16
Views
4
CrossRef citations to date
0
Altmetric
Review

Synaptic mechanisms in nociception: emerging targets for centrally-acting analgesics

Pages 173-189 | Published online: 25 Feb 2005

Bibliography

  • ELLIOTT AM, SMITH BH, PENNY KI, SMITH CW, CHAMBERS WA: The epidemiology of chronic pain in the community. Lancet (1999) 354:1248–1252.
  • ••Comprehensive study of the incidence of chronic pain in anaverage UK community.
  • SHIPTON EA: Pain: Acute and Chronic. Arnold, London, UK (1999).
  • BRASSEUR L: A review of current pharmacological treatment of pain. Drugs (1997) 53:10–17.
  • VANE JR, BAKHLE YS, BOTTING RM: Cyclooxygenases 1 and 2. Ann. Rev. Pharmacol. Toxicol (1998) 38:97–120.
  • LASHBROOK JM, OSSIPOVA MH, HUNTER JC, RAFFA RB, TALLARIDA RJ, PORRECA F: Synergistic antiallodynic effects of spinal morphine with ketorolac and selective COM-and COX2-inhibitors in nerve-injured rats. Pain (1999) 82:65–72.
  • QUARTARA L, MAGGI CA: The tachykinin NKi receptor. Part I: ligands and mechanisms of cellular activation. Neuropeptides (1997) 31:537–563.
  • QUARTARA L, MAGGI CA: The tachykinin NKi receptor. Part II: distribution and pathophysiological. Neuropep-tides (1998) 32:1–49.
  • SEWARD EM, SWAIN CJ: Neurokinin receptor antago-nists. Exp. Opin. Ther. Patents (1999) 9(5):571–582.
  • •Why NKi antagonists should work, and how they do not.
  • RUPNIAK NMJ, KRAMER MS: Discovery of the anti-depressant and anti-emetic efficacy of substance P (NKi) receptor antagonists. Trends Pharmacol. ScL (1999) 20:485–490.
  • •What NKI antagonists might be good for.
  • DIONNE RA, MAX MB, GORDON SM et al.: The substance Preceptor antagonist CP-99, 994 reduces acute postop-erative pain. Clin. Pharmacol. Therap. (1998) 64:562–568.
  • •The exception that proves the rule.
  • REXED B: The cytoarchitectonic organization of the spinal cord in the cat. J. Comp. Neurol (1952) 96:415–496.
  • SORKIN LS, CARLTON SM: Spinal anatomy and pharma-cology of afferent processing. In: Anesthesia: Biologic Foundations. Yaksh TL, Lynch C, Zapol WM, Maze M, Biebuyck JF, Saidman LJ (Eds.), Lippincott-Raven, New York, USA (1997):577–609.
  • •Good basic account of spinal nociceptive processing.
  • MANTYH PW, ROGERS SD, HONORE P et al: Inhibition of hyperalgesia by ablation of lamina I spinal neurons expressing the substance P receptor. Science (1997) 278:275–279.
  • ••Understanding these neurones is the key to chronic pain.
  • DUGGAN AW, HALL JG, HEADLEY PM: Enkephalins and dorsal horn neurones in the cat: effects on responses to noxious and innocuous stimulation. Br. J. Pharmacol. (1977) 61:399–408.
  • JENSEN TS: Opioids in the brain: supraspinal mechanisms in pain control. Acta Anaesth. Scand. (1997) 41:123–132.
  • BENNETT GJ: Opioids and painful peripheral neuropathy. In: Opioid Sensitivity of Chronic Non-Cancer Pain. Kalso E, McQuay HJ, Wiesenfeld-Hallin Z (Eds.), IASP Press, Seattle, USA (1999):319–326.
  • SINDRUP SH, JENSEN TS: Efficacy of pharmacological treatments of neuropathic pain: an update and effect related mechanism of drug action. Pain (1999) 83:389–400.
  • •Most recent overview of treatments for neuropathic pain.
  • MCQUAY HJ, MOORE RA: An Evidence-Based Resource for Pain Relief Oxford University Press, Oxford, UK (1998).
  • MCQUAY HJ, TRAMER M, NYE BA, CARROLL D, WIFFEN PJ, MOORE RA: Systematic review of antidepressants in neuropathic pain. Pain (1996) 68:217–227.
  • MCQUAY H, CARROLL D, JADAD AR, WIFFEN P, MOORE A: Anticonvulsant drugs for management of pain-a systematic review. Br. Med. J (1995) 311:1047–1052.
  • WOOLF CJ, BENNETT GJ, DOHERTY M et al.: Towards a mechanism-based classification of pain? Pain (1998) 77:227–229.
  • WOOLF CJ, MANNION RJ: Neuropathic pain: aetiology, symptoms, mechanisms, and management. Lancet (1999) 3 5 3:1959–1964.
  • •Why we need to go to a mechanism-based classification for chronic pain.
  • KOLTZENBURG M: The changing sensitivity in the life of the nociceptor. Pain (1999) S93–S102.
  • •Good overview of current knowledge of nociceptive primary afferent biology.
  • CODERRE TJ, KATZ J: Peripheral and central hyperex-citability: differential signs and symptoms in persis-tent pain. Behav. Brain Sci. (1997) 20:404–419.
  • •One of the best reviews of the processes that lead to central sensitisation.
  • YAKSH TL, HUA XY, KALCHEVA I, NOZAKITAGUCHI N, MARSALA M: The spinal biology in humans and animals of pain states generated by persistent small afferent input. Proc. Natl. Acad. Sci. USA (1999) 96:7680–7686.
  • TANG FR, SIM MK: Pre-and/or post-synaptic localisa-tion of metabotropic glutamate receptor 1 alpha (mGluR1 alpha) and 2/3 (mGluR2/3) in the rat spinal cord. NeuroscL Res. (1999) 34:73–78.
  • JIA H, RUSTIONI A, VALTSCHANOFF JG: Metabotropic glutamate receptors in superficial laminae of the rat dorsal horn. J. Comp. Neurol. (1999) 410:627–642.
  • YUNG KKL: Localization of glutamate receptors indorsal horn of rat spinal cord. Neuroreport (1998) 9:1639–1644.
  • BLEAZARD L, HILL RG, MORRIS R: The correlation between the distribution of the nk1 receptor and the actions of tachykinin agonists in the dorsal horn of the rat indicates that substance-P does not have a functional-role on substantia-gelatinosa (lamina-II) neurons. J. NeuroscL (1994) 14:7655–7664.
  • ZERARI F, KARPITSKIY V, KRAUSE J, DESCARRIES L, COUTURE R: Irnmunoelectron microscopic localiza-tion of NK3 receptor in the rat spinal cord. Neuroreport (1997) 8:2661–2664.
  • ZERARI F, KARPITSKIY V, KRAUSE J, DESCARRIES L, COUTURE R: Astroglial distribution of neurokinin-2 receptor immunoreactivity in the rat spinal cord. Neuroscience (1998) 84:1233–1246.
  • •There are no NK2 receptors on rat dorsal horn neurones.
  • SCHOEPP DD, JANE DE, MONN JA: Pharmacological agents acting at subtypes of metabotropic glutamate receptors. Neuropharmacology (1999) 38:1431–1476.
  • •Comprehensive review of metabotropic receptor pharmacology.
  • VANROSSUM D, HANISCH UK, QUIRION R: Neuroana-tomical localization, pharmacological characteriza-tion and functions of CGRP, related peptides and their receptors. NeuroscL Biobehav. Rev. (1997) 21:649–678.
  • BENNETT MR: The concept of long term potentiation of transmission at synapses. Prog. Neurobiol. (2000) 60:109–137.
  • BASBAUM Al: Distinct neurochemical features of acute and persistent pain. Proc. Natl. Acad. Sci. USA (1999) 96:7739–7743.
  • •Good introduction to second messengers in sensitisation.
  • MILLAN MJ: The induction of pain: an integrative review. Prog. Neurobiol. (1999) 57:1–164.
  • WOOLF CJ, COSTIGAN M: Transcriptional and posttranslational plasticity and the generation of inflammatory pain. Proc. Natl. Acad. Sci. USA (1999) 96:7723–7730.
  • THOMPSON SWN, BENNETT DLH, KERR BJ, BRADBURYEJ, MCMAHON SB: Brain-derived neurotrophic factor is an endogenous modulator of nocicep tive responses in the spinal cord. Proc. Natl. Acad. Sci. USA (1999) 96:7714–7718.
  • HOKFELT T, ZHANG X, XU ZQ et al.: Cellular and synaptic mechanisms in transition of pain from acute to chronic. In: Proceedings of the 8th World Congress on Pain. Jensen TS, Turner AJ, Wiesenfeld-Hallin Z Seattle, IASP Press (1997):133–153.
  • •Everything you ever wanted to know about which peptides do what after inflammation and nerve injury.
  • ATTAL N, BOUHASSIRA D: Mechanisms of pain in peripheral neuropathy. Acta Neurol. Scand. (1999) 100:12–24.
  • MAYER DJ, MAO JR, HOLT J, PRICE DD: Cellularmechanisms of neuropathic pain, morphine tolerance, and their interactions. Proc. Natl. Acad. ScL USA (1999) 96:7731–7736.
  • WOOLF CJ, SHORTLAND P, COGGESHALL RE: Peripheralnerve injury triggers central sprouting of myelinated afferents. Nature (1992) 355:75–78.
  • RAMER MS, THOMPSON SWN, MCMAHON SB: Causes andconsequences of sympathetic basket formation in dorsal root ganglia. Pain (1999) S111–S120.
  • SHI TJS, CUT JG, MEYERSON BA, LINDEROTH B, HOKFELTT: Regulation of galanin and neuropeptide Yin dorsal root ganglia and dorsal horn in rat mononeuropathic models: Possible relation to tactile hypersensitivity. NeuroscL (1999) 93:741–757.
  • DICKINSON T, FLEETWOOD-WALKER SM: VIP and PACAP: very important in pain? Trends Pharmacol. ScL (1999) 20:324–329.
  • OKUSE K, CHAPLAN SR, MCMAHON SB et al.: Regulationof expression of the sensory neuron-specific sodium channel SNS in inflammatory and neuropathic pain. Mol. Cell. NeuroscL (1997) 10:196–207.
  • MENDELL LM, ALBERS KM, DAVIS BM: Neurotrophins, nociceptors, and pain. Micros. Res. Tech. (1999) 45:252–261.
  • BURNSTOCK G: A unifying purinergic hypothesis for the initiation of pain. Lancet (1996) 347:1604–1605.
  • YAKSH TL: Spinal systems and pain processing: development of novel analgesic drugs with mechanis-tically defined models. Trends Pharmacol. Sci. (1999) 20:329–337.
  • FUENTES JA, RUIZGAYO M, MANZANARES J, VELA G, RECHE I, CORCHERO J: Cannabinoids as potential new analgesics. Life Sci. (1999) 65:675–685.
  • DECKER MW, MEYER MD: Therapeutic potential ofneuronal nicotinic acetylcholine receptor agonists as novel analgesics. Biochem. Pharmacol. (1 99 9) 58:917–923.
  • DONDIO G, RONZONI S, PETRILLO P: Non-peptide deltaopioid agonists and antagonists (Part II). Exp. Opin. Ther. Patents. (1999) 9(4):353–374.
  • MCCORMACK K: Signal transduction in neuropathic pain, with special emphasis on the analgesic role of opioids-Part II: Moving basic science towards a new pharmacotherapy. Pain Rev. (1999) 6:99–131.
  • POLGAR E, FOWLER JH, MCGILL MM, TODD AJ: The types of neuron which contain protein kinase C gamma in rat spinal cord. Brain Res. (1999) 833:71–80.
  • MARTIN WJ, LIU H, WANG H, MALMBERG AB, BASBAUMAT: Inflammation-induced up-regulation of protein kinase C gamma immunoreactivity in rat spinal cord correlates with enhanced nociceptive processing. Neuroscience (1999) 88:1267–1274.
  • PETERSEN-ZEITZ KR, BASBAUM AT: Second messengers, the substantia gelatinosa and injury-induced persis-tent pain. Pain (1999) (Suppl. 6):S5–S12.
  • MALMBERG AB, CHEN C, TONEGAWA S, BASBAUM AT:Preserved acute pain and reduced neuropathic pain in mice lacking PKC gamma. Science (1997) 278:279–283.
  • CHAPMAN V, BURITOVA J, HONORE P, BESSON JM: 7-nitro-indazole, a selective inhibitor of neuronal nitric-oxide synthase, reduces formalin evoked c-fos expression in dorsal horn neurons of the rat spinal-cord. Brain Res. (1995) 697:258–261.
  • HANDY RLC, WALLACE HP, GAFFEN ZA, WHITEHEAD KJ, MOORE PK: The antinociceptive effect of 1-(2-trifluoromethylphenyl) imidazole (trim), a potent inhibitor of neuronal nitric-oxide synthase in vitro, in the mouse. Br. J. Pharmacol. (1 99 5) 116:2349-2350.
  • HANDY RLC, MOORE PK: Effects of selective inhibitorsof neuronal nitric oxide synthase on carrageenan-induced mechanical and thermal hyperalgesia. Neuropharmacology (1998) 37:37–43.
  • CROSBY G, MAROTA JJA, HUANG PL: Intactnociception-induced neuroplasticity in transgenic mice deficient in neuronal nitric-oxide synthase. Neuroscience (1995) 69:1013–1017.
  • HUANG PL: Neuronal and endothelial nitric oxidesynthase gene knockout mice. Braz. J. Med. Biol. Res. (1999) 32:1353–1359.
  • EISENBERG E, KLEISER A, DORTORT A, HAIM T, YARNITSKY D: 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) 2:321–327.
  • SORENSEN J, BENGTSSON A, BACKMAN E, HENRIKSSONKG, BENGTSSON M: Pain analysis in patients with fibromyalgia-effects of intravenous morphine, lidocaine, and ketamine. Scand. j Rheumatol (1995) 24:360–365.
  • RABBEN T, SKJELBRED P, OYE I: Prolonged analgesic effect of ketamine, an N-methyl-D-aspartate receptor inhibitor, in patients with chronic pain./ Pharmacol Exp. Ther. (1999) 289:1060–1066.
  • ••NMDA antagonists are effective antihyperalgesics in man.
  • SCHMID RL, SANDLER AN, KATZ J: Use and efficacy oflow-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes. Pain (1999) 82:111–125.
  • BOYCE S, WYATT A, WEBB JK et al.: Selective NMDA NR2B antagonists induce antinociception without motor dysfunction: correlation with restricted localisation of NR2B subunit in dorsal horn. Neuropharmacology (1999) 38:611–623.
  • ••Selective block of NR2B-containing NMDA receptors givesantihyperalgesia without motor impairment.
  • MONYER H, SPRENGEL R, SCHOEPFER R et al.: Heteromeric NMDA receptors-molecular and functional distinction of subtypes. Science (1992) 256:1217–1221.
  • YAMAKURA T, SHIMOJI K: Subunit-and site-specific pharmacology of the NMDA receptor channel. Prog. Neurobiol (1999) 59:279–298.
  • •Useful overview of current knowledge of NMDA receptor subtypes.
  • WENZEL A, SCHEURER L, KUNZI R, FRITSCHY JM, MOHLER H, BENKE D: Distribution of NMDA receptor subunit proteins NR2A, 2B, 2C and 2D in rat brain. Neuroreport (1995) 7:45–48.
  • HESS SD, DAGGETT LP, CRONA J et al: Cloning and functional characterization of human heteromeric N-methyl-D-aspartate receptors. j Pharmacol. Exp. Ther. (1996) 278:808–816.
  • WET HB, JAKEMAN LB, HUNTER JC, BONHAUS DW: PharmacologicalcharacterizationofN-methyl-D-aspartate receptors in spinal cord of rats with a chronic peripheral mononeuropathy. Neuropharmacology (1997) 36:1561–1569.
  • BUTLER TW, BLAKE JF, BORDNER J et al.: (3R, 4S)-344-(4-fluoropheny1)-4-hydroxypiperidin--1-ylichroman-4, 7-diol: a conformationally restricted analogue of the NR2B subtype-selective NMDA antago-nist OS, 2, 9-1-(4-hy dr oxyph en y1)-2 (4-hy dr o x y-4-phenylpiperidino)-1-propanol. J Med. Chem. (1998) 41:1172–1184.
  • •Description of CP-283, 097, a selective, orally-available NR2B receptor antagonist.
  • FISCHER G, MUTEL V, TRUBE G et al.: Ro 25-6981, a highly potent and selective blocker of N-methyl-D-aspartate receptors containing the NR2B subunit. Characterization in vitro. J. Pharmacol. Exp. Ther. (1997) 283:1285–1292.
  • IMAMURA Y, BENNETT GJ: Felbamate relieves several abnormal pain sensations in rats with an experi-mental peripheral neuropathy.j Pharmacol. Exp. Ther. (1995) 275:177–182.
  • HUNTER JC, GOGAS KR, HEDLEY LR et al: The effect of novel anti-epileptic drugs in rat experimental models of acute and chronic pain. Eur. J. Pharmacol. (1997) 324:153–160.
  • KLECKNER NW, GLAZEWSKI JC, CHEN CC, MOSCRIP TD:Subtype-selective antagonism of N-methyl-D-aspartate receptors by felbamate: insights into the mechanism of action. J. Pharmacol Exp. Ther. (1999) 289:886–894.
  • ZHOU ZL, CAI SX, WHITTEMORE ER et al.: 4-hydroxy-142-(4-hydroxyphenoxy)ethy1]-4-(4-meth ylbenzyl) piperidine: a novel, potent, and selective NR1/2BNMDAreceptor antagonist. j Med. Chem. (1999) 42:2993–3000.
  • •Description of CO 101244/PD 174494, a selective NR2B NMDA antagonist.
  • WRIGHT JL, GREGORY TF, BIGGE CF et al: Subtype-selective N-methyl-D-aspartate receptor antagonists: synthesis and biological evaluation of 1-(arylal-kyny1)-4-benzylpiperidines. J. Med. Chem. (1999) 42:2469–2477.
  • TAMIZ AP, WHITTEMORE ER, WOODWARD RM, UPASANIRB, KEANA JFW: Structure-activity relationship for a series of 2-substituted 1, 2, 3, 4-tetrahydro-9H-pyrido[3, 4-b]indoles: potent subtype-selective inhibi-tors of N-methyl-D-aspar tate (NMDA) receptors. Bioorg. Med. Chem. Lett. (1999) 9:1619–1624.
  • TAMIZ AP, WHITTEMORE ER, ZHOU ZL et al: Structure-activity relationships for a series of bis(phenylalkyl)amines: potent subtype-selective inhibitors of N-methyl-D-aspartate receptors. J Med. Chem. (1998) 41:3499–3506.
  • HONER M, BENKE D, KUHSE J et al.: Differentiation ofNMDA receptor subtypes: preferential interaction of the glycine site antagonist [H-3]CGP 61594 with NR1 /2B receptors. Eur.j Neurosci. (1998) 10:5405.
  • TAMIZ AP, WHITTEMORE ER, SCHELKUN RM et al.: N-(2-(4-hydroxyphenyl)ethyl)-4-chlorocinn amide: a novel antagonist at the 1A/2B NMDA receptor subtype. Bioorg. Med. Chem. Lett. (1998) 8:199–200.
  • MONAGHAN DT, LARSEN H: NR1 and NR2 subunit contributions to N-methyl-D-aspartate receptor channel blocker pharmacology. J. Pharmacol. Exp. Ther. (1997) 280:614–620.
  • TAMIZ AP, CAI SX, ZHOU ZL et al.: Structure-activityrelationship of N-(phenylalkyl)cinnamides as novel NR2B sub typ e-selective NMDA receptor antagonists. J Med. Chem. (1999) 42:3412–3420.
  • WRIGHT JL, GREGORY TF, BOXER PA, MELTZER LT, SERPA KA, WISE LD: Discovery of subtype-selective NMDAreceptorligands:4-benzy1-1-pip eridinyalkynylpyrroles, pyrazoles and imidazoles as NR1 A/2B antagonists. Bioorg. Med. Chem. Lett. (1999) 9:2815–2818.
  • NIELSEN KJ, SKJAERBAEK N, DOOLEY M et al: Structure-activity studies of conantokins as human N-methyl-D-aspartate receptor modulators. J Med. Chem. (1999) 42:415–426.
  • MALMBERG AB, GILBERT H, ZHOU L-M, MCCABE T, BASBAUM Al: Antinociceptive and side-effect profile of subtype selective NMDA-receptor inhibitors derived from cone snail venoms. Proceedings of the IXth World Congress on Pain (1999) 288–288.
  • SUNDSTROM E, WHITTEMORE S, MO LL, SEIGER A: Analysis of NMDAreceptors in the human spinal cord. Exp. Neurol. (1997) 148:407–413.
  • ••NR2B units apparently absent from spinal cord.
  • WILCOX GL, SEYBOLD VS: Pharmacology of spinalafferent processing. In: Anesthesia: Biologic Foundations. Yaksh TL, Lynch C, Zapol WM, Maze M, Biebuyck JF, Saidman LJ (Eds.), Lippincott-Raven, New York, USA (1997):557–576.
  • DOUGHERTY PM, PALECEK J, PALECKOVA V, SORKIN LS, WILLIS WD: The role of NMDA and non-NMDA excita-tory amino acid receptors in the excitation of primate spinothalamic tract neurons by mechanical, chemical, thermal, and electrical stimuli. j Neurosci. (1992) 12:3025–3041.
  • BIGGE CF, BOXER PA, ORTWINE DF: AMPA/kainate receptors. Curr. Pharm. Des. (1996) 2:397–412.
  • CHITTAJALLU R, BRAITHVVAITE SP, CLARKE VRJ, HENLEYJM: Kainate receptors: subunits, synaptic localization and function. Trends Pharmacol ScL (1999) 20:26–35.
  • WOLLMUTH LP, SAKMANN B: Different mechanisms ofCa2+ transport in NNIDA and Ca2±-permeable AMPA glutamate receptor channels. J. Gen. Physiol. (1998) 112:623–636.
  • ENGELMAN HS, ALLEN TB, MACDERMOTT AB: The distri-bution of neurons expressing calcium-permeable AMPA receptors in the superficial laminae of the spinal cord dorsal horn. j NeuroscL (1999) 19:2081–2089.
  • ALBUQUERQUE C, LEE CJ, JACKSON AC, MACDERMOTTAB: Subpopulations of GABAergic and non-GABAergic rat dorsal horn neurons express Ca2+-permeable AMPA receptors. Eur.J. NeuroscL (1999) 11:2758–2766.
  • SPIKE RC, KERR R, MAXWELL DJ, TODD AJ: GluR1 andG1uR2/3 subunits of the AMPA-typ e glutamate receptor are associated with particular types of neurone in laminae I-III of the spinal dorsal horn of the rat. Eur. NeuroscL (1998) 10:324–333.
  • GROSSMAN SD, WOLFE BB, YASUDA RP, WRATHALL JR:Alterations in AMPA receptor subunit expression after experimental spinal cord contusion injury. jNeurosci. (1999) 19:5711–5720.
  • CHIMIRRI A, GITTO R, ZAPPALA M: AMPA receptor antagonists. Exp. Opin. Ther. Patents (1999) 9 (5):557–570. Update on AMPA receptor pharmacology.
  • LI P, WILDING TJ, KIM SJ, CALEJESAN AA, HUETTNER JE, ZHUO M: Kainate-receptor-mediated sensory synaptic transmission in mammalian spinal cord. Nature (1999) 397:161–164.
  • STANFA LC, DICKENSON AH: The role of non-N-methyl-D-aspartate ionotropic glutamate receptors in the spinal transmission of nociception in normal animals and animals with carrageenan inflammation. Neuroscience (1999) 93:1391–1398.
  • PROCTER MJ, HOUGHTON AK, FABER ESL et al.: Actions of kainate and AMPA selective glutamate receptor ligands on nociceptive processing in the spinal cord. Neuropharmacology (1998) 37:1287–1297.
  • IINO M, KOIKE M, ISA T, OZAWA S: Voltage-dependent blockage of Ca2±-permeable AMPA receptors by Joro spider toxin in cultured rat hippocamp al neurones. J. Physiol. (1996) 496:431–437.
  • KOIKE M, IINO M, OZAWA S: Blocking effect of 1-naphthyl acetyl spermine on Ca2±-permeable AMPA receptors in cultured rat hippocampal neurons. NeuroscL Res. (1997) 29:27–36.
  • BULDAKOVA SL, VOROBJEV VS, SHARONOVA IN, SAMOILOVA MV, MAGAZANIK LG: Characterization of AMPA receptor populations in rat brain cells by the use of subunit-specific open channel blocking drug, IENI-1460. Brain Res. (1999) 846:52–58.
  • •A selective Ca2+-permeable AMPA receptor antagonist.
  • SORKIN LS, YAKSH TL, DOOM CM: Mechanical allodynia in rats is blocked by a Ca2+ permeable AMPA receptor antagonist. Neuroreport (1999) 10:3523–3526.
  • ••Blocking Ca2+-permeable AMPA receptors isantihyperalgesic.
  • BOXALL SJ, THOMPSON SWN, DRAY A, DICKENSON AH, URBAN L: Metabotropic glutamate receptor activation contributes to nociceptive reflex activity in the rat spinal cord in vitro. Neuroscience (1996) 74:13–20.
  • YOUNG MR, FLEETWOOD-WALKER SM, DICKINSON 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) 777:161–169.
  • BUDAI D, LARSON AA: The involvement of metabotropic glutamate receptors in sensory transmission in dorsal horn of the rat spinal cord. Neuroscience (1998) 83:571–580.
  • BARANAUSKAS G, NISTRI A: Sensitization of pain pathways in the spinal cord: cellular mechanisms. Prog. Neurobiol. (1998) 54:349–365.
  • FISHER K, FUNDYTUS ME, CAHILL CM, CODERRE TJ: Intrathecal administration of the mGluR compound, (S)-4CPG, attenuates hyperalgesia and allodynia associated with sciatic nerve constriction injury in rats. Pain (1998) 77:59–66.
  • NEUGEBAUER V, CHEN PS, WILLIS WD: Role of metabotropic glutamate receptor subtype mGluR1 in brief nociception and central sensitization of primate SIT cells. J. Neurophysiol (1999) 82:272–282.
  • STANFA LC, DICKENSON AH: Inflammation alters the effects of mGlu receptor agonists on spinal nocicep-tive neurones. Eur.J. Pharmacol. (1998) 347:165–172.
  • ANNOURA H, FUKUNAGA A, UESUGI M, TATSUOKA T, HORIKAWA Y: A novel class of antagonists for metabo tropicglutamatereceptors, 7-(hydroxyimino)cyclopropa[b]chromen-1 a-carboxylates. Bioorg. Med. Chem. Lett. (1996) 6:763–766.
  • GASPARINI F, LINGENHOHL K, STOEHR N et al.: 2-methyl-6-(phenylethyny1)-pyridine (MPEP), a potent, selective and systemically active mG1u5 receptor antagonist. Neuropharmacology (1999) 38:1493–1503.
  • BEVAN S: Ion channels and receptors of peripheral neurons: their roles in nociception and antinocicep-tion. Proceedings of IBC Conference New Directions in the Control of Pain'. London, UK (October 1999).
  • LITTLEWOOD NK, TODD AJ, SPIKE RC, WATT C, SHEHAB SAS: The types of neuron in spinal dorsal horn which possess neurokinin-1 receptors. Neuroscience (1995) 66:597–608.
  • MARSHALL GE, SHEHAB SAS, SPIKE RC, TODD AJ: Neurokinin-1 receptors on lumbar spinothalamic neurons in the rat. Neuroscience (1996) 72:255–263.
  • THOMPSON SWN, 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 NeuroscL (1994) 14:3672–3687.
  • RUPNIAK NMJ, WEBB JK, WILLIAMS AR, CARLSON E, BOYCE S, HILL RG: Antinociceptive activity of the tachykinin NK1 receptor antagonist, CP-99, 994, in conscious gerbils. Br. J. Pharmacol. (1995) 116:1937–1943.
  • TRAUB RJ: The spinal contribution of substance P to the generation and maintenance of inflammatory hyperalgesia in the rat. Pain (1996) 67:151–161.
  • PARSONS AM, HONDA CN, JIA Y et al: Spinal NK1 receptors contribute to the increased excitability of the nociceptive flexor reflex during p ersistent periph-eral inflammation. Brain Res. (1996) 739:263–275.
  • MA QP, WOOLF CJ: Tachykinin NK1 receptor antago-nist RP67580 attenuates progressive hypersensitivity of flexor reflex during experimental inflammation in rats. Eur.J. Pharmacol. (1997) 322:165–171.
  • SLUKA KA, MILTON MA, WILLIS WD, WESTLUND KN: Differential roles of neurokinin 1 and neurokinin 2 receptors in the development and maintenance of heat hyperalgesia induced by acute inflammation. Br. Pharmacol (1997) 120:1263–1273.
  • HOUGHTON AK, CLARKE RW: NK1-tachykinin receptors and prolonged, stimulus-evoked alterations in the excitability of withdrawal reflexes in the decere-brated and spinalized rabbit. Neuroscience (1995) 66:673–683.
  • HOUGHTON AK, OGILVIE J, CLARKE RW: The involve-ment of NK2 and NK3 tachykinin receptors in central sensitization of withdrawal reflex in the decerebrated, sp in alized rabbit. Neuropharmacology (2000) 39:135–142.
  • MCCARSON KE, KRAUSE JE: Ni and NK3 type tachyki-nin receptor messenger-RNA expression in the rat spinal-cord dorsal horn is increased during adjuvant or formalin-induced nociception. j Neurosci. (1994) 14:712–720.
  • CROUL S, SVERSTIUK A, RADZIEVSKY A, MURRAY M: Modulation of neurotransmitter receptors following unilateral 11-52 deafferentation: NK1, NK3, NMDA, and 5HT1 a receptor binding autoradiography. j Comp. Neurol (1995) 361:633–644.
  • SAKURADA T, MANOME Y, KATSUMATA K et al.: Nalox one-reversible effect of spantide on the spinally mediated behavioral-response induced by neurokinin-2 and neurokinin-3 receptor agonists. Naunyn-Schmiedeberg's Arch. Pharmacol. (1992) 346:69–75.
  • CHIZH BA, CUMBERBATCH MJ, BIRCH PJ, HEADLEY PM: Endogenous modulation of excitatory amino acid responsiveness by tachykinin NK1 and NK2 receptors in the rat spinal cord. Br. J. Pharmacol (1995) 115:1013–1019.
  • •How tachykinins influence neuronal responses to glutamate receptor agonists.
  • SANTOS ARS, CALIXTO JB: Further evidence for the involvement of tachykinin receptor subtypes in formalin and capsaicin models of pain in mice. Neuropeptides (1997) 31:381–389.
  • SEYBOLD VS, GRKOVIC I, PORTBURY AL et al: Relation-ship of NK3 receptor-immunoreactivity to subpopula-tions of neurons in rat spinal cord. j Comp. Neurol (1997) 381:439–448.
  • ••NK3 receptors could influence release of NO.
  • LINDEN DR, JIA YP, SEYBOLD VS: Spinal neurokin(3) receptors facilitate the nociceptive flexor reflex via a pathway involving nitric oxide. Pain (1999) 80:301–308.
  • LINDEN DR, SEYBOLD VS: Spinal neurokinin(3) receptors mediate thermal but not mechanical hyperalgesia via nitric oxide. Pain (1999) 80:309–317.
  • •Functional evidence for a link between NK3 receptor activa-tion and NO.
  • MELLER ST, GEBHART GF: Nitric oxide (NO) and nociceptive processing in the spinal cord. Pain (1993) 52:127–136.
  • MCMAHON SB, LEWIN GR, WALL PD: Central hyp erex-citability triggered by noxious inputs. Curr. Opin. Neurobiol. (1993) 3:602–610.
  • LIN Q, PENG YB, WU J, WILLIS WD: Involvement of cGMP in nociceptive processing by and sensitization of spinothalamic neurons in primates. J NeuroscL (1997) 17:3293–3302.
  • KAWAMATA T, OMOTE K: Activation of spinal N-methyl-D-aspartate receptors stimulates a nitric oxide/cyclicguano sine3 ', 5 '-mon op ho-sphate/glutamate release cascade in nociceptive signaling. Anesthesiology (1999) 91:1415–1424.
  • DIETL MM, PALACIOS JM: Phylogeny of tachykinin receptor localization in the vertebrate central nervous system-apparent absence of neurokinin-2 and neurokinin-3 binding sites in the human brain. Brain Res. (1991) 539:211–222.
  • MILEUSNIC D, LEE JM, MAGNUSON DJ et al.: Neurokinin-3 receptor distribution in rat and human brain: An immunohistochemical study. Neuroscience (1999) 89:1269–1290.
  • ••NK3 receptors are in the human CNS.
  • OURYDONAT F, CARAYON P, THURNEYSSEN O et al.: Functional characterization of the nonpeptide neurokinin3 (NK3) receptor antagonist, SR142801 on the human NK3 receptor expressed in Chinese hamster ovary cells. J. Pharmacol. Exp. Ther. (1995) 274:148–154.
  • GIARDINA GAM, RAVEGLIA LF, GRUGNI M et al.: Discovery of a novel class of selective non-peptide antagonists for the human neurokinin-3 receptor. 2. Identification of (S)-N-(1-phenylpropy1)-3-hydroxy-2--phenylquinoline-4-carboxamide (SB 223412). J Med. Chem. (1999) 42:1053–1065.
  • HARRISON T, KORSGAARD MPG, SWAIN CJ, CASCIERI MA, SADOWSKI S, SEABROOK GR: High affinity, selective neurokinin 2 and neurokinin 3 receptor antagonists from a common structural template. Bioorg. Med. Chem. Lett. (1998) 8:1343–1348.
  • GIARDINA GAM, RAVEGLIA LF: Neurokinin-3 receptor antagonists. Exp. Opin. Ther. Patents (1997) 7 (4):307–323.
  • GAO ZL, PEET NP: Recent advances in neurokinin receptor antagonists. CUIT. Med. Chem. (1999) 6:375–388.
  • KUDLACZ EM, SHATZER SA, KNIPPENBERG RW et al: In vitro and in vivo characterization of MDL 105, 212A, a nonpeptide NK-1 /NK-2 tachykinin receptor antago-nist. J. Pharmacol. Exp. Ther. (1996) 277:840–851.
  • •A pan-tachykinin receptor antagonist.
  • EGLEN RM, HUNTER JC, DRAY A: Ions in the fire: recent ion-channel research and approaches to pain therapy. Trends Pharmacol Sci. (1999) 20:337–342.
  • •All the latest on ion channels in pain.
  • BRYANS JS, WUSTROW DJ: 3-substituted GABA analogs with central nervous system activity: a review. Med. Res. Rev. (1999) 19:149–177.
  • CHIZH BA, DICKENSON AH, WNENDT S: The race to control pain: more participants, more targets. Trends Pharmacol. Sci. (1999) 20:354–357.
  • BOWERSOX SS, LUTHER R: Pharmacotherapeutic potential of omega-conotoxin MVIIA (SNX-111), an N-type neuronal calcium channel blocker found in the venom of Conus magus. Toxkon (1998) 36: 1651-1658.
  • NEUGEBAUER V, RUMENAPP P, SCHAIBLE HG: Calcitonin gene-related peptide is involved in the spinal processing of mechanosensory input from the rat's knee joint and in the generation and maintenance of hyperexcitability of dorsal horn neurons during development of acute inflammation. Neuroscience (1996) 71:1095–1109.
  • XU XJ, WIESENFELD-HALLIN Z: Calcitonin gene-related peptide (8-37) does not antagonize calcitonin gene-related peptide in rat spinal cord. Neurosci. Lett. (1996) 204:185–188.
  • DAINES RA, SHAM KKC, TAGGART JJ et al: Quinine analogs as non-peptide calcitonin gene-related peptide (CGRP) receptor antagonists. Bioorg. Med. Chem. Lett. (1997) 7:2673–2676.
  • •The nearest we have to selective non-peptide CGRP receptor antagonists.
  • FOORD SM, MARSHALL FH: RAMPs: accessory proteins for seven transmembrane domain receptors. Trends Pharmacol. Sci. (1999) 20:184–187.
  • •Why it is going to be difficult to design CGRP receptor ligands.
  • NYBERG F, LE GREVES P, TERENIUS L: Modulation of endopeptidase activity by calcitonin gene related peptide: a mechanism affecting substance P action? Biochimie (1988) 70:65–68.
  • SCHAIBLE HG, HOPE PJ, LANG CW, DUGGAN AW: Calcitonin gene-related peptide causes intraspinal spreading of substance P released by peripheral stimulation. Eur.J. Neurosci. (1992) 4:750–757.
  • OWOLABI JB, RIZKALLA G, TEHIM A et al.: Characteriza-tion of antiallodynic actions of ALE-0540, a novel nerve growth factor receptor antagonist, in the rat. J. Pharmacol. Exp. Ther. (1999) 289:1271–1276.
  • •A non-peptide blocker for NGF.
  • PORTENOY RK: Current pharmacotherapy of chronic pain. J. Pain Symptom Manag. (2000) 19:S16–S20
  • DUGGAN AW: Release of neuropeptides in the spinal cord. Prog. Brain Res. (1994) 104:197–223.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.