Skip to main content
Top
Published in: CNS Drugs 1/2001

01-01-2001 | Review Article

Glutamate Receptors and Nociception

Implications for the Drug Treatment of Pain

Author: Dr Marian E. Fundytus

Published in: CNS Drugs | Issue 1/2001

Login to get access

Abstract

Evidence from the last several decades indicates that the excitatory amino acid glutamate plays a significant role in nociceptive processing. Glutamate and glutamate receptors are located in areas of the brain, spinal cord and periphery that are involved in pain sensation and transmission. Glutamate acts at several types of receptors, including ionotropic (directly coupled to ion channels) and metabotropic (directly coupled to intracellular second messengers). Ionotropic receptors include those selectively activated by N-methyl-D-aspartate, α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid and kainate. Metabotropic glutamate receptors are classified into 3 groups based on sequence homology, signal transduction mechanisms and receptor pharmacology.
Glutamate also interacts with the opioid system, and intrathecal or systemic coadministration of glutamate receptor antagonists with opioids may enhance analgesia while reducing the development of opioid tolerance and dependence. The actions of glutamate in the brain seem to be more complex. Activation of glutamate receptors in some brain areas seems to be pronociceptive (e.g. thalamus, trigeminal nucleus), although activation of glutamate receptors in other brain areas seems to be antinociceptive (e.g. periaqueductal grey, ventrolateral medulla).
Application of glutamate, or agonists selective for one of the several types of glutamate receptor, to the spinal cord or periphery induces nociceptive behaviours. Inhibition of glutamate release, or of glutamate receptors, in the spinal cord or periphery attenuates both acute and chronic pain in animal models. Similar benefits have been seen in studies involving humans (both patients and volunteers); however, results have been inconsistent. More research is needed to clearly define the role of existing treatment options and explore the possibilities for future drug development.
Literature
1.
go back to reference Nakanishi S. Molecular diversity of glutamate receptors and implications for brain function. Science 1992; 258: 597–603PubMedCrossRef Nakanishi S. Molecular diversity of glutamate receptors and implications for brain function. Science 1992; 258: 597–603PubMedCrossRef
2.
go back to reference MacDermott AB, Mayer ML, Westbrook GL, et al. NMDA-receptor activation increases cytoplasmic calcium concentration in cultured spinal cord neurones. Nature 1986; 321: 519–22PubMedCrossRef MacDermott AB, Mayer ML, Westbrook GL, et al. NMDA-receptor activation increases cytoplasmic calcium concentration in cultured spinal cord neurones. Nature 1986; 321: 519–22PubMedCrossRef
3.
go back to reference Mayer ML, MacDermott AB, Westbrook GL, et al. Agonist-and voltage-gated calcium entry in cultured mouse spinal cord neurons under voltage clamp measured using Arsenazo III. J Neurosci 1987; 7: 3230–44PubMed Mayer ML, MacDermott AB, Westbrook GL, et al. Agonist-and voltage-gated calcium entry in cultured mouse spinal cord neurons under voltage clamp measured using Arsenazo III. J Neurosci 1987; 7: 3230–44PubMed
4.
go back to reference Nowak L, Bregstovski P, Ascher P, et al. Magnesium gates glutamate-activated channels in mouse central neurones. Nature 1984; 307: 462–5PubMedCrossRef Nowak L, Bregstovski P, Ascher P, et al. Magnesium gates glutamate-activated channels in mouse central neurones. Nature 1984; 307: 462–5PubMedCrossRef
5.
go back to reference Mayer ML, Westbrook GL, Guthrie PB. Voltage-dependent block by Mg2+ of NMDA responses in spinal cord neurones. Nature 1984; 309: 261–3PubMedCrossRef Mayer ML, Westbrook GL, Guthrie PB. Voltage-dependent block by Mg2+ of NMDA responses in spinal cord neurones. Nature 1984; 309: 261–3PubMedCrossRef
6.
go back to reference Cotman CW, Monaghan DT. Excitatory amino acid neurotransmission: NMDA receptors and Hebb-type synaptic plasticity. Ann Rev Neurosci 1988; 11: 61–80PubMedCrossRef Cotman CW, Monaghan DT. Excitatory amino acid neurotransmission: NMDA receptors and Hebb-type synaptic plasticity. Ann Rev Neurosci 1988; 11: 61–80PubMedCrossRef
7.
go back to reference Holleman M, Heineman S. Cloned glutamate receptors. Ann Rev Neurosci 1994; 17: 31–108CrossRef Holleman M, Heineman S. Cloned glutamate receptors. Ann Rev Neurosci 1994; 17: 31–108CrossRef
8.
go back to reference Kemp JA, Leeson PD. The glycine site of the NMDA receptor: five years on. Trends Pharmacol Sci 1993; 14: 20–5PubMedCrossRef Kemp JA, Leeson PD. The glycine site of the NMDA receptor: five years on. Trends Pharmacol Sci 1993; 14: 20–5PubMedCrossRef
9.
go back to reference Lodge D, Johnson KM. Noncompetitive excitatory amino acid receptor antagonists. In: Lodge D, Collingridge GL, editors. Trends in pharmacological science, special report: The pharmacology of excitatory amino acids. Cambridge: Elsevier, 1991:13–8 Lodge D, Johnson KM. Noncompetitive excitatory amino acid receptor antagonists. In: Lodge D, Collingridge GL, editors. Trends in pharmacological science, special report: The pharmacology of excitatory amino acids. Cambridge: Elsevier, 1991:13–8
10.
go back to reference Scott RH, Sutton KG, Dolphin AC. Interactions of polyamines with neuronal ion channels. Trends Neurosci 1993; 16: 153–60PubMedCrossRef Scott RH, Sutton KG, Dolphin AC. Interactions of polyamines with neuronal ion channels. Trends Neurosci 1993; 16: 153–60PubMedCrossRef
11.
go back to reference Sucher NJ, Awobuluyi M, Choi Y-B, et al. NMDA receptors: from genes to channels. Trends Pharmacol Sci 1996; 17: 348–55PubMed Sucher NJ, Awobuluyi M, Choi Y-B, et al. NMDA receptors: from genes to channels. Trends Pharmacol Sci 1996; 17: 348–55PubMed
12.
go back to reference Moriyoshi K, Masu M, Ishiik T, et al. Molecular cloning and characterization of the rat NMDA receptor. Nature 1991; 354: 31–7PubMedCrossRef Moriyoshi K, Masu M, Ishiik T, et al. Molecular cloning and characterization of the rat NMDA receptor. Nature 1991; 354: 31–7PubMedCrossRef
13.
go back to reference Ishii I, Moriyoshi K, Sugihara H, et al. Molecular characterization of the family of the N-methyl-D-aspartate receptor subunits. J Biol Chem 1993; 268: 2836–43PubMed Ishii I, Moriyoshi K, Sugihara H, et al. Molecular characterization of the family of the N-methyl-D-aspartate receptor subunits. J Biol Chem 1993; 268: 2836–43PubMed
14.
go back to reference Meguro H, Mori H, Araki K, et al. Functional characterization of a heteromeric NMDA receptor channel expressed from cloned cDNAs. Nature 1992; 357: 70–4PubMedCrossRef Meguro H, Mori H, Araki K, et al. Functional characterization of a heteromeric NMDA receptor channel expressed from cloned cDNAs. Nature 1992; 357: 70–4PubMedCrossRef
15.
go back to reference Kutsuwada T, Kashiwabuchi N, Mori H, et al. Molecular diversity of the NMDA receptor channel. Nature 1992; 358: 36–41PubMedCrossRef Kutsuwada T, Kashiwabuchi N, Mori H, et al. Molecular diversity of the NMDA receptor channel. Nature 1992; 358: 36–41PubMedCrossRef
16.
go back to reference Monyer H, Sprengel R, Schoepfer R, et al. Heteromeric NMDA receptors: molecular and functional distinction of subtypes. Science 1992; 256: 1217–21PubMedCrossRef Monyer H, Sprengel R, Schoepfer R, et al. Heteromeric NMDA receptors: molecular and functional distinction of subtypes. Science 1992; 256: 1217–21PubMedCrossRef
17.
go back to reference Ikeda K, Nagasawa M, Mori H, et al. Cloning and expression of the epsilon 4 subunit of the NMDA receptor channel. FEBS Lett 1992; 313: 34–8PubMedCrossRef Ikeda K, Nagasawa M, Mori H, et al. Cloning and expression of the epsilon 4 subunit of the NMDA receptor channel. FEBS Lett 1992; 313: 34–8PubMedCrossRef
18.
go back to reference Mao J, Price DD, Mayer DJ. Mechanisms of hyperalgesia and morphine tolerance: a current view of their possible interactions. Pain 1995; 62: 259–74PubMedCrossRef Mao J, Price DD, Mayer DJ. Mechanisms of hyperalgesia and morphine tolerance: a current view of their possible interactions. Pain 1995; 62: 259–74PubMedCrossRef
19.
go back to reference Morris R, Southam E, Gittins SR, et al. The NO-cGMP pathway in neonatal rat dorsal horn. Eur J Neurosci 1994; 6: 876–9PubMedCrossRef Morris R, Southam E, Gittins SR, et al. The NO-cGMP pathway in neonatal rat dorsal horn. Eur J Neurosci 1994; 6: 876–9PubMedCrossRef
20.
go back to reference Murphy SN, Miller RJ. Regulation of Ca++ influx into striatal neurons by kainic acid. J Pharmacol Exp Ther 1989; 249: 184–93PubMed Murphy SN, Miller RJ. Regulation of Ca++ influx into striatal neurons by kainic acid. J Pharmacol Exp Ther 1989; 249: 184–93PubMed
22.
go back to reference Sommer B, Seeburg PH. Glutamate receptor channels: novel properties and new clones. Trends Pharmacol Sci 1992; 13: 291–6PubMedCrossRef Sommer B, Seeburg PH. Glutamate receptor channels: novel properties and new clones. Trends Pharmacol Sci 1992; 13: 291–6PubMedCrossRef
23.
go back to reference Cull-Candy SG, Ogden DC. Ion channels activated by L-glutamate and GABA in cultured cerebellar neurons of the rat. Proc R Soc London-Series B: Biol Sci 1985; 224, 367–73CrossRef Cull-Candy SG, Ogden DC. Ion channels activated by L-glutamate and GABA in cultured cerebellar neurons of the rat. Proc R Soc London-Series B: Biol Sci 1985; 224, 367–73CrossRef
24.
go back to reference Kiskin NI, Krishtal OA, Tsyndrenko AY. Excitatory amino acid receptors in hippocampal neurons: Kainate fails to desensitize them. Neurosci Lett 1986; 63: 225–30PubMedCrossRef Kiskin NI, Krishtal OA, Tsyndrenko AY. Excitatory amino acid receptors in hippocampal neurons: Kainate fails to desensitize them. Neurosci Lett 1986; 63: 225–30PubMedCrossRef
25.
go back to reference Tang C-M, Dichter M, Morad M. Quisqualate activates a rapidly inactivating high conductance ionic channel in hippocampal neurons. Science 1989; 243: 1474–7PubMedCrossRef Tang C-M, Dichter M, Morad M. Quisqualate activates a rapidly inactivating high conductance ionic channel in hippocampal neurons. Science 1989; 243: 1474–7PubMedCrossRef
26.
go back to reference Trussell LO, Thio LL, Zorumski CF, et al. Rapid desensitization of glutamate receptors in vertebrate central neurons. Proc Natl Acad Sci USA 1988; 85: 4562–6PubMedCrossRef Trussell LO, Thio LL, Zorumski CF, et al. Rapid desensitization of glutamate receptors in vertebrate central neurons. Proc Natl Acad Sci USA 1988; 85: 4562–6PubMedCrossRef
27.
go back to reference Gasic GP, Hollemann M. Molecular neurobiology of glutamate receptors. Ann Rev Physiol 1992; 54: 507–36CrossRef Gasic GP, Hollemann M. Molecular neurobiology of glutamate receptors. Ann Rev Physiol 1992; 54: 507–36CrossRef
28.
go back to reference Hollemann M, O’Shea-Greenfield A, Rogers SW, et al. Cloning by functional expression of a member of the glutamate receptor family. Nature 1989; 342: 643–8CrossRef Hollemann M, O’Shea-Greenfield A, Rogers SW, et al. Cloning by functional expression of a member of the glutamate receptor family. Nature 1989; 342: 643–8CrossRef
29.
go back to reference Keinanen K, Wisden W, Sommer B, et al. A family of AMPA-selective glutamate receptors. Science 1990; 249: 556–60PubMedCrossRef Keinanen K, Wisden W, Sommer B, et al. A family of AMPA-selective glutamate receptors. Science 1990; 249: 556–60PubMedCrossRef
30.
go back to reference Bettler B, Boulter J, Hermans-Borgmeyer I, et al. Cloning of a novel glutamate receptor subunit, GluR5-expression in the nervous system during development. Neuron 1990; 5: 583–95PubMedCrossRef Bettler B, Boulter J, Hermans-Borgmeyer I, et al. Cloning of a novel glutamate receptor subunit, GluR5-expression in the nervous system during development. Neuron 1990; 5: 583–95PubMedCrossRef
31.
go back to reference Bettler B, Egebjerg J, Sharma G, et al. Cloning of a putative glutamate receptor: a low affinity kainate-binding subunit. Neuron 1992; 8: 257–65PubMedCrossRef Bettler B, Egebjerg J, Sharma G, et al. Cloning of a putative glutamate receptor: a low affinity kainate-binding subunit. Neuron 1992; 8: 257–65PubMedCrossRef
32.
go back to reference Egebjerg J, Bettler B, Hermans-Borgmeyer I, et al. Cloning of a cDNA for a glutamate receptor subunit activated by kainate but not AMPA. Nature 1991; 351: 745–8PubMedCrossRef Egebjerg J, Bettler B, Hermans-Borgmeyer I, et al. Cloning of a cDNA for a glutamate receptor subunit activated by kainate but not AMPA. Nature 1991; 351: 745–8PubMedCrossRef
33.
go back to reference Okamoto N, Hori S, Akazawa C, et al. Molecular characterization of a new metabotropic glutamate receptor mGluR7 coupled to inhibitory cyclic AMP signal transduction. J Biol Chem 1994; 269: 1231–6PubMed Okamoto N, Hori S, Akazawa C, et al. Molecular characterization of a new metabotropic glutamate receptor mGluR7 coupled to inhibitory cyclic AMP signal transduction. J Biol Chem 1994; 269: 1231–6PubMed
34.
go back to reference Saugstad JA, Kinzie JM, Mulvihill ER, et al. Cloning and expression of a new member of the L-2-amino-4-phosphonobutyric acid-sensitive class of metabotropic glutamate receptors. Mol Pharmacol 1994; 45: 367–72PubMed Saugstad JA, Kinzie JM, Mulvihill ER, et al. Cloning and expression of a new member of the L-2-amino-4-phosphonobutyric acid-sensitive class of metabotropic glutamate receptors. Mol Pharmacol 1994; 45: 367–72PubMed
35.
go back to reference Duvoisin RM, Zhang C, Ramonell K. Glutamate receptor expressed in the retinal and olfactory bulb. J Neurosci 1995; 15: 3075–83PubMed Duvoisin RM, Zhang C, Ramonell K. Glutamate receptor expressed in the retinal and olfactory bulb. J Neurosci 1995; 15: 3075–83PubMed
36.
go back to reference Houamed KM, Kuijper JL, Gilbert TL, et al. Cloning, expression, and gene structure of a G protein-coupled glutamate receptor from rat brain. Science 1991; 252: 1318–21PubMedCrossRef Houamed KM, Kuijper JL, Gilbert TL, et al. Cloning, expression, and gene structure of a G protein-coupled glutamate receptor from rat brain. Science 1991; 252: 1318–21PubMedCrossRef
37.
go back to reference Abe T, Sugihara H, Nawa H, et al. Molecular characterization of a novel metabotropic glutamate receptor mGluR5 coupled to inositol phosphate/Ca2+ signal transduction. J Biol Chem 1992; 267: 13361–8PubMed Abe T, Sugihara H, Nawa H, et al. Molecular characterization of a novel metabotropic glutamate receptor mGluR5 coupled to inositol phosphate/Ca2+ signal transduction. J Biol Chem 1992; 267: 13361–8PubMed
38.
go back to reference Masu M, Tanabe Y, Tsuchida K, et al. Sequence and expression of a metabotropic glutamate receptor. Nature 1991; 349: 760–5PubMedCrossRef Masu M, Tanabe Y, Tsuchida K, et al. Sequence and expression of a metabotropic glutamate receptor. Nature 1991; 349: 760–5PubMedCrossRef
39.
go back to reference Daggett LP, Sacaan AI, Akong M, et al. Molecular and functional characterization of recombinant human metabotropic glutamate receptor subtype 5. Neuropharmacology 1995; 34: 871–86PubMedCrossRef Daggett LP, Sacaan AI, Akong M, et al. Molecular and functional characterization of recombinant human metabotropic glutamate receptor subtype 5. Neuropharmacology 1995; 34: 871–86PubMedCrossRef
40.
go back to reference Martin LJ, Blackstone CD, Huganir RL, et al. Cellular localization of a metabotropic glutamate receptor in rat brain. Neuron 1992; 9: 259–70PubMedCrossRef Martin LJ, Blackstone CD, Huganir RL, et al. Cellular localization of a metabotropic glutamate receptor in rat brain. Neuron 1992; 9: 259–70PubMedCrossRef
41.
go back to reference Joly C, Gomeza J, Brabet I, et al. Molecular, functional, and pharmacological characterization of the metabotropic glutamate receptor type 5 splice variants: comparison with mGluR1. J Neurosci 1995; 15: 3970–81PubMed Joly C, Gomeza J, Brabet I, et al. Molecular, functional, and pharmacological characterization of the metabotropic glutamate receptor type 5 splice variants: comparison with mGluR1. J Neurosci 1995; 15: 3970–81PubMed
42.
go back to reference Pin J-P, Waeber C, Prezeau L, et al. Alternative splicing generates metabotropic glutamate receptors inducing different patterns of calcium release in Xenopus oocytes. Proc Natl Acad Sci 1992; 89: 10331–5PubMedCrossRef Pin J-P, Waeber C, Prezeau L, et al. Alternative splicing generates metabotropic glutamate receptors inducing different patterns of calcium release in Xenopus oocytes. Proc Natl Acad Sci 1992; 89: 10331–5PubMedCrossRef
43.
go back to reference Conn PJ, Pin J-P. Pharmacology and function of metabotropic glutamate receptors. Ann Rev Pharmacol Toxicol 1997; 37: 205–37CrossRef Conn PJ, Pin J-P. Pharmacology and function of metabotropic glutamate receptors. Ann Rev Pharmacol Toxicol 1997; 37: 205–37CrossRef
44.
go back to reference Hayashi Y, Sekiyama N, Nakanishi S, et al. Analysis of agonist and antagonist activities of phenylglycine derivatives for different cloned metabotropic glutamate receptor subtypes. J Neurosci 1994; 14: 3370–7PubMed Hayashi Y, Sekiyama N, Nakanishi S, et al. Analysis of agonist and antagonist activities of phenylglycine derivatives for different cloned metabotropic glutamate receptor subtypes. J Neurosci 1994; 14: 3370–7PubMed
45.
go back to reference Ambrosini A, Meldolesi J. Muscarinic and quisqualate receptor-induced phosphoinositide hydrolysis in primary cultures of striatal and hippocampal neurons. Evidence for differential mechanisms of activation. J Neurochem 1989; 53: 825–33 Ambrosini A, Meldolesi J. Muscarinic and quisqualate receptor-induced phosphoinositide hydrolysis in primary cultures of striatal and hippocampal neurons. Evidence for differential mechanisms of activation. J Neurochem 1989; 53: 825–33
46.
go back to reference Manzoni OJJ, Finiels-Marlier F, Sassetti I, et al. The glutamate receptor of the Qp-type activates protein kinase C and is regulated by protein kinase C. Neurosci Lett 1990; 109: 146–51PubMedCrossRef Manzoni OJJ, Finiels-Marlier F, Sassetti I, et al. The glutamate receptor of the Qp-type activates protein kinase C and is regulated by protein kinase C. Neurosci Lett 1990; 109: 146–51PubMedCrossRef
47.
go back to reference Schoepp DD, Conn PJ. Metabotropic glutamate receptors in brain function and pathology. Trends Pharmacol Sci 1993; 14: 13–25PubMedCrossRef Schoepp DD, Conn PJ. Metabotropic glutamate receptors in brain function and pathology. Trends Pharmacol Sci 1993; 14: 13–25PubMedCrossRef
48.
go back to reference Kapcala LP, Weng C-F, Juang H-H. Protein kinase C activators stimulate beta-endorphin secretion from hypothalamic cells. Brain Res Bull 1992; 29: 553–7PubMedCrossRef Kapcala LP, Weng C-F, Juang H-H. Protein kinase C activators stimulate beta-endorphin secretion from hypothalamic cells. Brain Res Bull 1992; 29: 553–7PubMedCrossRef
49.
go back to reference Graham LT, Shank PR, Werman R, et al. Distribution of some synaptic transmitter suspects in cat spinal cord. J Neurochem 1967; 14: 465–72PubMedCrossRef Graham LT, Shank PR, Werman R, et al. Distribution of some synaptic transmitter suspects in cat spinal cord. J Neurochem 1967; 14: 465–72PubMedCrossRef
50.
go back to reference Johnson JL. The excitant amino acids glutamate and aspartic acid as transmitter candidates in the vertebrate central nervous system. Prog Neurobiol 1978; 10: 155–202PubMedCrossRef Johnson JL. The excitant amino acids glutamate and aspartic acid as transmitter candidates in the vertebrate central nervous system. Prog Neurobiol 1978; 10: 155–202PubMedCrossRef
51.
go back to reference Miller KE, Clements JR, Larson AA, et al. Organization of glutamate-like immunoreactivity in the rat superficial dorsal horn: light and electron microscopic observations. J Comp Neurol 1988; 277: 28–36 Miller KE, Clements JR, Larson AA, et al. Organization of glutamate-like immunoreactivity in the rat superficial dorsal horn: light and electron microscopic observations. J Comp Neurol 1988; 277: 28–36
52.
go back to reference Renno WM. Microdialysis of excitatory amino acids in the periaqueductal gray of the rat after unilateral peripheral inflammation. Amino Acids 1998; 14: 319–31PubMedCrossRef Renno WM. Microdialysis of excitatory amino acids in the periaqueductal gray of the rat after unilateral peripheral inflammation. Amino Acids 1998; 14: 319–31PubMedCrossRef
53.
go back to reference Sasaki M, Tohda C, Kuraishi Y. Region-specific increase in glutamate release from dorsal horn of rats with adjuvant inflammation. Neuroreport 1998; 9: 3219–22PubMedCrossRef Sasaki M, Tohda C, Kuraishi Y. Region-specific increase in glutamate release from dorsal horn of rats with adjuvant inflammation. Neuroreport 1998; 9: 3219–22PubMedCrossRef
54.
go back to reference Sluka KA, Willis WD. Increased spinal release of excitatory amino acids following intradermal injection of capsaicin is reduced by a protein kinase G inhibitor. Brain Res 1998; 798: 281–6PubMedCrossRef Sluka KA, Willis WD. Increased spinal release of excitatory amino acids following intradermal injection of capsaicin is reduced by a protein kinase G inhibitor. Brain Res 1998; 798: 281–6PubMedCrossRef
55.
go back to reference Al-Ghoul WM, Li Volsi G, Weinberg RJ, et al. Glutamate immunocytochemistry in the dorsal horn after injury or stimulation of the sciatic nerve of rats. Brain Res Bull 1993; 30: 453–9PubMedCrossRef Al-Ghoul WM, Li Volsi G, Weinberg RJ, et al. Glutamate immunocytochemistry in the dorsal horn after injury or stimulation of the sciatic nerve of rats. Brain Res Bull 1993; 30: 453–9PubMedCrossRef
56.
go back to reference Omote K, Kawamata T, Kawamata M, et al. Formalin-induced release of excitatory amino acids in the skin of the rat hindpaw. Brain Res 1998; 787: 161–4PubMedCrossRef Omote K, Kawamata T, Kawamata M, et al. Formalin-induced release of excitatory amino acids in the skin of the rat hindpaw. Brain Res 1998; 787: 161–4PubMedCrossRef
57.
go back to reference Greenamyre JT, Young AB, Penny JB. Quantitative autoradiographic distribution of L-[3H]glutamate-binding sites in the rat central nervous system. J Neurosci 1984; 4: 2133–44PubMed Greenamyre JT, Young AB, Penny JB. Quantitative autoradiographic distribution of L-[3H]glutamate-binding sites in the rat central nervous system. J Neurosci 1984; 4: 2133–44PubMed
58.
go back to reference Monaghan DT, Cotman CW. Distribution of N-methyl-D-aspartate-sensitive L-[3H]glutamate-binding sites in rat brain. J Neurosci 1985; 5: 2909–19PubMed Monaghan DT, Cotman CW. Distribution of N-methyl-D-aspartate-sensitive L-[3H]glutamate-binding sites in rat brain. J Neurosci 1985; 5: 2909–19PubMed
59.
go back to reference Jansen KL, Faull RL, Dragunow M, et al. Autoradiographic localisation of NMDA, quisqualate and kainic acid receptors in human spinal cord. Neurosci Lett 1990; 108: 53–7PubMedCrossRef Jansen KL, Faull RL, Dragunow M, et al. Autoradiographic localisation of NMDA, quisqualate and kainic acid receptors in human spinal cord. Neurosci Lett 1990; 108: 53–7PubMedCrossRef
60.
go back to reference Shaw PJ, Ince PG, Johnson M, et al. The quantitative autoradiographic distribution of [3H] MK-801 binding sites in the normal human spinal cord. Brain Res 1991; 539: 164–8PubMedCrossRef Shaw PJ, Ince PG, Johnson M, et al. The quantitative autoradiographic distribution of [3H] MK-801 binding sites in the normal human spinal cord. Brain Res 1991; 539: 164–8PubMedCrossRef
61.
go back to reference Carlton SM, Chung K, Ding Z, et al. Glutamate receptors on postganglionic sympathetic axons. Neurosci 1998; 83: 601–5CrossRef Carlton SM, Chung K, Ding Z, et al. Glutamate receptors on postganglionic sympathetic axons. Neurosci 1998; 83: 601–5CrossRef
62.
go back to reference Catania MV, De Socarraz H, Penney JB, et al. Metabotropic glutamate receptor heterogeneity in rat brain. Mol Pharmacol 1994; 45: 626–36PubMed Catania MV, De Socarraz H, Penney JB, et al. Metabotropic glutamate receptor heterogeneity in rat brain. Mol Pharmacol 1994; 45: 626–36PubMed
63.
go back to reference Carlton SM, Hargett GL, Coggeshall RE. Localization and activation of glutamate receptors in unmyelinated axons of rat glabrous skin. Neurosci Lett 1995; 197: 25–8PubMedCrossRef Carlton SM, Hargett GL, Coggeshall RE. Localization and activation of glutamate receptors in unmyelinated axons of rat glabrous skin. Neurosci Lett 1995; 197: 25–8PubMedCrossRef
64.
go back to reference Masu M, Nakajima Y, Moriyoshi K, et al. Molecular characterization of NMDA and metabotropic glutamate receptors. Ann NY Acad Sci 1994; 707: 153–64CrossRef Masu M, Nakajima Y, Moriyoshi K, et al. Molecular characterization of NMDA and metabotropic glutamate receptors. Ann NY Acad Sci 1994; 707: 153–64CrossRef
65.
go back to reference Koerner JF, Cotman CW. Micromolar L-2-amino-4-phosphonobutyric acid selectively inhibits perforant path synapses from lateral entorhinal cortex. Brain Res 1981; 216: 192–8PubMedCrossRef Koerner JF, Cotman CW. Micromolar L-2-amino-4-phosphonobutyric acid selectively inhibits perforant path synapses from lateral entorhinal cortex. Brain Res 1981; 216: 192–8PubMedCrossRef
66.
go back to reference Evans RH, Francis AA, Jones AW, et al. The effects of a series of-phosphonic —carboxylic amino acids on electrically evoked and amino acid induced responses in isolated spinal cord preparations. Br J Pharmacol 1982; 75: 65–75PubMedCrossRef Evans RH, Francis AA, Jones AW, et al. The effects of a series of-phosphonic —carboxylic amino acids on electrically evoked and amino acid induced responses in isolated spinal cord preparations. Br J Pharmacol 1982; 75: 65–75PubMedCrossRef
67.
go back to reference Davies J, Watkins JC. Actions of D- and L-forms of 2-amino-5-phosphonovalerate and 2-amino-4-phosphonobutyrate in the cat spinal cord. Brain Res 1982; 235: 378–86PubMedCrossRef Davies J, Watkins JC. Actions of D- and L-forms of 2-amino-5-phosphonovalerate and 2-amino-4-phosphonobutyrate in the cat spinal cord. Brain Res 1982; 235: 378–86PubMedCrossRef
68.
go back to reference Monaghan DT, Bridges RJ, Cotman CW. The excitatory amino acid receptors: their classes, pharmacology and distinct properties in the function of the central nervous system. Ann Rev Pharmacol Toxicol 1989; 29: 365–402CrossRef Monaghan DT, Bridges RJ, Cotman CW. The excitatory amino acid receptors: their classes, pharmacology and distinct properties in the function of the central nervous system. Ann Rev Pharmacol Toxicol 1989; 29: 365–402CrossRef
69.
go back to reference Furuyama T, Kiyama H, Sato K, et al. Region-specific expression of subunits of ionotropic glutamate receptors (AMPA-type, KA-type and NMDA receptors) in the rat spinal cord with special reference to nociception. Mol Brain Res 1993; 18: 141–51PubMedCrossRef Furuyama T, Kiyama H, Sato K, et al. Region-specific expression of subunits of ionotropic glutamate receptors (AMPA-type, KA-type and NMDA receptors) in the rat spinal cord with special reference to nociception. Mol Brain Res 1993; 18: 141–51PubMedCrossRef
70.
go back to reference Watanabe M, Mishina M, Inoue Y. Distinct spatiotemporal distributions of the N-methyl-D-aspartate receptor channel subunit mRNAs in the mouse cervical cord. J Comp Neurol 1994; 345: 314–9PubMedCrossRef Watanabe M, Mishina M, Inoue Y. Distinct spatiotemporal distributions of the N-methyl-D-aspartate receptor channel subunit mRNAs in the mouse cervical cord. J Comp Neurol 1994; 345: 314–9PubMedCrossRef
71.
go back to reference Tölle TR, Berthele A, Zieglgänsberger W, et al. The differential expression of 16 NMDA and non-NMDA receptor subunits in the rat spinal cord and in periaqueductal gray. J Neurosci 1993; 13: 5009–28PubMed Tölle TR, Berthele A, Zieglgänsberger W, et al. The differential expression of 16 NMDA and non-NMDA receptor subunits in the rat spinal cord and in periaqueductal gray. J Neurosci 1993; 13: 5009–28PubMed
72.
go back to reference Nagy I, Woolf CJ, Dray A, et al. Cobalt accumulation in neurons expressing ionotropic excitatory amino acid receptors in young rat spinal cord: morphology and distribution. J Comp Neurol 1994; 344: 321–35PubMedCrossRef Nagy I, Woolf CJ, Dray A, et al. Cobalt accumulation in neurons expressing ionotropic excitatory amino acid receptors in young rat spinal cord: morphology and distribution. J Comp Neurol 1994; 344: 321–35PubMedCrossRef
73.
go back to reference Tachibana M, Wenthold RJ, Morioka H, et al. Light and electron microscopic immunocytochemical localization of AMPA-selective glutamate receptors in the rat spinal cord. J Comp Neurol 1994; 344: 431–54PubMedCrossRef Tachibana M, Wenthold RJ, Morioka H, et al. Light and electron microscopic immunocytochemical localization of AMPA-selective glutamate receptors in the rat spinal cord. J Comp Neurol 1994; 344: 431–54PubMedCrossRef
74.
go back to reference Garcia-Ladona FJ, Palacios JM, Probst A, et al. Excitatory amino acid AMPA receptor mRNA localization in several regions of normal and neurological disease affected human brain. An in situ hybridization histochemistry study. Brain Res Mol Brain Res 1994; 21: 75–84 Garcia-Ladona FJ, Palacios JM, Probst A, et al. Excitatory amino acid AMPA receptor mRNA localization in several regions of normal and neurological disease affected human brain. An in situ hybridization histochemistry study. Brain Res Mol Brain Res 1994; 21: 75–84
75.
go back to reference Henley JM, Jenkins R, Hunt SP. Localization of glutamate receptor binding sites and mRNAs to the dorsal horn of the rat spinal cord. Neuropharmacology 1993; 32: 37–41PubMedCrossRef Henley JM, Jenkins R, Hunt SP. Localization of glutamate receptor binding sites and mRNAs to the dorsal horn of the rat spinal cord. Neuropharmacology 1993; 32: 37–41PubMedCrossRef
76.
go back to reference Blackstone CD, Moss SJ, Martin LJ, et al. Biochemical characterization and localization of a non-N-methyl-D-aspartate glutamate receptor in rat brain. J Neurochem 1992; 58: 1118–26PubMedCrossRef Blackstone CD, Moss SJ, Martin LJ, et al. Biochemical characterization and localization of a non-N-methyl-D-aspartate glutamate receptor in rat brain. J Neurochem 1992; 58: 1118–26PubMedCrossRef
77.
go back to reference Allaoua H, Chaudieu I, Krieger C, et al. Alterations in spinal cord excitatory amino acid receptors in amyotrophic lateral sclerosis patients. Brain Res 1992; 579: 169–72PubMedCrossRef Allaoua H, Chaudieu I, Krieger C, et al. Alterations in spinal cord excitatory amino acid receptors in amyotrophic lateral sclerosis patients. Brain Res 1992; 579: 169–72PubMedCrossRef
78.
go back to reference Mitchell JJ, Anderson KJ. Quantitative autoradiographic analysis of excitatory amino acid receptors in the cat spinal cord. Neurosci Lett 1991; 124: 269–72PubMedCrossRef Mitchell JJ, Anderson KJ. Quantitative autoradiographic analysis of excitatory amino acid receptors in the cat spinal cord. Neurosci Lett 1991; 124: 269–72PubMedCrossRef
79.
go back to reference Wenzel A, Scheurer L, Kunzi R, et al. Distribution of NMDA receptor subunit proteins NR2A, 2B, 2C and 2D in rat brain. Neuroreport 1995; 7: 45–8PubMed Wenzel A, Scheurer L, Kunzi R, et al. Distribution of NMDA receptor subunit proteins NR2A, 2B, 2C and 2D in rat brain. Neuroreport 1995; 7: 45–8PubMed
80.
go back to reference Farb CR, Aoki C, Ledoux JE. Differential localization of NMDA and AMPA receptor subunits in the lateral and basal nuclei of the amygdala: a light and electron microscopic study. J Comp Neurol 1995; 362: 86–108PubMedCrossRef Farb CR, Aoki C, Ledoux JE. Differential localization of NMDA and AMPA receptor subunits in the lateral and basal nuclei of the amygdala: a light and electron microscopic study. J Comp Neurol 1995; 362: 86–108PubMedCrossRef
81.
go back to reference Kus L, Saxon D, Beitz AJ. NMDAR1 mRNA distribution in motor and thalamic-projecting sensory neurons in the rat spinal cord and brain stem. Neurosci Lett 1995; 196: 201–4PubMedCrossRef Kus L, Saxon D, Beitz AJ. NMDAR1 mRNA distribution in motor and thalamic-projecting sensory neurons in the rat spinal cord and brain stem. Neurosci Lett 1995; 196: 201–4PubMedCrossRef
82.
go back to reference Liu H, Wang H, Sheng M, et al. Evidence for presynaptic N-methyl-D-aspartate autoreceptors in the spinal cord dorsal horn. Proc Natl Acad Sci 1994; 91: 8383–7PubMedCrossRef Liu H, Wang H, Sheng M, et al. Evidence for presynaptic N-methyl-D-aspartate autoreceptors in the spinal cord dorsal horn. Proc Natl Acad Sci 1994; 91: 8383–7PubMedCrossRef
83.
go back to reference Shaw PJ, Ince PG. A quantitative autoradiographic study of [3H]kainate binding sites in the normal human spinal cord, brainstem and motor cortex. Brain Res 1994; 641: 39–45PubMedCrossRef Shaw PJ, Ince PG. A quantitative autoradiographic study of [3H]kainate binding sites in the normal human spinal cord, brainstem and motor cortex. Brain Res 1994; 641: 39–45PubMedCrossRef
84.
go back to reference Huntley GW, Bickers JC, Jansson W, et al. Distribution and synaptic localization of immunocytochemically identified NMDA receptor subunit proteins in sensory-motor and visual cortices of monkey and human. J Neurosci 1994; 14: 3603–19PubMed Huntley GW, Bickers JC, Jansson W, et al. Distribution and synaptic localization of immunocytochemically identified NMDA receptor subunit proteins in sensory-motor and visual cortices of monkey and human. J Neurosci 1994; 14: 3603–19PubMed
85.
go back to reference Watanabe M, Mishina M, Inoue Y. Distinct gene expression of the N-methyl-D-aspartate receptor channel subunit in peripheral neurons of the mouse sensory ganglia and adrenal gland. Neurosci Lett 1994; 165: 183–6PubMedCrossRef Watanabe M, Mishina M, Inoue Y. Distinct gene expression of the N-methyl-D-aspartate receptor channel subunit in peripheral neurons of the mouse sensory ganglia and adrenal gland. Neurosci Lett 1994; 165: 183–6PubMedCrossRef
86.
go back to reference 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: 667–96PubMed 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: 667–96PubMed
87.
go back to reference Tallaksen-Breene SJ, Young AB, Penney JB, et al. Excitatory amino acid binding sites in the trigeminal principal sensory and spinal trigeminal nuclei of the rat. Neurosci Lett 1992; 141: 79–83CrossRef Tallaksen-Breene SJ, Young AB, Penney JB, et al. Excitatory amino acid binding sites in the trigeminal principal sensory and spinal trigeminal nuclei of the rat. Neurosci Lett 1992; 141: 79–83CrossRef
88.
go back to reference Young AB, Dauth GW, Hollingsworth Z, et al. Quisqualate and NMDA-sensitive [3H]glutamate binding in primate brain. J Neurosci Res 1990; 27: 512–21PubMedCrossRef Young AB, Dauth GW, Hollingsworth Z, et al. Quisqualate and NMDA-sensitive [3H]glutamate binding in primate brain. J Neurosci Res 1990; 27: 512–21PubMedCrossRef
89.
go back to reference Albin RL, Makowiec RL, Hollingsworth Z, et al. Excitatory amino acid binding sites in the periaqueductal gray of the rat. Neurosci Lett 1990; 118: 112–15PubMedCrossRef Albin RL, Makowiec RL, Hollingsworth Z, et al. Excitatory amino acid binding sites in the periaqueductal gray of the rat. Neurosci Lett 1990; 118: 112–15PubMedCrossRef
90.
go back to reference Bowery NG, Wong EH, Hudson AL. Quantitative autoradiography of [3H]-MK-801 binding sites in mammalian brain. Br J Pharmacol 1988; 93: 944–54PubMedCrossRef Bowery NG, Wong EH, Hudson AL. Quantitative autoradiography of [3H]-MK-801 binding sites in mammalian brain. Br J Pharmacol 1988; 93: 944–54PubMedCrossRef
91.
go back to reference Greenamyre JT, Olson JM, Penney Jr JB, et al. Autoradiographic characterization of N-methyl-D-aspartate-, quisqualate-and kainate-sensitive glutamate binding sites. J Pharmacol Exp Ther 1985; 233: 254–63PubMed Greenamyre JT, Olson JM, Penney Jr JB, et al. Autoradiographic characterization of N-methyl-D-aspartate-, quisqualate-and kainate-sensitive glutamate binding sites. J Pharmacol Exp Ther 1985; 233: 254–63PubMed
92.
go back to reference Olsen RW, Szamraj O, Houser CR. [3H]AMPA binding to glutamate receptor subpopulations in rat brain. Brain Res 1987; 402: 243–54PubMedCrossRef Olsen RW, Szamraj O, Houser CR. [3H]AMPA binding to glutamate receptor subpopulations in rat brain. Brain Res 1987; 402: 243–54PubMedCrossRef
93.
go back to reference Boxall SJ, Berthele A, Laurie DJ, et al. Enhanced expression of metabotropic glutamate receptor 3 messenger RNA in the rat spinal cord during ultraviolet irradiation induced peripheral inflammation. Neurosci 1998; 82: 591–602CrossRef Boxall SJ, Berthele A, Laurie DJ, et al. Enhanced expression of metabotropic glutamate receptor 3 messenger RNA in the rat spinal cord during ultraviolet irradiation induced peripheral inflammation. Neurosci 1998; 82: 591–602CrossRef
94.
go back to reference Vidnáyansky Z, Hamori J, Negyessy L, et al. Cellular and subcellular localization of the mGluR5a metabotropic glutamate receptor in rat spinal cord. Neuroreport 1994; 6: 209–13CrossRef Vidnáyansky Z, Hamori J, Negyessy L, et al. Cellular and subcellular localization of the mGluR5a metabotropic glutamate receptor in rat spinal cord. Neuroreport 1994; 6: 209–13CrossRef
95.
go back to reference Li H, Ohishi H, Kinoshita A, et al. Localization of a metabotropic glutamate receptor, mGluR7, in axon terminals of presumed nociceptive, primary afferent fibers in the superficial layers of the spinal dorsal horn: an electron microscope study in the rat. Neurosci Lett 1997; 223: 153–6PubMedCrossRef Li H, Ohishi H, Kinoshita A, et al. Localization of a metabotropic glutamate receptor, mGluR7, in axon terminals of presumed nociceptive, primary afferent fibers in the superficial layers of the spinal dorsal horn: an electron microscope study in the rat. Neurosci Lett 1997; 223: 153–6PubMedCrossRef
96.
go back to reference Ohishi H, Akazawa C, Shigemoto R, et al. Distribution of mRNAs for L-2-amino-phosphonobutyrate-sensitive metabotropic glutamate receptors, mGluR4 and mGluR7, in the rat brain. J Comp Neurol 1995; 360: 555–70PubMedCrossRef Ohishi H, Akazawa C, Shigemoto R, et al. Distribution of mRNAs for L-2-amino-phosphonobutyrate-sensitive metabotropic glutamate receptors, mGluR4 and mGluR7, in the rat brain. J Comp Neurol 1995; 360: 555–70PubMedCrossRef
97.
go back to reference Ohishi H, Nomura S, Ding YQ, et al. Presynaptic localization of a metabotropic glutamate receptor, mGluR7, in the primary afferent neurons: an immunohistochemical study in the rat. Neurosci Lett 1995; 202: 85–8PubMedCrossRef Ohishi H, Nomura S, Ding YQ, et al. Presynaptic localization of a metabotropic glutamate receptor, mGluR7, in the primary afferent neurons: an immunohistochemical study in the rat. Neurosci Lett 1995; 202: 85–8PubMedCrossRef
98.
go back to reference Ohishi H, Shigemoto R, Nakanishi S, et al. Distribution of the mRNA for a metabotropic glutamate receptor (mGluR3) in the rat brain: an in situ hybridization study. J Comp Neurol 1993; 335: 252–66PubMedCrossRef Ohishi H, Shigemoto R, Nakanishi S, et al. Distribution of the mRNA for a metabotropic glutamate receptor (mGluR3) in the rat brain: an in situ hybridization study. J Comp Neurol 1993; 335: 252–66PubMedCrossRef
99.
go back to reference Kinzie JM, Saugstad JA, Westbrook GL, et al. Distribution of metabotropic glutamate receptor 7 messenger RNA in the developing and adult rat brain. Neuroscience 1995; 69: 167–76PubMedCrossRef Kinzie JM, Saugstad JA, Westbrook GL, et al. Distribution of metabotropic glutamate receptor 7 messenger RNA in the developing and adult rat brain. Neuroscience 1995; 69: 167–76PubMedCrossRef
100.
go back to reference Ohishi H, Shigemoto R, Nakanishi S, et al. Distribution of the messenger RNA for a metabtropic glutamate receptor, mGluR2, in the central nervous system of the rat. Neuroscience 1993; 53: 1009–18PubMedCrossRef Ohishi H, Shigemoto R, Nakanishi S, et al. Distribution of the messenger RNA for a metabtropic glutamate receptor, mGluR2, in the central nervous system of the rat. Neuroscience 1993; 53: 1009–18PubMedCrossRef
101.
go back to reference Shigemoto R, Nakanishi S, Mizuno N. Distribution of the mRNA for a metabotropic receptor (mGluR1) in the central nervous system: an in situ hybridization study in adult and developing rat. J Comp Neurol 1992; 322: 121–35PubMedCrossRef Shigemoto R, Nakanishi S, Mizuno N. Distribution of the mRNA for a metabotropic receptor (mGluR1) in the central nervous system: an in situ hybridization study in adult and developing rat. J Comp Neurol 1992; 322: 121–35PubMedCrossRef
102.
go back to reference Curtis DR, Phillips JW, Watkins JC. Chemical excitation of spinal neurons. Nature 1959; 183: 611–2PubMedCrossRef Curtis DR, Phillips JW, Watkins JC. Chemical excitation of spinal neurons. Nature 1959; 183: 611–2PubMedCrossRef
103.
go back to reference Curtis DR, Watkins JC. Acidic amino acids with strong excitatory actions on mammalian neurons. J Physiol (Lond) 1963; 166: 1–14 Curtis DR, Watkins JC. Acidic amino acids with strong excitatory actions on mammalian neurons. J Physiol (Lond) 1963; 166: 1–14
104.
go back to reference Krnjevic K, Phillis JW. Iontophoretic studies of neurones in the mammalian cerebral cortex. J Physiol (Lond) 1963; 165: 274–305 Krnjevic K, Phillis JW. Iontophoretic studies of neurones in the mammalian cerebral cortex. J Physiol (Lond) 1963; 165: 274–305
105.
go back to reference McLennan H, Huffman RD, Marshall KC. Patterns of excitation of thalamic neurones by amino-acids and by acetylcholine. Nature 1968; 219: 387–8PubMedCrossRef McLennan H, Huffman RD, Marshall KC. Patterns of excitation of thalamic neurones by amino-acids and by acetylcholine. Nature 1968; 219: 387–8PubMedCrossRef
106.
go back to reference Nicoletti F, Bruno V, Copani A, et al. Metabotropic glutamate receptors: a new target for the therapy of neurodegenerative disorders? Trends Neurosci 1996; 19: 267–71PubMedCrossRef Nicoletti F, Bruno V, Copani A, et al. Metabotropic glutamate receptors: a new target for the therapy of neurodegenerative disorders? Trends Neurosci 1996; 19: 267–71PubMedCrossRef
107.
go back to reference Hopkin J, Neal MJ. Effect of electrical stimulation and high potassium concentrations on the efflux of 14C-glycine from slices of spinal cord. Br J Pharmacol 1971; 42: 215–23PubMedCrossRef Hopkin J, Neal MJ. Effect of electrical stimulation and high potassium concentrations on the efflux of 14C-glycine from slices of spinal cord. Br J Pharmacol 1971; 42: 215–23PubMedCrossRef
108.
go back to reference Ueda H, Miyamae T, Hayashi C, et al. Protein kinase C receptor coupled to Gil-phospholipase C activation in Xenopus oocytes. J Neurosci 1995; 15: 7485–99PubMed Ueda H, Miyamae T, Hayashi C, et al. Protein kinase C receptor coupled to Gil-phospholipase C activation in Xenopus oocytes. J Neurosci 1995; 15: 7485–99PubMed
109.
go back to reference Okano K, Ueda M, Kuraishi Y, et al. Effect of repeated cold stress on capsaicin-evoked release of glutamate from rat spinal dorsal horn slices. Neurosci Res 1997; 29: 319–24PubMedCrossRef Okano K, Ueda M, Kuraishi Y, et al. Effect of repeated cold stress on capsaicin-evoked release of glutamate from rat spinal dorsal horn slices. Neurosci Res 1997; 29: 319–24PubMedCrossRef
110.
go back to reference Buerkle H, Marsala M, Yaksh TL. Effect of continuous spinal remifentanil infusion on behavior and spinal glutamate release evoked by subcutaneous formalin in the rat. Br J Anaesth 1998; 80: 348–53PubMedCrossRef Buerkle H, Marsala M, Yaksh TL. Effect of continuous spinal remifentanil infusion on behavior and spinal glutamate release evoked by subcutaneous formalin in the rat. Br J Anaesth 1998; 80: 348–53PubMedCrossRef
111.
go back to reference Kawamata M, Omote K. Involvement of increased excitatory amino acids and intracellular Ca2+concentration in the spinal dorsal horn in an animal model of neuropathic pain. Pain 1996; 68: 85–96PubMedCrossRef Kawamata M, Omote K. Involvement of increased excitatory amino acids and intracellular Ca2+concentration in the spinal dorsal horn in an animal model of neuropathic pain. Pain 1996; 68: 85–96PubMedCrossRef
112.
go back to reference Klamt JG. Effects of intrathecally administered lamotrigine, a glutamate release inhibitor, on short- and long-term models of hyperalgesia in rats. Anesthesiology 1998; 88: 487–94PubMedCrossRef Klamt JG. Effects of intrathecally administered lamotrigine, a glutamate release inhibitor, on short- and long-term models of hyperalgesia in rats. Anesthesiology 1998; 88: 487–94PubMedCrossRef
113.
go back to reference Kangra I, Randic M. Outflow of endogenous aspartate and glutamate from the rat spinal dorsal horn in vitro by activation of low- and high-threshold primary afferent fibers. Modulation by μ-opioids. Brain Res 1991; 553: 347–52 Kangra I, Randic M. Outflow of endogenous aspartate and glutamate from the rat spinal dorsal horn in vitro by activation of low- and high-threshold primary afferent fibers. Modulation by μ-opioids. Brain Res 1991; 553: 347–52
114.
go back to reference Lawand NB, Willis WD, Westlund KN. Excitatory amino acid receptor involvement in peripheral nociceptive transmission in rats. Eur J Pharmacol 1997; 324: 169–77PubMedCrossRef Lawand NB, Willis WD, Westlund KN. Excitatory amino acid receptor involvement in peripheral nociceptive transmission in rats. Eur J Pharmacol 1997; 324: 169–77PubMedCrossRef
115.
go back to reference Jensen TS, Yaksh TL. Spinal monoamine and opiate systems partly mediate the antinociceptive effects produced by glutamate at brainstem sties. Brain Res 1984; 321: 287–97PubMedCrossRef Jensen TS, Yaksh TL. Spinal monoamine and opiate systems partly mediate the antinociceptive effects produced by glutamate at brainstem sties. Brain Res 1984; 321: 287–97PubMedCrossRef
116.
go back to reference Satoh M, Oku R, Akaiko A. Analgesia produced by microinjection of L-glutamate into the rostral ventromedial bulbar nuclei of the rat and its inhibition by intrathecal alpha-adrenergic blocking agents. Brain Res 1983; 261: 361–4PubMedCrossRef Satoh M, Oku R, Akaiko A. Analgesia produced by microinjection of L-glutamate into the rostral ventromedial bulbar nuclei of the rat and its inhibition by intrathecal alpha-adrenergic blocking agents. Brain Res 1983; 261: 361–4PubMedCrossRef
117.
go back to reference Ault B, Hildebrand LM. L-glutamate activates peripheral nociceptors. Agents Actions 1993; 39(Suppl. C): C142–4PubMedCrossRef Ault B, Hildebrand LM. L-glutamate activates peripheral nociceptors. Agents Actions 1993; 39(Suppl. C): C142–4PubMedCrossRef
118.
go back to reference Sher GD, Mitchell D. Intrathecal N-methyl-D-aspartate induces hyperexcitability in rat dorsal horn convergent neurons. Neurosci Lett 1990; 119: 199–202PubMedCrossRef Sher GD, Mitchell D. Intrathecal N-methyl-D-aspartate induces hyperexcitability in rat dorsal horn convergent neurons. Neurosci Lett 1990; 119: 199–202PubMedCrossRef
119.
go back to reference Aanonsen LM, Lei S, Wilcox GL. Excitatory amino acid receptors and nociceptive neurotransmission in rat spinal cord. Pain 1990; 41: 309–21PubMedCrossRef Aanonsen LM, Lei S, Wilcox GL. Excitatory amino acid receptors and nociceptive neurotransmission in rat spinal cord. Pain 1990; 41: 309–21PubMedCrossRef
120.
go back to reference Dougherty PM, Willis WD. Enhancement of spinothalamic neuron responses to chemical and mechanical stimuli following combined microiontophoretic application of N-methyl-D-aspartic acid and substance P. Pain 1991; 47: 85–93PubMedCrossRef Dougherty PM, Willis WD. Enhancement of spinothalamic neuron responses to chemical and mechanical stimuli following combined microiontophoretic application of N-methyl-D-aspartic acid and substance P. Pain 1991; 47: 85–93PubMedCrossRef
121.
go back to reference Cumberbatch MJ, Herrero JF, Headley PM. Exposure of rat spinal neurones to NMDA, AMPA and kainate produces only short-term enhancements of responses to noxious and non-noxious stimuli. Neurosci Lett 1994; 181: 98–102PubMedCrossRef Cumberbatch MJ, Herrero JF, Headley PM. Exposure of rat spinal neurones to NMDA, AMPA and kainate produces only short-term enhancements of responses to noxious and non-noxious stimuli. Neurosci Lett 1994; 181: 98–102PubMedCrossRef
122.
go back to reference Neugebauer V, Lucke T, Schaible HG. Differential effects of N-methyl-D-aspartate (NMDA) and non-NMDA receptor antagonists on the responses of rat spinal neurons with joint input. Neurosci Lett 1993; 155: 29–32PubMedCrossRef Neugebauer V, Lucke T, Schaible HG. Differential effects of N-methyl-D-aspartate (NMDA) and non-NMDA receptor antagonists on the responses of rat spinal neurons with joint input. Neurosci Lett 1993; 155: 29–32PubMedCrossRef
123.
go back to reference Sher GD, Mitchell D. N-methyl-D-aspartate receptors mediate responses of rat dorsal horn neurones to hindlimb ischemia. Brain Res 1990; 522: 55–62PubMedCrossRef Sher GD, Mitchell D. N-methyl-D-aspartate receptors mediate responses of rat dorsal horn neurones to hindlimb ischemia. Brain Res 1990; 522: 55–62PubMedCrossRef
124.
go back to reference Ault B, Hildebrand LM. Effects of excitatory amino acid receptor antagonists on a capsaicin-evoked nociceptive reflex: a comparison with morphine, clonidine and baclofen. Pain 1993; 52: 341–9PubMedCrossRef Ault B, Hildebrand LM. Effects of excitatory amino acid receptor antagonists on a capsaicin-evoked nociceptive reflex: a comparison with morphine, clonidine and baclofen. Pain 1993; 52: 341–9PubMedCrossRef
125.
go back to reference Thompson SW, Dray A, Urban L. Injury-induced plasticity of spinal reflex activity: NK1 neurokinin receptor activation and enhanced A- and C-fiber mediated responses in the rat spinal cord in vitro. J Neurosci 1994; 14: 3672–87PubMed Thompson SW, Dray A, Urban L. Injury-induced plasticity of spinal reflex activity: NK1 neurokinin receptor activation and enhanced A- and C-fiber mediated responses in the rat spinal cord in vitro. J Neurosci 1994; 14: 3672–87PubMed
126.
go back to reference Nagy I, Maggi CA, Dray A, et al. The role of neurokinin and N-methyl-D-aspartate receptors in synaptic transmission from capsaicin-sensitive primary afferents in the rat spinal cord in vitro. Neuroscience 1993; 52: 1029–37PubMedCrossRef Nagy I, Maggi CA, Dray A, et al. The role of neurokinin and N-methyl-D-aspartate receptors in synaptic transmission from capsaicin-sensitive primary afferents in the rat spinal cord in vitro. Neuroscience 1993; 52: 1029–37PubMedCrossRef
127.
go back to reference Urban L, Dray A. Synaptic activation of dorsal horn neurons by selective C fibre excitation with capsaicin in the mouse spinal cord in vitro. Neuroscience 1992; 47: 693–702PubMedCrossRef Urban L, Dray A. Synaptic activation of dorsal horn neurons by selective C fibre excitation with capsaicin in the mouse spinal cord in vitro. Neuroscience 1992; 47: 693–702PubMedCrossRef
128.
go back to reference Dickenson AH, Aydar E. Antagonism at the glycine site on the NMDA receptor reduces spinal nociception in the rat. Neurosci Lett 1991; 121: 263–6PubMedCrossRef Dickenson AH, Aydar E. Antagonism at the glycine site on the NMDA receptor reduces spinal nociception in the rat. Neurosci Lett 1991; 121: 263–6PubMedCrossRef
129.
go back to reference Dickenson AH, Chapman V, Green GM. The pharmacology of excitatory and inhibitory amino acid-mediated events in the transmission and modulation of pain in the spinal cord. Gen Pharmacol 1997; 28: 633–8PubMedCrossRef Dickenson AH, Chapman V, Green GM. The pharmacology of excitatory and inhibitory amino acid-mediated events in the transmission and modulation of pain in the spinal cord. Gen Pharmacol 1997; 28: 633–8PubMedCrossRef
130.
go back to reference Xu XJ, Zhang X, Hökfelt T, et al. Plasticity in spinal nociception after peripheral nerve section: reduced effectiveness of the NMDA receptor antagonist MK-801 in blocking wind-up and central sensitization of the flexor reflex. Brain Res 1995; 670: 342–6PubMedCrossRef Xu XJ, Zhang X, Hökfelt T, et al. Plasticity in spinal nociception after peripheral nerve section: reduced effectiveness of the NMDA receptor antagonist MK-801 in blocking wind-up and central sensitization of the flexor reflex. Brain Res 1995; 670: 342–6PubMedCrossRef
131.
go back to reference Haley JE, Sullivan AF, Dickenson AH. Evidence for spinal N-methyl-D-aspartate involvement in prolonged chemical nociception in the rat. Brain Res 1990; 518: 218–26PubMedCrossRef Haley JE, Sullivan AF, Dickenson AH. Evidence for spinal N-methyl-D-aspartate involvement in prolonged chemical nociception in the rat. Brain Res 1990; 518: 218–26PubMedCrossRef
132.
go back to reference Schaible H-G, Grubb BD, Neugebauer V, et al. The effects of NMDA antagonists on neuronal activity in cat spinal cord evoked by acute inflammation in the knee joint. Eur J Neurosci 1991; 3: 981–91PubMedCrossRef Schaible H-G, Grubb BD, Neugebauer V, et al. The effects of NMDA antagonists on neuronal activity in cat spinal cord evoked by acute inflammation in the knee joint. Eur J Neurosci 1991; 3: 981–91PubMedCrossRef
133.
go back to reference Dougherty PM, Palecek J, Paleckova V, et al. The role of NMDA and non-NMDA excitatory amino acid receptors in the excitation of primate spinothalamic tract neurons by mechanical, thermal and electrical stimuli. J Neurosci 1992; 12: 3025–41PubMed Dougherty PM, Palecek J, Paleckova V, et al. The role of NMDA and non-NMDA excitatory amino acid receptors in the excitation of primate spinothalamic tract neurons by mechanical, thermal and electrical stimuli. J Neurosci 1992; 12: 3025–41PubMed
134.
go back to reference Dougherty PM, Sluka KA, Sorkin LS, et al. Neural changes in acute arthritis in monkeys. I. Parallel enhancement of responses of spinothalamic tract neurons to mechanical stimulation and excitatory amino acids. Brain Res Brain Res Rev 1992; 17: 1–13 Dougherty PM, Sluka KA, Sorkin LS, et al. Neural changes in acute arthritis in monkeys. I. Parallel enhancement of responses of spinothalamic tract neurons to mechanical stimulation and excitatory amino acids. Brain Res Brain Res Rev 1992; 17: 1–13
135.
go back to reference Neugebauer V, Kornhuber J, Lucke T, et al. The clinically available NMDA receptor antagonist memantine is antinociceptive on rat spinal neurones. Neuroreport 1993; 4: 1259–62PubMedCrossRef Neugebauer V, Kornhuber J, Lucke T, et al. The clinically available NMDA receptor antagonist memantine is antinociceptive on rat spinal neurones. Neuroreport 1993; 4: 1259–62PubMedCrossRef
136.
go back to reference Neugebauer V, Lucke T, Grubb B, et al. The involvement of N-methyl-D-aspartate (NMDA) and non-NMDA receptors in the responsiveness of rat spinal neurons with input from the chronically inflamed ankle. Neurosci Lett 1994; 170: 237–40PubMedCrossRef Neugebauer V, Lucke T, Grubb B, et al. The involvement of N-methyl-D-aspartate (NMDA) and non-NMDA receptors in the responsiveness of rat spinal neurons with input from the chronically inflamed ankle. Neurosci Lett 1994; 170: 237–40PubMedCrossRef
137.
go back to reference Chapman V, Dickenson AH. Time related roles of excitatory amino acid receptors during persistent noxiously evoked responses of rat dorsal horn neurons. Brain Res 1995; 703: 45–50PubMedCrossRef Chapman V, Dickenson AH. Time related roles of excitatory amino acid receptors during persistent noxiously evoked responses of rat dorsal horn neurons. Brain Res 1995; 703: 45–50PubMedCrossRef
138.
go back to reference Dougherty PM, Willis WD. Enhanced responses of spinothalamic tract neurons to excitatory amino acids accompany capsaicin-induced sensitization in the monkey. J Neurosci 1992; 12: 883–94PubMed Dougherty PM, Willis WD. Enhanced responses of spinothalamic tract neurons to excitatory amino acids accompany capsaicin-induced sensitization in the monkey. J Neurosci 1992; 12: 883–94PubMed
139.
go back to reference Radhakrishnan V, Henry JL. Excitatory amino acid receptor mediation of sensory inputs to functionally identified dorsal horn neurons in cat spinal cord. Neuroscience 1993; 55: 531–44PubMedCrossRef Radhakrishnan V, Henry JL. Excitatory amino acid receptor mediation of sensory inputs to functionally identified dorsal horn neurons in cat spinal cord. Neuroscience 1993; 55: 531–44PubMedCrossRef
140.
go back to reference Song XJ, Zhao ZQ. Cooperative interaction among the various regulatory sites within the NMDA receptor-channel complex in modulating the evoked response to noxious thermal stimuli of spinal dorsal horn neurons in the cat. Exp Brain Res 1998; 120: 257–62PubMedCrossRef Song XJ, Zhao ZQ. Cooperative interaction among the various regulatory sites within the NMDA receptor-channel complex in modulating the evoked response to noxious thermal stimuli of spinal dorsal horn neurons in the cat. Exp Brain Res 1998; 120: 257–62PubMedCrossRef
141.
go back to reference Eaton SA, Salt TE. Thalamic NMDA receptors and nociceptive sensory synaptic transmission. Neurosci Lett 1990; 110: 297–302PubMedCrossRef Eaton SA, Salt TE. Thalamic NMDA receptors and nociceptive sensory synaptic transmission. Neurosci Lett 1990; 110: 297–302PubMedCrossRef
142.
go back to reference Chiang CY, Park SJ, Kwan CL, et al. NMDA receptor mechanisms contribute to neuroplasticity induced in caudalis nociceptive neurons by tooth pulp stimulation. J Neurophysiol 1998; 80: 2621–31PubMed Chiang CY, Park SJ, Kwan CL, et al. NMDA receptor mechanisms contribute to neuroplasticity induced in caudalis nociceptive neurons by tooth pulp stimulation. J Neurophysiol 1998; 80: 2621–31PubMed
143.
go back to reference Carlton SM, Rees H, Gondesen K, et al. Dextrophan attenuates responses of spinothalamic tract cells in normal and nerve-injured monkeys. Neurosci Lett 1997; 229: 169–72PubMedCrossRef Carlton SM, Rees H, Gondesen K, et al. Dextrophan attenuates responses of spinothalamic tract cells in normal and nerve-injured monkeys. Neurosci Lett 1997; 229: 169–72PubMedCrossRef
144.
go back to reference Aanonsen LM, Wilcox GL. Nociceptive action of excitatory amino acids in the mouse: effects of spinally administered opioids, phencyclidine and σ agonists. J Pharmacol Exp Ther 1987; 243: 9–19PubMed Aanonsen LM, Wilcox GL. Nociceptive action of excitatory amino acids in the mouse: effects of spinally administered opioids, phencyclidine and σ agonists. J Pharmacol Exp Ther 1987; 243: 9–19PubMed
145.
go back to reference Aanonsen LM, Wilcox GL. Muscimol, γ-aminobutyric acidA receptors and excitatory amino acids in the mouse spinal cord. J Pharmacol Exp Ther 1989; 248: 1034–8PubMed Aanonsen LM, Wilcox GL. Muscimol, γ-aminobutyric acidA receptors and excitatory amino acids in the mouse spinal cord. J Pharmacol Exp Ther 1989; 248: 1034–8PubMed
146.
go back to reference Kolhekar R, Meller ST, Gebhart GF. Characterization of the role of spinal N-methyl-D-aspartate receptors in thermal nociception in the rat. Neuroscience 1993; 57: 385–95PubMedCrossRef Kolhekar R, Meller ST, Gebhart GF. Characterization of the role of spinal N-methyl-D-aspartate receptors in thermal nociception in the rat. Neuroscience 1993; 57: 385–95PubMedCrossRef
147.
go back to reference Mjellem-Joly N, Lund A, Berge O-G, et al. Potentiation of a behavioural response in mice by spinal coadministration of substance and excitatory amino acid agonists. Neurosci Lett 1991; 133: 121–4PubMedCrossRef Mjellem-Joly N, Lund A, Berge O-G, et al. Potentiation of a behavioural response in mice by spinal coadministration of substance and excitatory amino acid agonists. Neurosci Lett 1991; 133: 121–4PubMedCrossRef
148.
go back to reference Okano K, Kuraishi Y, Satoh M. Effects of repeated cold stress on aversive responses produced by intrathecal excitatory amino acids in rats. Biol Pharm Bull 1995; 18: 1602–4PubMedCrossRef Okano K, Kuraishi Y, Satoh M. Effects of repeated cold stress on aversive responses produced by intrathecal excitatory amino acids in rats. Biol Pharm Bull 1995; 18: 1602–4PubMedCrossRef
149.
go back to reference Okano K, Kuraishi Y, Satoh M. Pharmacological evidence for involvement of excitatory amino acids in aversive responses induced by intrathecal substance P in rats. Biol Pharm Bull 1993; 16: 861–5PubMedCrossRef Okano K, Kuraishi Y, Satoh M. Pharmacological evidence for involvement of excitatory amino acids in aversive responses induced by intrathecal substance P in rats. Biol Pharm Bull 1993; 16: 861–5PubMedCrossRef
150.
go back to reference Raigorodsky G, Urca G. Intrathecal N-methyl-D-aspartate (NMDA) activates both nociceptive and antinociceptive systems. Brain Res 1987; 422: 158–62PubMedCrossRef Raigorodsky G, Urca G. Intrathecal N-methyl-D-aspartate (NMDA) activates both nociceptive and antinociceptive systems. Brain Res 1987; 422: 158–62PubMedCrossRef
151.
go back to reference Raigorodsky G, Urca G. Spinal antinociceptive effects of excitatory amino acid antagonists: quisqualate modulates the action of N-methyl-D-aspartate. Eur J Pharmacol 1990; 182: 37–47PubMedCrossRef Raigorodsky G, Urca G. Spinal antinociceptive effects of excitatory amino acid antagonists: quisqualate modulates the action of N-methyl-D-aspartate. Eur J Pharmacol 1990; 182: 37–47PubMedCrossRef
152.
go back to reference Sun X, Larson AA. Behavioral sensitization to kainic acid and quisqualic acid in mice: comparison to NMDA and substance P responses. J Neurosci 1991; 11: 3111–23PubMed Sun X, Larson AA. Behavioral sensitization to kainic acid and quisqualic acid in mice: comparison to NMDA and substance P responses. J Neurosci 1991; 11: 3111–23PubMed
153.
go back to reference Advokat C, Ghorpade A, Wolfe E. Intrathecal excitatory amino acid (EAA) agonists increase tail flick latencies (TFLs) of spinal rats. Pharmacol Biochem Behav 1994; 48: 693–8PubMedCrossRef Advokat C, Ghorpade A, Wolfe E. Intrathecal excitatory amino acid (EAA) agonists increase tail flick latencies (TFLs) of spinal rats. Pharmacol Biochem Behav 1994; 48: 693–8PubMedCrossRef
154.
go back to reference Kolhekar R, Meller ST, Gebhart GF. N-methyl-D-aspartate receptor-mediated changes in thermal nociception: allosteric modulation at glycine and polyamine recognition sites. Neuroscience 1994; 63: 925–36PubMedCrossRef Kolhekar R, Meller ST, Gebhart GF. N-methyl-D-aspartate receptor-mediated changes in thermal nociception: allosteric modulation at glycine and polyamine recognition sites. Neuroscience 1994; 63: 925–36PubMedCrossRef
155.
go back to reference Malmberg AB, Yaksh TL. Spinal nitric oxide synthesis inhibition blocks NMDA-induced thermal hyperalgesia and produces antinociception in the formalin test in rats. Pain 1993; 54: 291–300PubMedCrossRef Malmberg AB, Yaksh TL. Spinal nitric oxide synthesis inhibition blocks NMDA-induced thermal hyperalgesia and produces antinociception in the formalin test in rats. Pain 1993; 54: 291–300PubMedCrossRef
156.
go back to reference Meller ST, Dykstra CL, Gebhart GF. Acute thermal hyperalgesia in the rat is produced by activation of N-methyl-D-aspartate receptors and protein kinase C and production of nitric oxide. Neuroscience 1996; 71: 327–35PubMedCrossRef Meller ST, Dykstra CL, Gebhart GF. Acute thermal hyperalgesia in the rat is produced by activation of N-methyl-D-aspartate receptors and protein kinase C and production of nitric oxide. Neuroscience 1996; 71: 327–35PubMedCrossRef
157.
go back to reference Ferreira SH, Lorenzetti BB. Glutamate spinal retrograde sensitization of primary sensory neurons associated with nociception. Neuropharmacology 1994; 11: 1479–85CrossRef Ferreira SH, Lorenzetti BB. Glutamate spinal retrograde sensitization of primary sensory neurons associated with nociception. Neuropharmacology 1994; 11: 1479–85CrossRef
158.
go back to reference Björkman R, Hallman KM, Hedner J, et al. Acetaminophen blocks spinal hyperalgesia induced by NMDA and substance P. Pain 1994; 57: 259–64PubMedCrossRef Björkman R, Hallman KM, Hedner J, et al. Acetaminophen blocks spinal hyperalgesia induced by NMDA and substance P. Pain 1994; 57: 259–64PubMedCrossRef
159.
go back to reference Zochodne DW, Murray M, Nag S, et al. A segmental chronic pain syndrome in rats associated with intrathecal infusion of NMDA: evidence for selective action in the dorsal horn. Can J Neurol Sci 1994; 21: 24–8PubMed Zochodne DW, Murray M, Nag S, et al. A segmental chronic pain syndrome in rats associated with intrathecal infusion of NMDA: evidence for selective action in the dorsal horn. Can J Neurol Sci 1994; 21: 24–8PubMed
160.
go back to reference Zhou S, Bonasera L, Carlton SM. Peripheral administration of NMDA, AMPA or KA results in pain behavior in rats. Neuroreport 1996; 7: 895–900PubMedCrossRef Zhou S, Bonasera L, Carlton SM. Peripheral administration of NMDA, AMPA or KA results in pain behavior in rats. Neuroreport 1996; 7: 895–900PubMedCrossRef
161.
go back to reference Jackson DL, Graff CB, Richardson JD, et al. Glutamate participates in the peripheral modulation of thermal hyperalgesia in rats. Eur J Pharmacol 1995; 284: 321–5PubMedCrossRef Jackson DL, Graff CB, Richardson JD, et al. Glutamate participates in the peripheral modulation of thermal hyperalgesia in rats. Eur J Pharmacol 1995; 284: 321–5PubMedCrossRef
162.
go back to reference Woolf DJ, Thompson SWN. The induction and maintenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation: implications for post-injury pain hyper-sensitivity states. Pain 1991; 44: 293–9PubMedCrossRef Woolf DJ, Thompson SWN. The induction and maintenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation: implications for post-injury pain hyper-sensitivity states. Pain 1991; 44: 293–9PubMedCrossRef
163.
go back to reference Ma QP, Woolf CJ. Noxious stimuli induce an N-methyl-D-aspartate receptor-dependent hypersensitivity of the flexion withdrawal reflex to touch-implications for the treatment of mechanical allodynia. Pain 1995; 61: 383–90PubMedCrossRef Ma QP, Woolf CJ. Noxious stimuli induce an N-methyl-D-aspartate receptor-dependent hypersensitivity of the flexion withdrawal reflex to touch-implications for the treatment of mechanical allodynia. Pain 1995; 61: 383–90PubMedCrossRef
164.
go back to reference Silva E, Cleland CL, Gebhart GF. Contribution of glutamate receptors to the maintenance of mustard oil-induced hyperalgesia in spinalized rats. Exp Brain Res 1997; 117: 379–88PubMedCrossRef Silva E, Cleland CL, Gebhart GF. Contribution of glutamate receptors to the maintenance of mustard oil-induced hyperalgesia in spinalized rats. Exp Brain Res 1997; 117: 379–88PubMedCrossRef
165.
go back to reference Kolhekar R, Gebhart GF. NMDA and quisqualate modulation of visceral nociception in the rat. Brain Res 1994; 651: 215–26PubMedCrossRef Kolhekar R, Gebhart GF. NMDA and quisqualate modulation of visceral nociception in the rat. Brain Res 1994; 651: 215–26PubMedCrossRef
166.
go back to reference Rice ASC, McMahon SB. Pre-emptive intrathecal administration of an NMDA receptor antagonist (AP-5) prevents hyper-reflexia in a model of persistent visceral pain. Pain 1994; 335–40 Rice ASC, McMahon SB. Pre-emptive intrathecal administration of an NMDA receptor antagonist (AP-5) prevents hyper-reflexia in a model of persistent visceral pain. Pain 1994; 335–40
167.
go back to reference Coutinho SV, Urban MO, Gebhart GF. Role of glutamate receptors and nitric oxide in the rostral ventromedial medulla in visceral hyperalgesia. Pain 1998; 78: 59–69PubMedCrossRef Coutinho SV, Urban MO, Gebhart GF. Role of glutamate receptors and nitric oxide in the rostral ventromedial medulla in visceral hyperalgesia. Pain 1998; 78: 59–69PubMedCrossRef
168.
go back to reference Ide Y, Maehara Y, Tsukara S, et al. The effects of an intrathecal NMDA antagonist (AP5) on the behavioral changes induced by colorectal inflammation with turpentine in rats. Life Sci 1997; 60: 1359–63PubMedCrossRef Ide Y, Maehara Y, Tsukara S, et al. The effects of an intrathecal NMDA antagonist (AP5) on the behavioral changes induced by colorectal inflammation with turpentine in rats. Life Sci 1997; 60: 1359–63PubMedCrossRef
169.
go back to reference Olivar TO, Laird JMA. Differential effects of N-methyl-D-aspartate receptor blockade on nociceptive somatic and visceral reflexes. Pain 1999; 79: 67–73PubMedCrossRef Olivar TO, Laird JMA. Differential effects of N-methyl-D-aspartate receptor blockade on nociceptive somatic and visceral reflexes. Pain 1999; 79: 67–73PubMedCrossRef
170.
go back to reference Cahusac PMB, Evans RH, Hill RG, et al. The behavioral effects of an N-methylaspartate receptor antagonist following application to the lumbar spinal cord of conscious rats. Neuropharmacology 1984; 23: 719–24PubMedCrossRef Cahusac PMB, Evans RH, Hill RG, et al. The behavioral effects of an N-methylaspartate receptor antagonist following application to the lumbar spinal cord of conscious rats. Neuropharmacology 1984; 23: 719–24PubMedCrossRef
171.
go back to reference Forman LJ. NMDA receptor antagonism produces antinociception which is partially mediated by brain opioids and dopamine. Life Sci 1999; 64: 1877–87PubMedCrossRef Forman LJ. NMDA receptor antagonism produces antinociception which is partially mediated by brain opioids and dopamine. Life Sci 1999; 64: 1877–87PubMedCrossRef
172.
go back to reference Lutfy K, Weber E. Attenuation of nociceptive responses by ACEA-1021, a competitive NMDA receptor/glycine site antagonist, in the mice. Brain Res 1996; 743: 17–23PubMedCrossRef Lutfy K, Weber E. Attenuation of nociceptive responses by ACEA-1021, a competitive NMDA receptor/glycine site antagonist, in the mice. Brain Res 1996; 743: 17–23PubMedCrossRef
173.
go back to reference Bernardi M, Bertolini A, Szczawinska K, et al. Blockade of the polyamine site of NMDA receptors produces antinociception and enhances the effect of morphine, in mice. Eur J Pharmacol 1996; 298: 51–5PubMedCrossRef Bernardi M, Bertolini A, Szczawinska K, et al. Blockade of the polyamine site of NMDA receptors produces antinociception and enhances the effect of morphine, in mice. Eur J Pharmacol 1996; 298: 51–5PubMedCrossRef
174.
go back to reference Coderre TJ, Van Empel I. The utility of excitatory amino acid (EAA) antagonists as analgesic agents I. Comparison of the antinociceptive activity of various classes of EAA antagonists in mechanical, thermal and chemical nociceptive tests. Pain 1994; 59: 345–52 Coderre TJ, Van Empel I. The utility of excitatory amino acid (EAA) antagonists as analgesic agents I. Comparison of the antinociceptive activity of various classes of EAA antagonists in mechanical, thermal and chemical nociceptive tests. Pain 1994; 59: 345–52
175.
go back to reference Laughlin TM, Kitto KF, Wilcox GL. Redox manipulation of NMDA receptors hi vivo: alteration of acute pain and dynorphin-induced allodynia. Pain 1999; 80: 37–43PubMedCrossRef Laughlin TM, Kitto KF, Wilcox GL. Redox manipulation of NMDA receptors hi vivo: alteration of acute pain and dynorphin-induced allodynia. Pain 1999; 80: 37–43PubMedCrossRef
176.
go back to reference Jensen TS, Yaksh TL. The antinociceptive activity of excitatory amino acids in the rat brainstem: anatomical and pharmacological analysis. Brain Res 1992; 569: 255–67PubMedCrossRef Jensen TS, Yaksh TL. The antinociceptive activity of excitatory amino acids in the rat brainstem: anatomical and pharmacological analysis. Brain Res 1992; 569: 255–67PubMedCrossRef
177.
go back to reference Jacquet YF. The NMDA receptor: central role in pain inhibition in rat periaqueductal gray. Eur J Pharmacol 1988; 154: 271–6PubMedCrossRef Jacquet YF. The NMDA receptor: central role in pain inhibition in rat periaqueductal gray. Eur J Pharmacol 1988; 154: 271–6PubMedCrossRef
178.
go back to reference Coderre TJ. The role of excitatory amino acid receptors and intracellular messengers in persistent nociception after tissue injury in rats. Mol Neurobiol 1993; 7: 229–46PubMedCrossRef Coderre TJ. The role of excitatory amino acid receptors and intracellular messengers in persistent nociception after tissue injury in rats. Mol Neurobiol 1993; 7: 229–46PubMedCrossRef
179.
go back to reference Coderre TJ, Melzack R. Central neural mediators of secondary hyperalgesia following heat injury in rats: neuropeptides and excitatory amino acids. Neurosci Lett 1991; 131: 71–4PubMedCrossRef Coderre TJ, Melzack R. Central neural mediators of secondary hyperalgesia following heat injury in rats: neuropeptides and excitatory amino acids. Neurosci Lett 1991; 131: 71–4PubMedCrossRef
180.
go back to reference Coderre TJ, Melzack R. The contribution of excitatory amino acids to central sensitization and persistent nociception after formalin-induced tissue injury. J Neurosci 1992; 12: 3665–70PubMed Coderre TJ, Melzack R. The contribution of excitatory amino acids to central sensitization and persistent nociception after formalin-induced tissue injury. J Neurosci 1992; 12: 3665–70PubMed
181.
go back to reference Yamamoto T, Yaksh TL. Comparison of the antinociceptive effects of pre- and post-treatment with intrathecal morphine and MK-801, an NMDA antagonist, on the formalin test in the rat. Anesthesiology 1992; 77: 757–63PubMedCrossRef Yamamoto T, Yaksh TL. Comparison of the antinociceptive effects of pre- and post-treatment with intrathecal morphine and MK-801, an NMDA antagonist, on the formalin test in the rat. Anesthesiology 1992; 77: 757–63PubMedCrossRef
182.
go back to reference Elliot KJ, Brodsky M, Hynansky AD, et al. Dextromethorphan suppresses both formalin-induced nociceptive behavior and the formalin-induced increase in spinal cord c-fos mRNA. Pain 1995; 61: 401–9CrossRef Elliot KJ, Brodsky M, Hynansky AD, et al. Dextromethorphan suppresses both formalin-induced nociceptive behavior and the formalin-induced increase in spinal cord c-fos mRNA. Pain 1995; 61: 401–9CrossRef
183.
go back to reference Millan MJ, Seguin L. Chemically-diverse ligands at the glycine B site coupled to N-methyl-D-aspartate (NMDA) receptors selectively block the late phase of formalin-induced pain in mice. Neurosci Lett 1994; 178: 139–43PubMedCrossRef Millan MJ, Seguin L. Chemically-diverse ligands at the glycine B site coupled to N-methyl-D-aspartate (NMDA) receptors selectively block the late phase of formalin-induced pain in mice. Neurosci Lett 1994; 178: 139–43PubMedCrossRef
184.
go back to reference Murray CW, Cowan A, Larson AA. Neurokinin and NMDA antagonists (but not a kainic acid antagonist) are antinociceptive in the mouse formalin model. Pain 1991; 44: 179–85PubMedCrossRef Murray CW, Cowan A, Larson AA. Neurokinin and NMDA antagonists (but not a kainic acid antagonist) are antinociceptive in the mouse formalin model. Pain 1991; 44: 179–85PubMedCrossRef
185.
go back to reference Eisenberg E, Vos BP, Strassman AM. The NMDA antagonist memantine blocks pain behavior in a rat model of formalin-induced facial pain. Pain 1993; 54: 301–7PubMedCrossRef Eisenberg E, Vos BP, Strassman AM. The NMDA antagonist memantine blocks pain behavior in a rat model of formalin-induced facial pain. Pain 1993; 54: 301–7PubMedCrossRef
186.
go back to reference Nässtrom J, Karlsson U, Post C. Antinociceptive actions of different classes of excitatory amino acid receptor antagonists in mice. Eur J Pharmacol 1992; 212: 21–9PubMedCrossRef Nässtrom J, Karlsson U, Post C. Antinociceptive actions of different classes of excitatory amino acid receptor antagonists in mice. Eur J Pharmacol 1992; 212: 21–9PubMedCrossRef
187.
go back to reference Hunter JC, Singh L. Role of excitatory amino acid receptors in the mediation of nociceptive response to formalin in the rat. Neurosci Lett 1994; 174: 217–21PubMedCrossRef Hunter JC, Singh L. Role of excitatory amino acid receptors in the mediation of nociceptive response to formalin in the rat. Neurosci Lett 1994; 174: 217–21PubMedCrossRef
188.
go back to reference Kristensen JD, Karlsten R, Gordh T, et al. The NMDA antagonist 3-(20carboxypiperazin-4-yl)propyl-1-phosphonic acid (CPP) has antinociceptive effect after intrathecal injection in the rat. Pain 1994; 56: 59–67PubMedCrossRef Kristensen JD, Karlsten R, Gordh T, et al. The NMDA antagonist 3-(20carboxypiperazin-4-yl)propyl-1-phosphonic acid (CPP) has antinociceptive effect after intrathecal injection in the rat. Pain 1994; 56: 59–67PubMedCrossRef
189.
go back to reference Davidson EM, Carlton SM. Intraplantar injection of dextrorphan, ketamine or memantine attenuates formalin-induced behaviors. Brain Res 1998; 785: 136–42PubMedCrossRef Davidson EM, Carlton SM. Intraplantar injection of dextrorphan, ketamine or memantine attenuates formalin-induced behaviors. Brain Res 1998; 785: 136–42PubMedCrossRef
190.
go back to reference Davidson EM, Coggeshall RE, Carlton SM. Peripheral NMDA and non-NMDA receptors contribute to the nociceptive behaviors in the rat formalin test. Neuroreport 1997; 8: 941–6PubMedCrossRef Davidson EM, Coggeshall RE, Carlton SM. Peripheral NMDA and non-NMDA receptors contribute to the nociceptive behaviors in the rat formalin test. Neuroreport 1997; 8: 941–6PubMedCrossRef
191.
go back to reference Vaccarino AL, Marek P, Kest B, et al. NMDA receptor antagonists, MK-801 and ACEA-1011, prevent the development of tonic pain following subcutaneous formalin. Brain Res 1993; 615: 331–4PubMedCrossRef Vaccarino AL, Marek P, Kest B, et al. NMDA receptor antagonists, MK-801 and ACEA-1011, prevent the development of tonic pain following subcutaneous formalin. Brain Res 1993; 615: 331–4PubMedCrossRef
192.
go back to reference Shimoyama M, Shimoyama N, Gorman AL, et al. Oral ketamine is antinociceptive in the rat formalin test: role of the metabolite, norketamine. Pain 1999; 81: 85–93PubMedCrossRef Shimoyama M, Shimoyama N, Gorman AL, et al. Oral ketamine is antinociceptive in the rat formalin test: role of the metabolite, norketamine. Pain 1999; 81: 85–93PubMedCrossRef
193.
go back to reference Vaccarino AL, Clemmons HR, Mader Jr GJ, et al. A role of periaqueductal grey NMDA receptors in mediating formalin induced pain in the rat. Neurosci Lett 1997; 236: 117–9PubMedCrossRef Vaccarino AL, Clemmons HR, Mader Jr GJ, et al. A role of periaqueductal grey NMDA receptors in mediating formalin induced pain in the rat. Neurosci Lett 1997; 236: 117–9PubMedCrossRef
194.
go back to reference Chaplan SR, Malmberg AB, Yaksh TL. Efficacy of spinal NMDA receptor antagonism in formalin hyperalgesia and nerve injury evoked allodynia in the rat. J Pharmacol Exp Ther 1997; 280: 829–38PubMed Chaplan SR, Malmberg AB, Yaksh TL. Efficacy of spinal NMDA receptor antagonism in formalin hyperalgesia and nerve injury evoked allodynia in the rat. J Pharmacol Exp Ther 1997; 280: 829–38PubMed
195.
go back to reference Coderre TJ, Van Empel I. The utility of excitatory amino acid (EAA) antagonists as analgesic agents II. Assessment of the antinociceptive activity of combinations of competitive and non-competitive NMDA antagonists with agents acting at allosteric-glycine and polyamine receptor sites. Pain 1994; 59: 353–9 Coderre TJ, Van Empel I. The utility of excitatory amino acid (EAA) antagonists as analgesic agents II. Assessment of the antinociceptive activity of combinations of competitive and non-competitive NMDA antagonists with agents acting at allosteric-glycine and polyamine receptor sites. Pain 1994; 59: 353–9
196.
go back to reference Codere TJ. Potent analgesia induced in rats by combined action at PCP and polyamine recognition sites of the NMDA receptor complex. Eur J Neurosci 1993; 5: 390–3CrossRef Codere TJ. Potent analgesia induced in rats by combined action at PCP and polyamine recognition sites of the NMDA receptor complex. Eur J Neurosci 1993; 5: 390–3CrossRef
197.
go back to reference Wiertelak EP, Furness LE, Horan R, et al. Subcutaneous formalin produces centrifugal hyperalgesia at a non-injected site via the NMDA-nitric oxide cascade. Brain Res 1994; 649: 19–26PubMedCrossRef Wiertelak EP, Furness LE, Horan R, et al. Subcutaneous formalin produces centrifugal hyperalgesia at a non-injected site via the NMDA-nitric oxide cascade. Brain Res 1994; 649: 19–26PubMedCrossRef
198.
go back to reference Ren K, Dubner R. NMDA receptor antagonists attenuate mechanical hyperalgesia in rats with unilateral inflammation of the hindpaw. Neurosci Lett 1993; 163: 22–6PubMedCrossRef Ren K, Dubner R. NMDA receptor antagonists attenuate mechanical hyperalgesia in rats with unilateral inflammation of the hindpaw. Neurosci Lett 1993; 163: 22–6PubMedCrossRef
199.
go back to reference Eisenberg E, LaCross S, Strassman AM. The effects of the clinically tested NMDA receptor antagonist memantine on carrageenan-induced thermal hyperalgesia in rats. Eur J Pharmacol 1994; 255: 123–9PubMedCrossRef Eisenberg E, LaCross S, Strassman AM. The effects of the clinically tested NMDA receptor antagonist memantine on carrageenan-induced thermal hyperalgesia in rats. Eur J Pharmacol 1994; 255: 123–9PubMedCrossRef
200.
go back to reference Ren K, Hylden JLK, Williams GM, et al. The effects of a non-competitive NMDA receptor antagonist, MK-801, on behavioral hyperalgesia and dorsal horn neuronal activity in rats with unilateral inflammation. Pain 1992; 50: 331–44PubMedCrossRef Ren K, Hylden JLK, Williams GM, et al. The effects of a non-competitive NMDA receptor antagonist, MK-801, on behavioral hyperalgesia and dorsal horn neuronal activity in rats with unilateral inflammation. Pain 1992; 50: 331–44PubMedCrossRef
201.
go back to reference Ma QP, Allchorne AJ, Woolf CJ. Morphine, the NMDA receptor antagonist MK801 and the tachykinin NK1 receptor antagonist RP67580 attenuate the development of inflammation-induced progressive tactile hypersensitivity. Pain 1998; 77: 49–57PubMedCrossRef Ma QP, Allchorne AJ, Woolf CJ. Morphine, the NMDA receptor antagonist MK801 and the tachykinin NK1 receptor antagonist RP67580 attenuate the development of inflammation-induced progressive tactile hypersensitivity. Pain 1998; 77: 49–57PubMedCrossRef
202.
go back to reference Price DD, Mao J, Lu J, et al. Effects of the combined oral administration of NSAIDS and dextromethorphan on behavioral symptoms indicative of arthritic pain in rats. Pain 1996; 68: 119–27PubMedCrossRef Price DD, Mao J, Lu J, et al. Effects of the combined oral administration of NSAIDS and dextromethorphan on behavioral symptoms indicative of arthritic pain in rats. Pain 1996; 68: 119–27PubMedCrossRef
203.
go back to reference Meller ST, Cummings CP, Traub RJ, et al. The role of nitric oxide in the development and maintenance of the hyperalgesia produced by intraplantar injection of carrageenan in the rat. Neuroscience 1994; 60: 367–74PubMedCrossRef Meller ST, Cummings CP, Traub RJ, et al. The role of nitric oxide in the development and maintenance of the hyperalgesia produced by intraplantar injection of carrageenan in the rat. Neuroscience 1994; 60: 367–74PubMedCrossRef
204.
go back to reference Kolhekar R, Murphy S, Gebhart GF. Thalamic NMDA receptors modulate inflammation-produced hyperalgesia in the rat. Pain 1997; 71: 31–40PubMedCrossRef Kolhekar R, Murphy S, Gebhart GF. Thalamic NMDA receptors modulate inflammation-produced hyperalgesia in the rat. Pain 1997; 71: 31–40PubMedCrossRef
205.
go back to reference Kolhekar R, Gebhart GF. N-methyl-D-aspartate receptor-mediated changes in thermal nociception: allosteric modulation at glycine and polyamine recognition sites. Neuroscience 1994; 63: 925–36PubMedCrossRef Kolhekar R, Gebhart GF. N-methyl-D-aspartate receptor-mediated changes in thermal nociception: allosteric modulation at glycine and polyamine recognition sites. Neuroscience 1994; 63: 925–36PubMedCrossRef
206.
go back to reference Carlton SM, Coggeshall RE. Inflammation-induced changes in peripheral glutamate receptor populations. Brain Res 1999; 820: 63–70PubMedCrossRef Carlton SM, Coggeshall RE. Inflammation-induced changes in peripheral glutamate receptor populations. Brain Res 1999; 820: 63–70PubMedCrossRef
207.
go back to reference Renno WM. Prolonged noxious stimulation increases periaqueductal gray NMDA mRNA expression: a hybridization study using two different rat models for nociception. Neurobiology (Bp) 1998; 6: 333–57 Renno WM. Prolonged noxious stimulation increases periaqueductal gray NMDA mRNA expression: a hybridization study using two different rat models for nociception. Neurobiology (Bp) 1998; 6: 333–57
208.
go back to reference Wang H, Zhang RX, Wang R, et al. Decreased expression of N-methyl-D-aspartate (NMDA) receptors in rat dorsal root ganglion following complete Freund’s adjuvant-induced inflammation: an immunocytochemical study for NMDA NR1 subunit. Neurosci Lett 1999; 265: 195–8PubMedCrossRef Wang H, Zhang RX, Wang R, et al. Decreased expression of N-methyl-D-aspartate (NMDA) receptors in rat dorsal root ganglion following complete Freund’s adjuvant-induced inflammation: an immunocytochemical study for NMDA NR1 subunit. Neurosci Lett 1999; 265: 195–8PubMedCrossRef
209.
go back to reference Calcutt NA, Chaplan SR. Spinal pharmacology of tactile allodynia in diabetic rats. Br J Pharmacol 1997; 122: 1478–82PubMedCrossRef Calcutt NA, Chaplan SR. Spinal pharmacology of tactile allodynia in diabetic rats. Br J Pharmacol 1997; 122: 1478–82PubMedCrossRef
210.
go back to reference Carlton SM, Hargett GL. Treatment with the NMDA antagonist memantine attenuates nociceptive responses to mechanical stimulation in neuropathic rats. Neurosci Lett 1995; 198: 115–8PubMedCrossRef Carlton SM, Hargett GL. Treatment with the NMDA antagonist memantine attenuates nociceptive responses to mechanical stimulation in neuropathic rats. Neurosci Lett 1995; 198: 115–8PubMedCrossRef
211.
go back to reference Malcangio M, Tomlinson DR. A pharmacological analysis of mechanical hyperalgesia in streptozotocin/diabetic rats. Pain 1998; 76: 151–7PubMedCrossRef Malcangio M, Tomlinson DR. A pharmacological analysis of mechanical hyperalgesia in streptozotocin/diabetic rats. Pain 1998; 76: 151–7PubMedCrossRef
212.
go back to reference Mao J, Price DD, Hayes RL, et al. Intrathecal treatment with dextrorphan or ketamine potently reduces pain-related behaviors in a rat model of peripheral mononeuropathy. Brain Res 1993: 605: 164–8PubMedCrossRef Mao J, Price DD, Hayes RL, et al. Intrathecal treatment with dextrorphan or ketamine potently reduces pain-related behaviors in a rat model of peripheral mononeuropathy. Brain Res 1993: 605: 164–8PubMedCrossRef
213.
go back to reference Mao J, Price DD, Mayer DJ, et al. Intrathecal MK-801 and local nerve anesthesia synergistically reduce nociceptive behaviors in rats with experimental peripheral mononeuropathy. Brain 1992; 576: 254–62CrossRef Mao J, Price DD, Mayer DJ, et al. Intrathecal MK-801 and local nerve anesthesia synergistically reduce nociceptive behaviors in rats with experimental peripheral mononeuropathy. Brain 1992; 576: 254–62CrossRef
214.
go back to reference Mao J, Price DD, Hayes RL, et al. Differential roles of NMDA and non-NMDA receptor activation in induction and maintenance of thermal hyperalgesia in rats with painful peripheral mononeuropathy. Brain Res 1992; 598: 271–8PubMedCrossRef Mao J, Price DD, Hayes RL, et al. Differential roles of NMDA and non-NMDA receptor activation in induction and maintenance of thermal hyperalgesia in rats with painful peripheral mononeuropathy. Brain Res 1992; 598: 271–8PubMedCrossRef
215.
go back to reference Fisher K, Fundytus ME, Cahill CM, et al. Intrathecal administration of the mGluR compound, (S)-4CPG, attenuates hyperalgesia and allodynia associated with sciatic nerve constriction injury in rats. Pain 1998; 77: 59–66PubMedCrossRef Fisher K, Fundytus ME, Cahill CM, et al. Intrathecal administration of the mGluR compound, (S)-4CPG, attenuates hyperalgesia and allodynia associated with sciatic nerve constriction injury in rats. Pain 1998; 77: 59–66PubMedCrossRef
216.
go back to reference Kim YI, Na HS, Yoon YW, et al. NMDA receptors are important for both mechanical and thermal allodynia from peripheral nerve injury in rats. Neuroreport 1997; 8: 2149–53PubMedCrossRef Kim YI, Na HS, Yoon YW, et al. NMDA receptors are important for both mechanical and thermal allodynia from peripheral nerve injury in rats. Neuroreport 1997; 8: 2149–53PubMedCrossRef
217.
go back to reference Yamamoto T, Yaksh TL. Spinal pharmacology of thermal hyperesthesia induced by constriction injury of the sciatic nerve. Excitatory amino acids. Pain 1992; 49: 121–8 Yamamoto T, Yaksh TL. Spinal pharmacology of thermal hyperesthesia induced by constriction injury of the sciatic nerve. Excitatory amino acids. Pain 1992; 49: 121–8
218.
go back to reference Tal M, Bennett GJ. Neuropathic pain sensations are differentially sensitive to dextrorphan. Neuroreport 1994; 5: 1438–40PubMedCrossRef Tal M, Bennett GJ. Neuropathic pain sensations are differentially sensitive to dextrorphan. Neuroreport 1994; 5: 1438–40PubMedCrossRef
219.
go back to reference Tal M, Bennett GJ. Dextrorphan relieves neuropathic heat evoked hyperalgesia in the rat. Neurosci Lett 1993; 151: 107–10PubMedCrossRef Tal M, Bennett GJ. Dextrorphan relieves neuropathic heat evoked hyperalgesia in the rat. Neurosci Lett 1993; 151: 107–10PubMedCrossRef
220.
go back to reference Davar G, Hama A, Deykin A, et al. MK-801 blocks the development of thermal hyperalgesia in a rat model of experimental painful neuropathy. Brain Res 1991; 553: 327–30PubMedCrossRef Davar G, Hama A, Deykin A, et al. MK-801 blocks the development of thermal hyperalgesia in a rat model of experimental painful neuropathy. Brain Res 1991; 553: 327–30PubMedCrossRef
221.
go back to reference Boyce S, Wyatt A, Webb JK, et al. Selective NMDA NR2B antagonists induce antinociception without motor dysfunction: correlation with restricted localization of NR2B subunitin dorsal horn. Neuropharmacology 1999; 38: 611–23PubMedCrossRef Boyce S, Wyatt A, Webb JK, et al. Selective NMDA NR2B antagonists induce antinociception without motor dysfunction: correlation with restricted localization of NR2B subunitin dorsal horn. Neuropharmacology 1999; 38: 611–23PubMedCrossRef
222.
go back to reference Burton AW, Lee DH, Saab C, et al. Preemptive intrathecal ketamine injection produces a long-lasting decrease in neuropathic pain behaviors in a rat model. Reg Anesth Pain Med 1999; 24: 208–13PubMed Burton AW, Lee DH, Saab C, et al. Preemptive intrathecal ketamine injection produces a long-lasting decrease in neuropathic pain behaviors in a rat model. Reg Anesth Pain Med 1999; 24: 208–13PubMed
223.
go back to reference Munglani R, Hudspith MJ, Fleming B, et al. Effect of pre-emptive NMDA antagonist treatment on long-term Fos expression and hyperalgesia in a model of chronic neuropathic pain. Brain Res 1999; 822: 210–9PubMedCrossRef Munglani R, Hudspith MJ, Fleming B, et al. Effect of pre-emptive NMDA antagonist treatment on long-term Fos expression and hyperalgesia in a model of chronic neuropathic pain. Brain Res 1999; 822: 210–9PubMedCrossRef
224.
go back to reference Hao JX, Xu XJ. Treatment of a chronic allodynia-like response in spinally injured rats: effects of systemically administered excitatory amino acid receptor antagonists. Pain 1996; 66: 279–85PubMedCrossRef Hao JX, Xu XJ. Treatment of a chronic allodynia-like response in spinally injured rats: effects of systemically administered excitatory amino acid receptor antagonists. Pain 1996; 66: 279–85PubMedCrossRef
225.
go back to reference Qian J, Brown SD, Carlton SM. Systemic ketamine attenuates nociceptive behaviors in a rat model of peripheral neuropathy. Brain Res 1996; 715: 51–62PubMedCrossRef Qian J, Brown SD, Carlton SM. Systemic ketamine attenuates nociceptive behaviors in a rat model of peripheral neuropathy. Brain Res 1996; 715: 51–62PubMedCrossRef
226.
go back to reference Wong CS, Cherng CH, Tung CS. Intrathecal administration of excitatory amino acid receptor antagonists or nitric oxide synthase inhibitor reduced autotomy behavior in rats. Anesth Analg 1998; 87: 605–8PubMed Wong CS, Cherng CH, Tung CS. Intrathecal administration of excitatory amino acid receptor antagonists or nitric oxide synthase inhibitor reduced autotomy behavior in rats. Anesth Analg 1998; 87: 605–8PubMed
227.
go back to reference Tseng SH. Suppression of autotomy by N-methyl-D-aspartate receptor antagonist (MK-801) in the rat. Neurosci Lett 1998; 240: 17–20PubMedCrossRef Tseng SH. Suppression of autotomy by N-methyl-D-aspartate receptor antagonist (MK-801) in the rat. Neurosci Lett 1998; 240: 17–20PubMedCrossRef
228.
go back to reference Banos JE, Berslu E, Buti M, et al. Effects of dizocilpine on autotomy behavior after nerve section in mice. Brain Res 1994; 636: 107–10PubMedCrossRef Banos JE, Berslu E, Buti M, et al. Effects of dizocilpine on autotomy behavior after nerve section in mice. Brain Res 1994; 636: 107–10PubMedCrossRef
229.
go back to reference Seltzer Z, Cohn S, Ginzburg R, et al. Modulation of neuropathic pain behavior in rats by spinal disinhibition and NMDA receptor blockade of injury discharge. Pain 1991; 45: 69–75PubMedCrossRef Seltzer Z, Cohn S, Ginzburg R, et al. Modulation of neuropathic pain behavior in rats by spinal disinhibition and NMDA receptor blockade of injury discharge. Pain 1991; 45: 69–75PubMedCrossRef
230.
go back to reference Hao JX, Xu XJ, Aldskogius H, et al. the excitatory amino acid receptor antagonist MK-801 prevents the hypersensitivity induced by spinal cord ischemia in the rat. Exp Neurol 1991; 113: 182–91PubMedCrossRef Hao JX, Xu XJ, Aldskogius H, et al. the excitatory amino acid receptor antagonist MK-801 prevents the hypersensitivity induced by spinal cord ischemia in the rat. Exp Neurol 1991; 113: 182–91PubMedCrossRef
231.
go back to reference Hama AT, Unnerstall JR, Siegan JB, et al. Modulation of NMDA receptor expression in the rat spinal cord by peripheral nerve injury and adrenal medullary grafting. Brain Res 1995; 687: 103–13PubMedCrossRef Hama AT, Unnerstall JR, Siegan JB, et al. Modulation of NMDA receptor expression in the rat spinal cord by peripheral nerve injury and adrenal medullary grafting. Brain Res 1995; 687: 103–13PubMedCrossRef
232.
go back to reference Krenz NR, Weaver LC. Effect of spinal cord transection on N-methyl-D-aspartate receptors in the cord. J Neurotrauma 1998; 15: 1027–36PubMedCrossRef Krenz NR, Weaver LC. Effect of spinal cord transection on N-methyl-D-aspartate receptors in the cord. J Neurotrauma 1998; 15: 1027–36PubMedCrossRef
233.
go back to reference Li P, Wilding TJ, Kim SJ, et al. Kainate-receptor mediated sensory synaptic transmission in mammalian spinal cord. Nature 1999; 397: 161–4PubMedCrossRef Li P, Wilding TJ, Kim SJ, et al. Kainate-receptor mediated sensory synaptic transmission in mammalian spinal cord. Nature 1999; 397: 161–4PubMedCrossRef
234.
go back to reference Procter MJ, Houghton AK, Faber ES, et al. Actions of kainate and AMPA selective glutamate receptor ligands on nociceptive processing in the spinal cord. Neuropharmacology 1998; 37: 1287–97PubMedCrossRef Procter MJ, Houghton AK, Faber ES, et al. Actions of kainate and AMPA selective glutamate receptor ligands on nociceptive processing in the spinal cord. Neuropharmacology 1998; 37: 1287–97PubMedCrossRef
235.
go back to reference Ault B, Hildebrand LM. Activation of nociceptive reflexes by peripheral kainate receptors. J Pharmacol Exp Ther 1993; 265: 927–32PubMed Ault B, Hildebrand LM. Activation of nociceptive reflexes by peripheral kainate receptors. J Pharmacol Exp Ther 1993; 265: 927–32PubMed
236.
go back to reference Brambilla A, Prudentino A, Grippa N, et al. Pharmacological characterization of AMPA-induced biting behaviour in mice. Eur J Pharmacol 1996; 305: 115–7PubMedCrossRef Brambilla A, Prudentino A, Grippa N, et al. Pharmacological characterization of AMPA-induced biting behaviour in mice. Eur J Pharmacol 1996; 305: 115–7PubMedCrossRef
237.
go back to reference Yezierski RP, Liu S, Ruenes GL, et al. Excitotoxic spinal cord injury: behavioral and morphological characteristics of a central pain model. Pain 1998; 75: 141–55PubMedCrossRef Yezierski RP, Liu S, Ruenes GL, et al. Excitotoxic spinal cord injury: behavioral and morphological characteristics of a central pain model. Pain 1998; 75: 141–55PubMedCrossRef
238.
go back to reference Meller ST, Dykstra C, Gebhart GF. Acute mechanical hyperalgesia in the rat can be produced by coactivation of spinal ionotropic AMPA and metabotropic glutamate receptors, activation of phospholipase A2 and generation of cyclooxygenase products. Prog Brain Res 1996; 110: 177–92PubMedCrossRef Meller ST, Dykstra C, Gebhart GF. Acute mechanical hyperalgesia in the rat can be produced by coactivation of spinal ionotropic AMPA and metabotropic glutamate receptors, activation of phospholipase A2 and generation of cyclooxygenase products. Prog Brain Res 1996; 110: 177–92PubMedCrossRef
239.
go back to reference Meller ST, Dykstra CL, Gebhart GF. Acute mechanical hyperalgesia is produced by coactivation of AMPA and metabotropic glutamate receptors. Neuroreport 1993; 4: 879–82PubMedCrossRef Meller ST, Dykstra CL, Gebhart GF. Acute mechanical hyperalgesia is produced by coactivation of AMPA and metabotropic glutamate receptors. Neuroreport 1993; 4: 879–82PubMedCrossRef
240.
go back to reference Lutfy K, Cai SX, Woodward RM, et al. Antinociceptive effects of NMDA and non-NMDA receptor antagonists in the tail flick test in mice. Pain 1997; 70: 31–40PubMedCrossRef Lutfy K, Cai SX, Woodward RM, et al. Antinociceptive effects of NMDA and non-NMDA receptor antagonists in the tail flick test in mice. Pain 1997; 70: 31–40PubMedCrossRef
241.
go back to reference Szekely JI, Kedves R, Mate I, et al. Apparent antinociceptive and anti-inflammatory effects of GYKI 52466. Eur J Pharmacol 1997; 336: 143–54PubMedCrossRef Szekely JI, Kedves R, Mate I, et al. Apparent antinociceptive and anti-inflammatory effects of GYKI 52466. Eur J Pharmacol 1997; 336: 143–54PubMedCrossRef
242.
go back to reference Nishiyama T, Gyermek L, Lee C, et al. The spinal antinociceptive effects of a novel competitive AMPA receptor antagonist, YM872, on thermal or formalin-induced pain in rats. Anesth Analg 1999; 89: 143–7PubMed Nishiyama T, Gyermek L, Lee C, et al. The spinal antinociceptive effects of a novel competitive AMPA receptor antagonist, YM872, on thermal or formalin-induced pain in rats. Anesth Analg 1999; 89: 143–7PubMed
243.
go back to reference Simmons RMA, Li DL, Hoo KH, et al. Kainate GluR5 receptor subtype mediates the nociceptive response to formalin in the rat. Neuropharmacology 1998; 37: 25–36PubMedCrossRef Simmons RMA, Li DL, Hoo KH, et al. Kainate GluR5 receptor subtype mediates the nociceptive response to formalin in the rat. Neuropharmacology 1998; 37: 25–36PubMedCrossRef
244.
go back to reference Zahn PV, Umali E, Brennan TJ. Intrathecal non-NMDA excitatory amino acid receptor antagonists inhibit pain behaviors in a rat model of post operative pain. Pain 1998; 74: 213–23PubMedCrossRef Zahn PV, Umali E, Brennan TJ. Intrathecal non-NMDA excitatory amino acid receptor antagonists inhibit pain behaviors in a rat model of post operative pain. Pain 1998; 74: 213–23PubMedCrossRef
245.
go back to reference Pellegrini-Giampietro DE, Fan S, Ault B, et al. Glutamate receptor gene expression in spinal cord of arthritic rats. J Neurosci 1994; 14: 1576–83PubMed Pellegrini-Giampietro DE, Fan S, Ault B, et al. Glutamate receptor gene expression in spinal cord of arthritic rats. J Neurosci 1994; 14: 1576–83PubMed
246.
go back to reference Carlton SM, Hargett GL, Coggeshall RE. Plasticity in alpha-amino-3-hydroxy-5-methyl-4-isooxazolepropionic acid receptor subunits in the rat dorsal horn following deafferentation. Neurosci Lett 1998; 242: 21–4PubMedCrossRef Carlton SM, Hargett GL, Coggeshall RE. Plasticity in alpha-amino-3-hydroxy-5-methyl-4-isooxazolepropionic acid receptor subunits in the rat dorsal horn following deafferentation. Neurosci Lett 1998; 242: 21–4PubMedCrossRef
247.
go back to reference Xu XJ, Hao JX, Seiger A, et al. Systemic excitatory amino acid receptor antagonists of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor and of the N-methyl-D-aspartate (NMDA) receptor relieve mechanical hypersensitivity after transient spinal cord ischemia in rats. J Pharmacol Exp Ther 1993; 267: 140–4PubMed Xu XJ, Hao JX, Seiger A, et al. Systemic excitatory amino acid receptor antagonists of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor and of the N-methyl-D-aspartate (NMDA) receptor relieve mechanical hypersensitivity after transient spinal cord ischemia in rats. J Pharmacol Exp Ther 1993; 267: 140–4PubMed
248.
go back to reference Advokat C, Rutherford D. Selective antinociceptive effect of excitatory amino acid antagonists in intact and acute spinal rats. Pharmacol Biochem Behav 1995; 51: 855–60PubMedCrossRef Advokat C, Rutherford D. Selective antinociceptive effect of excitatory amino acid antagonists in intact and acute spinal rats. Pharmacol Biochem Behav 1995; 51: 855–60PubMedCrossRef
249.
go back to reference Popratiloff A, Weinberg RJ, Rustioni A. AMPA receptors at primary afferent synapses in substantia gelatinosa after sciatic nerve section. Eur J Neurosci 1998; 10: 3220–30PubMedCrossRef Popratiloff A, Weinberg RJ, Rustioni A. AMPA receptors at primary afferent synapses in substantia gelatinosa after sciatic nerve section. Eur J Neurosci 1998; 10: 3220–30PubMedCrossRef
250.
go back to reference Harris JA, Corsi M, Quartaroli M, et al. Up regulation of spinal glutamate receptors in chronic pain. Neuroscience 1996; 74: 7–12PubMedCrossRef Harris JA, Corsi M, Quartaroli M, et al. Up regulation of spinal glutamate receptors in chronic pain. Neuroscience 1996; 74: 7–12PubMedCrossRef
251.
go back to reference Coutinho SV, Meller ST, Gebhart GF. Intracolonic zymosan produces visceral hyperalgesia in the rat that is mediated by spinal NMDA and non-NMDA receptors. Brain Res 1996; 736: 7–15PubMedCrossRef Coutinho SV, Meller ST, Gebhart GF. Intracolonic zymosan produces visceral hyperalgesia in the rat that is mediated by spinal NMDA and non-NMDA receptors. Brain Res 1996; 736: 7–15PubMedCrossRef
252.
go back to reference Charpak S, Gahwiler BH, Do KQ, et al. Potassium conductances in hippocampal neurons blocked by excitatory amino-acid transmitters. Nature 1990; 347: 765–7PubMedCrossRef Charpak S, Gahwiler BH, Do KQ, et al. Potassium conductances in hippocampal neurons blocked by excitatory amino-acid transmitters. Nature 1990; 347: 765–7PubMedCrossRef
253.
go back to reference Stratton KR, Worley PF, Baraban JM. Excitation of hippocampal neurons by stimulation of glutamate Qp receptors. Eur J Pharmacol 1989; 173: 235–7PubMedCrossRef Stratton KR, Worley PF, Baraban JM. Excitation of hippocampal neurons by stimulation of glutamate Qp receptors. Eur J Pharmacol 1989; 173: 235–7PubMedCrossRef
254.
go back to reference Libri V, Constanti A, Zibetti M, et al. Metabotropic glutamate receptor subtypes mediating slow inward tail current (IADP) induction and inhibition of synaptic transmission in olfactory cortical neurones. Br J Pharmacol 1997; 120: 1083–95PubMedCrossRef Libri V, Constanti A, Zibetti M, et al. Metabotropic glutamate receptor subtypes mediating slow inward tail current (IADP) induction and inhibition of synaptic transmission in olfactory cortical neurones. Br J Pharmacol 1997; 120: 1083–95PubMedCrossRef
255.
go back to reference Eaton SA, Jane DE, Jones PL St J, et al. Competitive antagonism at metabotropic glutamate receptors by (S)-4-carboxy-phenylglycine and (RS)-α-methyl-carboxyphenylglycine. Eur J Pharmacol 1993; 244: 195–7PubMedCrossRef Eaton SA, Jane DE, Jones PL St J, et al. Competitive antagonism at metabotropic glutamate receptors by (S)-4-carboxy-phenylglycine and (RS)-α-methyl-carboxyphenylglycine. Eur J Pharmacol 1993; 244: 195–7PubMedCrossRef
256.
go back to reference Jane DE, Jones PL St J, Pook PC-K, et al. Stereospecific antagonism by (+)-α-methyl-4-carboxyphenylglycine (MCPG) of (1S,3R)-ACPD-induced effects in neonatal rat motoneurones and rat thalamic neurones. Neuropharmacology 1993; 32: 725–7PubMedCrossRef Jane DE, Jones PL St J, Pook PC-K, et al. Stereospecific antagonism by (+)-α-methyl-4-carboxyphenylglycine (MCPG) of (1S,3R)-ACPD-induced effects in neonatal rat motoneurones and rat thalamic neurones. Neuropharmacology 1993; 32: 725–7PubMedCrossRef
257.
go back to reference Salt TE, Eaton SA. Modulation of sensory neurone excitatory and inhibitory responses in the ventrobasal thalamus by activation of metabotropic excitatory amino acid receptors. Neuropharmacology 1995; 34: 1043–51PubMedCrossRef Salt TE, Eaton SA. Modulation of sensory neurone excitatory and inhibitory responses in the ventrobasal thalamus by activation of metabotropic excitatory amino acid receptors. Neuropharmacology 1995; 34: 1043–51PubMedCrossRef
258.
go back to reference Zheng F, Gallagher JP. Metabotropic glutamate receptor agonists potentiate a slow afterdepolarization in CNS neurons. Neuroreport 1992; 3: 622–4PubMedCrossRef Zheng F, Gallagher JP. Metabotropic glutamate receptor agonists potentiate a slow afterdepolarization in CNS neurons. Neuroreport 1992; 3: 622–4PubMedCrossRef
259.
go back to reference Zheng F, Gallagher JP, Connor JA. Activation of a metabotropic excitatory amino acid receptor potentiates spike-driven calcium increases in neurons of the dorsolateral septum. J Neurosci 1996; 16: 6079–88PubMed Zheng F, Gallagher JP, Connor JA. Activation of a metabotropic excitatory amino acid receptor potentiates spike-driven calcium increases in neurons of the dorsolateral septum. J Neurosci 1996; 16: 6079–88PubMed
260.
go back to reference Zheng F, Hasuo H, Gallagher JP. 1S,3R-ACPD-preferring inward current in rat dorsolateral septal neurons is mediated by a novel excitatory amino acid receptor. Neuropharmacology 1995; 34: 905–17PubMedCrossRef Zheng F, Hasuo H, Gallagher JP. 1S,3R-ACPD-preferring inward current in rat dorsolateral septal neurons is mediated by a novel excitatory amino acid receptor. Neuropharmacology 1995; 34: 905–17PubMedCrossRef
261.
go back to reference Desai MA, Smith TS, Conn PJ. Multiple metabotropic glutamate receptors regulate hippocampal function. Synapse 1992; 12: 206–13PubMedCrossRef Desai MA, Smith TS, Conn PJ. Multiple metabotropic glutamate receptors regulate hippocampal function. Synapse 1992; 12: 206–13PubMedCrossRef
262.
go back to reference Glaum SR, Miller RJ. Metabotropic glutamate receptors mediate excitatory transmission in the nucleus of the solitary tract. J Neurosci 1992; 12: 2251–8PubMed Glaum SR, Miller RJ. Metabotropic glutamate receptors mediate excitatory transmission in the nucleus of the solitary tract. J Neurosci 1992; 12: 2251–8PubMed
263.
go back to reference Shen K-Z, Johnson SW. A slow excitatory postsynaptic current mediated by G-protein-coupled metabotropic glutamate receptors in rat ventral tegmental dopamine neurons. Eur J Neurosci 1997; 9: 48–54PubMedCrossRef Shen K-Z, Johnson SW. A slow excitatory postsynaptic current mediated by G-protein-coupled metabotropic glutamate receptors in rat ventral tegmental dopamine neurons. Eur J Neurosci 1997; 9: 48–54PubMedCrossRef
264.
go back to reference Glaum SR, Slater NT, Rossi DJ, et al. Role of metabotropic glutamate (ACPD) receptors at the parallel fiber-Purkinje cell synapse. J Neurophysiol 1992; 68: 1453–62PubMed Glaum SR, Slater NT, Rossi DJ, et al. Role of metabotropic glutamate (ACPD) receptors at the parallel fiber-Purkinje cell synapse. J Neurophysiol 1992; 68: 1453–62PubMed
265.
go back to reference Boxall SJ, Thompson SW, Dray A, et al. Metabotropic glutamate receptor activation contributes to nociceptive reflex activity in the rat spinal cord in vitro. Neuroscience 1996; 74: 13–20PubMedCrossRef Boxall SJ, Thompson SW, Dray A, et al. Metabotropic glutamate receptor activation contributes to nociceptive reflex activity in the rat spinal cord in vitro. Neuroscience 1996; 74: 13–20PubMedCrossRef
266.
go back to reference Thompson GA, Jones PL St J, Kilpatrick IC. The actions of a range of excitatory amino acids at (1S,3R)-1-aminocyclopentane-1,3-dicarboxylic acid-depolarizing receptors on neonatal rat motoneurones. Neuropharmacology 1995; 34: 857–63PubMedCrossRef Thompson GA, Jones PL St J, Kilpatrick IC. The actions of a range of excitatory amino acids at (1S,3R)-1-aminocyclopentane-1,3-dicarboxylic acid-depolarizing receptors on neonatal rat motoneurones. Neuropharmacology 1995; 34: 857–63PubMedCrossRef
267.
go back to reference Young MR, Fleetwood-Walker SM, Mitchell R, et al. Evidence for a role of metabotropic glutamate receptors in sustained nociceptive inputs to rat dorsal horn neurons. Neuropharmacology 1994; 33: 141–4PubMedCrossRef Young MR, Fleetwood-Walker SM, Mitchell R, et al. Evidence for a role of metabotropic glutamate receptors in sustained nociceptive inputs to rat dorsal horn neurons. Neuropharmacology 1994; 33: 141–4PubMedCrossRef
268.
go back to reference Young MR, Fleetwood-Walker SM, Mitchell R, et al. The involvement of metabotropic glutamate receptors and their intracellular signaling pathways in sustained nociceptive transmission in rat dorsal horn neurons. Neuropharmacology 1995; 34: 1033–41PubMedCrossRef Young MR, Fleetwood-Walker SM, Mitchell R, et al. The involvement of metabotropic glutamate receptors and their intracellular signaling pathways in sustained nociceptive transmission in rat dorsal horn neurons. Neuropharmacology 1995; 34: 1033–41PubMedCrossRef
269.
go back to reference Young MR, Fleetwood-Walker SM, Dickenson T, et al. Behavioural and electrophysiological evidence supporting a role for group I metabotropic glutamate receptors in the mediation of nociceptive inputs to the rat spinal cord. Brain Res 1997; 777: 161–9PubMedCrossRef Young MR, Fleetwood-Walker SM, Dickenson T, et al. Behavioural and electrophysiological evidence supporting a role for group I metabotropic glutamate receptors in the mediation of nociceptive inputs to the rat spinal cord. Brain Res 1997; 777: 161–9PubMedCrossRef
270.
go back to reference Young MR, Blackburn-Munro G, Dickinson T, et al. Antisense ablation of type I metabotropic glutamate receptor mGluR1 inhibits spinal nociceptive transmission. J Neurosci 1998; 18: 10180–8PubMed Young MR, Blackburn-Munro G, Dickinson T, et al. Antisense ablation of type I metabotropic glutamate receptor mGluR1 inhibits spinal nociceptive transmission. J Neurosci 1998; 18: 10180–8PubMed
271.
go back to reference Spanswick D, Pickering AE, Gibson IC, et al. Excitation of sympathetic preganglionic neurons via metabotropic excitatory amino acid receptors. Neuroscience 1995; 68: 1247–61PubMedCrossRef Spanswick D, Pickering AE, Gibson IC, et al. Excitation of sympathetic preganglionic neurons via metabotropic excitatory amino acid receptors. Neuroscience 1995; 68: 1247–61PubMedCrossRef
272.
go back to reference Stanfa LC, Dickenson AH. Inflammation alters the effects of mGlu receptor agonists in spinal nociceptive neurones. Eur J Pharmacol 1998; 347: 165–72PubMedCrossRef Stanfa LC, Dickenson AH. Inflammation alters the effects of mGlu receptor agonists in spinal nociceptive neurones. Eur J Pharmacol 1998; 347: 165–72PubMedCrossRef
273.
go back to reference Neugebauer V, Lucke T, Schaible HG. Requirement of metabotropic glutamate receptors for the generation of inflammationevoked hyperexcitability in rat spinal cord neurons. Eur J Neurosci 1994; 6: 1179–86PubMedCrossRef Neugebauer V, Lucke T, Schaible HG. Requirement of metabotropic glutamate receptors for the generation of inflammationevoked hyperexcitability in rat spinal cord neurons. Eur J Neurosci 1994; 6: 1179–86PubMedCrossRef
274.
go back to reference Fisher K, Coderre TJ. Comparison of nociceptive effects produced by intrathecal administration of mGluR agonists. Neuroreport 1996; 7: 2743–7PubMedCrossRef Fisher K, Coderre TJ. Comparison of nociceptive effects produced by intrathecal administration of mGluR agonists. Neuroreport 1996; 7: 2743–7PubMedCrossRef
275.
go back to reference Fundytus ME, Fisher K, Dray A, et al. In vivo antinociceptive activity of anti-rat mGluR1 and mGluR5 antibodies in rats. Neuroreport 1998; 9: 731–5PubMedCrossRef Fundytus ME, Fisher K, Dray A, et al. In vivo antinociceptive activity of anti-rat mGluR1 and mGluR5 antibodies in rats. Neuroreport 1998; 9: 731–5PubMedCrossRef
276.
go back to reference Fundytus ME, Yashpal K, Chabot J-G, et al. Knockdown of spinal metabotropic glutamate receptor 1 (mGluR1) alleviates pain and restores opioid efficacy after nerve injury in rats. Br J Pharmacol. In press Fundytus ME, Yashpal K, Chabot J-G, et al. Knockdown of spinal metabotropic glutamate receptor 1 (mGluR1) alleviates pain and restores opioid efficacy after nerve injury in rats. Br J Pharmacol. In press
277.
go back to reference Maione S, Marabese I, Leyva J, et al. Characterization of mGluRs which modulate nociception in the PAG of the mouse. Neuropharmacology 1998; 37: 1475–83PubMedCrossRef Maione S, Marabese I, Leyva J, et al. Characterization of mGluRs which modulate nociception in the PAG of the mouse. Neuropharmacology 1998; 37: 1475–83PubMedCrossRef
278.
go back to reference Maione S, Oliva P, Marabese I, et al. Periaqueductal gray matter metabotropic glutamate receptors modulate formalin-induced nociception. Pain 2000; 85: 183–9PubMedCrossRef Maione S, Oliva P, Marabese I, et al. Periaqueductal gray matter metabotropic glutamate receptors modulate formalin-induced nociception. Pain 2000; 85: 183–9PubMedCrossRef
279.
go back to reference Fisher K, Coderre TJ. The contribution of mGluRs to formalin-induced nociception. Pain 1996; 68: 255–63PubMedCrossRef Fisher K, Coderre TJ. The contribution of mGluRs to formalin-induced nociception. Pain 1996; 68: 255–63PubMedCrossRef
280.
go back to reference Watkins JC, Collingridge GL. Phenylglycine derivatives as antagonists of metabotropic glutamate receptors. Trends Pharmacol Sci 1994; 15: 333–42PubMedCrossRef Watkins JC, Collingridge GL. Phenylglycine derivatives as antagonists of metabotropic glutamate receptors. Trends Pharmacol Sci 1994; 15: 333–42PubMedCrossRef
281.
go back to reference Fundytus ME, Fisher K, Dray A, et al. Antisense oligonucleotides targeting group I mGluRs attenuate nerve constriction-induced hyperalgesia and allodynia. Soc Neurosci Abstracts 1997; 23: 1013 Fundytus ME, Fisher K, Dray A, et al. Antisense oligonucleotides targeting group I mGluRs attenuate nerve constriction-induced hyperalgesia and allodynia. Soc Neurosci Abstracts 1997; 23: 1013
282.
go back to reference Fundytus ME, Henry JL, Dray A, et al. Antisense knockdown of mGluR1 reverses hyperalgesia and allodynia associated with an established neuropathic injury in rats. Proceedings of the 9th World Congress on Pain. Prog Pain Res Manag 2000; 16: 343–9 Fundytus ME, Henry JL, Dray A, et al. Antisense knockdown of mGluR1 reverses hyperalgesia and allodynia associated with an established neuropathic injury in rats. Proceedings of the 9th World Congress on Pain. Prog Pain Res Manag 2000; 16: 343–9
283.
go back to reference Zahn PK, Brennan TJ. Intrathecal metabotropic glutamate receptor antagonists do not decrease mechanical hyperalgesia in a rat model of post operative pain. Anesth Analg 1998; 87: 1354–9PubMed Zahn PK, Brennan TJ. Intrathecal metabotropic glutamate receptor antagonists do not decrease mechanical hyperalgesia in a rat model of post operative pain. Anesth Analg 1998; 87: 1354–9PubMed
284.
go back to reference Casabona G, Catania MV, Storto M, et al. Deafferentation upregulates the expression of the mGlu1a metabotropic glutamate receptor protein in the olfactory bulb. Eur J Neurosci 1998; 10: 771–6PubMedCrossRef Casabona G, Catania MV, Storto M, et al. Deafferentation upregulates the expression of the mGlu1a metabotropic glutamate receptor protein in the olfactory bulb. Eur J Neurosci 1998; 10: 771–6PubMedCrossRef
285.
go back to reference Yashpal K, Pitcher GM, Parent A, et al. Noxious thermal and chemical stimulation induce increases in 3H-phorbol 12,13- dibutyrate binding in spinal cord dorsal horn as well as persistent pain and hyperalgesia, which is reduced by inhibition of protein kinase C. J Neurosci 1995; 15: 3263–72PubMed Yashpal K, Pitcher GM, Parent A, et al. Noxious thermal and chemical stimulation induce increases in 3H-phorbol 12,13- dibutyrate binding in spinal cord dorsal horn as well as persistent pain and hyperalgesia, which is reduced by inhibition of protein kinase C. J Neurosci 1995; 15: 3263–72PubMed
286.
go back to reference Coderre TJ, Yashpal K. Intracellular messengers contributing to persistent nociception and hyperalgesia induced by L-glutamate and substance P in the rat formalin pain model. Eur J Neurosci 1994; 6: 1328–34PubMedCrossRef Coderre TJ, Yashpal K. Intracellular messengers contributing to persistent nociception and hyperalgesia induced by L-glutamate and substance P in the rat formalin pain model. Eur J Neurosci 1994; 6: 1328–34PubMedCrossRef
287.
go back to reference Coderre TJ, Contribution of protein kinase C to central sensitization and persistent pain following tissue injury. Neurosci Lett 1992; 140: 181–4PubMedCrossRef Coderre TJ, Contribution of protein kinase C to central sensitization and persistent pain following tissue injury. Neurosci Lett 1992; 140: 181–4PubMedCrossRef
288.
go back to reference Nakanishi O, Ishikawa T, Imamura Y. Modulation of formalin-evoked hyperalgesia by intrathecal N-type Ca channel and protein kinase C inhibitor in the rat. Cell Mol Neurobiol 1999; 19: 191–7PubMedCrossRef Nakanishi O, Ishikawa T, Imamura Y. Modulation of formalin-evoked hyperalgesia by intrathecal N-type Ca channel and protein kinase C inhibitor in the rat. Cell Mol Neurobiol 1999; 19: 191–7PubMedCrossRef
289.
go back to reference Mao J, Price DD, Phillips LL, et al. Increases in protein kinase C gamma immunoreactivity in the spinal cord dorsal horn of rats with painful mononeuropathy. Neurosci Lett 1995; 198: 75–8PubMedCrossRef Mao J, Price DD, Phillips LL, et al. Increases in protein kinase C gamma immunoreactivity in the spinal cord dorsal horn of rats with painful mononeuropathy. Neurosci Lett 1995; 198: 75–8PubMedCrossRef
290.
go back to reference Martin WJ, Liu H, Wang H, et al. Inflammation-induced upregulation of protein kinase C gamma immunoreactivity in rat spinal cord correlates with enhanced nociceptive processing. Neuroscience 1999; 88: 1267–74PubMedCrossRef Martin WJ, Liu H, Wang H, et al. Inflammation-induced upregulation of protein kinase C gamma immunoreactivity in rat spinal cord correlates with enhanced nociceptive processing. Neuroscience 1999; 88: 1267–74PubMedCrossRef
291.
go back to reference Fundytus ME. Activity at group I mGluRs: a common mechanism underlying hyperalgesia/allodynia, opioid sensitivity and NMDA sensitivity in neuropathic rats. Can Pain Soc Abstracts 1999. Pain Res Manag 1999: 4: 34 Fundytus ME. Activity at group I mGluRs: a common mechanism underlying hyperalgesia/allodynia, opioid sensitivity and NMDA sensitivity in neuropathic rats. Can Pain Soc Abstracts 1999. Pain Res Manag 1999: 4: 34
292.
go back to reference Mayer DJ, Mao J, Price DD. The association of neuropathic pain, morphine tolerance and dependence, and the translocation of protein kinase C. NIDA Res Monogr 1995; 147: 269–98PubMed Mayer DJ, Mao J, Price DD. The association of neuropathic pain, morphine tolerance and dependence, and the translocation of protein kinase C. NIDA Res Monogr 1995; 147: 269–98PubMed
293.
go back to reference Mao J, Price DD, Mayer DJ, et al. Pain-related increases in spinal cord membrane-bound protein kinase C following peripheral nerve injury. Brain Res 1992; 588: 144–9PubMedCrossRef Mao J, Price DD, Mayer DJ, et al. Pain-related increases in spinal cord membrane-bound protein kinase C following peripheral nerve injury. Brain Res 1992; 588: 144–9PubMedCrossRef
294.
go back to reference Malmberg AB, Chen CC, Tonegawa S, et al. Preserved acute pain and reduced neuropathic pain in mice lacking PKC gamma. Science 1997; 278: 279–83PubMedCrossRef Malmberg AB, Chen CC, Tonegawa S, et al. Preserved acute pain and reduced neuropathic pain in mice lacking PKC gamma. Science 1997; 278: 279–83PubMedCrossRef
295.
go back to reference Anikstejn L, Otani S, Ben-Ari Y. Quisqualate metabotropic receptors modulate NMDA currents and facilitate induction of long-term potentiation through protein kinase C. Eur J Neurosci 1992; 4: 500–5CrossRef Anikstejn L, Otani S, Ben-Ari Y. Quisqualate metabotropic receptors modulate NMDA currents and facilitate induction of long-term potentiation through protein kinase C. Eur J Neurosci 1992; 4: 500–5CrossRef
296.
go back to reference Bleakman D, Rusin KI, Chard PS, et al. Metabotropic glutamate receptors potentiate ionotropic glutamate responses in the rat dorsal horn. Mol Pharmacol 1992; 42: 192–6PubMed Bleakman D, Rusin KI, Chard PS, et al. Metabotropic glutamate receptors potentiate ionotropic glutamate responses in the rat dorsal horn. Mol Pharmacol 1992; 42: 192–6PubMed
297.
go back to reference Chen L, Huang L-YM. Protein kinase C reduces Mg2+ block of NMDA-receptor channels as a mechanism of modulation. Nature 1992; 356: 521–3PubMedCrossRef Chen L, Huang L-YM. Protein kinase C reduces Mg2+ block of NMDA-receptor channels as a mechanism of modulation. Nature 1992; 356: 521–3PubMedCrossRef
298.
go back to reference Harvey J, Collingridge GL. Signal transduction pathways involved in the acute potentiation of NMDA responses by 1S,3R-ACPD in rat hippocampal slices. Br J Pharmacol 1993; 109: 1085–90PubMedCrossRef Harvey J, Collingridge GL. Signal transduction pathways involved in the acute potentiation of NMDA responses by 1S,3R-ACPD in rat hippocampal slices. Br J Pharmacol 1993; 109: 1085–90PubMedCrossRef
299.
go back to reference Kelso SR, Nelson TE, Leonard JP. Protein kinase C-mediated enhancement of NMDA currents by metabotropic glutamate receptors in Xenopus oocytes. J Physiol 1992; 449: 705–18PubMed Kelso SR, Nelson TE, Leonard JP. Protein kinase C-mediated enhancement of NMDA currents by metabotropic glutamate receptors in Xenopus oocytes. J Physiol 1992; 449: 705–18PubMed
300.
go back to reference Kitamura Y, Miyazaki A, Yamanaka Y, et al. Stimulatory effects of protein kinase C and calmodulin kinase II on N-methyl-D-aspartate receptor/channels in the post-synaptic density of rat brain. J Neurochem 1993; 61: 100–9PubMedCrossRef Kitamura Y, Miyazaki A, Yamanaka Y, et al. Stimulatory effects of protein kinase C and calmodulin kinase II on N-methyl-D-aspartate receptor/channels in the post-synaptic density of rat brain. J Neurochem 1993; 61: 100–9PubMedCrossRef
301.
go back to reference Raymond LA, Tingley WG, Blackstone CD, et al. Glutamate receptor modulation by protein kinase phosphorylation. J Physiol Paris 1994; 88: 181–92PubMedCrossRef Raymond LA, Tingley WG, Blackstone CD, et al. Glutamate receptor modulation by protein kinase phosphorylation. J Physiol Paris 1994; 88: 181–92PubMedCrossRef
302.
go back to reference Swope SL, Moss SI, Raymond LA, et al. Regulation of ligand-gated ion channels by protein phosphorylation. Adv Second Messenger Phosphoprotein Res 1999; 33: 49–78PubMedCrossRef Swope SL, Moss SI, Raymond LA, et al. Regulation of ligand-gated ion channels by protein phosphorylation. Adv Second Messenger Phosphoprotein Res 1999; 33: 49–78PubMedCrossRef
303.
go back to reference Bruno V, Copani A, Knopfel T, et al. Activation of metabotropic glutamate receptors coupled to inositol phospholipid hydrolysis amplifies NMDA-induced neuronal degeneration in cultured cortical cells. Neuropharmacology 1995; 34: 1089–98PubMedCrossRef Bruno V, Copani A, Knopfel T, et al. Activation of metabotropic glutamate receptors coupled to inositol phospholipid hydrolysis amplifies NMDA-induced neuronal degeneration in cultured cortical cells. Neuropharmacology 1995; 34: 1089–98PubMedCrossRef
304.
go back to reference Martin G, Nie Z, Siggins GR. Metabotropic glutamate receptors regulate N-methyl-D-aspartate-mediated synaptic transmission in nucleus accumbens. J Neurophysiol 1997; 78: 3028–38PubMed Martin G, Nie Z, Siggins GR. Metabotropic glutamate receptors regulate N-methyl-D-aspartate-mediated synaptic transmission in nucleus accumbens. J Neurophysiol 1997; 78: 3028–38PubMed
305.
go back to reference Martin G, Przewlocki R, Siggins GR. Chronic morphine treatment selectively augments metabotropic glutamate receptor-induced inhibition of N-methyl-D-aspartate receptor-mediated neurotransmission in nucleus accumbens. J Pharmacol Exp Ther 1998; 288: 30–5 Martin G, Przewlocki R, Siggins GR. Chronic morphine treatment selectively augments metabotropic glutamate receptor-induced inhibition of N-methyl-D-aspartate receptor-mediated neurotransmission in nucleus accumbens. J Pharmacol Exp Ther 1998; 288: 30–5
306.
go back to reference Bruno V, Battaglia G, Copani A, et al. Activation of class II or III metabotropic glutamate receptors protects cultured cortical neurons against excitotoxic degeneration. Eur J Neurosci 1995; 7: 1906–13PubMedCrossRef Bruno V, Battaglia G, Copani A, et al. Activation of class II or III metabotropic glutamate receptors protects cultured cortical neurons against excitotoxic degeneration. Eur J Neurosci 1995; 7: 1906–13PubMedCrossRef
307.
go back to reference Fundytus ME, Yashpal K, Dray A, et al. Antisense oligonucleotide knockdown of mGluR1 reverses enhanced NMDA sensitivity in neuropathic rats. Soc Neurosci Abstracts, 1999; 25: 449 Fundytus ME, Yashpal K, Dray A, et al. Antisense oligonucleotide knockdown of mGluR1 reverses enhanced NMDA sensitivity in neuropathic rats. Soc Neurosci Abstracts, 1999; 25: 449
308.
go back to reference Abou-Samira AB, Harwood JP, Catt KJ, et al. Mechanisms of action of CRF and other regulators of ACTH release in pituitary corticotrophs. Ann NY Acad Sci 1987; 512: 67–84CrossRef Abou-Samira AB, Harwood JP, Catt KJ, et al. Mechanisms of action of CRF and other regulators of ACTH release in pituitary corticotrophs. Ann NY Acad Sci 1987; 512: 67–84CrossRef
309.
go back to reference Zieglgansberger W, Tulloch IF. The effects of methionine- and leucine-enkephalin on spinal neurons of the cat. Brain Res 1979; 167: 53–64PubMedCrossRef Zieglgansberger W, Tulloch IF. The effects of methionine- and leucine-enkephalin on spinal neurons of the cat. Brain Res 1979; 167: 53–64PubMedCrossRef
310.
go back to reference Hill RG, Pepper CM. Selective effects of morphine on the no-ciceptive responses of thalamic neurones in the rat. Br J Pharmacol 1978; 64: 137–43PubMedCrossRef Hill RG, Pepper CM. Selective effects of morphine on the no-ciceptive responses of thalamic neurones in the rat. Br J Pharmacol 1978; 64: 137–43PubMedCrossRef
311.
go back to reference Jhamandas KH, Marsala M, Ibuki T, et al. Spinal amino acid release and precipitated withdrawal in rats chronically infused with spinal morphine. J Neurosci 1996; 15: 2758–66 Jhamandas KH, Marsala M, Ibuki T, et al. Spinal amino acid release and precipitated withdrawal in rats chronically infused with spinal morphine. J Neurosci 1996; 15: 2758–66
312.
go back to reference Hong M, Milne B, Jhamandas K. Evidence for the involvement of excitatory amino acid pathways in the development of precipitated withdrawal from acute and chronic morphine: an in vivo voltammetric study in the rat locus coeruleus. Brain Res 1993; 623: 131–41PubMedCrossRef Hong M, Milne B, Jhamandas K. Evidence for the involvement of excitatory amino acid pathways in the development of precipitated withdrawal from acute and chronic morphine: an in vivo voltammetric study in the rat locus coeruleus. Brain Res 1993; 623: 131–41PubMedCrossRef
313.
go back to reference Kogan JH, Aghajanian GK. Long-term glutamate desensitization in locus coeruleus neurons and its role in opiate withdrawal. Brain Res 1995; 689: 111–21PubMedCrossRef Kogan JH, Aghajanian GK. Long-term glutamate desensitization in locus coeruleus neurons and its role in opiate withdrawal. Brain Res 1995; 689: 111–21PubMedCrossRef
314.
go back to reference Aghajanian GK, Kogan JH, Moghaddam B. Opiate withdrawal increases glutamate and aspartate efflux in the locus coeruleus an in vivo microdialysis study. Brain Res 1994; 636: 126–30PubMedCrossRef Aghajanian GK, Kogan JH, Moghaddam B. Opiate withdrawal increases glutamate and aspartate efflux in the locus coeruleus an in vivo microdialysis study. Brain Res 1994; 636: 126–30PubMedCrossRef
315.
go back to reference Tokuyama S, Wakabayashi H, Ho IK. Direct evidence for a role of glutamate in the expression of the opioid withdrawal syndrome. Eur J Pharmacol 1996; 295: 123–9PubMedCrossRef Tokuyama S, Wakabayashi H, Ho IK. Direct evidence for a role of glutamate in the expression of the opioid withdrawal syndrome. Eur J Pharmacol 1996; 295: 123–9PubMedCrossRef
316.
go back to reference Lizasoain I, Leza JC, Cuellar B, et al. Inhibition of morphine withdrawal by lamotrigine: involvement of nitric oxide. Eur J Pharmacol 1996; 299: 41–5PubMedCrossRef Lizasoain I, Leza JC, Cuellar B, et al. Inhibition of morphine withdrawal by lamotrigine: involvement of nitric oxide. Eur J Pharmacol 1996; 299: 41–5PubMedCrossRef
317.
go back to reference 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: 549–59PubMedCrossRef 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: 549–59PubMedCrossRef
318.
go back to reference Gracy KN, Svingos AL, Pickel VM. Dual ultrastructural localization of mu-opioid receptors and NMDA-type glutamate receptors in the shell of the rat nucleus accumbens. J Neurosci 1997; 17: 4839–48PubMed Gracy KN, Svingos AL, Pickel VM. Dual ultrastructural localization of mu-opioid receptors and NMDA-type glutamate receptors in the shell of the rat nucleus accumbens. J Neurosci 1997; 17: 4839–48PubMed
319.
go back to reference Angulo JA, Williams A, Ledoux M, et al. Elevation of striatal and accumbal preproenkephalin, preprotachykinin and preprodynorphin mRNA abundance subsequent to N-methyl-D-aspartate receptor blockade with MK-801. Brain Res Mol Brain Res 1995; 29: 15–22PubMedCrossRef Angulo JA, Williams A, Ledoux M, et al. Elevation of striatal and accumbal preproenkephalin, preprotachykinin and preprodynorphin mRNA abundance subsequent to N-methyl-D-aspartate receptor blockade with MK-801. Brain Res Mol Brain Res 1995; 29: 15–22PubMedCrossRef
320.
go back to reference Angulo JA, Watanabe Y, Cadet J, et al. Upregulation of fore-brain proenkephalin mRNA subsequent to NMDA receptor blockade. Eur J Pharmacol 1993; 244: 317–8PubMedCrossRef Angulo JA, Watanabe Y, Cadet J, et al. Upregulation of fore-brain proenkephalin mRNA subsequent to NMDA receptor blockade. Eur J Pharmacol 1993; 244: 317–8PubMedCrossRef
321.
go back to reference Chen L, Gu Y, Huang LY. The mechanism of action for the block of NMDA receptor channels by the opioid peptide dynorphin. J Neurosci 1995; 15: 4602–11PubMed Chen L, Gu Y, Huang LY. The mechanism of action for the block of NMDA receptor channels by the opioid peptide dynorphin. J Neurosci 1995; 15: 4602–11PubMed
322.
go back to reference 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: 208–12PubMedCrossRef 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: 208–12PubMedCrossRef
323.
go back to reference Hocherman SD, Randic M. Reduction of NMDA-induced Ca2+ transients by a mu-opioid receptor agonist in dorsal horn neurons. Neuroreport 1997; 8: 3061–5PubMedCrossRef Hocherman SD, Randic M. Reduction of NMDA-induced Ca2+ transients by a mu-opioid receptor agonist in dorsal horn neurons. Neuroreport 1997; 8: 3061–5PubMedCrossRef
324.
go back to reference Feng J, Kendig JJ. The NMDA receptor antagonist MK-801 differentially modulates mu and kappa opioid actions in spinal cord in vitro. Pain 1996; 66: 343–9PubMedCrossRef Feng J, Kendig JJ. The NMDA receptor antagonist MK-801 differentially modulates mu and kappa opioid actions in spinal cord in vitro. Pain 1996; 66: 343–9PubMedCrossRef
325.
go back to reference Oleskevich S, Clements JD, Williams JT. Opioid-glutamate intereactions in rat locus coeruleus neurons. J Neurophysiol 1993; 70: 931–7PubMed Oleskevich S, Clements JD, Williams JT. Opioid-glutamate intereactions in rat locus coeruleus neurons. J Neurophysiol 1993; 70: 931–7PubMed
326.
go back to reference Martin G, Nie Z, Siggins GR. Mu-Opioid receptors modulate NMDA receptor-mediated responses in nucleus accumbens neurons. J Neurosci 1997; 17: 11–22PubMed Martin G, Nie Z, Siggins GR. Mu-Opioid receptors modulate NMDA receptor-mediated responses in nucleus accumbens neurons. J Neurosci 1997; 17: 11–22PubMed
327.
go back to reference Vaughan CW, Christie MJ. Presynaptic inhibitory action of opioids on synaptic transmission in the rat periaqueductal grey in vitro. J Physiol (Lond) 1997; 498: 463–72 Vaughan CW, Christie MJ. Presynaptic inhibitory action of opioids on synaptic transmission in the rat periaqueductal grey in vitro. J Physiol (Lond) 1997; 498: 463–72
328.
go back to reference Zhang KM, Wang XM, Mokha SS. Opioids modulate N-methyl-D-aspartic acid (NMDA)-evoked responses of neurons in the superficial and deeper dorsal horn of the medulla (trigeminal nucleus caudalis). Brain Res 1996; 719: 229–33PubMedCrossRef Zhang KM, Wang XM, Mokha SS. Opioids modulate N-methyl-D-aspartic acid (NMDA)-evoked responses of neurons in the superficial and deeper dorsal horn of the medulla (trigeminal nucleus caudalis). Brain Res 1996; 719: 229–33PubMedCrossRef
329.
go back to reference Cai YC, Ma L, Fan GH, et al. Activation of N-methyl-D-aspartate receptor attenuates acute responsiveness of delta-opioid receptors. Mol Pharmacol 1997; 51: 583–7PubMed Cai YC, Ma L, Fan GH, et al. Activation of N-methyl-D-aspartate receptor attenuates acute responsiveness of delta-opioid receptors. Mol Pharmacol 1997; 51: 583–7PubMed
330.
go back to reference Yukhananov RYu, Larson AA. Morphine modulates excitatory amino acid-induced activity in the mouse spinal cord: short-term effects on N-methyl-D-aspartate (NMDA) and long-term effects on kainic acid. Brain Res 1994; 646: 194–200PubMedCrossRef Yukhananov RYu, Larson AA. Morphine modulates excitatory amino acid-induced activity in the mouse spinal cord: short-term effects on N-methyl-D-aspartate (NMDA) and long-term effects on kainic acid. Brain Res 1994; 646: 194–200PubMedCrossRef
331.
go back to reference DeLander GE, Wahl JJ. Morphine (intracerebroventricular) activates spinal systems to inhibit behavior induced by putative pain neurotransmitters. J Pharmacol Exp Ther 1989; 251: 1090–5PubMed DeLander GE, Wahl JJ. Morphine (intracerebroventricular) activates spinal systems to inhibit behavior induced by putative pain neurotransmitters. J Pharmacol Exp Ther 1989; 251: 1090–5PubMed
332.
go back to reference Lutfy K, Doan P, Nguyen M, et al. ACEA-1328, an NMDA receptor antagonist, increased the potency of morphine and U50,488H in the tail flick test in mice. Pharmacol Res 1998; 38: 453–60PubMedCrossRef Lutfy K, Doan P, Nguyen M, et al. ACEA-1328, an NMDA receptor antagonist, increased the potency of morphine and U50,488H in the tail flick test in mice. Pharmacol Res 1998; 38: 453–60PubMedCrossRef
333.
go back to reference McCarthy RJ, Kroin JS, Tuman KJ, et al. Antinociceptive potentiation and attenuation of tolerance by intrathecal co-infusion of magnesium sulfate and morphine in rats. Anesth Analg 1998; 86: 830–6PubMed McCarthy RJ, Kroin JS, Tuman KJ, et al. Antinociceptive potentiation and attenuation of tolerance by intrathecal co-infusion of magnesium sulfate and morphine in rats. Anesth Analg 1998; 86: 830–6PubMed
334.
go back to reference Bhargava HN. Enhancement of morphine actions in morphinenaïve and morphine-tolerant mice by LY235959, a competitive antagonist of the NMDA receptor. Gen Pharmacol 1997; 28: 61–4PubMedCrossRef Bhargava HN. Enhancement of morphine actions in morphinenaïve and morphine-tolerant mice by LY235959, a competitive antagonist of the NMDA receptor. Gen Pharmacol 1997; 28: 61–4PubMedCrossRef
335.
go back to reference Grass S, Hoffman O, Xu XJ, et al. N-methyl-D-aspartate receptor antagonists potentiate morphine’s antinociceptive effect in the rat. Acta Physiol Scand 1996; 158: 269–73PubMedCrossRef Grass S, Hoffman O, Xu XJ, et al. N-methyl-D-aspartate receptor antagonists potentiate morphine’s antinociceptive effect in the rat. Acta Physiol Scand 1996; 158: 269–73PubMedCrossRef
336.
go back to reference Advokat C, Rhein FQ. Potentiation of morphine-induced anti-nociception in acute spinal rats by the NMDA antagonist dextrorphan. Brain Res 1995; 699: 157–60PubMedCrossRef Advokat C, Rhein FQ. Potentiation of morphine-induced anti-nociception in acute spinal rats by the NMDA antagonist dextrorphan. Brain Res 1995; 699: 157–60PubMedCrossRef
337.
go back to reference Suh HW, Song DK, Choi YS, et al. Multiplicative interaction between intrathecally and intracerebroventricularly administered morphine for antinociception in the mouse: involvement of supraspinal NMDA but not non-NMD A receptors. Life Sci 1995:56:PL181–5PubMedCrossRef Suh HW, Song DK, Choi YS, et al. Multiplicative interaction between intrathecally and intracerebroventricularly administered morphine for antinociception in the mouse: involvement of supraspinal NMDA but not non-NMD A receptors. Life Sci 1995:56:PL181–5PubMedCrossRef
338.
go back to reference Hoffmann O, Wiesenfeld-Hallin Z. Dextromethorphan potentiates morphine antinociception, but dos not reverse tolerance in rats. Neuroreport 1996; 7: 838–40PubMedCrossRef Hoffmann O, Wiesenfeld-Hallin Z. Dextromethorphan potentiates morphine antinociception, but dos not reverse tolerance in rats. Neuroreport 1996; 7: 838–40PubMedCrossRef
339.
go back to reference Bhargava HN, Matwyshyn GA, Gudehithlu KP. Effects of acute and chronic administration of dizocilpine on the pharmacological responses to U-50,488H and brain and spinal cord kappa-opioid receptors in the rat. Pharmacol 1995; 51: 323–30CrossRef Bhargava HN, Matwyshyn GA, Gudehithlu KP. Effects of acute and chronic administration of dizocilpine on the pharmacological responses to U-50,488H and brain and spinal cord kappa-opioid receptors in the rat. Pharmacol 1995; 51: 323–30CrossRef
340.
go back to reference Bespalov A, Kudryashova M, Zvartau E. Prolongation of morphine analgesia by competitive NMDA receptor antagonist D-CPPene (SDZ EAA 494) in rats. Eur J Pharmacol 1998; 351: 299–305PubMedCrossRef Bespalov A, Kudryashova M, Zvartau E. Prolongation of morphine analgesia by competitive NMDA receptor antagonist D-CPPene (SDZ EAA 494) in rats. Eur J Pharmacol 1998; 351: 299–305PubMedCrossRef
341.
go back to reference Spinella M, Znamensky V, Moroz M, et al. Actions of NMDA and cholinergic receptor antagonists in the rostral ventromedial medulla upon beta-endorphin analgesia elicited from the ventrolateral periaqueductal gray. Brain Res 1999;829: 151–9PubMedCrossRef Spinella M, Znamensky V, Moroz M, et al. Actions of NMDA and cholinergic receptor antagonists in the rostral ventromedial medulla upon beta-endorphin analgesia elicited from the ventrolateral periaqueductal gray. Brain Res 1999;829: 151–9PubMedCrossRef
342.
go back to reference Spinella M, Cooper ML, Bodnar RJ. Excitatory amino acid antagonists in the rostral ventromedial medulla inhibit mesencephalic morphine analgesia in rats. Pain 1996; 64: 545–52PubMedCrossRef Spinella M, Cooper ML, Bodnar RJ. Excitatory amino acid antagonists in the rostral ventromedial medulla inhibit mesencephalic morphine analgesia in rats. Pain 1996; 64: 545–52PubMedCrossRef
343.
go back to reference Suh HW, Choi YS, Yoo JS, et al. Non-NMDA receptor antagonist attenuates antinociception induced by morphine but not beta-endorphin, D-Pen2-D-Pen5-enkephalin, and U50,488H administered intracerebroventricularly. Neuropeptides 1995; 28: 125–9PubMedCrossRef Suh HW, Choi YS, Yoo JS, et al. Non-NMDA receptor antagonist attenuates antinociception induced by morphine but not beta-endorphin, D-Pen2-D-Pen5-enkephalin, and U50,488H administered intracerebroventricularly. Neuropeptides 1995; 28: 125–9PubMedCrossRef
344.
go back to reference Koyuncuoglu H, Güngör M, Sagduyu H, et al. The antagonistic effects of aspartic acid on some effects of morphine on rats. Eur J Pharmacol 1974; 27: 148–50PubMedCrossRef Koyuncuoglu H, Güngör M, Sagduyu H, et al. The antagonistic effects of aspartic acid on some effects of morphine on rats. Eur J Pharmacol 1974; 27: 148–50PubMedCrossRef
345.
go back to reference Koyuncuoglu H, Güngör M, Sagduyu H, et al. Antagonizing effect of aspartic acid on the development of physical dependence on and tolerance to morphine in the rat. Drug Res 1977; 27: 1676–9 Koyuncuoglu H, Güngör M, Sagduyu H, et al. Antagonizing effect of aspartic acid on the development of physical dependence on and tolerance to morphine in the rat. Drug Res 1977; 27: 1676–9
346.
go back to reference Koyuncuoglu H, Güngör M, Sagduyu H, et al. Suppression by ketamine and dextromethorphan of precipitated abstinence syndrome in rats. Pharmacol Biochem Behav 1990; 35: 829–32PubMedCrossRef Koyuncuoglu H, Güngör M, Sagduyu H, et al. Suppression by ketamine and dextromethorphan of precipitated abstinence syndrome in rats. Pharmacol Biochem Behav 1990; 35: 829–32PubMedCrossRef
347.
go back to reference Trujillo KA, Akil H. Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science 1991; 251: 85–7PubMedCrossRef Trujillo KA, Akil H. Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science 1991; 251: 85–7PubMedCrossRef
348.
go back to reference Trujillo KA, Akil H. Inhibition of opiate tolerance by non-competitive N-methyl-D-aspartate receptor antagonists. Brain Res 1994; 633: 178–88PubMedCrossRef Trujillo KA, Akil H. Inhibition of opiate tolerance by non-competitive N-methyl-D-aspartate receptor antagonists. Brain Res 1994; 633: 178–88PubMedCrossRef
349.
go back to reference Marek P, Ben-Eliyahu S, Gold M, et al. Excitatory amino acid antagonists (kynurenic acid and MK-801 attenuate the development of morphine tolerance in rats. Brain Res 1991; 547: 77–81PubMedCrossRef Marek P, Ben-Eliyahu S, Gold M, et al. Excitatory amino acid antagonists (kynurenic acid and MK-801 attenuate the development of morphine tolerance in rats. Brain Res 1991; 547: 77–81PubMedCrossRef
350.
go back to reference Marek P, Ben-Eliyahu S, Vaccarino AL, et al. Delayed application of MK-801 attenuates development of morphine tolerance in rats. Brain Res 1991; 558: 163–5PubMedCrossRef Marek P, Ben-Eliyahu S, Vaccarino AL, et al. Delayed application of MK-801 attenuates development of morphine tolerance in rats. Brain Res 1991; 558: 163–5PubMedCrossRef
351.
go back to reference Gonzalez P, Cabello P, Germany A, et al. Decrease of tolerance to, and physical dependence on morphine by, glutamate receptor antagonists. Eur J Pharmacol 1997; 332: 257–62PubMedCrossRef Gonzalez P, Cabello P, Germany A, et al. Decrease of tolerance to, and physical dependence on morphine by, glutamate receptor antagonists. Eur J Pharmacol 1997; 332: 257–62PubMedCrossRef
352.
go back to reference Makimura M, Sugimoto H, Shinomiya K, et al. Inhibitory effect of the NMDA receptor antagonist, dizocilpine (MK-801), on the development of morphine dependence. J Toxicol Sci 1996; 21: 135–41PubMedCrossRef Makimura M, Sugimoto H, Shinomiya K, et al. Inhibitory effect of the NMDA receptor antagonist, dizocilpine (MK-801), on the development of morphine dependence. J Toxicol Sci 1996; 21: 135–41PubMedCrossRef
353.
go back to reference 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 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
354.
go back to reference Bhargava HN. Non-competitive antagonism of N-methyl-D-aspartate receptor inhibits tolerance to the analgesic action of U-50,488H, a kappa-opiate receptor agonist in the rat. Gen Pharmacol 1995; 26: 1055–60PubMedCrossRef Bhargava HN. Non-competitive antagonism of N-methyl-D-aspartate receptor inhibits tolerance to the analgesic action of U-50,488H, a kappa-opiate receptor agonist in the rat. Gen Pharmacol 1995; 26: 1055–60PubMedCrossRef
355.
go back to reference Elliott K, Hynansky A, Inturrisi CE. Dextromethorphan attenuates and reverses analgesic tolerance to morphine. Pain 1994; 59: 361–8PubMedCrossRef Elliott K, Hynansky A, Inturrisi CE. Dextromethorphan attenuates and reverses analgesic tolerance to morphine. Pain 1994; 59: 361–8PubMedCrossRef
356.
go back to reference Mao J, Price DD, Lu J, et al. Antinociceptive tolerance to the mu-opioid agonist DAMGO is dose-dependently reduced by MK-801 in rats. Neurosci Lett 1998; 250: 193–6PubMedCrossRef Mao J, Price DD, Lu J, et al. Antinociceptive tolerance to the mu-opioid agonist DAMGO is dose-dependently reduced by MK-801 in rats. Neurosci Lett 1998; 250: 193–6PubMedCrossRef
357.
go back to reference Manning BH, Mao J, Frenk H, et al. Continuous co-administration of dextromethorphan or MK-801 with morphine: attenuation of morphine dependence and naloxone-reversible attenuation of morphine tolerance. Pain 1996; 67: 79–88PubMedCrossRef Manning BH, Mao J, Frenk H, et al. Continuous co-administration of dextromethorphan or MK-801 with morphine: attenuation of morphine dependence and naloxone-reversible attenuation of morphine tolerance. Pain 1996; 67: 79–88PubMedCrossRef
358.
go back to reference Lutfy K, Shen KZ, Woodward RM, et al. Inhibition of morphine tolerance by NMDA receptor antagonists in the formalin test. Brain Res 1996; 731: 171–81PubMedCrossRef Lutfy K, Shen KZ, Woodward RM, et al. Inhibition of morphine tolerance by NMDA receptor antagonists in the formalin test. Brain Res 1996; 731: 171–81PubMedCrossRef
359.
go back to reference Lutfy K, Shen KZ, Kwon IS, et al. blockade of morphine tolerance by ACEA-1328, a novel NMDA receptor/glycine site antagonist. Eur J Pharmacol 1995; 273: 187–9PubMedCrossRef Lutfy K, Shen KZ, Kwon IS, et al. blockade of morphine tolerance by ACEA-1328, a novel NMDA receptor/glycine site antagonist. Eur J Pharmacol 1995; 273: 187–9PubMedCrossRef
360.
go back to reference Popik P, Skolnick P. The NMDA antagonist memantine blocks the expression and maintenance of morphine dependence. Pharmacol Biochem Behav 1996; 53: 791–7PubMedCrossRef Popik P, Skolnick P. The NMDA antagonist memantine blocks the expression and maintenance of morphine dependence. Pharmacol Biochem Behav 1996; 53: 791–7PubMedCrossRef
361.
go back to reference Zhao GM, Bhargava HN. Effect of antagonism of the NMDA receptor on tolerance to [D-Pen2,D-Pen5]enkephalin, a delta 1-opioid receptor agonist. Peptides 1996; 17: 233–6PubMedCrossRef Zhao GM, Bhargava HN. Effect of antagonism of the NMDA receptor on tolerance to [D-Pen2,D-Pen5]enkephalin, a delta 1-opioid receptor agonist. Peptides 1996; 17: 233–6PubMedCrossRef
362.
go back to reference Fundytus ME, Coderre TJ. Effect of activity at metabotropic, as well as ionotropic (NMDA), glutamate receptors on morphine dependence. Br J Pharmacol 1994; 113: 1215–20PubMedCrossRef Fundytus ME, Coderre TJ. Effect of activity at metabotropic, as well as ionotropic (NMDA), glutamate receptors on morphine dependence. Br J Pharmacol 1994; 113: 1215–20PubMedCrossRef
363.
go back to reference Allen RM, Dykstra LA. The competitive NMDA receptor antagonist LY235959 modulates the progression of morphine tolerance in rats. Psychopharmacology(Berl) 1999; 142: 209–14CrossRef Allen RM, Dykstra LA. The competitive NMDA receptor antagonist LY235959 modulates the progression of morphine tolerance in rats. Psychopharmacology(Berl) 1999; 142: 209–14CrossRef
364.
go back to reference Kolesnikov Y, Pasternak GW. Topical opioids in mice: analgesia and reversal of tolerance by a topical N-methyl-D-aspartate antagonist. J Pharmacol Exp Ther 1999; 290: 247–52PubMed Kolesnikov Y, Pasternak GW. Topical opioids in mice: analgesia and reversal of tolerance by a topical N-methyl-D-aspartate antagonist. J Pharmacol Exp Ther 1999; 290: 247–52PubMed
365.
go back to reference Wong CS, Cherng CS, Luk HN, et al. Effects of NMDA receptor antagonists on inhibition of morphine tolerance in rats: binding at mu-opioid receptors. Eur J Pharmacol 1996; 297: 27–33PubMedCrossRef Wong CS, Cherng CS, Luk HN, et al. Effects of NMDA receptor antagonists on inhibition of morphine tolerance in rats: binding at mu-opioid receptors. Eur J Pharmacol 1996; 297: 27–33PubMedCrossRef
366.
go back to reference Wang L, Milne B, Jhamandas K. Involvement of excitatory amino acid pathways in the expression of precipitated opioid withdrawal in the rostral ventrolateral medulla: an in vivo voltammetric study. Brain Res 1995; 697: 130–42PubMedCrossRef Wang L, Milne B, Jhamandas K. Involvement of excitatory amino acid pathways in the expression of precipitated opioid withdrawal in the rostral ventrolateral medulla: an in vivo voltammetric study. Brain Res 1995; 697: 130–42PubMedCrossRef
367.
go back to reference Popik P, Mamczarz J, Fraczek M, et al. Inhibition of reinforcing effects of morphine and naloxone precipitated opioid withdrawal by novel glycine site and uncompetitive NMDA receptor antagonists. Neuropharmacology 1998; 37: 1033–42PubMedCrossRef Popik P, Mamczarz J, Fraczek M, et al. Inhibition of reinforcing effects of morphine and naloxone precipitated opioid withdrawal by novel glycine site and uncompetitive NMDA receptor antagonists. Neuropharmacology 1998; 37: 1033–42PubMedCrossRef
368.
go back to reference Kosten TA, DeCaprio JL, Rosen MI. The severity of naloxone precipitated withdrawal is attenuated by felbamate, a possible glycine antagonist. Neuropsychopharmacology 1995; 13: 323–33PubMedCrossRef Kosten TA, DeCaprio JL, Rosen MI. The severity of naloxone precipitated withdrawal is attenuated by felbamate, a possible glycine antagonist. Neuropsychopharmacology 1995; 13: 323–33PubMedCrossRef
369.
go back to reference Zhu H, Ho IK. NMDA-R1 antisense oligonucleotide attenuates withdrawal signs from morphine. Eur J Pharmacol 1998; 52: 151–6CrossRef Zhu H, Ho IK. NMDA-R1 antisense oligonucleotide attenuates withdrawal signs from morphine. Eur J Pharmacol 1998; 52: 151–6CrossRef
370.
go back to reference Bilsky EJ, Inturrisi CE, Sadee W, et al. Competitive and non-competitive NMDA antagonists block the development of anti-nociceptive tolerance to morphine, but not to selective mu or delta opioid agonists in mice. Pain 1996; 68: 229–37PubMedCrossRef Bilsky EJ, Inturrisi CE, Sadee W, et al. Competitive and non-competitive NMDA antagonists block the development of anti-nociceptive tolerance to morphine, but not to selective mu or delta opioid agonists in mice. Pain 1996; 68: 229–37PubMedCrossRef
371.
go back to reference Elliott K, Kest B, Man A, et al. N-methyl-D-aspartate (NMDA) receptors, mu and kappa opioid tolerance, and perspectives on new analgesic drug development. Neuropsychopharmacology 1995; 13: 347–56PubMedCrossRef Elliott K, Kest B, Man A, et al. N-methyl-D-aspartate (NMDA) receptors, mu and kappa opioid tolerance, and perspectives on new analgesic drug development. Neuropsychopharmacology 1995; 13: 347–56PubMedCrossRef
372.
go back to reference Pasternak GW, Kolesnikov YA, Babey AM. Perspectives on the N-methyl-D-aspartate/nitric oxide cascade and opioid tolerance. Neuropsychopharmacology 1995; 13: 309–13PubMedCrossRef Pasternak GW, Kolesnikov YA, Babey AM. Perspectives on the N-methyl-D-aspartate/nitric oxide cascade and opioid tolerance. Neuropsychopharmacology 1995; 13: 309–13PubMedCrossRef
373.
go back to reference Bhargava HN, Zhao GM. Effects of competitive and noncompetitive antagonists of the N-methyl-D-aspartate receptor on the analgesic action of delta 1- and delta-2-opioid receptor agonists in mice. Br J Pharmacol 1996; 119: 1586–90CrossRef Bhargava HN, Zhao GM. Effects of competitive and noncompetitive antagonists of the N-methyl-D-aspartate receptor on the analgesic action of delta 1- and delta-2-opioid receptor agonists in mice. Br J Pharmacol 1996; 119: 1586–90CrossRef
374.
go back to reference Bhargava HN, Zhao GM. Effects of N-methyl-D-aspartate receptor antagonists on the analgesia and tolerance to D-Ala2, Glu4 deltorphin II, a delta 2-opioid receptor agonist in mice. Brain Res 1996; 719: 56–61PubMedCrossRef Bhargava HN, Zhao GM. Effects of N-methyl-D-aspartate receptor antagonists on the analgesia and tolerance to D-Ala2, Glu4 deltorphin II, a delta 2-opioid receptor agonist in mice. Brain Res 1996; 719: 56–61PubMedCrossRef
375.
go back to reference Bell JA, Beglan CL. Co-treatment with MK-801 potentiates naloxone-precipitated morphine withdrawal in the isolated spinal cord of the neonatal rat. Eur J Pharmacol 1995; 294: 297–301PubMedCrossRef Bell JA, Beglan CL. Co-treatment with MK-801 potentiates naloxone-precipitated morphine withdrawal in the isolated spinal cord of the neonatal rat. Eur J Pharmacol 1995; 294: 297–301PubMedCrossRef
376.
go back to reference Zhu H, Jang CG, Ma T, et al. Region specific expression of NMDA receptor NR1 subunit mRNA in hypothalamus and pons following chronic morphine treatment. Eur J Pharmacol 1999; 365: 47–54PubMedCrossRef Zhu H, Jang CG, Ma T, et al. Region specific expression of NMDA receptor NR1 subunit mRNA in hypothalamus and pons following chronic morphine treatment. Eur J Pharmacol 1999; 365: 47–54PubMedCrossRef
377.
go back to reference Bhargava HN, Kumar S. Modification of the binding of [3H]MK-801 to brain regions and spinal cord of rats treated chronically with U-50,488H, a kappa-opioid receptor agonist. Brain Res 1997; 749: 347–50PubMedCrossRef Bhargava HN, Kumar S. Modification of the binding of [3H]MK-801 to brain regions and spinal cord of rats treated chronically with U-50,488H, a kappa-opioid receptor agonist. Brain Res 1997; 749: 347–50PubMedCrossRef
378.
go back to reference Gudehithlu KP, Bhargava HN. Differential binding of [3H]MK-801 to brain regions and spinal cord of mice treated chronically with morphine. Gen Pharmacol 1996; 27: 91–4PubMedCrossRef Gudehithlu KP, Bhargava HN. Differential binding of [3H]MK-801 to brain regions and spinal cord of mice treated chronically with morphine. Gen Pharmacol 1996; 27: 91–4PubMedCrossRef
379.
go back to reference Bhargava HN, Reddy PL, Gudehithlu KP. Down-regulation of N-methyl-D-aspartate (NMDA) receptors of brain regions and spinal cord of rats treated chronically with morphine. Gen Pharmacol 1995; 26: 131–6PubMedCrossRef Bhargava HN, Reddy PL, Gudehithlu KP. Down-regulation of N-methyl-D-aspartate (NMDA) receptors of brain regions and spinal cord of rats treated chronically with morphine. Gen Pharmacol 1995; 26: 131–6PubMedCrossRef
380.
go back to reference Kolaj M, Randic M. mu-Opioid receptor-mediated reduction of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-activated current in dorsal horn neurons. Neurosci Lett 1996; 204: 133–7PubMedCrossRef Kolaj M, Randic M. mu-Opioid receptor-mediated reduction of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-activated current in dorsal horn neurons. Neurosci Lett 1996; 204: 133–7PubMedCrossRef
381.
go back to reference Nishiyama T, Yaksh TL, Weber E. Effects of intrathecal NMDA and non-NMDA antagonists on acute thermal nociception and their interaction with morphine. Anesthesiology 1998; 89: 715–22PubMedCrossRef Nishiyama T, Yaksh TL, Weber E. Effects of intrathecal NMDA and non-NMDA antagonists on acute thermal nociception and their interaction with morphine. Anesthesiology 1998; 89: 715–22PubMedCrossRef
382.
go back to reference Van Praag H, Frenk H. The role of glutamate in opiate descending inhibition of nociceptive spinal reflexes. Brain Res 1990; 524: 101–5PubMedCrossRef Van Praag H, Frenk H. The role of glutamate in opiate descending inhibition of nociceptive spinal reflexes. Brain Res 1990; 524: 101–5PubMedCrossRef
383.
go back to reference Taylor JR, Punch LJ, Elsworth JD. A comparison of the effects of clonidine and CNQX infusion into the locus coeruleus and the amygdala on naloxone-precipitated opiate withdrawal in the rat. Psychopharmacology (Berl) 1998; 138: 133–42CrossRef Taylor JR, Punch LJ, Elsworth JD. A comparison of the effects of clonidine and CNQX infusion into the locus coeruleus and the amygdala on naloxone-precipitated opiate withdrawal in the rat. Psychopharmacology (Berl) 1998; 138: 133–42CrossRef
384.
go back to reference Rasmussen K, Kendrick WT, Kogan JH, et al. A selective AMPA antagonist, LY293558, suppresses morphine withdrawal-induced activation of locus coeruleus neurons and behavioral signs of morphine withdrawal. Neuropsychopharmacology 1996; 15: 497–505PubMedCrossRef Rasmussen K, Kendrick WT, Kogan JH, et al. A selective AMPA antagonist, LY293558, suppresses morphine withdrawal-induced activation of locus coeruleus neurons and behavioral signs of morphine withdrawal. Neuropsychopharmacology 1996; 15: 497–505PubMedCrossRef
385.
go back to reference McLemore GL, Kest B, Inturrisi CE. The effects of LY293558, an AMPA receptor antagonist, on acute and chronic morphine dependence. Brain Res 1997; 778: 120–6PubMedCrossRef McLemore GL, Kest B, Inturrisi CE. The effects of LY293558, an AMPA receptor antagonist, on acute and chronic morphine dependence. Brain Res 1997; 778: 120–6PubMedCrossRef
386.
go back to reference Kest B, McLemore G, Kao B, et al. The competitive alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor antagonist LY293558 attenuates and reverses analgesic tolerance to morphine but not to delta or kappa opioids. J Pharmacol Exp Ther 1997; 283: 1249–55PubMed Kest B, McLemore G, Kao B, et al. The competitive alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor antagonist LY293558 attenuates and reverses analgesic tolerance to morphine but not to delta or kappa opioids. J Pharmacol Exp Ther 1997; 283: 1249–55PubMed
387.
go back to reference Fitzgerald LW, Ortiz J, Hamedani AG, et al. Drugs of abuse and stress increase the expression of GluR1 and NMDAR1 glutamate receptor subunits in the rat ventral tegmental area: common adaptations among cross-sensitizing agents. J Neurosci 1996; 16: 274–82PubMed Fitzgerald LW, Ortiz J, Hamedani AG, et al. Drugs of abuse and stress increase the expression of GluR1 and NMDAR1 glutamate receptor subunits in the rat ventral tegmental area: common adaptations among cross-sensitizing agents. J Neurosci 1996; 16: 274–82PubMed
388.
go back to reference Fundytus ME, Coderre TJ. Opioid tolerance and dependence: a new model highlighting the role of metabotropic glutamate receptors. Pain Forum 1999; 8(1): 3–13CrossRef Fundytus ME, Coderre TJ. Opioid tolerance and dependence: a new model highlighting the role of metabotropic glutamate receptors. Pain Forum 1999; 8(1): 3–13CrossRef
389.
go back to reference Fundytus ME, Coderre TJ. mGluRs and opioid dependence: a further examination of the mechanisms. Pain Forum 1999; 8(2): 59–63CrossRef Fundytus ME, Coderre TJ. mGluRs and opioid dependence: a further examination of the mechanisms. Pain Forum 1999; 8(2): 59–63CrossRef
390.
go back to reference Fundytus ME, Ritchie J, Coderre TJ. Attenuation of morphine withdrawal symptoms by subtype selective metabotropic glutamate receptor antagonists. Br J Pharmacol 1997; 120: 1015–20PubMedCrossRef Fundytus ME, Ritchie J, Coderre TJ. Attenuation of morphine withdrawal symptoms by subtype selective metabotropic glutamate receptor antagonists. Br J Pharmacol 1997; 120: 1015–20PubMedCrossRef
391.
go back to reference Vandergriff J, Rasmussen K. The selective mGlu2/3 receptor agonist LY354740 attenuates morphine-withdrawal-induced activation of locus coeruleus neurons and behavioral signs of morphine withdrawal. Neuropharmacology 1999; 38: 217–22PubMedCrossRef Vandergriff J, Rasmussen K. The selective mGlu2/3 receptor agonist LY354740 attenuates morphine-withdrawal-induced activation of locus coeruleus neurons and behavioral signs of morphine withdrawal. Neuropharmacology 1999; 38: 217–22PubMedCrossRef
392.
go back to reference Fundytus ME, Coderre TJ. Chronic inhibition of intracellular Ca2+ release or PKC activation significantly reduces the development of morphine dependence. Eur J Pharmacol 1996; 300: 173–81PubMedCrossRef Fundytus ME, Coderre TJ. Chronic inhibition of intracellular Ca2+ release or PKC activation significantly reduces the development of morphine dependence. Eur J Pharmacol 1996; 300: 173–81PubMedCrossRef
393.
go back to reference Mao J, Price DD, Mayer DJ. Thermal hyperalgesia in association with the development of morphine tolerance in rats: roles of excitatory amino acid receptors and protein kinase C. J Neurosci 1994; 14: 2301–12PubMed Mao J, Price DD, Mayer DJ. Thermal hyperalgesia in association with the development of morphine tolerance in rats: roles of excitatory amino acid receptors and protein kinase C. J Neurosci 1994; 14: 2301–12PubMed
394.
go back to reference Mayer DJ, Mao J, Price DD. The development of morphine tolerance and dependence is associated with translocation of protein kinase C. Pain 1995; 61: 365–74PubMedCrossRef Mayer DJ, Mao J, Price DD. The development of morphine tolerance and dependence is associated with translocation of protein kinase C. Pain 1995; 61: 365–74PubMedCrossRef
395.
go back to reference Maldonado R, Valverde O, Garbay C, et al. Protein kinases in the locus coeruleus and periaqueductal gray matter are involved in the expression of opiate withdrawal. Naunyn-Schmiedeberg’s Arch Pharmacol 1995; 352: 565–75 Maldonado R, Valverde O, Garbay C, et al. Protein kinases in the locus coeruleus and periaqueductal gray matter are involved in the expression of opiate withdrawal. Naunyn-Schmiedeberg’s Arch Pharmacol 1995; 352: 565–75
396.
go back to reference Narita M, Feng Y, Makimura M, et al. A protein kinase inhibitor, H-7, inhibits the development of tolerance to opioid antinociception. Eur J Pharmacol 1994; 271: 543–5PubMedCrossRef Narita M, Feng Y, Makimura M, et al. A protein kinase inhibitor, H-7, inhibits the development of tolerance to opioid antinociception. Eur J Pharmacol 1994; 271: 543–5PubMedCrossRef
397.
go back to reference Mao J, Price DD, Phillips LL, et al. 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: 257–67PubMedCrossRef Mao J, Price DD, Phillips LL, et al. 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: 257–67PubMedCrossRef
398.
go back to reference Kramer HK, Simon EJ. Role of protein kinase C (PKC) in agonist-induced mu-opioid receptor down-regulation: II. Activation and involvement of the alpha, epsilon and zeta isoforms of PKC. J Neurochem 1999; 72: 594–604 Kramer HK, Simon EJ. Role of protein kinase C (PKC) in agonist-induced mu-opioid receptor down-regulation: II. Activation and involvement of the alpha, epsilon and zeta isoforms of PKC. J Neurochem 1999; 72: 594–604
399.
go back to reference Fundytus ME, Coderre TJ. Attenuation of precipitated morphine withdrawal symptoms by acute i.c.v. administration of a group II mGluR agonist. Br J Pharmacol 1997; 121: 511–4 Fundytus ME, Coderre TJ. Attenuation of precipitated morphine withdrawal symptoms by acute i.c.v. administration of a group II mGluR agonist. Br J Pharmacol 1997; 121: 511–4
400.
go back to reference Fundytus ME, Dray A, Henry JL, et al. An antisense oligonucleotide targeting mGluR1 restores opioid sensitivity in neuropathic rats [abstract]. INRC’98 (International Narcotics Research Conference) 1998; 42 Fundytus ME, Dray A, Henry JL, et al. An antisense oligonucleotide targeting mGluR1 restores opioid sensitivity in neuropathic rats [abstract]. INRC’98 (International Narcotics Research Conference) 1998; 42
401.
go back to reference Ossipov MH, Lopez Y, Nichols ML, et al. Inhibition by spinal morphine of the tail-flick response is attenuated in rats with nerve ligation injury. Neurosci Lett 1995; 199: 83–6PubMedCrossRef Ossipov MH, Lopez Y, Nichols ML, et al. Inhibition by spinal morphine of the tail-flick response is attenuated in rats with nerve ligation injury. Neurosci Lett 1995; 199: 83–6PubMedCrossRef
402.
go back to reference Ossipov MH, Lopez Y, Nichols ML, et al. The loss of anti-nociceptive efficacy of spinal morphine in rats with nerve ligation injury is prevented by reducing spinal afferent drive. Neurosci Lett 1995; 199: 87–90PubMedCrossRef Ossipov MH, Lopez Y, Nichols ML, et al. The loss of anti-nociceptive efficacy of spinal morphine in rats with nerve ligation injury is prevented by reducing spinal afferent drive. Neurosci Lett 1995; 199: 87–90PubMedCrossRef
403.
go back to reference Mao J, Price DD, Mayer DJ. Experimental mononeuropathy reduces the antinociceptive effects of morphine: implications for common intracellular mechanisms involved in morphine tolerance and neuropathic pain. Pain 1995; 61: 353–64PubMedCrossRef Mao J, Price DD, Mayer DJ. Experimental mononeuropathy reduces the antinociceptive effects of morphine: implications for common intracellular mechanisms involved in morphine tolerance and neuropathic pain. Pain 1995; 61: 353–64PubMedCrossRef
404.
go back to reference Cherney NI, Thaler HT, Friedlander-Klar H, et al. Opioid responsiveness of cancer pain syndromes caused by neuropathic or nociceptive mechanisms: a combined analysis of controlled, single-dose studies. Neurology 1994; 44: 857–61CrossRef Cherney NI, Thaler HT, Friedlander-Klar H, et al. Opioid responsiveness of cancer pain syndromes caused by neuropathic or nociceptive mechanisms: a combined analysis of controlled, single-dose studies. Neurology 1994; 44: 857–61CrossRef
405.
go back to reference MacDonald N. Opiate resistant pain: a therapeutic dilemma. Recent Results Cancer Res 1991; 121: 24–35PubMedCrossRef MacDonald N. Opiate resistant pain: a therapeutic dilemma. Recent Results Cancer Res 1991; 121: 24–35PubMedCrossRef
406.
go back to reference McQuay HJ, Jadad AR, Carroll D, et al. Opioid sensitivity of chronic pain: a patient-controlled analgesia method. Anesthesia 1992; 47: 757–67CrossRef McQuay HJ, Jadad AR, Carroll D, et al. Opioid sensitivity of chronic pain: a patient-controlled analgesia method. Anesthesia 1992; 47: 757–67CrossRef
407.
go back to reference Christensen D, Idanpaan-Heikkila JJ, Guilbaud G, et al. The antinociceptive effect of combined systemic administration of morphine and the glycine/NMDA receptor antagonist, (+)- HA966 in a rat model of peripheral neuropathy. Br J Pharmacol 1998; 125: 1641–50PubMedCrossRef Christensen D, Idanpaan-Heikkila JJ, Guilbaud G, et al. The antinociceptive effect of combined systemic administration of morphine and the glycine/NMDA receptor antagonist, (+)- HA966 in a rat model of peripheral neuropathy. Br J Pharmacol 1998; 125: 1641–50PubMedCrossRef
408.
go back to reference Kauppila T, Xu XJ, Yu W, et al. Dextromethorphan potentiates the effect of morphine in rats with peripheral neuropathy. Neuroreport 1998; 9: 1071–4PubMedCrossRef Kauppila T, Xu XJ, Yu W, et al. Dextromethorphan potentiates the effect of morphine in rats with peripheral neuropathy. Neuroreport 1998; 9: 1071–4PubMedCrossRef
409.
go back to reference Nichols ML, Lopez Y, Ossipov MH, et al. Enhancement of the antiallodynic and antinociceptive efficacy of spinal morphine by antisera to dynorphin A(l–13)or MK-801 in a nerve-ligation model of peripheral neuropathy. Pain 1997; 69: 317–22PubMedCrossRef Nichols ML, Lopez Y, Ossipov MH, et al. Enhancement of the antiallodynic and antinociceptive efficacy of spinal morphine by antisera to dynorphin A(l–13)or MK-801 in a nerve-ligation model of peripheral neuropathy. Pain 1997; 69: 317–22PubMedCrossRef
410.
go back to reference Yung KK. Localization of glutamate receptors in dorsal horn of rat spinal cord. Neuroreport 1998; 9: 1639–44PubMedCrossRef Yung KK. Localization of glutamate receptors in dorsal horn of rat spinal cord. Neuroreport 1998; 9: 1639–44PubMedCrossRef
411.
go back to reference Liu XB. Subcellular distribution of AMPA and NMDA receptor subunit immunoreactivity in ventral posterior and reticular nuclei of rat and cat thalamus. J Comp Neurol 1997; 388: 587–602PubMedCrossRef Liu XB. Subcellular distribution of AMPA and NMDA receptor subunit immunoreactivity in ventral posterior and reticular nuclei of rat and cat thalamus. J Comp Neurol 1997; 388: 587–602PubMedCrossRef
412.
go back to reference Lambert DG, Wojcikiewicz RJH, Safrany ST, et al. Muscarinic receptors, phosphoinositide metabolism and intracellular calcium in neuronal cells. Prog Neuropsychopharmacol Biol Psychiatry 1992; 16: 253–70PubMedCrossRef Lambert DG, Wojcikiewicz RJH, Safrany ST, et al. Muscarinic receptors, phosphoinositide metabolism and intracellular calcium in neuronal cells. Prog Neuropsychopharmacol Biol Psychiatry 1992; 16: 253–70PubMedCrossRef
413.
414.
415.
go back to reference Smart D, Lambert DG. The stimulatory effects of opioids and their possible role in the development of tolerance. Trends Pharmacol Sci 1996; 17: 717–9CrossRef Smart D, Lambert DG. The stimulatory effects of opioids and their possible role in the development of tolerance. Trends Pharmacol Sci 1996; 17: 717–9CrossRef
416.
417.
go back to reference Fan GH, Zhao J, Wu YL, et al. N-methyl-D-aspartate attenuates opioid receptor-mediated G protein activation and this process involves protein kinase C. Mol Pharmacol 1998; 53: 684–90PubMed Fan GH, Zhao J, Wu YL, et al. N-methyl-D-aspartate attenuates opioid receptor-mediated G protein activation and this process involves protein kinase C. Mol Pharmacol 1998; 53: 684–90PubMed
418.
go back to reference Narita M, Mizoguchi H, Kampine JP, et al. Role of protein kinase C in desensitization of spinal delta-opioid-mediated antinociception in the mouse. BrJ Pharmacol 1996; 118: 1829–35CrossRef Narita M, Mizoguchi H, Kampine JP, et al. Role of protein kinase C in desensitization of spinal delta-opioid-mediated antinociception in the mouse. BrJ Pharmacol 1996; 118: 1829–35CrossRef
419.
go back to reference Wang L, Medina VM, Rivera M, et al. Relevance of phosphorylation state to opioid responsiveness in opiate naive and tolerant/dependent tissue. Brain Res 1996; 723: 61–9PubMedCrossRef Wang L, Medina VM, Rivera M, et al. Relevance of phosphorylation state to opioid responsiveness in opiate naive and tolerant/dependent tissue. Brain Res 1996; 723: 61–9PubMedCrossRef
420.
go back to reference Ingram SL, Williams JT. Modulation of the hyperpolarization-activated current (Ih) by cyclic nucleotides in guinea-pig primary afferent neurons. J Physiol (Lond) 1996; 492: 97–106 Ingram SL, Williams JT. Modulation of the hyperpolarization-activated current (Ih) by cyclic nucleotides in guinea-pig primary afferent neurons. J Physiol (Lond) 1996; 492: 97–106
421.
go back to reference Kingston PA, Zufall F, Barnstable CJ. Rat hippocampal neurons express genes for both rod retinal and olfactory cyclic nucleotide-gated channels: novel targets for cAMP/cGMP function. Proc Natl Acad Sci USA 1996; 93: 10440–5PubMedCrossRef Kingston PA, Zufall F, Barnstable CJ. Rat hippocampal neurons express genes for both rod retinal and olfactory cyclic nucleotide-gated channels: novel targets for cAMP/cGMP function. Proc Natl Acad Sci USA 1996; 93: 10440–5PubMedCrossRef
422.
go back to reference Pedarzani P, Storm JF. Protein kinase A-independent modulation of ion channels in the brain by cyclic AMP. Proc Natl Acad Sci USA 1995; 92: 11716–20PubMedCrossRef Pedarzani P, Storm JF. Protein kinase A-independent modulation of ion channels in the brain by cyclic AMP. Proc Natl Acad Sci USA 1995; 92: 11716–20PubMedCrossRef
423.
go back to reference Wiesenfeld-Hallin Z. Combined opioid-NMDA antagonist therapies: what advantages do they offer for the control of pain syndromes? Drugs 1998; 55: 1–4PubMedCrossRef Wiesenfeld-Hallin Z. Combined opioid-NMDA antagonist therapies: what advantages do they offer for the control of pain syndromes? Drugs 1998; 55: 1–4PubMedCrossRef
424.
go back to reference Wong C-S, Cherng C-H, Ho S-T. Clinical applications of excitatory amino acid antagonists in pain management. Acta Anaesthesiol Sin 1995; 33: 227–32PubMed Wong C-S, Cherng C-H, Ho S-T. Clinical applications of excitatory amino acid antagonists in pain management. Acta Anaesthesiol Sin 1995; 33: 227–32PubMed
425.
go back to reference Dickenson AH. NMDA receptor antagonists: interactions with opioids. Acta Anaesth Scand 1997; 41: 112–5PubMedCrossRef Dickenson AH. NMDA receptor antagonists: interactions with opioids. Acta Anaesth Scand 1997; 41: 112–5PubMedCrossRef
426.
427.
go back to reference Sosnowski M. Pain management: physiopathology, future research and endpoints. Support Care Cancer 1993; 1: 79–88PubMedCrossRef Sosnowski M. Pain management: physiopathology, future research and endpoints. Support Care Cancer 1993; 1: 79–88PubMedCrossRef
428.
go back to reference Gebhardt B. Pharmacology and clinical results with peridural and intrathecal administration of ketamine. Anaesthesiology 1994; 43: S34–40 Gebhardt B. Pharmacology and clinical results with peridural and intrathecal administration of ketamine. Anaesthesiology 1994; 43: S34–40
429.
go back to reference Abram SE. Continuous spinal anesthesia for cancer and chronic pain. Reg Anesth 1993; 189: 406–13 Abram SE. Continuous spinal anesthesia for cancer and chronic pain. Reg Anesth 1993; 189: 406–13
430.
go back to reference Hewitt DJ. The use of NMDA-receptor antagonists in the treatment of chronic pain. Clin J Pain 2000; 16(2 Suppl.); S73–9PubMedCrossRef Hewitt DJ. The use of NMDA-receptor antagonists in the treatment of chronic pain. Clin J Pain 2000; 16(2 Suppl.); S73–9PubMedCrossRef
431.
go back to reference Sang CN. NMDA-receptor antagonists in neuropathic pain: experimental methods to clinical trials. J Pain Symptom Manage 2000; 19(1 Suppl.): S21–5PubMedCrossRef Sang CN. NMDA-receptor antagonists in neuropathic pain: experimental methods to clinical trials. J Pain Symptom Manage 2000; 19(1 Suppl.): S21–5PubMedCrossRef
432.
go back to reference Dickenson AH. A cure for wind up: NMDA receptor antagonists as potential analgesics. Trends Pharmacol Sci 1990; 11: 307–9PubMedCrossRef Dickenson AH. A cure for wind up: NMDA receptor antagonists as potential analgesics. Trends Pharmacol Sci 1990; 11: 307–9PubMedCrossRef
433.
go back to reference Eide PK. Wind-up and the NMDA receptor complex from a clinical perspective. Eur J Pain 2000; 4: 5–15PubMedCrossRef Eide PK. Wind-up and the NMDA receptor complex from a clinical perspective. Eur J Pain 2000; 4: 5–15PubMedCrossRef
434.
go back to reference Mercadante S. Ketamine in cancer pain: an update. Palliat Med 1996; 10: 225–30PubMed Mercadante S. Ketamine in cancer pain: an update. Palliat Med 1996; 10: 225–30PubMed
435.
go back to reference Webb J, Kamali F. Analgesic effects of lamotrigine and phenytoin on cold-induced pain: a crossover placebo-controlled study in healthy volunteers. Pain 1998; 76: 357–63PubMedCrossRef Webb J, Kamali F. Analgesic effects of lamotrigine and phenytoin on cold-induced pain: a crossover placebo-controlled study in healthy volunteers. Pain 1998; 76: 357–63PubMedCrossRef
436.
go back to reference Eisenberg E, Alon N, Ishay A, et al. Lamotrigine in the treatment of painful diabetic neuropathy. Eur J Neurol 1998; 5: 167–73PubMedCrossRef Eisenberg E, Alon N, Ishay A, et al. Lamotrigine in the treatment of painful diabetic neuropathy. Eur J Neurol 1998; 5: 167–73PubMedCrossRef
437.
go back to reference Arendt-Nielsen L, Petersen-Felix S, Fischer M, et al. The effect of N-methyl-D-aspartate antagonist (ketamine) on single and repeated nociceptive stimuli- a placebo-controlled experimental human study. Anesth Analg 1995; 81: 63–8PubMed Arendt-Nielsen L, Petersen-Felix S, Fischer M, et al. The effect of N-methyl-D-aspartate antagonist (ketamine) on single and repeated nociceptive stimuli- a placebo-controlled experimental human study. Anesth Analg 1995; 81: 63–8PubMed
438.
go back to reference Arendt-Nielsen L, Nielsen J, Petersen-Felix S, et al. Effect of racemic mixture and the (+S)-isomer of ketamine on temporal and spatial summation of pain. Br J Anaesth 1996; 77: 625–31PubMedCrossRef Arendt-Nielsen L, Nielsen J, Petersen-Felix S, et al. Effect of racemic mixture and the (+S)-isomer of ketamine on temporal and spatial summation of pain. Br J Anaesth 1996; 77: 625–31PubMedCrossRef
439.
go back to reference Klepstad P, Maurset A, Moberg ER, et al. Evidence for a role for NMDA receptors in pain perception. Eur J Pharmacol 1990; 187: 513–8PubMedCrossRef Klepstad P, Maurset A, Moberg ER, et al. Evidence for a role for NMDA receptors in pain perception. Eur J Pharmacol 1990; 187: 513–8PubMedCrossRef
440.
go back to reference Maurset A, Skoglund LA, Hustveit O, et al. Comparison of ketamine and pethidine in experimental and post-operative pain. Pain 1989: 36: 37–41PubMedCrossRef Maurset A, Skoglund LA, Hustveit O, et al. Comparison of ketamine and pethidine in experimental and post-operative pain. Pain 1989: 36: 37–41PubMedCrossRef
441.
go back to reference Ilkjaer S, Petersen KL, Brennum J, et al. Effect of systemic N-methyl-D-aspartate receptor antagonist (ketamine) on primary and secondary hyperalgesia in humans. Br J Anaesth 1996; 76: 829–34PubMedCrossRef Ilkjaer S, Petersen KL, Brennum J, et al. Effect of systemic N-methyl-D-aspartate receptor antagonist (ketamine) on primary and secondary hyperalgesia in humans. Br J Anaesth 1996; 76: 829–34PubMedCrossRef
442.
go back to reference Ilkjaer S, Dirks J, Brennum J, et al. Effect of systemic N-methyl-D-aspartate receptor antagonist (dextromethorphan) on primary and secondary hyperalgesia in humans. Br J Anaesth 1997; 79: 600–5PubMedCrossRef Ilkjaer S, Dirks J, Brennum J, et al. Effect of systemic N-methyl-D-aspartate receptor antagonist (dextromethorphan) on primary and secondary hyperalgesia in humans. Br J Anaesth 1997; 79: 600–5PubMedCrossRef
443.
go back to reference Park KM, Max MB, Robinovitz E, et al. Effects of intravenous ketamine, alfentanil, or placebo on pain, pinprick hyperalgesia, and allodynia produced by intradermal capsaicin in human subjects. Pain 1995; 63: 163–72PubMedCrossRef Park KM, Max MB, Robinovitz E, et al. Effects of intravenous ketamine, alfentanil, or placebo on pain, pinprick hyperalgesia, and allodynia produced by intradermal capsaicin in human subjects. Pain 1995; 63: 163–72PubMedCrossRef
444.
go back to reference Andersen OK, Felsby S, Nicolaisen L, et al. The effect of ketamine on stimulation of primary and secondary hyperalgesic areas induced by capsaicin — a double-blind, placebo-controlled, human experimental study. Pain 1996; 66: 51–62PubMedCrossRef Andersen OK, Felsby S, Nicolaisen L, et al. The effect of ketamine on stimulation of primary and secondary hyperalgesic areas induced by capsaicin — a double-blind, placebo-controlled, human experimental study. Pain 1996; 66: 51–62PubMedCrossRef
445.
go back to reference Price DD, Mao J, Frenk H, et al. The N-methyl-D-aspartate receptor antagonist dextromethorphan selectively reduces temporal summation of second pain in man. Pain 1994; 59: 165–74PubMedCrossRef Price DD, Mao J, Frenk H, et al. The N-methyl-D-aspartate receptor antagonist dextromethorphan selectively reduces temporal summation of second pain in man. Pain 1994; 59: 165–74PubMedCrossRef
446.
go back to reference Mikkelsen S, Ilkjaer S, Brennum J, et al. The effect of naloxone on ketamine-induced effects on hyperalgesia and ketamine-induced side effects in humans. Anesthesiology 1999; 90: 1539–45PubMedCrossRef Mikkelsen S, Ilkjaer S, Brennum J, et al. The effect of naloxone on ketamine-induced effects on hyperalgesia and ketamine-induced side effects in humans. Anesthesiology 1999; 90: 1539–45PubMedCrossRef
447.
go back to reference Warncke T, Stubhaug A, Jørum E. Ketamine, an NMDA receptor antagonist, suppresses spatial and temporal properties of burn-induced secondary hyperalgesia in man: a double-blind, cross-over comparison with morphine and placebo. Pain 1997; 72: 99–106PubMedCrossRef Warncke T, Stubhaug A, Jørum E. Ketamine, an NMDA receptor antagonist, suppresses spatial and temporal properties of burn-induced secondary hyperalgesia in man: a double-blind, cross-over comparison with morphine and placebo. Pain 1997; 72: 99–106PubMedCrossRef
448.
go back to reference Warncke T, Stubhaug A, Jørum E. Preinjury treatment with morphine or ketamine inhibits the development of experimentally induced secondary hyperalgesia in man. Pain 2000; 86: 293–303PubMedCrossRef Warncke T, Stubhaug A, Jørum E. Preinjury treatment with morphine or ketamine inhibits the development of experimentally induced secondary hyperalgesia in man. Pain 2000; 86: 293–303PubMedCrossRef
449.
go back to reference Kauppila T, Gronroos M, Pertovaara A. An attempt to attenuate experimental pain in humans by dextromethorphan, an NMDA receptor antagonist. Pharmacol Biochem Behav 1995; 52: 641–4PubMedCrossRef Kauppila T, Gronroos M, Pertovaara A. An attempt to attenuate experimental pain in humans by dextromethorphan, an NMDA receptor antagonist. Pharmacol Biochem Behav 1995; 52: 641–4PubMedCrossRef
450.
go back to reference Schepelmann K, Schugens MM, Loschman PA, et al. The non-competitive N-methyl-D-aspartate-antagonist memantine does not affect segmental mono- and polysynaptic reflexes in man. Neurosci Lett 1998; 257: 159–61PubMedCrossRef Schepelmann K, Schugens MM, Loschman PA, et al. The non-competitive N-methyl-D-aspartate-antagonist memantine does not affect segmental mono- and polysynaptic reflexes in man. Neurosci Lett 1998; 257: 159–61PubMedCrossRef
451.
go back to reference Kinnman E, Nygards EB, Hansson P. Effects of dextromethorphan in clinical doses on capsaicin-induced ongoing pain and mechanical hypersensitivity. J Pain Symptom Manage 1997; 14: 195–201PubMedCrossRef Kinnman E, Nygards EB, Hansson P. Effects of dextromethorphan in clinical doses on capsaicin-induced ongoing pain and mechanical hypersensitivity. J Pain Symptom Manage 1997; 14: 195–201PubMedCrossRef
452.
go back to reference Rabben T. Effects of the NMDA receptor antagonist ketamine in electrically induced A delta-fiber pain. Methods Find Exp Clin Pharmacol 2000; 22(3): 185–9PubMed Rabben T. Effects of the NMDA receptor antagonist ketamine in electrically induced A delta-fiber pain. Methods Find Exp Clin Pharmacol 2000; 22(3): 185–9PubMed
453.
go back to reference Sethna NF, Liu M, Gracely R, et al. Analgesic and cognitive effects of intravenous ketamine-alfentanil combinations versus either drug alone after intradermal capsaicin in normal subjects. Anesth Analg 1998; 86: 1250–6PubMed Sethna NF, Liu M, Gracely R, et al. Analgesic and cognitive effects of intravenous ketamine-alfentanil combinations versus either drug alone after intradermal capsaicin in normal subjects. Anesth Analg 1998; 86: 1250–6PubMed
454.
go back to reference Eide PK, Stubhaug A. Relief of glossopharyngeal neuralgia by ketamine-induced N-methyl-aspartate receptor blockade. Neurosurgery 1997; 41: 505–8PubMedCrossRef Eide PK, Stubhaug A. Relief of glossopharyngeal neuralgia by ketamine-induced N-methyl-aspartate receptor blockade. Neurosurgery 1997; 41: 505–8PubMedCrossRef
455.
go back to reference Nikolajsen L, Hansen PO, Jensen TS. Oral ketamine therapy in the treatment of postamputation stump pain. Acta Anaesthesiol Scand 1997; 41: 427–9PubMedCrossRef Nikolajsen L, Hansen PO, Jensen TS. Oral ketamine therapy in the treatment of postamputation stump pain. Acta Anaesthesiol Scand 1997; 41: 427–9PubMedCrossRef
456.
go back to reference Stannard CF, Porter GE. Ketamine hydrochloride in the treatment of phantom limb pain. Pain 1993; 54: 227–30PubMedCrossRef Stannard CF, Porter GE. Ketamine hydrochloride in the treatment of phantom limb pain. Pain 1993; 54: 227–30PubMedCrossRef
457.
go back to reference Hoffmann V, Coppejans H, Vercauteren M, et al. Successful treatment of postherpetic neuralgia with oral ketamine. Clin J Pain 1994; 10: 240–2PubMedCrossRef Hoffmann V, Coppejans H, Vercauteren M, et al. Successful treatment of postherpetic neuralgia with oral ketamine. Clin J Pain 1994; 10: 240–2PubMedCrossRef
458.
go back to reference Mercadante S, Lodi F, Sapio M, et al. Long-term ketamine subcutaneous continuous infusion in neuropathic cancer pain. J Pain Symptom Manage 1995; 10: 564–8PubMedCrossRef Mercadante S, Lodi F, Sapio M, et al. Long-term ketamine subcutaneous continuous infusion in neuropathic cancer pain. J Pain Symptom Manage 1995; 10: 564–8PubMedCrossRef
459.
go back to reference Takahashi H, Miyazaki M, Nanbu T, et al. The NMDA-receptor antagonist ketamine abolishes neuropathic pain after epidural administration in a clinical case. Pain 1998; 75: 391–4PubMedCrossRef Takahashi H, Miyazaki M, Nanbu T, et al. The NMDA-receptor antagonist ketamine abolishes neuropathic pain after epidural administration in a clinical case. Pain 1998; 75: 391–4PubMedCrossRef
460.
go back to reference Eisenberg E, Pud D. Can patients with chronic neuropathic pain be cured by acute administration of the NMDA receptor antagonist amantadine? Pain 1998; 74: 337–9PubMedCrossRef Eisenberg E, Pud D. Can patients with chronic neuropathic pain be cured by acute administration of the NMDA receptor antagonist amantadine? Pain 1998; 74: 337–9PubMedCrossRef
461.
go back to reference Persson J, Axelsson G, Hallin RG, et al. Beneficial effects of ketamine in a chronic pain state with allodynia, possibly due to central sensitization. Pain 1995; 60: 217–22PubMedCrossRef Persson J, Axelsson G, Hallin RG, et al. Beneficial effects of ketamine in a chronic pain state with allodynia, possibly due to central sensitization. Pain 1995; 60: 217–22PubMedCrossRef
462.
go back to reference Rabben T, Skjelbred P, Øye I. Prolonged analgesic effect of ketamine, an N-methyl-D-aspartate receptor inhibitor, in patients with chronic pain. JPET 1999; 289: 1060–6 Rabben T, Skjelbred P, Øye I. Prolonged analgesic effect of ketamine, an N-methyl-D-aspartate receptor inhibitor, in patients with chronic pain. JPET 1999; 289: 1060–6
463.
go back to reference Enarson MC, Hays H, Woodroffe MA. Clinical experience with oral ketamine. J Pain Symptom Manage 1999; 17(5): 384–6PubMedCrossRef Enarson MC, Hays H, Woodroffe MA. Clinical experience with oral ketamine. J Pain Symptom Manage 1999; 17(5): 384–6PubMedCrossRef
464.
go back to reference Pud D, Eisenberg E, Spitzer A, et al. The NMDA receptor antagonist amantadine reduces surgical neuropathic pain in cancer patients: a double-blind, randomized, placebo controlled trial. Pain 1998; 75: 349–54PubMedCrossRef Pud D, Eisenberg E, Spitzer A, et al. The NMDA receptor antagonist amantadine reduces surgical neuropathic pain in cancer patients: a double-blind, randomized, placebo controlled trial. Pain 1998; 75: 349–54PubMedCrossRef
465.
go back to reference Eide PK, Stubhaug A, Stenehjem AE. Central dysesthesia pain after traumatic spinal cord injury is dependent on N-methyl-D-aspartate receptor activation. Neurosurgery 1995;37: 1080–7PubMedCrossRef Eide PK, Stubhaug A, Stenehjem AE. Central dysesthesia pain after traumatic spinal cord injury is dependent on N-methyl-D-aspartate receptor activation. Neurosurgery 1995;37: 1080–7PubMedCrossRef
466.
go back to reference Eide PK, Jørum E, Stubhaug A, et al. Relief of post-herpetic neuralgia with the N-methyl-D-aspartic acid receptor antagonist ketamine: a double-blind, cross-over comparison with morphine and placebo. Pain 1994; 58: 347–54PubMedCrossRef Eide PK, Jørum E, Stubhaug A, et al. Relief of post-herpetic neuralgia with the N-methyl-D-aspartic acid receptor antagonist ketamine: a double-blind, cross-over comparison with morphine and placebo. Pain 1994; 58: 347–54PubMedCrossRef
467.
go back to reference Nikolajsen L, Hansen CL, Nielsen J, et al. The effect of ketamine on phantom pain: a central neuropathic disorder maintained by peripheral input. Pain 1996; 67: 69–77PubMedCrossRef Nikolajsen L, Hansen CL, Nielsen J, et al. The effect of ketamine on phantom pain: a central neuropathic disorder maintained by peripheral input. Pain 1996; 67: 69–77PubMedCrossRef
468.
go back to reference Graven-Nielsen T, Aspegren Kendall S, Henriksson KG, et al. Ketamine reduces muscle pain, temporal summation, and referred pain in fibromyalgia patients. Pain 2000; 85: 483–91PubMedCrossRef Graven-Nielsen T, Aspegren Kendall S, Henriksson KG, et al. Ketamine reduces muscle pain, temporal summation, and referred pain in fibromyalgia patients. Pain 2000; 85: 483–91PubMedCrossRef
469.
go back to reference Sorensen J, Bengtsson A, Backman E, et al. Pain analysis in patients with fibromyalgia. Effects of intravenous morphine, lidocaine, and ketamine. Scand J Rheumatol 1995; 24: 360–5 Sorensen J, Bengtsson A, Backman E, et al. Pain analysis in patients with fibromyalgia. Effects of intravenous morphine, lidocaine, and ketamine. Scand J Rheumatol 1995; 24: 360–5
470.
go back to reference Backonja M, Arndt G, Gombar KA, et al. Response of chronic neuropathic pain syndromes to ketamine: a preliminary report. Pain 1994; 56: 51–7PubMedCrossRef Backonja M, Arndt G, Gombar KA, et al. Response of chronic neuropathic pain syndromes to ketamine: a preliminary report. Pain 1994; 56: 51–7PubMedCrossRef
471.
go back to reference Felsby S, Nielsen J, Arendt-Nielsen L, et al. NMDA receptor blockade in chronic neuropathic pain: a comparison of ketamine and magnesium chloride. Pain 1995; 64: 283–91CrossRef Felsby S, Nielsen J, Arendt-Nielsen L, et al. NMDA receptor blockade in chronic neuropathic pain: a comparison of ketamine and magnesium chloride. Pain 1995; 64: 283–91CrossRef
472.
go back to reference Medrik-Goldberg T, Lifschitz D, Pud D, et al. Intravenous lidocaine, amantadine, and placebo in the treatment of sciatica: a double-blind, randomized, controlled study. Reg Anesth Pain Med 1999; 24: 534–40PubMedCrossRef Medrik-Goldberg T, Lifschitz D, Pud D, et al. Intravenous lidocaine, amantadine, and placebo in the treatment of sciatica: a double-blind, randomized, controlled study. Reg Anesth Pain Med 1999; 24: 534–40PubMedCrossRef
473.
go back to reference Eisenberg E, Kleiser A, Dortort A, et al. The NMDA (N-methyl-D-aspartate) receptor antagonist memantine in the treatment of postherpetic neuralgia: a double-blind, placebo-controlled study. Eur J Pain 1998; 2: 321–7PubMedCrossRef Eisenberg E, Kleiser A, Dortort A, et al. The NMDA (N-methyl-D-aspartate) receptor antagonist memantine in the treatment of postherpetic neuralgia: a double-blind, placebo-controlled study. Eur J Pain 1998; 2: 321–7PubMedCrossRef
474.
go back to reference Max MB, Byas-Smith MG, Gracely RH, et al. Intravenous infusion of the NMDA antagonist, ketamine, in chronic post-traumatic pain with allodynia: a double-blind comparison to alfentanil and placebo. Clin Neuropharmacol 1995; 18: 360–8PubMedCrossRef Max MB, Byas-Smith MG, Gracely RH, et al. Intravenous infusion of the NMDA antagonist, ketamine, in chronic post-traumatic pain with allodynia: a double-blind comparison to alfentanil and placebo. Clin Neuropharmacol 1995; 18: 360–8PubMedCrossRef
475.
go back to reference Nicolodi M, Sicuteri F. Negative modulators of excitatory amino acids in episodic and chronic migraine: preventing and reverting chronic migraine. Special lecture 7th INWIN Congress. Int J Clin Pharmacol Res 1998; 18: 93–100 Nicolodi M, Sicuteri F. Negative modulators of excitatory amino acids in episodic and chronic migraine: preventing and reverting chronic migraine. Special lecture 7th INWIN Congress. Int J Clin Pharmacol Res 1998; 18: 93–100
476.
go back to reference Nicolodi M, Del Bianco PL, Sicuteri F. Modulation of excitatory amino acids pathway: a possible therapeutic approach to chronic daily headache associated with analgesic drugs abuse. Int J Clin Pharmacol Res 1997; 17: 97–100PubMed Nicolodi M, Del Bianco PL, Sicuteri F. Modulation of excitatory amino acids pathway: a possible therapeutic approach to chronic daily headache associated with analgesic drugs abuse. Int J Clin Pharmacol Res 1997; 17: 97–100PubMed
477.
go back to reference Nicolodi M, Sicuteri F. Exploration of NMDA receptors in migraine: therapeutic and theoretic implications. Int J Clin Pharmacol Res 1995; 15: 181–9PubMed Nicolodi M, Sicuteri F. Exploration of NMDA receptors in migraine: therapeutic and theoretic implications. Int J Clin Pharmacol Res 1995; 15: 181–9PubMed
478.
go back to reference Cherry DA, Plummer JL, Gourlay GK, et al. Ketamine as an adjunct to morphine in the treatment of pain. Pain 1995; 62: 119–21PubMedCrossRef Cherry DA, Plummer JL, Gourlay GK, et al. Ketamine as an adjunct to morphine in the treatment of pain. Pain 1995; 62: 119–21PubMedCrossRef
479.
go back to reference Yang CY, Wong CS, Chang JY, et al. Intrathecal ketamine reduces morphine requirements in patients with terminal cancer pain. Can J Anaesth 1996; 43: 379–83PubMedCrossRef Yang CY, Wong CS, Chang JY, et al. Intrathecal ketamine reduces morphine requirements in patients with terminal cancer pain. Can J Anaesth 1996; 43: 379–83PubMedCrossRef
480.
go back to reference Fu ES, Miguel R, Scharf JE. Preemptive ketamine decreases postoperative narcotic requirements in patients undergoing abdominal surgery. Anesth Analg 1997; 84: 1086–90PubMed Fu ES, Miguel R, Scharf JE. Preemptive ketamine decreases postoperative narcotic requirements in patients undergoing abdominal surgery. Anesth Analg 1997; 84: 1086–90PubMed
481.
go back to reference Menigaux C, Fletcher D, Dupont X, et al. The benefits of intraoperative small-dose ketamine on postoperative pain after anterior cruciate ligament repair. Anesth Analg 2000; 90: 129–35PubMedCrossRef Menigaux C, Fletcher D, Dupont X, et al. The benefits of intraoperative small-dose ketamine on postoperative pain after anterior cruciate ligament repair. Anesth Analg 2000; 90: 129–35PubMedCrossRef
482.
go back to reference Stubhaug A, Breivik H, Eide PK, et al. Mapping of punctuate hyperalgesia around a surgical incision demonstrates that ketamine is a powerful suppressor of central sensitization to pain following surgery. Acta Anaesthesiol Scand 1997; 41: 1124–32PubMedCrossRef Stubhaug A, Breivik H, Eide PK, et al. Mapping of punctuate hyperalgesia around a surgical incision demonstrates that ketamine is a powerful suppressor of central sensitization to pain following surgery. Acta Anaesthesiol Scand 1997; 41: 1124–32PubMedCrossRef
483.
go back to reference Wong CS, Liaw WJ, Tung CS, et al. Ketamine potentiates analgesic effect of morphine in postoperative epidural pain control. Reg Anesth 1996; 21: 534–41PubMed Wong CS, Liaw WJ, Tung CS, et al. Ketamine potentiates analgesic effect of morphine in postoperative epidural pain control. Reg Anesth 1996; 21: 534–41PubMed
484.
go back to reference Dahl V, Ernoe PE, Steen T, et al. Does ketamine have preemptive effects in women undergoing abdominal hysterectomy procedures. Anesth Analg 2000; 90: 1419–22PubMedCrossRef Dahl V, Ernoe PE, Steen T, et al. Does ketamine have preemptive effects in women undergoing abdominal hysterectomy procedures. Anesth Analg 2000; 90: 1419–22PubMedCrossRef
485.
go back to reference Adam F, Libier M, Oszustowicz T, et al. Preoperative small-dose ketamine has no preemptive analgesic effect in patients undergoing total mastectomy. Anesth Analg 1999; 89: 444–7PubMed Adam F, Libier M, Oszustowicz T, et al. Preoperative small-dose ketamine has no preemptive analgesic effect in patients undergoing total mastectomy. Anesth Analg 1999; 89: 444–7PubMed
486.
go back to reference Ilkjaer S, Nikolajsen L, Hansen TM, et al. Effect of i.V. ketamine in combination with epidural bupivacaine or epidural morphine on postoperative pain and wound tenderness after renal surgery. Br J Anaesth 1998; 81: 707–12PubMed Ilkjaer S, Nikolajsen L, Hansen TM, et al. Effect of i.V. ketamine in combination with epidural bupivacaine or epidural morphine on postoperative pain and wound tenderness after renal surgery. Br J Anaesth 1998; 81: 707–12PubMed
487.
go back to reference Adriaenssens G, Vermeyen KM, Hoffmann VL, et al. Postoperative analgesia with i.v. patient-controlled morphine: effect of adding ketamine. Br J Anaesth 1999; 83: 393–6 Adriaenssens G, Vermeyen KM, Hoffmann VL, et al. Postoperative analgesia with i.v. patient-controlled morphine: effect of adding ketamine. Br J Anaesth 1999; 83: 393–6
488.
go back to reference Chia YY, Liu K, Liu YC, et al. Adding ketamine in a multi-modal patient-controlled epidural regimen reduces postoperative pain and analgesic consumption. Anesth Analg 1998; 86: 1245–9PubMed Chia YY, Liu K, Liu YC, et al. Adding ketamine in a multi-modal patient-controlled epidural regimen reduces postoperative pain and analgesic consumption. Anesth Analg 1998; 86: 1245–9PubMed
489.
go back to reference Wilder-Smith OH, Arendt-Nielsen L, Gaumann D, et al. Sensory changes and pain after abdominal hysterectomy: a comparison of anesthetic supplementation with fentanyl versus magnesium or ketamine. Anesth Analg 1998; 86: 95–101PubMed Wilder-Smith OH, Arendt-Nielsen L, Gaumann D, et al. Sensory changes and pain after abdominal hysterectomy: a comparison of anesthetic supplementation with fentanyl versus magnesium or ketamine. Anesth Analg 1998; 86: 95–101PubMed
490.
go back to reference Wilder-Smith CH, Knopfli R, Wilder-Smith OH. Perioperative magnesium infusion and postoperative pain. Acta Anaesthesiol Scand 1997; 41: 1023–7PubMedCrossRef Wilder-Smith CH, Knopfli R, Wilder-Smith OH. Perioperative magnesium infusion and postoperative pain. Acta Anaesthesiol Scand 1997; 41: 1023–7PubMedCrossRef
491.
go back to reference Tanaka M, Shimizu S, Nishimura W, et al. Relief of neuropathic pain with intravenous magnesium. Masui 1998; 47: 1109–13PubMed Tanaka M, Shimizu S, Nishimura W, et al. Relief of neuropathic pain with intravenous magnesium. Masui 1998; 47: 1109–13PubMed
492.
go back to reference Birch PJ. Clinical relevance of receptor pharmacology in the nociceptive pathway. Pain Rev 1995; 2: 13–27 Birch PJ. Clinical relevance of receptor pharmacology in the nociceptive pathway. Pain Rev 1995; 2: 13–27
493.
go back to reference Schugens MM, Egerter R, Daum I, et al. The NMDA antagonist memantine impairs classical eyeblink conditioning in humans. Neurosci Lett 1997; 224: 57–60PubMedCrossRef Schugens MM, Egerter R, Daum I, et al. The NMDA antagonist memantine impairs classical eyeblink conditioning in humans. Neurosci Lett 1997; 224: 57–60PubMedCrossRef
494.
go back to reference Muir KW, Grosset DG, Lees KR. Effects of prolonged infusions of the NMDA antagonist aptiganel hydrochloride (CNS 1102) in normal volunteers. Clin Neuropharmacol 1997; 20: 311–21PubMedCrossRef Muir KW, Grosset DG, Lees KR. Effects of prolonged infusions of the NMDA antagonist aptiganel hydrochloride (CNS 1102) in normal volunteers. Clin Neuropharmacol 1997; 20: 311–21PubMedCrossRef
495.
go back to reference Murman DL, Giordani B, Mellow AM, et al. Cognitive, behavioral, and motor effects of the NMDA antagonist ketamine in Huntington’s disease. Neurology 1997; 49: 153–61PubMedCrossRef Murman DL, Giordani B, Mellow AM, et al. Cognitive, behavioral, and motor effects of the NMDA antagonist ketamine in Huntington’s disease. Neurology 1997; 49: 153–61PubMedCrossRef
496.
go back to reference Oye I, Paulsen O, Maurset A. Effects of ketamine on sensory perception: evidence for a role of N-methyl-D-aspartate receptors. J Pharmacol Exp Ther 1992; 260: 1209–13PubMed Oye I, Paulsen O, Maurset A. Effects of ketamine on sensory perception: evidence for a role of N-methyl-D-aspartate receptors. J Pharmacol Exp Ther 1992; 260: 1209–13PubMed
497.
go back to reference Nelson KA, Park KM, Robinovitz E, et al. High-dose oral dextromethorphan versus placebo in painful diabetic neuropathy and postherpetic neuralgia. Neurology 1997; 48: 1212–8PubMedCrossRef Nelson KA, Park KM, Robinovitz E, et al. High-dose oral dextromethorphan versus placebo in painful diabetic neuropathy and postherpetic neuralgia. Neurology 1997; 48: 1212–8PubMedCrossRef
498.
go back to reference McQuay HJ, Carroll D, Jadad AR, et al. Dextromethorphan for the treatment of neuropathic pain: a double-blind randomised controlled crossover trial with integral n-of-1 design. Pain 1994; 59: 127–33PubMedCrossRef McQuay HJ, Carroll D, Jadad AR, et al. Dextromethorphan for the treatment of neuropathic pain: a double-blind randomised controlled crossover trial with integral n-of-1 design. Pain 1994; 59: 127–33PubMedCrossRef
499.
go back to reference Mercadante S, Casuccio A, Genovese G. Ineffectiveness of dextromethorphan in cancer pain. J Pain Symptom Manage 1998; 16: 317–22PubMedCrossRef Mercadante S, Casuccio A, Genovese G. Ineffectiveness of dextromethorphan in cancer pain. J Pain Symptom Manage 1998; 16: 317–22PubMedCrossRef
500.
go back to reference Kristensen JD, Svensson B, Gordh Jr T. The NMDA-receptor antagonist CPP abolishes neurogenic ‘wind-up pain’ after intrathecal administration. Pain 1992; 51: 249–53PubMedCrossRef Kristensen JD, Svensson B, Gordh Jr T. The NMDA-receptor antagonist CPP abolishes neurogenic ‘wind-up pain’ after intrathecal administration. Pain 1992; 51: 249–53PubMedCrossRef
501.
go back to reference Wu C-T, Yu J-C, Yeh C-C, et al. Preincisional dextromethorphan treatment decreases postoperative pain and opioid requirement after laparoscopic cholecystectomy. Anesth Analg 1999; 88: 1331–4PubMed Wu C-T, Yu J-C, Yeh C-C, et al. Preincisional dextromethorphan treatment decreases postoperative pain and opioid requirement after laparoscopic cholecystectomy. Anesth Analg 1999; 88: 1331–4PubMed
502.
go back to reference Kawamata T, Omote K, Kawamata M, et al. Premedication with oral dextromethorphan reduces postoperative pain after tonsillectomy. Anesth Analg 1998; 86: 594–7PubMed Kawamata T, Omote K, Kawamata M, et al. Premedication with oral dextromethorphan reduces postoperative pain after tonsillectomy. Anesth Analg 1998; 86: 594–7PubMed
503.
go back to reference Wu CT, Yu JC, Liu ST, et al. Preincisional dextromethorphan treatment for postoperative pain management after upper abdominal surgery. World J Surg 2000; 24: 512–7PubMedCrossRef Wu CT, Yu JC, Liu ST, et al. Preincisional dextromethorphan treatment for postoperative pain management after upper abdominal surgery. World J Surg 2000; 24: 512–7PubMedCrossRef
504.
go back to reference Grace RF, Power I, Umedaly H, et al. Preoperative dextromethorphan reduces intraoperative but not postoperative morphine requirements after laparotomy. Anesth Analg 1998; 87: 1135–8PubMed Grace RF, Power I, Umedaly H, et al. Preoperative dextromethorphan reduces intraoperative but not postoperative morphine requirements after laparotomy. Anesth Analg 1998; 87: 1135–8PubMed
505.
go back to reference Ilkjaer S, Bach LF, Nielsen PA, et al. Effect of preoperative oral dextromethorphan on immediate and late postoperative pain and hyperalgesia after total abdominal hysterectomy. Pain 2000; 86: 19–24PubMedCrossRef Ilkjaer S, Bach LF, Nielsen PA, et al. Effect of preoperative oral dextromethorphan on immediate and late postoperative pain and hyperalgesia after total abdominal hysterectomy. Pain 2000; 86: 19–24PubMedCrossRef
506.
go back to reference Henderson DJ, Withington BS, Wilson JA, et al. Perioperative dextromethorphan reduces postoperative pain after hysterectomy. Anesth Analg 1999; 89: 399–402PubMed Henderson DJ, Withington BS, Wilson JA, et al. Perioperative dextromethorphan reduces postoperative pain after hysterectomy. Anesth Analg 1999; 89: 399–402PubMed
507.
go back to reference McConaghy PM, McSorley P, McCaughey W, et al. Dextromethorphan and pain after total abdominal hysterectomy. Br J Anaesth 1998; 81: 731–6PubMedCrossRef McConaghy PM, McSorley P, McCaughey W, et al. Dextromethorphan and pain after total abdominal hysterectomy. Br J Anaesth 1998; 81: 731–6PubMedCrossRef
508.
go back to reference Sang CN, Hostetter MP, Gracely RH, et al. AMPA/KA antagonist LY293558 reduces capsaicin-evoked hyperalgesia but not pain in normal skin in humans. Anesthesiology 1998; 89: 1060-7PubMedCrossRef Sang CN, Hostetter MP, Gracely RH, et al. AMPA/KA antagonist LY293558 reduces capsaicin-evoked hyperalgesia but not pain in normal skin in humans. Anesthesiology 1998; 89: 1060-7PubMedCrossRef
Metadata
Title
Glutamate Receptors and Nociception
Implications for the Drug Treatment of Pain
Author
Dr Marian E. Fundytus
Publication date
01-01-2001
Publisher
Springer International Publishing
Published in
CNS Drugs / Issue 1/2001
Print ISSN: 1172-7047
Electronic ISSN: 1179-1934
DOI
https://doi.org/10.2165/00023210-200115010-00004

Other articles of this Issue 1/2001

CNS Drugs 1/2001 Go to the issue

Current Opinion

The Star Systems

Therapy in Practice

Compulsive Buying Disorder