    Babey, A.M.3
  • 253
    • 77953807418 scopus 로고    scopus 로고
    • Modulation of mu opioid receptor desensitization in peripheral sensory neurons by phosphoinositide 3-kinase gamma
    • König C, Gavrilova-Ruch O, von Banchet GS, et al. Modulation of mu opioid receptor desensitization in peripheral sensory neurons by phosphoinositide 3-kinase gamma. Neuroscience 2010; 169: 449-54.
    • (2010) Neuroscience , vol.169 , pp. 449-454
    • König, C.1    Gavrilova-Ruch, O.2    von Banchet, G.S.3
  • 254
    • 0025192474 scopus 로고
    • Gz, a guanine nucleotide-binding protein with unique biochemical properties
    • Casey PJ, Fong HK, Simon MI, Gilman AG. Gz, a guanine nucleotide-binding protein with unique biochemical properties. J Biol Chem 1990; 265: 2383-90.
    • (1990) J Biol Chem , vol.265 , pp. 2383-2390
    • Casey, P.J.1    Fong, H.K.2    Simon, M.I.3    Gilman, A.G.4
  • 256
    • 0036847009 scopus 로고    scopus 로고
    • Identity of adenylyl cyclase isoform determines the G protein mediating chronic opioid-induced adenylyl cyclase supersensitivity
    • Ammer H, Christ TE. Identity of adenylyl cyclase isoform determines the G protein mediating chronic opioid-induced adenylyl cyclase supersensitivity. J Neurochem 2002; 83: 818-27.
    • (2002) J Neurochem , vol.83 , pp. 818-827
    • Ammer, H.1    Christ, T.E.2
  • 257
    • 33644868454 scopus 로고    scopus 로고
    • Adenylyl cyclase type 5 (AC5) is an essential mediator of morphine action
    • Kim KS, Lee KW, Lee KW, et al. Adenylyl cyclase type 5 (AC5) is an essential mediator of morphine action. Proc Natl Acad Sci U S A 2006; 103: 3908-13.
    • (2006) Proc Natl Acad Sci U S A , vol.103 , pp. 3908-3913
    • Kim, K.S.1    Lee, K.W.2    Lee, K.W.3
  • 258
    • 0031950047 scopus 로고    scopus 로고
    • Influence of Gz and Gi2 transducer proteins in the affinity of opioid agonists to mu receptors
    • Garzón J, Castro M, Sánchez-Blazquez P, Sánchez-Blázquez P. Influence of Gz and Gi2 transducer proteins in the affinity of opioid agonists to mu receptors. Eur J Neurosci 1998; 10: 2557-64.
    • (1998) Eur J Neurosci , vol.10 , pp. 2557-2564
    • Garzón, J.1    Castro, M.2    Sánchez-Blazquez, P.3    Sánchez-Blázquez, P.4
  • 259
    • 0034617376 scopus 로고    scopus 로고
    • Hypertolerance to morphine in G(z alpha)-deficient mice
    • Hendry IA, Kelleher KL, Bartlett SE, et al. Hypertolerance to morphine in G(z alpha)-deficient mice. Brain Res 2000; 870: 10-9.
    • (2000) Brain Res , vol.870 , pp. 10-19
    • Hendry, I.A.1    Kelleher, K.L.2    Bartlett, S.E.3
  • 260
    • 1642363345 scopus 로고    scopus 로고
    • Deletion of guanine nucleotide binding protein alpha z subunit in mice induces a gene dose dependent tolerance to morphine
    • Leck KJ, Bartlett SE, Smith MT, et al. Deletion of guanine nucleotide binding protein alpha z subunit in mice induces a gene dose dependent tolerance to morphine. Neuropharmacology 2004; 46: 836-46.
    • (2004) Neuropharmacology , vol.46 , pp. 836-846
    • Leck, K.J.1    Bartlett, S.E.2    Smith, M.T.3
  • 261
    • 14044273637 scopus 로고    scopus 로고
    • R7BP, a novel neuronal protein interacting with RGS proteins of the R7 family
    • Martemyanov KA, Yoo PJ, Skiba NP, Arshavsky VY. R7BP, a novel neuronal protein interacting with RGS proteins of the R7 family. J Biol Chem 2005; 280: 5133-6.
    • (2005) J Biol Chem , vol.280 , pp. 5133-5136
    • Martemyanov, K.A.1    Yoo, P.J.2    Skiba, N.P.3    Arshavsky, V.Y.4
  • 262
    • 0025907203 scopus 로고
    • Excitatory amino acid antagonists (kynurenic acid and MK-801) attenuate the development of morphine tolerance in the rat
    • Marek P, Ben-Eliyahu S, Gold M, Liebeskind JC. Excitatory amino acid antagonists (kynurenic acid and MK-801) attenuate the development of morphine tolerance in the rat. Brain Res 1991; 547: 77-81.
    • (1991) Brain Res , vol.547 , pp. 77-81
    • Marek, P.1    Ben-Eliyahu, S.2    Gold, M.3    Liebeskind, J.C.4
  • 263
    • 13244292311 scopus 로고    scopus 로고
    • An antisense oligonucleotide to the N-methyl-D-aspartate (NMDA) subunit NMDAR1 attenuates NMDA-induced nociception, hyperalgesia, and morphine tolerance
    • Shimoyama N, Shimoyama M, Davis AM, Monaghan DT, Inturrisi CE. An antisense oligonucleotide to the N-methyl-D-aspartate (NMDA) subunit NMDAR1 attenuates NMDA-induced nociception, hyperalgesia, and morphine tolerance. J Pharmacol Exp Ther 2005; 312: 834-40.
    • (2005) J Pharmacol Exp Ther , vol.312 , pp. 834-840
    • Shimoyama, N.1    Shimoyama, M.2    Davis, A.M.3    Monaghan, D.T.4    Inturrisi, C.E.5
  • 264
    • 84863727622 scopus 로고    scopus 로고
    • Tolerance and opioid-induced hyperalgesia. Is a divorce imminent?
    • Richebe P, Cahana A, Rivat C. Tolerance and opioid-induced hyperalgesia. Is a divorce imminent? Pain 2012.
    • (2012) Pain
    • Richebe, P.1    Cahana, A.2    Rivat, C.3
  • 265
    • 0033957306 scopus 로고    scopus 로고
    • Antagonists of excitatory opioid receptor functions enhance morphine's analgesic potency and attenuate opioid tolerance/dependence liability
    • Crain SM, Shen KF. Antagonists of excitatory opioid receptor functions enhance morphine's analgesic potency and attenuate opioid tolerance/dependence liability. Pain 2000; 84: 121-31.
    • (2000) Pain , vol.84 , pp. 121-131
    • Crain, S.M.1    Shen, K.F.2
  • 266
    • 0035900180 scopus 로고    scopus 로고
    • Cholera toxin-B subunit blocks excitatory opioid receptor-mediated hyperalgesic effects in mice, thereby unmasking potent opioid analgesia and attenuating opioid tolerance/dependence
    • Shen KF, Crain SM. Cholera toxin-B subunit blocks excitatory opioid receptor-mediated hyperalgesic effects in mice, thereby unmasking potent opioid analgesia and attenuating opioid tolerance/dependence. Brain Res 2001; 919: 20-30.
    • (2001) Brain Res , vol.919 , pp. 20-30
    • Shen, K.F.1    Crain, S.M.2
  • 268
    • 0028952242 scopus 로고
    • The human mu opioid receptor: Modulation of functional desensitization by calcium/calmodulin-dependent protein kinase and protein kinase C
    • Mestek A, Hurley JH, Bye LS, et al. The human mu opioid receptor: modulation of functional desensitization by calcium/calmodulin-dependent protein kinase and protein kinase C. J Neurosci 1995; 15: 2396-406.
    • (1995) J Neurosci , vol.15 , pp. 2396-2406
    • Mestek, A.1    Hurley, J.H.2    Bye, L.S.3
  • 269
    • 2942657437 scopus 로고    scopus 로고
    • Role of the calcium/calmodulin-dependent protein kinase II (CaMKII) in the morphine-induced pharmacological effects in the mouse
    • Narita M, Matsumura Y, Ozaki S, et al. Role of the calcium/calmodulin-dependent protein kinase II (CaMKII) in the morphine-induced pharmacological effects in the mouse. Neuroscience 2004; 126: 415-21.
    • (2004) Neuroscience , vol.126 , pp. 415-421
    • Narita, M.1    Matsumura, Y.2    Ozaki, S.3
  • 270
    • 0030922877 scopus 로고    scopus 로고
    • Site mutation in the rat mu-opioid receptor demonstrates the involvement of calcium/calmodulin-dependent protein kinase II in agonistmediated desensitization
    • Koch T, Kroslak T, Mayer P, Raulf E, Höllt V. Site mutation in the rat mu-opioid receptor demonstrates the involvement of calcium/calmodulin-dependent protein kinase II in agonistmediated desensitization. J Neurochem 1997; 69: 1767-70.
    • (1997) J Neurochem , vol.69 , pp. 1767-1770
    • Koch, T.1    Kroslak, T.2    Mayer, P.3    Raulf, E.4    Höllt, V.5
  • 271
    • 1842790618 scopus 로고    scopus 로고
    • Bidirectional regulation of neuronal nitric-oxide synthase phosphorylation at serine 847 by the N-methyl-D-aspartate receptor
    • Rameau GA, Chiu LY, Ziff EB. Bidirectional regulation of neuronal nitric-oxide synthase phosphorylation at serine 847 by the N-methyl-D-aspartate receptor. J Biol Chem 2004; 279: 14307-14.
    • (2004) J Biol Chem , vol.279 , pp. 14307-14314
    • Rameau, G.A.1    Chiu, L.Y.2    Ziff, E.B.3
  • 272
    • 0035152963 scopus 로고    scopus 로고
    • Glutamate receptors and nociception: Implications for the drug treatment of pain
    • Fundytus ME. Glutamate receptors and nociception: implications for the drug treatment of pain. CNS Drugs 2001; 15: 29-58.
    • (2001) CNS Drugs , vol.15 , pp. 29-58
    • Fundytus, M.E.1
  • 273
    • 69249149994 scopus 로고    scopus 로고
    • Selective s-1 (sigma1) receptor antagonists: Emerging target for the treatment of neuropathic pain
    • Diaz JL, Zamanillo D, Corbera J, et al. Selective s-1 (sigma1) receptor antagonists: emerging target for the treatment of neuropathic pain. Cent Nerv Syst Agents Med Chem 2009; 9: 172-83.
    • (2009) Cent Nerv Syst Agents Med Chem , vol.9 , pp. 172-183
    • Diaz, J.L.1    Zamanillo, D.2    Corbera, J.3
  • 274
    • 84862786212 scopus 로고    scopus 로고
    • Blockade of PDGFR-beta activation eliminates morphine analgesic tolerance
    • Wang Y, Barker K, Shi S, et al. Blockade of PDGFR-beta activation eliminates morphine analgesic tolerance. Nat Med 2012; 18: 385-7.
    • (2012) Nat Med , vol.18 , pp. 385-387
    • Wang, Y.1    Barker, K.2    Shi, S.3
  • 275
    • 0037115025 scopus 로고    scopus 로고
    • Genetic dissociation of opiate tolerance and physical dependence in d-opioid receptor-1 and preproenkephalin knock-out mice
    • Nitsche JF, Schuller AG, King MA, et al. Genetic dissociation of opiate tolerance and physical dependence in d-opioid receptor-1 and preproenkephalin knock-out mice. J Neurosci 2002; 22: 10906-13.
    • (2002) J Neurosci , vol.22 , pp. 10906-10913
    • Nitsche, J.F.1    Schuller, A.G.2    King, M.A.3
  • 276
    • 9444274109 scopus 로고    scopus 로고
    • Protein kinase C activation enhances morphine-induced rapid desensitization of m-opioid receptors in mature rat locus ceruleus neurons
    • Bailey CP, Kelly E, Henderson G. Protein kinase C activation enhances morphine-induced rapid desensitization of m-opioid receptors in mature rat locus ceruleus neurons. Mol Pharmacol 2004; 66: 1592-8.
    • (2004) Mol Pharmacol , vol.66 , pp. 1592-1598
    • Bailey, C.P.1    Kelly, E.2    Henderson, G.3
  • 277
    • 77249125513 scopus 로고    scopus 로고
    • The effect of protein kinase C and G protein-coupled receptor kinase inhibition on tolerance induced by mu-opioid agonists of different efficacy
    • Hull LC, Llorente J, Gabra BH, et al. The effect of protein kinase C and G protein-coupled receptor kinase inhibition on tolerance induced by mu-opioid agonists of different efficacy. J Pharmacol Exp Ther 2010; 332: 1127-35.
    • (2010) J Pharmacol Exp Ther , vol.332 , pp. 1127-1135
    • Hull, L.C.1    Llorente, J.2    Gabra, B.H.3
  • 278
    • 0035056184 scopus 로고    scopus 로고
    • Activation of brainstem Nmethyl-D-aspartate receptors is required for the analgesic actions of morphine given systemically
    • Heinricher MM, Schouten JC, Jobst EE. Activation of brainstem Nmethyl-D-aspartate receptors is required for the analgesic actions of morphine given systemically. Pain 2001; 92: 129-38.
    • (2001) Pain , vol.92 , pp. 129-138
    • Heinricher, M.M.1    Schouten, J.C.2    Jobst, E.E.3
  • 279
    • 0242495833 scopus 로고    scopus 로고
    • A novel endocytic recycling signal that distinguishes the membrane trafficking of naturally occurring opioid receptors
    • Tanowitz M, von Zastrow M. A novel endocytic recycling signal that distinguishes the membrane trafficking of naturally occurring opioid receptors. J Biol Chem 2003; 278: 45978-86.
    • (2003) J Biol Chem , vol.278 , pp. 45978-45986
    • Tanowitz, M.1    von Zastrow, M.2
  • 281
    • 0026002567 scopus 로고
    • Magnitude of acute tolerance to opioids is not related to their potency
    • Kissin I, Brown PT, Bradley EL, Jr. Magnitude of acute tolerance to opioids is not related to their potency. Anesthesiology 1991; 75: 813-6.
    • (1991) Anesthesiology , vol.75 , pp. 813-816
    • Kissin, I.1    Brown, P.T.2    Bradley Jr., E.L.3
  • 282
    • 80053276069 scopus 로고    scopus 로고
    • Analgesic tolerance to highefficacy agonists but not to morphine is diminished in phosphorylation-deficient S375A mu-opioid receptor knock-in mice
    • Grecksch G, Just S, Pierstorff C, et al. Analgesic tolerance to highefficacy agonists but not to morphine is diminished in phosphorylation-deficient S375A mu-opioid receptor knock-in mice. J Neurosci 2011; 31: 13890-6.
    • (2011) J Neurosci , vol.31 , pp. 13890-13896
    • Grecksch, G.1    Just, S.2    Pierstorff, C.3
  • 283
    • 0345305690 scopus 로고    scopus 로고
    • Mu-opioid receptor desensitization in mature rat neurons: Lack of interaction between DAMGO and morphine
    • Bailey CP, Couch D, Johnson E, et al. Mu-opioid receptor desensitization in mature rat neurons: lack of interaction between DAMGO and morphine. J Neurosci 2003; 23: 10515-20.
    • (2003) J Neurosci , vol.23 , pp. 10515-10520
    • Bailey, C.P.1    Couch, D.2    Johnson, E.3
  • 284
    • 0024391747 scopus 로고
    • Intracerebroventricular N-ethylmaleimide differentially reduces supraspinal opioid analgesia in mice
    • Sánchez-Blazquez P, Ulibarri I, Garzón J. Intracerebroventricular N-ethylmaleimide differentially reduces supraspinal opioid analgesia in mice. Eur J Pharmacol 1989; 166: 193-200.
    • (1989) Eur J Pharmacol , vol.166 , pp. 193-200
    • Sánchez-Blazquez, P.1    Ulibarri, I.2    Garzón, J.3
  • 285
    • 0027953839 scopus 로고
    • Dissimilar efficacy of opioids to produce mu-mediated analgesia: Role of Gx/z and Gi2 transducer proteins
    • Garzón J, Martínez-Peña Y, Sánchez-Blazquez P. Dissimilar efficacy of opioids to produce mu-mediated analgesia: role of Gx/z and Gi2 transducer proteins. Life Sci 1994; 55: L205-L212.
    • (1994) Life Sci , vol.55
    • Garzón, J.1    Martínez-Peña, Y.2    Sánchez-Blazquez, P.3
  • 286
    • 0030910987 scopus 로고    scopus 로고
    • Opioids binding mu and delta receptors exhibit diverse efficacy in the activation of Gi2 and G(x/z) transducer proteins in mouse periaqueductal gray matter
    • Garzón J, García-España A, Sánchez-Blazquez P. Opioids binding mu and delta receptors exhibit diverse efficacy in the activation of Gi2 and G(x/z) transducer proteins in mouse periaqueductal gray matter. J Pharmacol Exp Ther 1997; 281: 549-57.
    • (1997) J Pharmacol Exp Ther , vol.281 , pp. 549-557
    • Garzón, J.1    García-España, A.2    Sánchez-Blazquez, P.3
  • 287
    • 0031808936 scopus 로고    scopus 로고
    • Delta opioid receptor subtypes activate inositol-signaling pathways in the production of antinociception
    • Sánchez-Blázquez P, Garzón J. Delta opioid receptor subtypes activate inositol-signaling pathways in the production of antinociception. J Pharmacol Exp Ther 1998; 285: 820-7.
    • (1998) J Pharmacol Exp Ther , vol.285 , pp. 820-827
    • Sánchez-Blázquez, P.1    Garzón, J.2
  • 288
    • 0031950047 scopus 로고    scopus 로고
    • Influence of Gz and Gi2 transducer proteins in the affinity of opioid agonists to mu receptors
    • Garzón J, Castro M, Sánchez-Blázquez P. Influence of Gz and Gi2 transducer proteins in the affinity of opioid agonists to mu receptors. Eur J Neurosci 1998; 10: 2557-64.
    • (1998) Eur J Neurosci , vol.10 , pp. 2557-2564
    • Garzón, J.1    Castro, M.2    Sánchez-Blázquez, P.3
  • 289
    • 70349122557 scopus 로고    scopus 로고
    • Role of N-methyl-D-aspartate receptor antagonists in postoperative pain management
    • Suzuki M. Role of N-methyl-D-aspartate receptor antagonists in postoperative pain management. Curr Opin Anaesthesiol 2009; 22: 618-22.
    • (2009) Curr Opin Anaesthesiol , vol.22 , pp. 618-622
    • Suzuki, M.1
  • 290
    • 69249229607 scopus 로고    scopus 로고
    • Role of protein kinase C in functional selectivity for desensitization at the mu-opioid receptor: From pharmacological curiosity to therapeutic potential
    • Ingram SL, Traynor JR. Role of protein kinase C in functional selectivity for desensitization at the mu-opioid receptor: from pharmacological curiosity to therapeutic potential. Br J Pharmacol 2009; 158: 154-6.
    • (2009) Br J Pharmacol , vol.158 , pp. 154-156
    • Ingram, S.L.1    Traynor, J.R.2
  • 292
    • 0019410287 scopus 로고
    • Inhibition of tolerance to the pharmacological effects of human b-endorphin by prolyl-leucyl-glycinamide and cyclo(leucylglycine) in the rat
    • Bhargava HN. Inhibition of tolerance to the pharmacological effects of human b-endorphin by prolyl-leucyl-glycinamide and cyclo(leucylglycine) in the rat. J Pharmacol Exp Ther 1981; 218: 404-8.
    • (1981) J Pharmacol Exp Ther , vol.218 , pp. 404-408
    • Bhargava, H.N.1
  • 293
    • 0023788179 scopus 로고
    • Endogenous oxytocin inhibits morphine tolerance through limbic forebrain oxytocin receptors
    • Sarnyai Z, Viski S, Krivan M, et al. Endogenous oxytocin inhibits morphine tolerance through limbic forebrain oxytocin receptors. Brain Res 1988; 463: 284-8.
    • (1988) Brain Res , vol.463 , pp. 284-288
    • Sarnyai, Z.1    Viski, S.2    Krivan, M.3
  • 294
    • 0023836208 scopus 로고
    • Enhancement of morphine analgesia and prevention of morphine tolerance in the rat by the cholecystokinin antagonist L-364,718
    • Dourish CT, Hawley D, Iversen SD. Enhancement of morphine analgesia and prevention of morphine tolerance in the rat by the cholecystokinin antagonist L-364,718. Eur J Pharmacol 1988; 147: 469-72.
    • (1988) Eur J Pharmacol , vol.147 , pp. 469-472
    • Dourish, C.T.1    Hawley, D.2    Iversen, S.D.3
  • 295
    • 0023898750 scopus 로고
    • Calcium channel antagonists increase morphine-induced analgesia and antagonize morphine tolerance
    • Contreras E, Tamayo L, Amigo M. Calcium channel antagonists increase morphine-induced analgesia and antagonize morphine tolerance. Eur J Pharmacol 1988; 148: 463-6.
    • (1988) Eur J Pharmacol , vol.148 , pp. 463-466
    • Contreras, E.1    Tamayo, L.2    Amigo, M.3
  • 296
    • 0024330196 scopus 로고
    • Modification of morphineinduced analgesia, tolerance and dependence by bromocriptine
    • Gomaa AA, Mohamed LH, Ahmed HN. Modification of morphineinduced analgesia, tolerance and dependence by bromocriptine. Eur J Pharmacol 1989; 170: 129-35.
    • (1989) Eur J Pharmacol , vol.170 , pp. 129-135
    • Gomaa, A.A.1    Mohamed, L.H.2    Ahmed, H.N.3
  • 297
    • 0026465706 scopus 로고
    • Analog of neuropeptide FF attenuates morphine tolerance
    • Lake JR, Hebert KM, Payza K, et al. Analog of neuropeptide FF attenuates morphine tolerance. Neurosci Lett 1992; 146: 203-6.
    • (1992) Neurosci Lett , vol.146 , pp. 203-206
    • Lake, J.R.1    Hebert, K.M.2    Payza, K.3
  • 299
    • 0029092919 scopus 로고
    • Effects of calcitonin gene-related peptide on acute and chronic effects of morphine
    • Azarov AV, Szabo G, Czako L, Telegdy G. Effects of calcitonin gene-related peptide on acute and chronic effects of morphine. Pharmacol Biochem Behav 1995; 52: 595-9.
    • (1995) Pharmacol Biochem Behav , vol.52 , pp. 595-599
    • Azarov, A.V.1    Szabo, G.2    Czako, L.3    Telegdy, G.4
  • 301
    • 0030426798 scopus 로고    scopus 로고
    • The substance P amino-terminal metabolite substance P(1-7), administered peripherally, prevents the development of acute morphine tolerance and attenuates the expression of withdrawal in mice
    • Kreeger JS, Larson AA. The substance P amino-terminal metabolite substance P(1-7), administered peripherally, prevents the development of acute morphine tolerance and attenuates the expression of withdrawal in mice. J Pharmacol Exp Ther 1996; 279: 662-7.
    • (1996) J Pharmacol Exp Ther , vol.279 , pp. 662-667
    • Kreeger, J.S.1    Larson, A.A.2
  • 302
    • 0030859694 scopus 로고    scopus 로고
    • Chronic neurosteroid treatment prevents the development of morphine tolerance and attenuates abstinence behavior in mice
    • Reddy DS, Kulkarni SK. Chronic neurosteroid treatment prevents the development of morphine tolerance and attenuates abstinence behavior in mice. Eur J Pharmacol 1997; 337: 19-25.
    • (1997) Eur J Pharmacol , vol.337 , pp. 19-25
    • Reddy, D.S.1    Kulkarni, S.K.2
  • 303
    • 0033542790 scopus 로고    scopus 로고
    • Reversal of morphine tolerance and dependence by melatonin: Possible role of central and peripheral benzodiazepine receptors
    • Raghavendra V, Kulkarni SK. Reversal of morphine tolerance and dependence by melatonin: possible role of central and peripheral benzodiazepine receptors. Brain Res 1999; 834: 178-81.
    • (1999) Brain Res , vol.834 , pp. 178-181
    • Raghavendra, V.1    Kulkarni, S.K.2
  • 304
    • 0034667560 scopus 로고    scopus 로고
    • Enhanced spinal nociceptin receptor expression develops morphine tolerance and dependence
    • Ueda H, Inoue M, Takeshima H, Iwasawa Y. Enhanced spinal nociceptin receptor expression develops morphine tolerance and dependence. J Neurosci 2000; 20: 7640-7.
    • (2000) J Neurosci , vol.20 , pp. 7640-7647
    • Ueda, H.1    Inoue, M.2    Takeshima, H.3    Iwasawa, Y.4


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.