References

Aanonsen L.M., Wilcox G.L. Nociceptive action of excitatory amino acids in the mouse: effects of spinally administered opioids, phencyclidine and sigma agonists. Journal of Pharmacology and Experimental Therapeutics. 1987;243:9–19.

Aantaa R., Marjamaki A., Scheinin M. Molecular pharmacology of alpha 2-adrenoceptor subtypes. Annals of Medicine. 1995;27:439–449.

Abrams J.K., Johnson P.L., Hollis J.H., et al. Anatomic and functional topography of the dorsal raphe nucleus. Annals of the New York Academy of Sciences. 2004;1018:46–57.

Ahluwalia J., Urban L., Capogna M., et al. Cannabinoid 1 receptors are expressed in nociceptive primary sensory neurons. Neuroscience. 2000;100:685–688.

Aimi Y., Fujimura M., Vincent S.R., et al. Localization of NADPH-diaphorase–containing neurons in sensory ganglia of the rat. Journal of Comparative Neurology. 1991;306:382–392.

Albuquerque C., Lee C.J., Jackson A.C., et al. Subpopulations of GABAergic and non-GABAergic rat dorsal horn neurons express Ca2+-permeable AMPA receptors. European Journal of Neuroscience. 1999;11:2758–2766.

Alhaider A.A., Lei S.Z., Wilcox G.L. Spinal 5-HT3 receptor–mediated antinociception: possible release of GABA. Journal of Neuroscience. 1991;11:1881–1888.

Allen B.J., Rogers S.D., Ghilardi J.R., et al. Noxious cutaneous thermal stimuli induce a graded release of endogenous substance P in the spinal cord: imaging peptide action in vivo. Journal of Neuroscience. 1997;17:5921–5927.

Allen J.W., Yaksh T.L. Assessment of acute thermal nociception in laboratory animals. Methods in Molecular Medicine. 2004;99:11–24.

Almeida T.A., Rojo J., Nieto P.M., et al. Tachykinins and tachykinin receptors: structure and activity relationships. Current Medicinal Chemistry. 2004;11:2045–2081.

Anden N.E., Jukes M.G., Lundberg A. The effect of DOPA on the spinal cord. 2. A pharmacological analysis. Acta Physiologica Scandinavica. 1966;67:387–397.

Anderson C.R. NADPH diaphorase–positive neurons in the rat spinal cord include a subpopulation of autonomic preganglionic neurons. Neuroscience Letters. 1992;139:280–284.

Atlas D. Signaling role of the voltage-gated calcium channel as the molecular on/off switch of secretion. Cell Signal. 2010;22(11):1597–1603.

Baba H., Ji R.R., Kohno T., et al. Removal of GABAergic inhibition facilitates polysynaptic A fiber–mediated excitatory transmission to the superficial spinal dorsal horn. Molecular and Cellular Neurosciences. 2003;24:818–830.

Baba H., Kohno T., Okamoto M., et al. Muscarinic facilitation of GABA release in substantia gelatinosa of the rat spinal dorsal horn. Journal of Physiology. 1998;508:83–93.

Baba H., Shimoji K., Yoshimura M., et al. Norepinephrine facilitates inhibitory transmission in substantia gelatinosa of adult rat spinal cord (part 1): effects on axon terminals of GABAergic and glycinergic neurons. Anesthesiology. 2000;92:473–484.

Baber N.S., Dourish C.T., Hill D.R. The role of CCK caerulein, and CCK antagonists in nociception. Pain. 1989;39:307–328.

Balazs R., Miller S., Romano C., et al. Metabotropic glutamate receptor mGluR5 in astrocytes: pharmacological properties and agonist regulation. Journal of Neurochemistry. 1997;69:151–163.

Ballet S., Mauborgne A., Harmon M., et al. Altered opioid-mediated control of the spinal release of dynorphin and met-enkephalin in polyarthritic rats. Synapse. 2000;37:262–272.

Bandler R., Carrive P., Zhang S.P. Integration of somatic and autonomic reactions within the midbrain periaqueductal grey: viscerotopic, somatotopic and functional organization. Progress in Brain Research. 1991;87:269–305.

Barasi S., Duggal K.N. The effect of local and systemic application of dopaminergic agents on tail flick latency in the rat. European Journal of Pharmacology. 1985;117:287–294.

Barber R.P., Vaughn J.E., Saito K., et al. GABAergic terminals are presynaptic to primary afferent terminals in the substantia gelatinosa of the rat spinal cord. Brain Research. 1978;141:35–55.

Barbieri M., Nistri A. Depression of windup of spinal neurons in the neonatal rat spinal cord in vitro by an NK3 tachykinin receptor antagonist. Journal of Neurophysiology. 2001;85:1502–1511.

Bardin L., Bardin M., Lavarenne J., et al. Effect of intrathecal serotonin on nociception in rats: influence of the pain test used. Experimental Brain Research. 1997;113:81–87.

Bardoni R., Goldstein P.A., Lee C.J., et al. ATP P2X receptors mediate fast synaptic transmission in the dorsal horn of the rat spinal cord. Journal of Neuroscience. 1997;17:5297–5304.

Barnes N.M., Sharp T. A review of central 5-HT receptors and their function. Neuropharmacology. 1999;38:1083–1152.

Battaglia G., Monn J.A., Schoepp D.D. In vivo inhibition of veratridine-evoked release of striatal excitatory amino acids by the group II metabotropic glutamate receptor agonist LY354740 in rats. Neuroscience Letters. 1997;229:161–164.

Battaglia G., Rustioni A. Coexistence of glutamate and substance P in dorsal root ganglion neurons of the rat and monkey. Journal of Comparative Neurology. 1988;277:302–312.

Battaglia G., Rustioni A. Substance P innervation of the rat and cat thalamus. II. Cells of origin in the spinal cord. Journal of Comparative Neurology. 1992;315:473–486.

Battaglia G., Spreafico R., Rustioni A., et al. Substance P innervation of the rat and cat thalamus. I. Distribution and relation to ascending spinal pathways. Journal of Comparative Neurology. 1992;315:457–472.

Bauer M.K., Lieb K., Schulze-Osthoff K., et al. Expression and regulation of cyclooxygenase-2 in rat microglia. European Journal of Biochemistry. 1997;243:726–731.

Behbehani M.M., Fields H.L. Evidence that an excitatory connection between the periaqueductal gray and nucleus raphe magnus mediates stimulation produced analgesia. Brain Research. 1979;170:85–93.

Bennett A.D., Chastain K.M., Hulsebosch C.E. Alleviation of mechanical and thermal allodynia by CGRP(8–37) in a rodent model of chronic central pain. Pain. 2000;86:163–175.

Bernardini N., Levey A.I., Augusti-Tocco G., et al. Rat dorsal root ganglia express m1-m4 muscarinic receptor proteins. Journal of the Peripheral Nervous System. 1999;4:222–232.

Berridge C.W., Waterhouse B.D. The locus coeruleus–noradrenergic system: modulation of behavioral state and state-dependent cognitive processes. Brain Research. Brain Research Reviews. 2003;42:33–84.

Berthele A., Boxall S.J., Urban A., et al. Distribution and developmental changes in metabotropic glutamate receptor messenger RNA expression in the rat lumbar spinal cord. Brain Research. Developmental Brain Research. 1999;112:39–53.

Biella G., Panara C., Pecile A., et al. Facilitatory role of calcitonin gene–related peptide (CGRP) on excitation induced by substance P (SP) and noxious stimuli in rat spinal dorsal horn neurons. An iontophoretic study in vivo. Brain Research. 1991;559:352–356.

Bird G.C., Han J.S., Fu Y., et al. Pain-related synaptic plasticity in spinal dorsal horn neurons: role of CGRP. Molecular Pain. 2006;2:31.

Bird G.C., Lash L.L., Han J.S., et al. Protein kinase A–dependent enhanced NMDA receptor function in pain-related synaptic plasticity in rat amygdala neurones. Journal of Physiology. 2005;564:907–921.

Birder L.A., Perl E.R. Expression of alpha2-adrenergic receptors in rat primary afferent neurones after peripheral nerve injury or inflammation. Journal of Physiology. 1999;515:533–542.

Birnbaumer L., Abramowitz J., Brown A.M. Receptor-effector coupling by G proteins. Biochimica et Biophysica Acta. 1990;1031:163–224.

Blessing W.W. Lower brainstem pathways regulating sympathetically mediated changes in cutaneous blood flow. Cellular and molecular neurobiology. 2003;23(4-5):527–538.

Block C.H., Hoffman G., Kapp B.S., et al. Peptide-containing pathways from the parabrachial complex to the central nucleus of the amygdala. Peptides. 1989;10:465–471.

Boehm S. ATP stimulates sympathetic transmitter release via presynaptic P2X purinoceptors. Journal of Neuroscience. 1999;19:737–746.

Bohlhalter S., Weinmann O., Mohler H., et al. Laminar compartmentalization of GABAA-receptor subtypes in the spinal cord: an immunohistochemical study. Journal of Neuroscience. 1996;16:283–297.

Borges L.F., Iversen S.D. Topography of choline acetyltransferase immunoreactive neurons and fibers in the rat spinal cord. Brain Research. 1986;362:140–148.

Bourgoin S., Le Bars D., Clot A.M., et al. Subcutaneous formalin induces a segmental release of Met-enkephalin–like material from the rat spinal cord. Pain. 1990;41:323–329.

Bowker R.M., Westlund K.N., Sullivan M.C., et al. Descending serotonergic, peptidergic and cholinergic pathways from the raphe nuclei: a multiple transmitter complex. Brain Research. 1983;288:33–48.

Bowsher D. Role of the reticular formation in responses to noxious stimulation. Pain. 1976;2:361–378.

Boxall S.J., Berthele A., Laurie D.J., et al. Enhanced expression of metabotropic glutamate receptor 3 messenger RNA in the rat spinal cord during ultraviolet irradiation induced peripheral inflammation. Neuroscience. 1998;82:591–602.

Boyce S., Wyatt A., Webb J.K., 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.

Branchek T.A., Smith K.E., Gerald C., et al. Galanin receptor subtypes. Trends in Pharmacological Sciences. 2000;21:109–117.

Breitinger H.G., Becker C.M. The inhibitory glycine receptor—simple views of a complicated channel. Chembiochem. 2002;3:1042–1052.

Broman J., Anderson S., Ottersen O.P., et al. Enrichment of glutamate-like immunoreactivity in primary afferent terminals throughout the spinal cord dorsal horn. European Journal of Neuroscience. 1993;5:1050–1061.

Buczynski M.W., Dumlao D.S., et al. Thematic Review Series: Proteomics. An integrated omics analysis of eicosanoid biology. Journal of lipid research. 2009;50(6):1015–1038.

Buczynski M.W., Svensson C.I., Dumlao D.S., et al. Inflammatory hyperalgesia induces essential bioactive lipid production in the spinal cord. Journal of Neurochemistry. 2010;114:981–993.

Burke J.E., Dennis E.A. Phospholipase A2 structure/function, mechanism, and signaling. Journal of lipid research. 2009;50:S237–S242. Suppl

Bylund D.B. Pharmacological characteristics of alpha-2 adrenergic receptor subtypes. Annals of the New York Academy of Sciences. 1995;763:1–7.

Camarata P.J., Yaksh T.L. Characterization of the spinal adrenergic receptors mediating the spinal effects produced by the microinjection of morphine into the periaqueductal gray. Brain Research. 1985;336:133–142.

Cardenas C.G., Del Mar L.P., Scroggs R.S. Two parallel signaling pathways couple 5HT1A receptors to N- and L-type calcium channels in C-like rat dorsal root ganglion cells. Journal of Neurophysiology. 1997;77:3284–3296.

Carlton S.M., Hargett G.L., Coggeshall R.E. Plasticity in alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunits in the rat dorsal horn following deafferentation. Neuroscience Letters. 1998;242:21–24.

Carlton S.M., Hayes E.S. Light microscopic and ultrastructural analysis of GABA-immunoreactive profiles in the monkey spinal cord. Journal of Comparative Neurology. 1990;300:162–182.

Carlton S.M., Westlund K.N., Zhang D., et al. GABA-immunoreactive terminals synapse on primate spinothalamic tract cells. Journal of Comparative Neurology. 1992;322:528–537.

Casey K.L. Escape elicited by bulboreticular stimulation in the cat. International Journal of Neuroscience. 1971;2:29–34.

Caulfield M.P., Brown D.A. Cannabinoid receptor agonists inhibit Ca current in NG108-15 neuroblastoma cells via a pertussis toxin-sensitive mechanism. Br J Pharmacol. 1992;106(2):231–232.

Caulfield M.P. Muscarinic receptors—characterization, coupling and function. Pharmacology and Therapeutics. 1993;58:319–379.

Caulfield M.P., Birdsall N.J. International Union of Pharmacology. XVII. Classification of muscarinic acetylcholine receptors. Pharmacological Reviews. 1998;50:279–290.

Cesselin F., Le Bars D., et al. Spontaneous and evoked release of methionine-enkephalin-like material from the rat spinal cord in vivo. Brain Res. 1985;339(2):305–313.

Cesselin F., Bourgoin S., Clot A.M., et al. Segmental release of Met-enkephalin–like material from the spinal cord of rats, elicited by noxious thermal stimuli. Brain Research. 1989;484:71–77.

Chain D.G., Casadio A., et al. Mechanisms for generating the autonomous cAMP-dependent protein kinase required for long-term facilitation in Aplysia. Neuron. 1999;22(1):147–156.

Chaplan S.R., Malmberg A.B., Yaksh T.L., et al. Efficacy of spinal NMDA receptor antagonism in formalin hyperalgesia and nerve injury evoked allodynia in the rat. Journal of Pharmacology and Experimental Therapeutics. 1997;280:829–838.

Chapman V. The cannabinoid CB1 receptor antagonist, SR141716A, selectively facilitates nociceptive responses of dorsal horn neurones in the rat. British Journal of Pharmacology. 1999;127:1765–1767.

Chapman V., Dickenson A.H. The effects of Sandostatin and somatostatin on nociceptive transmission in the dorsal horn of the rat spinal cord. Neuropeptides. 1992;23:147–152.

Chapman V., Dickenson A.H. The effect of intrathecal administration of RP67580, a potent neurokinin 1 antagonist on nociceptive transmission in the rat spinal cord. Neuroscience Letters. 1993;157:149–152.

Chapman V., Dickenson A.H. Time-related roles of excitatory amino acid receptors during persistent noxiously evoked responses of rat dorsal horn neurones. Brain Research. 1995;703:45–50.

Chebib M., Johnston G.A. The “ABC” of GABA receptors: a brief review. Clinical and Experimental Pharmacology and Physiology. 1999;26:937–940.

Chen S.R., Pan H.L. Activation of muscarinic receptors inhibits spinal dorsal horn projection neurons: role of GABAB receptors. Neuroscience. 2004;125:141–148.

Choca J.I., Green R.D., Proudfit H.K., et al. Adenosine A1 and A2 receptors of the substantia gelatinosa are located predominantly on intrinsic neurons: an autoradiography study. Journal of Pharmacology and Experimental Therapeutics. 1988;247:757–764.

Choi J.I., Svensson C.I., Koehrn F.J., et al. Peripheral inflammation induces tumor necrosis factor dependent AMPA receptor trafficking and Akt phosphorylation in spinal cord in addition to pain behavior. Pain. 2010;149:243–253.

Chung J.M., Lee K.H., Surmeier D.J., et al. Response characteristics of neurons in the ventral posterior lateral nucleus of the monkey thalamus. Journal of Neurophysiology. 1986;56:370–390.

Clark A.K., Staniland A.A., Malcongio M., et al. Fractalkine/CX3CR1 signalling in chronic pain and inflammation. Current Pharmaceutical Biotechnology. 2011;12:1707–1714.

Coderre T.J., Gonzales R., Goldyne M.E., et al. Noxious stimulus–induced increase in spinal prostaglandin E2 is noradrenergic terminal–dependent. Neuroscience Letters. 1990;115:253–258.

Coderre T.J., Melzack R. The contribution of excitatory amino acids to central sensitization and persistent nociception after formalin-induced tissue injury. Journal of Neuroscience. 1992;12:3665–3670.

Coderre T.J., Melzack R. The role of NMDA receptor–operated calcium channels in persistent nociception after formalin-induced tissue injury. Journal of Neuroscience. 1992;12:3671–3675.

Conn P.J., Pin J.P. Pharmacology and functions of metabotropic glutamate receptors. Annual Review of Pharmacology and Toxicology. 1997;37:205–237.

Cordero-Erausquin M., Changeux J.P. Tonic nicotinic modulation of serotoninergic transmission in the spinal cord. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:2803–2807.

Cordero-Erausquin M., Pons S., Faure P., et al. Nicotine differentially activates inhibitory and excitatory neurons in the dorsal spinal cord. Pain. 2004;109:308–318.

Craig A.D. The functional anatomy of lamina I and its role in post-stroke central pain. Progress in Brain Research. 2000;129:137–151.

Cridland R.A., Henry J.L. Comparison of the effects of substance P, neurokinin A, physalaemin and eledoisin in facilitating a nociceptive reflex in the rat. Brain Research. 1986;381:93–99.

Cridland R.A., Henry J.L. Effects of intrathecal administration of neuropeptides on a spinal nociceptive reflex in the rat: VIP, galanin, CGRP, TRH, somatostatin and angiotensin II. Neuropeptides. 1988;11:23–32.

Cui M., Feng Y., McAdoo D.J., et al. Periaqueductal gray stimulation–induced inhibition of nociceptive dorsal horn neurons in rats is associated with the release of norepinephrine, serotonin, and amino acids. Journal of Pharmacology and Experimental Therapeutics. 1999;289:868–876.

Curtis D.R., Hosli L., Johnston G.A., et al. Inhibition of spinal neurons by glycine. Nature. 1967;215:1502–1503.

Cozzi A., Attucci S., et al. Type 2 metabotropic glutamate (mGlu) receptors tonically inhibit transmitter release in rat caudate nucleus: in vivo studies with (2S,1’S,2’S,3’R)-2-(2’-carboxy-3’-phenylcyclopropyl)glycine, a new potent and selective antagonist. The European journal of neuroscience. 1997;9(7):1350–1355.

Dahlstrom A., Fuxe K. Evidence for the existence of monoamine containing neurons in the central nervous system. I. Demonstration of monoamines in the cell bodies of brain stem neurons. Acta Physiologica Scandinavica. Supplementum. 1964;232:1–55.

Damaj M.I., Meyer E.M., Martin B.R., et al. The antinociceptive effects of alpha7 nicotinic agonists in an acute pain model. Neuropharmacology. 2000;39:2785–2791.

Davies J., Watkins J.C. Role of excitatory amino acid receptors in mono- and polysynaptic excitation in the cat spinal cord. Experimental Brain Research. 1983;49:280–290.

Davies S.N., Lodge D. Evidence for involvement of N-methylaspartate receptors in “wind-up” of class 2 neurones in the dorsal horn of the rat. Brain Research. 1987;424:402–406.

De Biasi S., Rustioni A. Glutamate and substance P coexist in primary afferent terminals in the superficial laminae of spinal cord. Proceedings of the National Academy of Sciences of the United States of America. 1988;85:7820–7824.

De Koninck Y. Altered chloride homeostasis in neurological disorders: a new target. Current Opinion in Pharmacology. 2007;7:93–99.

De Koninck Y., Henry J.L. Substance P–mediated slow excitatory postsynaptic potential elicited in dorsal horn neurons in vivo by noxious stimulation. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:11344–11348.

De Koninck Y., Henry J.L. Prolonged GABAA-mediated inhibition following single hair afferent input to single spinal dorsal horn neurones in cats. Journal of Physiology. 1994;476:89–100.

Deadwyler S.A., Hampson R.E., et al. Cannabinoids modulate voltage sensitive potassium A-current in hippocampal neurons via a cAMP-dependent process. J Pharmacol Exp Ther. 1995;273(2):734–743.

Delfs J.R., Dichter M.A. Effects of somatostatin on mammalian cortical neurons in culture: physiological actions and unusual dose–response characteristics. Journal of Neuroscience. 1983;3:1176–1188.

Denny-Brown D., Kirk E.J., Yanagisawa N. The tract of Lissauer in relation to sensory transmission in the dorsal horn of spinal cord in the macaque monkey. Journal of Comparative Neurology. 1973;151:175–200.

Deuchars S.A., Atkinson L., Brooke R.E., et al. Neuronal P2X7 receptors are targeted to presynaptic terminals in the central and peripheral nervous systems. Journal of Neuroscience. 2001;21:7143–7152.

Deuchars S.A., Brooke R.E., Deuchars J., et al. Adenosine A1 receptors reduce release from excitatory but not inhibitory synaptic inputs onto lateral horn neurons. Journal of Neuroscience. 2001;21:6308–6320.

Devane W.A., Hanus L., et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992;258(5090):1946–1949.

Dickenson A., Where and how do opioids act?. Gebhart G., Hammond D., Jensen T., eds. Proceedings of the 7th World Congress on Pain. Progress in pain research and management, vol 2. Seattle: IASP Press; 1994:525–552.

Dickenson A.H., Aydar E. Antagonism at the glycine site on the NMDA receptor reduces spinal nociception in the rat. Neuroscience Letters. 1991;121:263–266.

Dickenson A.H., Sullivan A.F. Evidence for a role of the NMDA receptor in the frequency dependent potentiation of deep rat dorsal horn nociceptive neurones following C fibre stimulation. Neuropharmacology. 1987;26:1235–1238.

Dickenson A.H., Sullivan A.F. Differential effects of excitatory amino acid antagonists on dorsal horn nociceptive neurones in the rat. Brain Research. 1990;506:31–39.

Dickinson T., Fleetwood-Walker S.M. VIP and PACAP: very important in pain? Trends in Pharmacological Sciences. 1999;20:324–329.

Dickinson T., Fleetwood-Walker S.M., Mitchell R., et al. Evidence for roles of vasoactive intestinal polypeptide (VIP) and pituitary adenylate cyclase activating polypeptide (PACAP) receptors in modulating the responses of rat dorsal horn neurons to sensory inputs. Neuropeptides. 1997;31:175–185.

Dickinson T., Mitchell R., Robberecht P., et al. The role of VIP/PACAP receptor subtypes in spinal somatosensory processing in rats with an experimental peripheral mononeuropathy. Neuropharmacology. 1999;38:167–180.

Diez Guerra F.J., Zaidi M., Bevis P., et al. Evidence for release of calcitonin gene–related peptide and neurokinin A from sensory nerve endings in vivo. Neuroscience. 1988;25:839–846.

Ding Y.Q., Lu C.R., Wang H., et al. Two major distinct subpopulations of neurokinin-3 receptor–expressing neurons in the superficial dorsal horn of the rat spinal cord. European Journal of Neuroscience. 2002;16:551–556.

Di Luca M., Gardoni F., Finardi A., et al. NMDA receptor subunits are phosphorylated by activation of neurotrophin receptors in PSD of rat spinal cord. Neuroreport. 2001;12:1301–1305.

Dirig D.M., Yaksh T.L. Intrathecal baclofen and muscimol, but not midazolam, are antinociceptive using the rat-formalin model. Journal of Pharmacology and Experimental Therapeutics. 1995;275:219–227.

Dirig D.M., Yaksh T.L. In vitro prostanoid release from spinal cord following peripheral inflammation: effects of substance P, NMDA and capsaicin. British Journal of Pharmacology. 1999;126:1333–1340.

Djouhri L., Lawson S.N. Abeta-fiber nociceptive primary afferent neurons: a review of incidence and properties in relation to other A-fiber neurons in mammals. Brain Research Reviews. 2004;46:131–145.

Dogrul A., Ossipov M.H., Porreca F. Differential mediation of descending pain facilitation and inhibition by spinal 5HT-3 and 5HT-7 receptors. Brain Research. 2009;1280:52–59.

Dolan S., Nolan A.M. Behavioral evidence supporting a differential role for spinal group I and II metabotropic glutamate receptors in inflammatory hyperalgesia in sheep. Neuropharmacology. 2002;43:319–326.

Doly S., Madeira A., Fischer J., et al. The 5-HT2A receptor is widely distributed in the rat spinal cord and mainly localized at the plasma membrane of postsynaptic neurons. Journal of Comparative Neurology. 2004;472:496–511.

Donaldson L.F., Humphrey P.S., Oldfield S., et al. Expression and regulation of prostaglandin E receptor subtype mRNAs in rat sensory ganglia and spinal cord in response to peripheral inflammation. Prostaglandins and Other Lipid Mediators. 2001;63:109–122.

Dopp J.M., Mackenzie-Graham A., Otero G.C., et al. Differential expression, cytokine modulation, and specific functions of type-1 and type-2 tumor necrosis factor receptors in rat glia. Journal of Neuroimmunology. 1997;75:104–112.

Dougherty P.M., 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, chemical, thermal, and electrical stimuli. Journal of Neuroscience. 1992;12:3025–3041.

Dougherty P.M., Palecek J., Zorn S., et al. Combined application of excitatory amino acids and substance P produces long-lasting changes in responses of primate spinothalamic tract neurons. Brain Research. Brain Research Reviews. 1993;18:227–246.

Du D., Eisenach J.C., Ririe D.G., et al. The antinociceptive effects of spinal cyclooxygenase inhibitors on uterine cervical distension. Brain Research. 2004;1024:130–136.

Du J., Zhou S., Carlton S.M. Group II metabotropic glutamate receptor activation attenuates peripheral sensitization in inflammatory states. Neuroscience. 2008;154:754–766.

Duggan A.W., Hendry I.A., Morton C.R., et al. Cutaneous stimuli releasing immunoreactive substance P in the dorsal horn of the cat. Brain Research. 1988;451:261–273.

Duggan A.W., Hope P.J., Jarrot B., et al. Release, spread and persistence of immunoreactive neurokinin A in the dorsal horn of the cat following noxious cutaneous stimulation. Studies with antibody microprobes. Neuroscience. 1990;35:195–202.

Dun N.J., Miyazaki T., Tang H., et al. Pituitary adenylate cyclase activating polypeptide immunoreactivity in the rat spinal cord and medulla: implication of sensory and autonomic functions. Neuroscience. 1996;73:677–686.

Dunwiddie T.V., Masino S.A. The role and regulation of adenosine in the central nervous system. Annual Review of Neuroscience. 2001;24:31–55.

Edsall S.A., Knapp R.J., et al. Antisense oligodeoxynucleotide treatment to the brain cannabinoid receptor inhibits antinociception. Neuroreport. 1996;7(2):593–596.

Edwards F.A., Gibb A.J., Colquhoun D., et al. ATP receptor–mediated synaptic currents in the central nervous system. Nature. 1992;359:144–147.

Eisenach J.C., De Kock M., Klimscha W., et al. Alpha(2)-adrenergic agonists for regional anesthesia. A clinical review of clonidine (1984-1995). Anesthesiology. 1996;85:655–674.

Emmers R. Thalamic mechanisms that process a temporal pulse code for pain. Brain Research. 1976;103:425–441.

Engelman H.S., Allen T.B., MacDermott A.B. The distribution of neurons expressing calcium-permeable AMPA receptors in the superficial laminae of the spinal cord dorsal horn. Journal of Neuroscience. 1999;19:2081–2089.

Ericson A.C., Blomqvist A., Craig A.D., et al. Evidence for glutamate as neurotransmitter in trigemino- and spinothalamic tract terminals in the nucleus submedius of cats. European Journal of Neuroscience. 1995;7:305–317.

Eschalier A., Kayser V., Guilbaud G., et al. Influence of a specific 5-HT3 antagonist on carrageenan-induced hyperalgesia in rats. Pain. 1989;36:249–255.

Fagni L., Ango F., Perroy J., et al. Identification and functional roles of metabotropic glutamate receptor–interacting proteins. Seminars in Cell and Developmental Biology. 2004;15:289–298.

Fagni L., Chavis P., Ango F., et al. Complex interactions between mGluRs, intracellular Ca2+ stores and ion channels in neurons. Trends in Neurosciences. 2000;23:80–88.

Fairbanks C.A., Stone L.S., Kitto K.F., et al. Alpha(2C)-adrenergic receptors mediate spinal analgesia and adrenergic-opioid synergy. Journal of Pharmacology and Experimental Therapeutics. 2002;300:282–290.

Fang L., Wu J., Lin Q., et al. Calcium-calmodulin–dependent protein kinase II contributes to spinal cord central sensitization. Journal of Neuroscience. 2002;22:4196–4204.

Fang L., Wu J., Lin Q., et al. Protein kinases regulate the phosphorylation of the GluR1 subunit of AMPA receptors of spinal cord in rats following noxious stimulation. Brain Research. Molecular Brain Research. 2003;118:160–165.

Fardin V., Oliveras J.L., Besson J.M. A reinvestigation of the analgesic effects induced by stimulation of the periaqueductal gray matter in the rat. II. Differential characteristics of the analgesia induced by ventral and dorsal PAG stimulation. Brain Research. 1984;306:125–139.

Faris P.L., Komisaruk B.R., Watkins L.R., et al. Evidence for the neuropeptide cholecystokinin as an antagonist of opiate analgesia. Science. 1983;219:310–312.

Farquhar-Smith W.P., Egertova M., et al. Cannabinoid CB(1) receptor expression in rat spinal cord. Mol Cell Neurosci. 2000;15(6):510–521.

Felder C.C., Joyce K.E., Briley E.M., et al. Comparison of the pharmacology and signal transduction of the human cannabinoid CB1 and CB2 receptors. Molecular Pharmacology. 1995;48:443–450.

Fellin T., Carmignoto G. Neurone-to-astrocyte signalling in the brain represents a distinct multifunctional unit. Journal of Physiology. 2004;559:3–15.

Fields R.D. Volume transmission in activity-dependent regulation of myelinating glia. Molecular Pharmacology. 2004;45:503–509.

Fisher K., Coderre T.J. The contribution of metabotropic glutamate receptors (mGluRs) to formalin-induced nociception. Pain. 1996;68:255–263.

Fisher K., Coderre T.J. Hyperalgesia and allodynia induced by intrathecal (RS)-dihydroxyphenylglycine in rats. Neuroreport. 1998;9:1169–1172.

Fleetwood-Walker S.M., Hope P.J., Mitchell R., et al. The influence of opioid receptor subtypes on the processing of nociceptive inputs in the spinal dorsal horn of the cat. Brain Research. 1988;451:213–226.

Fleetwood-Walker S.M., Mitchell R., Hope P.J., et al. An alpha 2 receptor mediates the selective inhibition by noradrenaline of nociceptive responses of identified dorsal horn neurones. Brain Research. 1985;334:243–254.

Flores C.M. The promise and pitfalls of a nicotinic cholinergic approach to pain management. Pain. 2000;88:1–6.

Fredholm B.B., Ap I.J., Jacobson K.A., et al. International Union of Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacological Reviews. 2001;53:527–552.

Fujimoto J.M., Arts K.S., Rady J.J., et al. Spinal dynorphin A (1–17): possible mediator of antianalgesic action. Neuropharmacology. 1990;29:609–617.

Fyffe R.E., Perl E.R. Is ATP a central synaptic mediator for certain primary afferent fibers from mammalian skin? Proceedings of the National Academy of Sciences of the United States of America. 1984;81:6890–6893.

Game C.J., Lodge D. The pharmacology of the inhibition of dorsal horn neurones by impulses in myelinated cutaneous afferents in the cat. Experimental Brain Research. 1975;23:75–84.

Gamse R., Lackner D., Gamse G., et al. Effect of capsaicin pretreatment on capsaicin-evoked release of immunoreactive somatostatin and substance P from primary sensory neurons. Naunyn-Schmiedeberg’s Archives of Pharmacology. 1981;316:38–41.

Gao X., Kim H.K., Chung J.M., et al. Enhancement of NMDA receptor phosphorylation of the spinal dorsal horn and nucleus gracilis neurons in neuropathic rats. Pain. 2005;116:62–72.

Gao Y.J., Zhang Y.Q., Zhao Z.Q. Involvement of spinal neurokinin-1 receptors in the maintenance but not induction of carrageenan-induced thermal hyperalgesia in the rat. Brain Research Bulletin. 2003;61:587–593.

Garcia P.S., Gulati A., Levy J.H. The role of thrombin and protease-activated receptors in pain mechanisms. Thrombosis and Haemostasis. 2010;103:1145–1151.

Garry M.G., Hargreaves K.M. Enhanced release of immunoreactive CGRP and substance P from spinal dorsal horn slices occurs during carrageenan inflammation. Brain Research. 1992;582:139–142.

Garthwaite J., Charles S.L., Chess-Williams R. Endothelium-derived relaxing factor release on activation of NMDA receptors suggests role as intercellular messenger in the brain. Nature. 1988;336:385–388.

Gaudreau G.A., Plourde V. Role of tachykinin NK1, NK2 and NK3 receptors in the modulation of visceral hypersensitivity in the rat. Neuroscience Letters. 2003;351:59–62.

Gaudreau G.A., Plourde V. Involvement of N-methyl-d-aspartate (NMDA) receptors in a rat model of visceral hypersensitivity. Behavioural Brain Research. 2004;150:185–189.

Gaumann D.M., Yaksh T.L. Intrathecal somatostatin in rats: antinociception only in the presence of toxic effects. Anesthesiology. 1988;68:733–742.

Gaumann D.M., Yaksh T.L., Post C., et al. Intrathecal somatostatin in cat and mouse studies on pain, motor behavior, and histopathology. Anesthesia and Analgesia. 1989;68:623–632.

Gaumann D.M., Yaksh T.L., Tyce G.M., et al. Effects of intrathecal morphine, clonidine, and midazolam on the somato-sympathoadrenal reflex response in halothane-anesthetized cats. Anesthesiology. 1990;73:42–432.

Gebhart G.F., Sandkuhler J., Thalhammer J.G., et al. Quantitative comparison of inhibition in spinal cord of nociceptive information by stimulation in periaqueductal gray or nucleus raphe magnus of the cat. Journal of Neurophysiology. 1983;50:1433–1445.

Gebhart G.F., Sandkuhler J., Thalhammer J.G., et al. Inhibition in spinal cord of nociceptive information by electrical stimulation and morphine microinjection at identical sites in midbrain of the cat. Journal of Neurophysiology. 1984;51:75–89.

Geiger J.D., LaBella F.S., Nagy J.I. Characterization and localization of adenosine receptors in rat spinal cord. Journal of Neuroscience. 1984;4:2303–2310.

Genzen J.R., McGehee D.S. Short- and long-term enhancement of excitatory transmission in the spinal cord dorsal horn by nicotinic acetylcholine receptors. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:6807–6812.

Gerber G., Randic M. Excitatory amino acid–mediated components of synaptically evoked input from dorsal roots to deep dorsal horn neurons in the rat spinal cord slice. Neuroscience Letters. 1989;106:211–219.

Gerber G., Randic M. Participation of excitatory amino acid receptors in the slow excitatory synaptic transmission in the rat spinal dorsal horn in vitro. Neuroscience Letters. 1989;106:220–228.

Gerber G., Youn D.H., Hsu C.H., et al. Spinal dorsal horn synaptic plasticity: involvement of group I metabotropic glutamate receptors. Progress in Brain Research. 2000;129:115–134.

Gerber G., Zhong J., Youn D., et al. Group II and group III metabotropic glutamate receptor agonists depress synaptic transmission in the rat spinal cord dorsal horn. Neuroscience. 2000;100:393–406.

Ghilardi J.R., Svensson C.I., Rogers S.D., et al. Constitutive spinal cyclooxygenase-2 participates in the initiation of tissue injury-induced hyperalgesia. Journal of Neuroscience. 2004;24:2727–2732.

Gmelin G., Zimmermann M. Effects of gamma-aminobutyrate and bicuculline on primary afferent depolarization of cutaneous fibres in the cat spinal cord. Neuroscience. 1983;10:869–874.

Go V.L., Yaksh T.L. Release of substance P from the cat spinal cord. Journal of Physiology. 1987;391:141–167.

Gold M.S., Levine J.D., Correa A.M. Modulation of TTX-R INa by PKC and PKA and their role in PGE2-induced sensitization of rat sensory neurons in vitro. Journal of Neuroscience. 1998;18:10345–10355.

Gomez-Villafuertes R., Gualix J., Miras-Portugal M.T. Single GABAergic synaptic terminals from rat midbrain exhibit functional P2X and dinucleotide receptors, able to induce GABA secretion. Journal of Neurochemistry. 2001;77:84–93.

Gormsen L., Finnerup N.B., et al. “The efficacy of the AMPA receptor antagonist NS1209 and lidocaine in nerve injury pain: a randomized, double-blind, placebo-controlled, three-way crossover study. Anesthesia and analgesia. 2009;108(4):1311–1319.

Gregus A.M., Doolen S., et al. Spinal 12-lipoxygenase-derived hepoxilin A3 contributes to inflammatory hyperalgesia via activation of TRPV1 and TRPA1 receptors. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(17):6721–6726.

Grudt T.J., Williams J.T., Travagli R.A. Inhibition by 5-hydroxytryptamine and noradrenaline in substantia gelatinosa of guinea-pig spinal trigeminal nucleus. Journal of Physiology. 1995;485:113–120.

Gu J.G. P2X receptor–mediated modulation of sensory transmission to the spinal cord dorsal horn. Neuroscientist. 2003;9:370–378.

Gu J.G., Albuquerque C., Lee C.J., et al. Synaptic strengthening through activation of Ca2+-permeable AMPA receptors. Nature. 1996;381:793–796.

Guindon J., Hohmann A.G. The endocannabinoid system and pain. CNS & Neurological Disorders Drug Targets. 2009;8:403–421.

Gundlach A.L., Dodd P.R., Grabara C.S., et al. Deficit of spinal cord glycine/strychnine receptors in inherited myoclonus of poll Hereford calves. Science. 1988;241:1807–1810.

Guo W., Wei F., Zou S., et al. Group I metabotropic glutamate receptor NMDA receptor coupling and signaling cascade mediate spinal dorsal horn NMDA receptor 2B tyrosine phosphorylation associated with inflammatory hyperalgesia. Journal of Neuroscience. 2004;24:9161–9173.

Haberberger R.V., Bernardini N., Kress M., et al. Nicotinic acetylcholine receptor subtypes in nociceptive dorsal root ganglion neurons of the adult rat. Autonomic Neuroscience: Basic & Clinical. 2004;113:32–42.

Haberberger R.V., Henrich M., Couraud J.Y., et al. Muscarinic M2-receptors in rat thoracic dorsal root ganglia. Neuroscience Letters. 1999;266:177–180.

Hague C., Chen Z., Uberti M., et al. Alpha(1)-adrenergic receptor subtypes: non-identical triplets with different dancing partners? Life Sciences. 2003;74:411–418.

Haley J.E., Dickenson A.H., Schachter M., et al. Electrophysiological evidence for a role of nitric oxide in prolonged chemical nociception in the rat. Neuropharmacology. 1992;31:251–258.

Hama A., Woon Lee J., Sagen J. Differential efficacy of intrathecal NMDA receptor antagonists on inflammatory mechanical and thermal hyperalgesia in rats. European Journal of Pharmacology. 2003;459:49–58.

Hammond D.L. GABA(B) receptors: new tricks by an old dog. Current Opinion in Pharmacology. 2001;1:26–30.

Hammond D.L., Tyce G.M., Yaksh T.L., et al. Efflux of 5-hydroxytryptamine and noradrenaline into spinal cord superfusates during stimulation of the rat medulla. Journal of Physiology. 1985;359:151–162.

Hammond D.L., Washington J.D. Antagonism of L-baclofen–induced antinociception by CGP 35348 in the spinal cord of the rat. European Journal of Pharmacology. 1993;234:255–262.

Hamon M., Gallissot M.C., Menard F., et al. 5-HT3 receptor binding sites are on capsaicin-sensitive fibres in the rat spinal cord. European Journal of Pharmacology. 1989;164:315–322.

Hantman A.W., van den Pol A.N., Perl E.R. Morphological and physiological features of a set of spinal substantia gelatinosa neurons defined by green fluorescent protein expression. Journal of Neuroscience. 2004;24:836–842.

Hao J.X., Xu X.J., Yu Y.X., et al. Transient spinal cord ischemia induces temporary hypersensitivity of dorsal horn wide dynamic range neurons to myelinated, but not unmyelinated, fiber input. Journal of Neurophysiology. 1992;68:384–391.

Hao J.X., Xu X.J., Yu X.Y., et al. Baclofen reverses the hypersensitivity of dorsal horn wide dynamic range neurons to mechanical stimulation after transient spinal cord ischemia; implications for a tonic GABAergic inhibitory control of myelinated fiber input. Journal of Neurophysiology. 1992;68:392–396.

Hartmann B., Ahmadi S., Heppenstall P.A., et al. The AMPA receptor subunits GluR-A and GluR-B reciprocally modulate spinal synaptic plasticity and inflammatory pain. Neuron. 2004;44:637–650.

Harvey R.J., Depner U.B., Wassle H., et al. GlyR alpha3: an essential target for spinal PGE2-mediated inflammatory pain sensitization. Science. 2004;304:884–887.

Hay D.L., Conner A.C., Howitt S.G., et al. The pharmacology of adrenomedullin receptors and their relationship to CGRP receptors. Journal of Molecular Neuroscience. 2004;22:105–113.

Headley P.M., Parsons C.G., West D.C. The role of N-methylaspartate receptors in mediating responses of rat and cat spinal neurones to defined sensory stimuli. Journal of Physiology. 1987;385:169–188.

Heapy C.G., Jamieson A., Russell N.J.W. Afferent C-fiber and A-delta activity in models of inflammation [abstract]. British Journal of Pharmacology. 1987;90:164P.

Hecker M., Mulsch A., Busse R. Subcellular localization and characterization of neuronal nitric oxide synthase. Journal of Neurochemistry. 1994;62:1524–1529.

Herkenham M., Lynn A.B., et al. Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci. 1991;11(2):563–583.

Hevers W., Luddens H. The diversity of GABAA receptors. Pharmacological and electrophysiological properties of GABAA channel subtypes. Molecular Neurobiology. 1998;18:35–86.

Hide I., Tanaka M., Inoue A., et al. Extracellular ATP triggers tumor necrosis factor-alpha release from rat microglia. Journal of Neurochemistry. 2000;75:965–972.

Hieble J.P., Bylund D.B., Clarke D.E., et al. International Union of Pharmacology. X. Recommendation for nomenclature of alpha 1-adrenoceptors: consensus update. Pharmacological Reviews. 1995;47:267–270.

Hislop J.N., Henry A.G., et al. Ubiquitination regulates proteolytic processing of G protein-coupled receptors after their sorting to lysosomes. The Journal of biological chemistry. 2009;284(29):19361–19370.

Hiura A., Nasu F., Ishizuka H. Relationship of substance P– and CGRP-immunoreactive central endings of the primary afferent neurons to GABAergic interneurons in the guinea pig substantia gelatinosa. Okajimas Folia Anatomica Japonica. 1998;74:231–235.

Hoglund A.U., Baghdoyan H.A. M2, M3 and M4, but not M1, muscarinic receptor subtypes are present in rat spinal cord. Journal of Pharmacology and Experimental Therapeutics. 1997;281:470–477.

Hohmann A.G., Herkenham M. Regulation of cannabinoid and mu opioid receptors in rat lumbar spinal cord following neonatal capsaicin treatment. Neurosci Lett. 1998;252(1):13–16.

Hohmann A.G., Briley E.M., et al. Pre- and postsynaptic distribution of cannabinoid and mu opioid receptors in rat spinal cord. Brain Res. 1999;822(1-2):17–25.

Hohmann A.G. Spinal and peripheral mechanisms of cannabinoid antinociception: behavioral, neurophysiological and neuroanatomical perspectives. Chemistry and Physics of Lipids. 2002;121:173–190.

Hokfelt T. Neuropeptides in perspective: the last ten years. Neuron. 1991;7:867–879.

Hokfelt T., Arvidsson U., Cullheim S., et al. Multiple messengers in descending serotonin neurons: localization and functional implications. Journal of Chemical Neuroanatomy. 2000;18:75–86.

Hokfelt T., Wiesenfeld-Hallin Z., Villar M., et al. Increase of galanin-like immunoreactivity in rat dorsal root ganglion cells after peripheral axotomy. Neuroscience Letters. 1987;83:217–220.

Hollis J.H., Lightman S.L., Lowry C.A., et al. Integration of systemic and visceral sensory information by medullary catecholaminergic systems during peripheral inflammation. Annals of the New York Academy of Sciences. 2004;1018:71–75.

Hollmann M., Hartley M., Heinemann S. Ca2+ permeability of KA-AMPA–gated glutamate receptor channels depends on subunit composition. Science. 1991;252:851–853.

Honda K., Harada A., Takano Y., et al. Involvement of M3 muscarinic receptors of the spinal cord in formalin-induced nociception in mice. Brain Research. 2000;859:38–44.

Honda M., Tanabe M., Ono H., et al. Serotonergic depression of spinal monosynaptic transmission is mediated by 5-HT1B receptors. European Journal of Pharmacology. 2003;482:155–161.

Hongo T., Jankowska E., Lundberg A. Post-synaptic excitation and inhibition from primary afferents in neurones of the spinocervical tract. Journal of Physiology. 1968;199:569–592.

Honor P., Menning P.M., Rogers S.D., et al. Spinal substance P receptor expression and internalization in acute, short-term, and long-term inflammatory pain states. Journal of Neuroscience. 1999;19:7670–7678.

Houghton A.K., Ogilvie J., Clarke R.W. The involvement of tachykinin NK2 and NK3 receptors in central sensitization of a spinal withdrawal reflex in the decerebrated, spinalized rabbit. Neuropharmacology. 2000;39:133–140.

Hoyer D., Martin G. 5-HT receptor classification and nomenclature: towards a harmonization with the human genome. Neuropharmacology. 1997;36:419–428.

Hoyer D., Clarke D.E., Fozard J.R., et al. International Union of Pharmacology classification of receptors for 5-hydroxytryptamine (serotonin). Pharmacological Reviews. 1994;46:157–203.

Hsieh G.C., Pai M., Chandran P., et al. Central and peripheral sites of action for CB receptor mediated analgesic activity in chronic inflammatory and neuropathic pain models in rats. British Journal of Pharmacology. 2011;162:428–440.

Hua X.Y., Chen P., Marsala M., et al. Intrathecal substance P–induced thermal hyperalgesia and spinal release of prostaglandin E2 and amino acids. Neuroscience. 1999;89:525–534.

Hua X.Y., Chen P., Polgár E., et al. Spinal neurokinin NK1 receptor down-regulation and antinociception: effects of spinal NK1 receptor antisense oligonucleotides and NK1 receptor occupancy. Journal of Neurochemistry. 1998;70:688–698.

Hua X.Y., Hayes C.S., Hofer A., et al. Galanin acts at GalR1 receptors in spinal antinociception: synergy with morphine and AP-5. Journal of Pharmacology and Experimental Therapeutics. 2004;308:574–582.

Hua X.Y., Salgado K.F., Gu G., et al. Mechanisms of antinociception of spinal galanin: how does galanin inhibit spinal sensitization? Neuropeptides. 2005;39:211–216.

Huettner J.E. Kainate receptors and synaptic transmission. Progress in Neurobiology. 2003;70:387–407.

Hugel S., Schlichter R. Presynaptic P2X receptors facilitate inhibitory GABAergic transmission between cultured rat spinal cord dorsal horn neurons. Journal of Neuroscience. 2000;20:2121–2130.

Hughes D.I., Polgár E., Shehab S.A., et al. Peripheral axotomy induces depletion of the vesicular glutamate transporter VGLUT1 in central terminals of myelinated afferent fibres in the rat spinal cord. Brain Research. 2004;1017:69–76.

Hutchison W.D., Morton C.R., Terenius L. Dynorphin A: in vivo release in the spinal cord of the cat. Brain Research. 1990;532:299–306.

Hwang S.J., Pagliardini S., Rustioni A., et al. Presynaptic kainate receptors in primary afferents to the superficial laminae of the rat spinal cord. Journal of Comparative Neurology. 2001;436:275–289.

Hylden J.L., Wilcox G.L. Intrathecal substance P elicits a caudally-directed biting and scratching behavior in mice. Brain Research. 1981;217:212–215.

Iadarola M.J., Brady L.S., Draisci G., et al. Enhancement of dynorphin gene expression in spinal cord following experimental inflammation: stimulus specificity, behavioral parameters and opioid receptor binding. Pain. 1988;35:313–326.

Inoue K. Microglial activation by purines and pyrimidines. Glia. 2002;40:156–163.

Inoue K., Koizumi S., Tsuda M., et al. Signaling of ATP receptors in glia-neuron interaction and pain. Life Sciences. 2003;74:189–197.

Iwamoto E.T., Marion L. Characterization of the antinociception produced by intrathecally administered muscarinic agonists in rats. Journal of Pharmacology and Experimental Therapeutics. 1993;266:329–338.

Jang I.S., Rhee J.S., Kubota H., et al. Developmental changes in P2X purinoceptors on glycinergic presynaptic nerve terminals projecting to rat substantia gelatinosa neurones. Journal of Physiology. 2001;536:505–519.

Jeftinija S., Jeftinija K., Liu F., et al. Excitatory amino acids are released from rat primary afferent neurons in vitro. Neuroscience Letters. 1991;125:191–194.

Jennings E.A., Vaughan C.W., et al. Cannabinoid actions on rat superficial medullary dorsal horn neurons in vitro. J Physiol. 2001;534(Pt 3):805–812.

Jensen T.S., Yaksh T.L. Effects of an intrathecal dopamine agonist, apomorphine, on thermal and chemical evoked noxious responses in rats. Brain Research. 1984;296:285–293.

Jensen T.S., Yaksh T.L. Examination of spinal monoamine receptors through which brainstem opiate-sensitive systems act in the rat. Brain Research. 1986;363:114–127.

Jensen T.S., Yaksh T.L. Brainstem excitatory amino acid receptors in nociception: microinjection mapping and pharmacological characterization of glutamate-sensitive sites in the brainstem associated with algogenic behavior. Neuroscience. 1992;46:535–547.

Jessell T.M., Yoshioka K., Jahr C.E. Amino acid receptor–mediated transmission at primary afferent synapses in rat spinal cord. Journal of Experimental Biology. 1986;124:239–258.

Jhamandas K., Yaksh T.L., Harty G., et al. Action of intrathecal capsaicin and its structural analogues on the content and release of spinal substance P: selectivity of action and relationship to analgesia. Brain Research. 1984;306:215–225.

Ji R.R., Gereau R.W., 4th., Malcangio M., et al. MAP kinase and pain. Brain Research Reviews. 2009;60:135–148.

Jia H., Rustioni A., Valtschanoff J.G., et al. Metabotropic glutamate receptors in superficial laminae of the rat dorsal horn. Journal of Comparative Neurology. 1999;410:627–642.

Jin X., Gereau R.W., 4th. Acute p38-mediated modulation of tetrodotoxin-resistant sodium channels in mouse sensory neurons by tumor necrosis factor-alpha. Journal of Neuroscience. 2006;26:246–255.

Jones E.G. Modern views of cellular thalamic mechanisms. In: Bentivoglio M., Spreafico R., eds. Cellular thalamic mechanisms. Amsterdam: Elsevier; 1988:1–22.

Jones E.G. A new view of specific and nonspecific thalamocortical connections. Advances in Neurology. 1998;77:49–71. discussion 72–73

Jones S.L. Descending noradrenergic influences on pain. Progress in brain research. 1991;88:381–394.

Jones T.L., Sorkin L.S. Calcium-permeable alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/kainate receptors mediate development, but not maintenance, of secondary allodynia evoked by first-degree burn in the rat. Journal of Pharmacology and Experimental Therapeutics. 2004;310:223–229.

Jones T.L., Sorkin L.S. Activated PKA and PKC, but not CaMKIIalpha, are required for AMPA/kainate-mediated pain behavior in the thermal stimulus model. Pain. 2005;117:259–270.

Joo Choi E., Hong M.P., Kyoo Shin Y., et al. ATP modulation of sodium currents in rat dorsal root ganglion neurons. Brain Research. 2003;968:15–25.

Ju G., Hokfelt T., Brodin E., et al. Primary sensory neurons of the rat showing calcitonin gene–related peptide immunoreactivity and their relation to substance P–, somatostatin-, galanin-, vasoactive intestinal polypeptide– and cholecystokinin-immunoreactive ganglion cells. Cell and Tissue Research. 1987;247:417–431.

Ju G., Melander T., Ceccatelli S., et al. Immunohistochemical evidence for a spinothalamic pathway co-containing cholecystokinin- and galanin-like immunoreactivities in the rat. Neuroscience. 1987;20:439–456.

Kaelin-Lang A., Lauterburg T., Burgunder J.M. Expression of adenosine A2a receptor gene in rat dorsal root and autonomic ganglia. Neuroscience Letters. 1998;246:21–24.

Kaelin-Lang A., Lauterburg T., Burgunder J.M. Expression of adenosine A2a receptors gene in the olfactory bulb and spinal cord of rat and mouse. Neuroscience Letters. 1999;261:189–191.

Kamisaki Y., Hamada T., Maeda K., et al. Presynaptic alpha 2 adrenoceptors inhibit glutamate release from rat spinal cord synaptosomes. Journal of Neurochemistry. 1993;60:522–526.

Kaneko M., Hammond D.L. Role of spinal gamma-aminobutyric acid A receptors in formalin-induced nociception in the rat. Journal of Pharmacology and Experimental Therapeutics. 1997;282:928–938.

Karim F., Wang C.C., Gereau R.W., 4th. Metabotropic glutamate receptor subtypes 1 and 5 are activators of extracellular signal–regulated kinase signaling required for inflammatory pain in mice. Journal of Neuroscience. 2001;21:3771–3779.

Kawamata T., Omote K., Toriyabe M., et al. The activation of 5-HT(3) receptors evokes GABA release in the spinal cord. Brain Research. 2003;978:250–255.

Kawamura T., Yamauchi T., Koyama M., et al. Expression of prostaglandin EP2 receptor mRNA in the rat spinal cord. Life Sciences. 1997;61:2111–2116.

Kawaraguchi Y., Kawaguchi M., Takahashi M., et al. Delta-opioid agonist SNC80 can attenuate the development of dynorphin A–induced tactile allodynia in rats. Anesthesiology. 2004;101:546–549.

Kawasaki Y., Kumamoto E., Furue H., et al. Alpha 2 adrenoceptor–mediated presynaptic inhibition of primary afferent glutamatergic transmission in rat substantia gelatinosa neurons. Anesthesiology. 2003;98:682–689.

Kawatani M., Nagel J., de Groat W.C. Identification of neuropeptides in pelvic and pudendal nerve afferent pathways to the sacral spinal cord of the cat. Journal of Comparative Neurology. 1986;249:117–132.

Keil G.J., 2nd., DeLander G.E. Spinally-mediated antinociception is induced in mice by an adenosine kinase–, but not by an adenosine deaminase–, inhibitor. Life Sciences. 1992;51:PL171–PL176.

Keller A.F., Coull J.A., Cherry N., et al. Region-specific developmental specialization of GABA-glycine cosynapses in laminas I-II of the rat spinal dorsal horn. Journal of Neuroscience. 2001;21:7871–7880.

Kellstein D.E., Price D.D., Mayer D.J., et al. Cholecystokinin and its antagonist lorglumide respectively attenuate and facilitate morphine-induced inhibition of C-fiber evoked discharges of dorsal horn nociceptive neurons. Brain Research. 1991;540:302–306.

Kennedy C., Assis T.S., Currie A.J., et al. Crossing the pain barrier: P2 receptors as targets for novel analgesics. Journal of Physiology. 2003;553:683–694.

Kerchner G.A., Li P., Zhuo M. Speaking out of turn: a role for silent synapses in pain. IUBMB Life. 1999;48:251–256.

Kerchner G.A., Wang G.D., Qiu C.S., et al. Direct presynaptic regulation of GABA/glycine release by kainate receptors in the dorsal horn: an ionotropic mechanism. Neuron. 2001;32:477–488.

Kerr B.J., Bradbury E.J., Bennett D.J., et al. Brain-derived neurotrophic factor modulates nociceptive sensory inputs and NMDA-evoked responses in the rat spinal cord. Journal of Neuroscience. 1999;19:5138–5148.

Kerr B.J., Cafferty W.B., Gupta Y.K., et al. Galanin knockout mice reveal nociceptive deficits following peripheral nerve injury. European Journal of Neuroscience. 2000;12:793–802.

Khasabova I.A., Harding-Rose C., et al. Differential effects of CB1 and opioid agonists on two populations of adult rat dorsal root ganglion neurons. J Neurosci. 2004;24(7):1744–1753.

Khakh B.S., Burnstock G., Kennedy C., et al. International Union of Pharmacology. XXIV. Current status of the nomenclature and properties of P2X receptors and their subunits. Pharmacological Reviews. 2001;53:107–118.

Khakh B.S., Henderson G. ATP receptor–mediated enhancement of fast excitatory neurotransmitter release in the brain. Molecular Pharmacology. 1998;54:372–378.

Khan I., Osaka H., Stanislaus S., et al. Nicotinic acetylcholine receptor distribution in relation to spinal neurotransmission pathways. Journal of Comparative Neurology. 2003;467:44–59.

Khan I.M., Buerkle H., Taylor P., et al. Nociceptive and antinociceptive responses to intrathecally administered nicotinic agonists. Neuropharmacology. 1998;37:1515–1525.

Khan I.M., Marsala M., Printz M.P., et al. Intrathecal nicotinic agonist–elicited release of excitatory amino acids as measured by in vivo spinal microdialysis in rats. Journal of Pharmacology and Experimental Therapeutics. 1996;278:97–106.

Khandwala H., Zhang Z., Loomis C.W. Inhibition of strychnine-allodynia is mediated by spinal adenosine A1- but not A2-receptors in the rat. Brain Research. 1998;808:106–109.

Kharazia V.N., Weinberg R.J. Glutamate in thalamic fibers terminating in layer IV of primary sensory cortex. Journal of Neuroscience. 1994;14:6021–6032.

Khasar S.G., Lin Y.H., Martin A., et al. A novel nociceptor signaling pathway revealed in protein kinase C epsilon mutant mice. Neuron. 1999;24:253–260.

Khayyat G.F., Yu U.J., King R.B. Response patterns to noxious and non-noxious stimuli in rostral trigeminal relay nuclei. Brain Research. 1975;97:47–60.

Kia H.K., Miquel M.C., McKernan R.M., et al. Localization of 5-HT3 receptors in the rat spinal cord: immunohistochemistry and in situ hybridization. Neuroreport. 1995;6:257–261.

Kidd E.J., Laporte A.M., Langlois X., et al. 5-HT3 receptors in the rat central nervous system are mainly located on nerve fibres and terminals. Brain Research. 1993;612:289–298.

Kim S.J., Chung W.H., Rhim H., et al. Postsynaptic action mechanism of somatostatin on the membrane excitability in spinal substantia gelatinosa neurons of juvenile rats. Neuroscience. 2002;114:1139–1148.

King A.E., Lopez-Garcia J.A. Excitatory amino acid receptor–mediated neurotransmission from cutaneous afferents in rat dorsal horn in vitro. Journal of Physiology. 1993;472:443–457.

King A.E., Thompson S.W., Urban L., et al. An intracellular analysis of amino acid induced excitations of deep dorsal horn neurones in the rat spinal cord slice. Neuroscience Letters. 1988;89:286–292.

King T.E., Heath M.J., Debs P., et al. The development of the nociceptive responses in neurokinin-1 receptor knockout mice. Neuroreport. 2000;11:587–591.

Kiser R.S., Lebovitz R.M., German D.C., et al. Anatomic and pharmacologic differences between two types of aversive midbrain stimulation. Brain Research. 1978;155:331–342.

Kiyama H., Emson P.C. Distribution of somatostatin mRNA in the rat nervous system as visualized by a novel non-radioactive in situ hybridization histochemistry procedure. Neuroscience. 1990;38:223–244.

Kiyosawa A., Katsurabayashi S., Akaike N., et al. Nicotine facilitates glycine release in the rat spinal dorsal horn. Journal of Physiology. 2001;536:101–110.

Klein C.M., Coggeshall R.E., Carlton S.M., et al. The effects of A- and C-fiber stimulation on patterns of neuropeptide immunostaining in the rat superficial dorsal horn. Brain Research. 1992;580:121–128.

Koetzner L., Gregory J.A., Yaksh T.L., et al. Intrathecal protease-activated receptor stimulation produces thermal hyperalgesia through spinal cyclooxygenase activity. Journal of Pharmacology and Experimental Therapeutics. 2004;311:356–363.

Koetzner L., Hua X.Y., Lai J., et al. Nonopioid actions of intrathecal dynorphin evoke spinal excitatory amino acid and prostaglandin E2 release mediated by cyclooxygenase-1 and -2. Journal of Neuroscience. 2004;24:1451–1458.

Kohno T., Kumamoto E., Baba H., et al. Actions of midazolam on GABAergic transmission in substantia gelatinosa neurons of adult rat spinal cord slices. Anesthesiology. 2000;92:507–515.

Kontinen V.K., Meert T.F. Vocalization responses after intrathecal administration of ionotropic glutamate receptor agonists in rats. Anesthesia and Analgesia. 2002;95:997–1001.

Krisch B. Somatostatin-immunoreactive fiber projections into the brain stem and the spinal cord of the rat. Cell and Tissue Research. 1981;217:531–552.

Kumar N., Laferriere A., Yu J.S., et al. Metabotropic glutamate receptors (mGluRs) regulate noxious stimulus–induced glutamate release in the spinal cord dorsal horn of rats with neuropathic and inflammatory pain. Journal of Neurochemistry. 2010;114:281–290.

Kuo D.C., Kawatani M., de Groat W.C. Vasoactive intestinal polypeptide identified in the thoracic dorsal root ganglia of the cat. Brain Research. 1985;330:178–182.

Kuraishi Y., Harada Y., Satoh M., et al. Antagonism by phenoxybenzamine of the analgesic effect of morphine injected into the nucleus reticularis gigantocellularis of the rat. Neuropharmacology. 1979;18:107–110.

Kuraishi Y., Harada Y., Takagi H., et al. Noradrenaline regulation of pain-transmission in the spinal cord mediated by alpha-adrenoceptors. Brain Research. 1979;174:333–336.

Kuraishi Y., Hirota N., Sato Y., et al. Evidence that substance P and somatostatin transmit separate information related to pain in the spinal dorsal horn. Brain Research. 1985;325:294–298.

Kuraishi Y., Hirota N., Sato Y., et al. Stimulus specificity of peripherally evoked substance P release from the rabbit dorsal horn in situ. Neuroscience. 1989;30:241–250.

Kyles A.E., Waterman A.E., Livingston A. Antinociceptive activity of midazolam in sheep. Journal of Veterinary Pharmacology and Therapeutics. 1995;18:54–60.

Lai J., Ossipov M.H., Vanderah T.W., et al. Neuropathic pain: the paradox of dynorphin. Molecular Interventions. 2001;1:160–167.

Laird J.M., Roza C., De Filipe C., et al. Role of central and peripheral tachykinin NK1 receptors in capsaicin-induced pain and hyperalgesia in mice. Pain. 2001;90:97–103.

Laneuville O., Dorais J., Couture R. Characterization of the effects produced by neurokinins and three agonists selective for neurokinin receptor subtypes in a spinal nociceptive reflex of the rat. Life Sciences. 1988;42:1295–1305.

Lao L.J., Kumamoto E., Luo C., et al. Adenosine inhibits excitatory transmission to substantia gelatinosa neurons of the adult rat spinal cord through the activation of presynaptic A(1) adenosine receptor. Pain. 2001;94:315–324.

Larsson M. Ionotropic glutamate receptors in spinal nociceptive processing. Journal of Neuroscience. 2009;28:7084–7090.

Larsson M., Broman J. Translocation of GluR1-containing AMPA receptors to a spinal nociceptive synapse during acute noxious stimulation. Journal of Neuroscience. 2008;28:7084–7090.

Laughlin T.M., Larson A.A., Wilcox G.L. Mechanisms of induction of persistent nociception by dynorphin. Journal of Pharmacology and Experimental Therapeutics. 2001;299:6–11.

Laughlin T.M., Vanderah T.W., Lashbrook J., et al. Spinally administered dynorphin A produces long-lasting allodynia: involvement of NMDA but not opioid receptors. Pain. 1997;72:253–260.

Lavand’homme P.M., Eisenach J.C. Exogenous and endogenous adenosine enhance the spinal antiallodynic effects of morphine in a rat model of neuropathic pain. Pain. 1999;80:31–36.

Lawson S.N., Perry M.J., Prabhakar E., et al. Primary sensory neurones: neurofilament, neuropeptides, and conduction velocity. Brain Research Bulletin. 1993;30:239–243.

Leah J., Menetrey D., de Pommery J., et al. Neuropeptides in long ascending spinal tract cells in the rat: evidence for parallel processing of ascending information. Neuroscience. 1988;24:195–207.

Le Bars D., Bourgoin S., Clot A.M., et al. Noxious mechanical stimuli increase the release of Met-enkephalin–like material heterosegmentally in the rat spinal cord. Brain Research. 1987;402:188–192.

Lee C.J., Bardoni R., Tong C.K., et al. Functional expression of AMPA receptors on central terminals of rat dorsal root ganglion neurons and presynaptic inhibition of glutamate release. Neuron. 2002;35:135–146.

Lee C.J., Engelman H.S., MacDermott A.B. Activation of kainate receptors on rat sensory neurons evokes action potential firing and may modulate transmitter release. Annals of the New York Academy of Sciences. 1999;868:546–549.

Lee C.J., Kong H., Manzini M.C., et al. Kainate receptors expressed by a subpopulation of developing nociceptors rapidly switch from high to low Ca2+ permeability. Journal of Neuroscience. 2001;21:4572–4581.

Lee D.H., Liu X., Kim H.T., et al. Receptor subtype mediating the adrenergic sensitivity of pain behavior and ectopic discharges in neuropathic Lewis rats. Journal of Neurophysiology. 1999;81:2226–2233.

Lee K.M., Jeon S.M., Cho H.J. Tumor necrosis factor receptor 1 induces interleukin-6 upregulation through NF-kappaB in a rat neuropathic pain model. European Journal of Pain. 2009;13:794–806.

Lee Y.W., Yaksh T.L. Pharmacology of the spinal adenosine receptor which mediates the antiallodynic action of intrathecal adenosine agonists. Journal of Pharmacology and Experimental Therapeutics. 1996;277:1642–1648.

Leem J.W., Gwak Y.S., Lee E.H., et al. Effects of iontophoretically applied substance P, calcitonin gene–related peptide on excitability of dorsal horn neurones in rats. Yonsei Medical Journal. 2001;42:74–83.

Ledent C., Valverde O., et al. Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice. Science. 1999;283(5400):401–404.

Leong M.L., Gu M., Speltz-Paiz R., et al. Neuronal loss in the rostral ventromedial medulla in a rat model of neuropathic pain. Journal of Neuroscience. 2011;31:17028–17039.

Leung L., Cahill C.M. TNF-alpha and neuropathic pain–a review. Journal of neuroinflammation. 2010;7:27.

Lever I.J., Bradbury E.J., Cunningham J.R., et al. Brain-derived neurotrophic factor is released in the dorsal horn by distinctive patterns of afferent fiber stimulation. Journal of Neuroscience. 2001;21:4469–4477.

Li D.P., Chen S.R., Pan Y.Z., et al. Role of presynaptic muscarinic and GABA(B) receptors in spinal glutamate release and cholinergic analgesia in rats. Journal of Physiology. 2002;543:807–818.

Li J., McRoberts J.A., Nie J., et al. Electrophysiological characterization of N-methyl-D-aspartate receptors in rat dorsal root ganglia neurons. Pain. 2004;109:443–452.

Li P., Calejesan A.A., Zhuo M., et al. ATP P2x receptors and sensory synaptic transmission between primary afferent fibers and spinal dorsal horn neurons in rats. Journal of Neurophysiology. 1998;80:3356–3360.

Li P., Wilding T.J., Kim S.J., et al. Kainate-receptor–mediated sensory synaptic transmission in mammalian spinal cord. Nature. 1999;397:161–164.

Li X., Eisenach J.C. Nicotinic acetylcholine receptor regulation of spinal norepinephrine release. Anesthesiology. 2002;96:1450–1456.

Liaw W.J., Stephens R.L., Jr., Binns B.C., et al. Spinal glutamate uptake is critical for maintaining normal sensory transmission in rat spinal cord. Pain. 2005;115:60–70.

Light A.R. The organization of nociceptive neurons in the spinal grey matter. In: The initial processing of pain and its descending control: spinal and trigeminal system. New York: Karger; 1992:109–168.

Lin Q., Peng Y.B., Willis W.D., et al. Role of GABA receptor subtypes in inhibition of primate spinothalamic tract neurons: difference between spinal and periaqueductal gray inhibition. Journal of Neurophysiology. 1996;75:109–123.

Link R.E., Desai K., Hein L., et al. Cardiovascular regulation in mice lacking alpha2-adrenergic receptor subtypes b and c. Science. 1996;273:803–805.

Liu H., Mantyh P.W., Basbaum A.I., et al. NMDA-receptor regulation of substance P release from primary afferent nociceptors. Nature. 1997;386:721–724.

Liu H.X., Brumovsky P., Schmidt R., et al. Receptor subtype-specific pronociceptive and analgesic actions of galanin in the spinal cord: selective actions via GalR1 and GalR2 receptors. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:9960–9964.

Liu J., Zhao X., Cao J., et al. Differential roles of PKA and epac on the production of cytokines in the endotoxin-stimulated primary cultured microglia. Journal of Molecular Neuroscience. 2011;45:186–193.

Liu X.J., Salter M.W. Glutamate receptor phosphorylation and trafficking in pain plasticity in spinal cord dorsal horn. European Journal of Neuroscience. 2010;32:278–289.

Lohse M.J., Forstermann U., Schmidt H.H., et al. Pharmacology of NO:cGMP signal transduction. Naunyn–Schmiedeberg’s Archives of Pharmacology. 1998;358:111–112.

Lu C.R., Willcockson H.H., et al. Ionotropic glutamate receptors are expressed in GABAergic terminals in the rat superficial dorsal horn. The Journal of comparative neurology. 2005;486(2):169–178.

Lucifora S., Willcockson H.H., et al. Presynaptic low- and high-affinity kainate receptors in nociceptive spinal afferents. Pain. 2006;120(1-2):97–105.

Lujan R., Roberts J.D., Shigemoto R., et al. Differential plasma membrane distribution of metabotropic glutamate receptors mGluR1 alpha, mGluR2 and mGluR5, relative to neurotransmitter release sites. Journal of Chemical Neuroanatomy. 1997;13:219–241.

Lundberg J.M., Rudehill A., Sollevi A., Fried G., Wallin G. Co-release of neuropeptide Y and noradrenaline from pig spleen in vivo: importance of subcellular storage, nerve impulse frequency and pattern, feedback regulation and resupply by axonal transport. Neuroscience. 1989;28:475–486.

Lutz E.M., Sheward W.J., West K.M., et al. The VIP2 receptor: molecular characterisation of a cDNA encoding a novel receptor for vasoactive intestinal peptide. FEBS Letters. 1993;334:3–8.

Mabuchi T., Shintani N., Matsumura S., et al. Pituitary adenylate cyclase–activating polypeptide is required for the development of spinal sensitization and induction of neuropathic pain. Journal of Neuroscience. 2004;24:9283–9291.

Mackie K., Lai Y., et al. Cannabinoids activate an inwardly rectifying potassium conductance and inhibit Q-type calcium currents in AtT20 cells transfected with rat brain cannabinoid receptor. J Neurosci. 1995;15(10):6552–6561.

MacKinnon A.C., Spedding M., Brown C.M., et al. Alpha 2-adrenoceptors: more subtypes but fewer functional differences. Trends in Pharmacological Sciences. 1994;15:119–123.

Maeurer C., Holland S., Pierre S., et al. Sphingosine-1-phosphate induced mTOR-activation is mediated by the E3-ubiquitin ligase PAM. Cellular Signalling. 2009;21:293–300.

Magnusson K.R., Clements J.R., Larson A.A., et al. Localization of glutamate in trigeminothalamic projection neurons: a combined retrograde transport–immunohistochemical study. Somatosensory Research. 1987;4:177–190.

Mailleux P., Vanderhaeghen J.J. Distribution of neuronal cannabinoid receptor in the adult rat brain: a comparative receptor binding radioautography and in situ hybridization histochemistry. Neuroscience. 1992;48:655–668.

Makhinson M., Chotiner J.K., Watson J.B., et al. Adenylyl cyclase activation modulates activity-dependent changes in synaptic strength and Ca2+/calmodulin-dependent kinase II autophosphorylation. Journal of Neuroscience. 1999;19:2500–2510.

Malan T.P., Mata H.P., Porreca F., et al. Spinal GABA(A) and GABA(B) receptor pharmacology in a rat model of neuropathic pain. Anesthesiology. 2002;96:1161–1167.

Malcangio M., Fernandes K., Tomlinson D.R. NMDA receptor activation modulates evoked release of substance P from rat spinal cord. British Journal of Pharmacology. 1998;125:1625–1626.

Malmberg A.B., Chen C., Tonegawa S., et al. Preserved acute pain and reduced neuropathic pain in mice lacking PKCgamma. Science. 1997;278:279–283.

Malmberg A.B., Yaksh T.L. Antinociceptive actions of spinal nonsteroidal anti-inflammatory agents on the formalin test in the rat. Journal of Pharmacology and Experimental Therapeutics. 1992;263:136–146.

Malmberg A.B., Yaksh T.L. Hyperalgesia mediated by spinal glutamate or substance P receptor blocked by spinal cyclooxygenase inhibition. Science. 1992;257:1276–1279.

Malmberg A.B., Yaksh T.L. Pharmacology of the spinal action of ketorolac, morphine, ST-91, U50488H, and L-PIA on the formalin test and an isobolographic analysis of the NSAID interaction. Anesthesiology. 1993;79:270–281.

Malmberg A.B., Yaksh T.L. Spinal nitric oxide synthesis inhibition blocks NMDA-induced thermal hyperalgesia and produces antinociception in the formalin test in rats. Pain. 1993;54:291–300.

Malmberg A.B., Yaksh T.L. The effect of morphine on formalin-evoked behaviour and spinal release of excitatory amino acids and prostaglandin E2 using microdialysis in conscious rats. British Journal of Pharmacology. 1995;114:1069–1075.

Malmberg A.B., Yaksh T.L. Cyclooxygenase inhibition and the spinal release of prostaglandin E2 and amino acids evoked by paw formalin injection: a microdialysis study in unanesthetized rats. Journal of Neuroscience. 1995;15:2768–2776.

Mantyh P.W. Neurobiology of substance P and the NK1 receptor. Journal of Clinical Psychiatry. 2002;63(Suppl 11):6–10.

Mantyh P.W., DeMaster E., Malhotra A., et al. Receptor endocytosis and dendrite reshaping in spinal neurons after somatosensory stimulation. Science. 1995;268:1629–1632.

Mao J., Price D.D., Mayer D.J., et al. Thermal hyperalgesia in association with the development of morphine tolerance in rats: roles of excitatory amino acid receptors and protein kinase C. Journal of Neuroscience. 1994;14:2301–2312.

Marchese A., Benovic J.L. Agonist-promoted ubiquitination of the G protein–coupled receptor CXCR4 mediates lysosomal sorting. Journal of Biological Chemistry. 2001;276:45509–45512.

Martin R., Torres M., Sánchez-Prieto J. mGluR7 inhibits glutamate release through a PKC-independent decrease in the activity of P/Q-type Ca2+ channels and by diminishing cAMP in hippocampal nerve terminals. European Journal of Neuroscience. 2007;26:312–322.

Martin W.J., Malmberg A.B., Basbaum A.I. PKCgamma contributes to a subset of the NMDA-dependent spinal circuits that underlie injury-induced persistent pain. Journal of Neuroscience. 2001;21:5321–5327.

Marsala M., Malmberg A.B., Yaksh T.L. The spinal loop dialysis catheter: characterization of use in the unanesthetized rat. Journal of Neuroscience Methods. 1995;62:43–53.

Marsala M., Yaksh T.L. Reversible aortic occlusion in rats: post-reflow hyperesthesia and motor effects blocked by spinal NMDA antagonism [abstract]. Anesthesiology. 1992;77:A664.

Marvizon J.C., Martinez V., Grady E.F., et al. Neurokinin 1 receptor internalization in spinal cord slices induced by dorsal root stimulation is mediated by NMDA receptors. Journal of Neuroscience. 1997;17:8129–8136.

Matsuka Y., Neubert J.K., Maidment N.T., et al. Concurrent release of ATP and substance P within guinea pig trigeminal ganglia in vivo. Brain Research. 2001;915:248–255.

Matsumura K., Watanabe Y., Onoe H. Prostacyclin receptor in the brain and central terminals of the primary sensory neurons: an autoradiographic study using a stable prostacyclin analogue [3H]iloprost. Neuroscience. 1995;65:493–503.

Mauborgne A., Polienor H., Hamon M., et al. Adenosine receptor–mediated control of in vitro release of pain-related neuropeptides from the rat spinal cord. European Journal of Pharmacology. 2002;441:47–55.

Maxwell D.J., Kerr R., Rashid S., et al. Characterisation of axon terminals in the rat dorsal horn that are immunoreactive for serotonin 5-HT3A receptor subunits. Experimental Brain Research. 2003;149:114–124.

Mayer M.L., Armstrong N. Structure and function of glutamate receptor ion channels. Annual Review of Physiology. 2004;66:161–181.

Maze M., Tranquilli W. Alpha-2 adrenoceptor agonists: defining the role in clinical anesthesia. Anesthesiology. 1991;74:581–605.

McCarthy P.W., Lawson S.N. Cell type and conduction velocity of rat primary sensory neurons with substance P–like immunoreactivity. Neuroscience. 1989;28:745–753.

McCarthy P.W., Lawson S.N. Cell type and conduction velocity of rat primary sensory neurons with calcitonin gene–related peptide–like immunoreactivity. Neuroscience. 1990;34:623–632.

McGaraughty S., Chu K.L., Wismer C.T., et al. Effects of A-134974, a novel adenosine kinase inhibitor, on carrageenan-induced inflammatory hyperalgesia and locomotor activity in rats: evaluation of the sites of action. Journal of Pharmacology and Experimental Therapeutics. 2001;296:501–509.

McGeehan G.M., Becherer J.D., Bast R.C., Jr., et al. Regulation of tumour necrosis factor-alpha processing by a metalloproteinase inhibitor. Nature. 1994;370:558–561.

McGowan M.K., Hammond D.L. Antinociception produced by microinjection of L-glutamate into the ventromedial medulla of the rat: mediation by spinal GABAA receptors. Brain Research. 1993;620:86–96.

Mechoulam R., Ben-Shabat S., et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol. 1995;50(1):83–90.

Melien O., Winsnes R., Refsnes M., et al. Pertussis toxin abolishes the inhibitory effects of prostaglandins E1, E2, I2 and F2 alpha on hormone-induced cAMP accumulation in cultured hepatocytes. European Journal of Biochemistry. 1988;172:293–297.

Meller S.T., Dykstra C., Gebhart G.F., et al. Production of endogenous nitric oxide and activation of soluble guanylate cyclase are required for N-methyl-D-aspartate–produced facilitation of the nociceptive tail-flick reflex. European Journal of Pharmacology. 1992;214:93–96.

Melzack R., Wall P.D. Pain mechanisms: a new theory. Science. 1965;150:971–979.

Mendell L.M. Physiological properties of unmyelinated fiber projection to the spinal cord. Experimental Neurology. 1966;16:316–332.

Mendell L.M., Wall P.D. Responses of single dorsal cord cells to peripheral cutaneous unmyelinated fibers. Nature. 1965;206:97–99.

Meuser T., Pietruck C., Gabriel A., et al. 5-HT7 receptors are involved in mediating 5-HT–induced activation of rat primary afferent neurons. Life Sciences. 2002;71:2279–2289.

Michael G.J., Averill S., Nitkunan A., et al. Nerve growth factor treatment increases brain-derived neurotrophic factor selectively in TrkA-expressing dorsal root ganglion cells and in their central terminations within the spinal cord. Journal of Neuroscience. 1997;17:8476–8490.

Michener S.R., Aimone L.D., Yaksh T.L., et al. Distribution of galanin-like immunoreactivity in the pig, rat and human central nervous system. Peptides. 1990;11:1217–1223.

Miletic V., Bowen K.K., et al. Loose ligation of the rat sciatic nerve is accompanied by changes in the subcellular content of protein kinase C beta II and gamma in the spinal dorsal horn. Neuroscience letters. 2000;288(3):199–202.

Millan M.J. The induction of pain: an integrative review. Progress in Neurobiology. 1999;57:1–164.

Milligan E.D., Sloane E.M., Watkins L.R. Glia in pathological pain: a role for fractalkine. Journal of Neuroimmunology. 2008;198:113–120.

Minson J.B., Arnolda L.F., et al. Neurochemistry of nerve fibers apposing sympathetic preganglionic neurons activated by sustained hypotension. The Journal of comparative neurology. 2002;449(4):307–318.

Mitchell J.J., Anderson K.J. Quantitative autoradiographic analysis of excitatory amino acid receptors in the cat spinal cord. Neuroscience Letters. 1991;124:269–272.

Miyazawa A., Fujiyoshi Y., Unwin N. Structure and gating mechanism of the acetylcholine receptor pore. Nature. 2003;423:949–955.

Mizukawa K., Vincent S.R., McGeer P.L., et al. Distribution of reduced-nicotinamide-adenine-dinucleotide-phosphate diaphorase–positive cells and fibers in the cat central nervous system. Journal of Comparative Neurology. 1989;279:281–311.

Mjellem-Joly N., Lund A., Berge O.G., et al. Potentiation of a behavioural response in mice by spinal coadministration of substance P and excitatory amino acid agonists. Neuroscience Letters. 1991;133:121–124.

Mollenholt P., Post C., Rawal N., et al. Antinociceptive and “neurotoxic” actions of somatostatin in rat spinal cord after intrathecal administration. Pain. 1988;32:95–105.

Momiyama A. Distinct synaptic and extrasynaptic NMDA receptors identified in dorsal horn neurones of the adult rat spinal cord. Journal of Physiology. 2000;523:621–628.

Montana M.C., Cavallone L.F., et al. The metabotropic glutamate receptor subtype 5 antagonist fenobam is analgesic and has improved in vivo selectivity compared with the prototypical antagonist 2-methyl-6-(phenylethynyl)-pyridine. The Journal of pharmacology and experimental therapeutics. 2009;330(3):834–843.

Moochhala S.M., Sawynok J. Hyperalgesia produced by intrathecal substance P and related peptides: desensitization and cross desensitization. British Journal of Pharmacology. 1984;82:381–388.

Morales M., Battenberg E., Bloom F.E. Distribution of neurons expressing immunoreactivity for the 5HT3 receptor subtype in the rat brain and spinal cord. Journal of Comparative Neurology. 1998;402:385–401.

Mori H., Mishina M. Structure and function of the NMDA receptor channel. Neuropharmacology. 1995;34:1219–1237.

Morisset V., Urban L. Cannabinoid-induced presynaptic inhibition of glutamatergic EPSCs in substantia gelatinosa neurons of the rat spinal cord. Journal of Neurophysiology. 2001;86:40–48.

Moroni F., Cozzi A., Lombardi G., et al. Presynaptic mGlu1 type receptors potentiate transmitter output in the rat cortex. European Journal of Pharmacology. 1998;347:189–195.

Morris R., Cheunsuang O., Stewart A., et al. Spinal dorsal horn neurone targets for nociceptive primary afferents: do single neurone morphological characteristics suggest how nociceptive information is processed at the spinal level? Brain Research. Brain Research Reviews. 2004;46:173–190.

Morris R., Southam E., Braid D.J., et al. Nitric oxide may act as a messenger between dorsal root ganglion neurones and their satellite cells. Neuroscience Letters. 1992;137:29–32.

Morton C.R., Hutchison W.D. Release of sensory neuropeptides in the spinal cord: studies with calcitonin gene–related peptide and galanin. Neuroscience. 1989;31:807–815.

Morton C.R., Hutchison W.D. Morphine does not reduce the intraspinal release of calcitonin gene–related peptide in the cat. Neuroscience Letters. 1990;117:319–324.

Morton C.R., Hutchison W.D., Hendry I.A. Release of immunoreactive somatostatin in the spinal dorsal horn of the cat. Neuropeptides. 1988;12:189–197.

Morton C.R., Hutchison W.D., Hendry I.A., et al. Somatostatin: evidence for a role in thermal nociception. Brain Research. 1989;488:89–96.

Moss A., Blackburn-Munro G., Garry E.M., et al. A role of the ubiquitin-proteasome system in neuropathic pain. Journal of Neuroscience. 2002;22:1363–1372.

Murase K., Ryu P.D., Randic M., et al. Tachykinins modulate multiple ionic conductances in voltage-clamped rat spinal dorsal horn neurons. Journal of Neurophysiology. 1989;61:854–865.

Naguib M., Yaksh T.L. Antinociceptive effects of spinal cholinesterase inhibition and isobolographic analysis of the interaction with mu and alpha 2 receptor systems. Anesthesiology. 1994;80:1338–1348.

Naguib M., Yaksh T.L. Characterization of muscarinic receptor subtypes that mediate antinociception in the rat spinal cord. Anesthesia and Analgesia. 1997;85:847–853.

Nahin R.L. Immunocytochemical identification of long ascending peptidergic neurons contributing to the spinoreticular tract in the rat. Neuroscience. 1987;23:859–869.

Nahin R.L., Hylden J.L., Iadarola M.J., et al. Peripheral inflammation is associated with increased dynorphin immunoreactivity in both projection and local circuit neurons in the superficial dorsal horn of the rat lumbar spinal cord. Neuroscience Letters. 1989;96:247–252.

Nahin R.L., Ren K., De Leon M., et al. Primary sensory neurons exhibit altered gene expression in a rat model of neuropathic pain. Pain. 1994;58:95–108.

Nai Q., McIntosh J.M., Margiotta J.F. Relating neuronal nicotinic acetylcholine receptor subtypes defined by subunit composition and channel function. Molecular Pharmacology. 2003;63:311–324.

Nakatsuka T., Gu J.G. ATP P2X receptor–mediated enhancement of glutamate release and evoked EPSCs in dorsal horn neurons of the rat spinal cord. Journal of Neuroscience. 2001;21:6522–6531.

Narikawa K., Furue H., Kumamoto E., et al. In vivo patch-clamp analysis of IPSCs evoked in rat substantia gelatinosa neurons by cutaneous mechanical stimulation. Journal of Neurophysiology. 2000;84:2171–2174.

Narita M., Dun S.L., Dun N.J., et al. Hyperalgesia induced by pituitary adenylate cyclase–activating polypeptide in the mouse spinal cord. European Journal of Pharmacology. 1996;311:121–126.

Narumiya S., Sugimoto Y., Ushikubi F. Prostanoid receptors: structures, properties, and functions. Physiological Reviews. 1999;79:1193–1226.

Natura G., von Banchet G.S., et al. Calcitonin gene-related peptide enhances TTX-resistant sodium currents in cultured dorsal root ganglion neurons from adult rats. Pain. 2005;116(3):194–204.

Negishi M., Ito S., Hayaishi O. Prostaglandin E receptors in bovine adrenal medulla are coupled to adenylate cyclase via Gi and to phosphoinositide metabolism in a pertussis toxin–insensitive manner. Journal of Biological Chemistry. 1989;264:3916–3923.

Negishi M., Sugimoto Y., Ichikawa A. Molecular mechanisms of diverse actions of prostanoid receptors. Biochimica et Biophysica Acta. 1995;1259:109–119.

Neugebauer V., Chen P.S., Willis W.D., et al. Groups II and III metabotropic glutamate receptors differentially modulate brief and prolonged nociception in primate STT cells. Journal of Neurophysiology. 2000;84:2998–3009.

Niclou S.P., Suidan H.S., Pavlik A., et al. Changes in the expression of protease-activated receptor 1 and protease nexin-1 mRNA during rat nervous system development and after nerve lesion. European Journal of Neuroscience. 1998;10:159–1607.

Nicol G.D., Klingberg D.K., Vasko M.R. Prostaglandin E2 increases calcium conductance and stimulates release of substance P in avian sensory neurons. Journal of Neuroscience. 1992;12:1917–1927.

Niederberger E., Schmidtko A., Rothstein J.D., et al. Modulation of spinal nociceptive processing through the glutamate transporter GLT-1. Neuroscience. 2003;116:81–87.

Nishiyama K., Kwak S., Murayama S., et al. Substance P is a possible neurotransmitter in the rat spinothalamic tract. Neuroscience Research. 1995;21:261–266.

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. Anesthesia and Analgesia. 1999;89:143–147.

Nishiyama T., Hanaoka K. Midazolam can potentiate the analgesic effects of intrathecal bupivacaine on thermal- or inflammatory-induced pain. Anesthesia and Analgesia. 2003;96:1386–1391.

Nishiyama T., Yokoyama T., Hanaoka K. Midazolam improves postoperative epidural analgesia with continuous infusion of local anaesthetics. Canadian Journal of Anaesthesia. 1998;45:551–555.

Niv D., Whitwam J.G., Loh L., et al. Depression of nociceptive sympathetic reflexes by the intrathecal administration of midazolam. British Journal of Anaesthesia. 1983;55:541–547.

Noguchi K., Ruda M.A. Gene regulation in an ascending nociceptive pathway: inflammation-induced increase in preprotachykinin mRNA in rat lamina I spinal projection neurons. Journal of Neuroscience. 1992;12:2563–2572.

North R.A. Molecular physiology of P2X receptors. Physiological Reviews. 2002;82:1013–1067.

North R.A., Williams J.T., Surprenant A., et al. Mu and delta receptors belong to a family of receptors that are coupled to potassium channels. Proceedings of the National Academy of Sciences of the United States of America. 1987;84:5487–5491.

Nozaki-Taguchi N., Yaksh T.L. Pharmacology of spinal glutamatergic receptors in post-thermal injury–evoked tactile allodynia and thermal hyperalgesia. Anesthesiology. 2002;96:617–626.

Nyberg F., Yaksh T.L., Terenius L. Opioid activity released from cat spinal cord by sciatic nerve stimulation. Life Sciences. 1983;33(Suppl 1):17–20.

O’Banion M.K. Cyclooxygenase-2: molecular biology, pharmacology, and neurobiology. Critical Reviews in Neurobiology. 1999;13:45–82.

Obata K., Noguchi K. MAPK activation in nociceptive neurons and pain hypersensitivity. Life Sciences. 2004;74:2643–2653.

O’Brien C., Woolf C.J., Fitzgerald M., et al. Differences in the chemical expression of rat primary afferent neurons which innervate skin, muscle or joint. Neuroscience. 1989;32:493–502.

O’Dell T.J., Hawkins R.D., Kandel E.R., et al. Tests of the roles of two diffusible substances in long-term potentiation: evidence for nitric oxide as a possible early retrograde messenger. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:11285–11289.

Ohara S., Lenz F.A. Medial lateral extent of thermal and pain sensations evoked by microstimulation in somatic sensory nuclei of human thalamus. Journal of Neurophysiology. 2003;90:2367–2377.

Ohishi H., Nomura S., Ding Y.Q., et al. Presynaptic localization of a metabotropic glutamate receptor, mGluR7, in the primary afferent neurons: an immunohistochemical study in the rat. Neuroscience Letters. 1995;202:85–88.

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. Journal of Comparative Neurology. 1993;335:252–266.

Ohishi H., Shigemoto R., Nakanishi S., et al. Distribution of the messenger RNA for a metabotropic glutamate receptor, mGluR2, in the central nervous system of the rat. Neuroscience. 1993;53:1009–1018.

Oida H., Namba T., Sugimoto Y., et al. In situ hybridization studies of prostacyclin receptor mRNA expression in various mouse organs. British Journal of Pharmacology. 1995;116:2828–2837.

Okano S., Ikeura Y., Inatomi M., et al. Effects of tachykinin NK1 receptor antagonists on the viscerosensory response caused by colorectal distention in rabbits. Journal of Pharmacology and Experimental Therapeutics. 2002;300:925–931.

Oku R., Satoh M., Takagi H., et al. Release of substance P from the spinal dorsal horn is enhanced in polyarthritic rats. Neuroscience Letters. 1987;74:315–319.

Olave M.J., Maxwell D.J. Neurokinin-1 projection cells in the rat dorsal horn receive synaptic contacts from axons that possess alpha2C-adrenergic receptors. Journal of Neuroscience. 2003;23:6837–6846.

Olias G., Viollet C., Kusserow H., et al. Regulation and function of somatostatin receptors. Journal of Neurochemistry. 2004;89:1057–1091.

Oliveira A.L., Hydling F., Olsson E., et al. Cellular localization of three vesicular glutamate transporter mRNAs and proteins in rat spinal cord and dorsal root ganglia. Synapse. 2003;50:117–129.

Omkumar R.V., Kiely M.J., Rosenstein A.J., et al. Identification of a phosphorylation site for calcium/calmodulin dependent protein kinase II in the NR2B subunit of the N-methyl-D-aspartate receptor. Journal of Biological Chemistry. 1996;271:31670–31678.

Onaka M., Minami T., Nishihara I., et al. Involvement of glutamate receptors in strychnine- and bicuculline-induced allodynia in conscious mice. Anesthesiology. 1996;84:1215–1222.

O’Neill M.F., Dourish C.T., Iversen S.D., et al. Morphine-induced analgesia in the rat paw pressure test is blocked by CCK and enhanced by the CCK antagonist MK-329. Neuropharmacology. 1989;28:243–247.

Osborne M.G., Coderre T.J. Effects of intrathecal administration of nitric oxide synthase inhibitors on carrageenan-induced thermal hyperalgesia. British Journal of Pharmacology. 1999;126:1840–1846.

Ossipov M.H., Bazov I., Gardell L.R., et al. Control of chronic pain by he ubiquitin proteasome system in the spinal cord. Journal of Neuroscience. 2007;27:8226–8237.

Ossipov M.H., Lai J., King T., et al. Antinociceptive and nociceptive actions of opioids. Journal of Neurobiology. 2004;61:126–148.

Oyama T., Ueda M., Kuraishi Y., et al. Dual effect of serotonin on formalin-induced nociception in the rat spinal cord. Neuroscience Research. 1996;25:129–135.

Palecek J., Paleckova V., Dougherty P.M., et al. The effect of phorbol esters on the responses of primate spinothalamic neurons to mechanical and thermal stimuli. Journal of Neurophysiology. 1994;71:529–537.

Pan H.L., Khan G.M., Alloway K.D., et al. Resiniferatoxin induces paradoxical changes in thermal and mechanical sensitivities in rats: mechanism of action. Journal of Neuroscience. 2003;23:2911–2919.

Papir-Kricheli D., Frey J., Laufer R., et al. Behavioural effects of receptor-specific substance P agonists. Pain. 1987;31:263–276.

Park Y.K., Galik J., Ryu P.D., et al. Activation of presynaptic group I metabotropic glutamate receptors enhances glutamate release in the rat spinal cord substantia gelatinosa. Neuroscience Letters. 2004;361:220–224.

Parker E.M., Izzarelli D.G., Nowak H.P., et al. Cloning and characterization of the rat GALR1 galanin receptor from Rin14B insulinoma cells. Brain Research. Molecular Brain Research. 1995;34:179–189.

Patel M.K., Pinnock R.D., Lee K., et al. Adenosine exerts multiple effects in dorsal horn neurones of the adult rat spinal cord. Brain Research. 2001;920:19–26.

Paternain A.V., Rodriguez-Moreno A., Villarroel A., et al. Activation and desensitization properties of native and recombinant kainate receptors. Neuropharmacology. 1998;37:1249–1259.

Patwardhan A.M., Scotland P.E., Akopian A.N., et al. Activation of TRPV1 in the spinal cord by oxidized linoleic acid metabolites contributes to inflammatory hyperalgesia. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:18820–18824.

Pehl U., Schmid H.A. Electrophysiological responses of neurons in the rat spinal cord to nitric oxide. Neuroscience. 1997;77:563–573.

Peng Y.B., Lin Q., Willis W.D., et al. Involvement of alpha-2 adrenoceptors in the periaqueductal gray–induced inhibition of dorsal horn cell activity in rats. Journal of Pharmacology and Experimental Therapeutics. 1996;278:125–135.

Persson S., Broman J. Glutamate, but not aspartate, is enriched in trigeminothalamic tract terminals and associated with their synaptic vesicles in the rat nucleus submedius. Experimental Brain Research. 2004;157:152–161.

Pertwee R.G. Cannabinoid receptors and pain. Prog Neurobiol. 2001;63(5):569–611.

Petralia R.S., Wang Y.X., Niedzielski A.S., et al. The metabotropic glutamate receptors, mGluR2 and mGluR3, show unique postsynaptic, presynaptic and glial localizations. Neuroscience. 1996;71:949–976.

Petruska J.C., Cooper B.Y., Gu J.G., et al. Distribution of P2X1, P2X2, and P2X3 receptor subunits in rat primary afferents: relation to population markers and specific cell types. Journal of Chemical Neuroanatomy. 2000;20:141–162.

Piascik M.T., Soltis E.E., Piacik M.M., et al. Alpha-adrenoceptors and vascular regulation: molecular, pharmacologic and clinical correlates. Pharmacology & Therapeutics. 1996;72:215–241.

Pierce P.A., Xie G.X., Levine J.D., et al. 5-Hydroxytryptamine receptor subtype messenger RNAs in rat peripheral sensory and sympathetic ganglia: a polymerase chain reaction study. Neuroscience. 1996;70:553–559.

Pintor A., Pezzola A., et al. The mGlu5 receptor agonist CHPG stimulates striatal glutamate release: possible involvement of A2A receptors. Neuroreport. 2000;11(16):3611–3614.

Pogatzki E.M., Niemeier J.S., Sorkin L.S., et al. Spinal glutamate receptor antagonists differentiate primary and secondary mechanical hyperalgesia caused by incision. Pain. 2003;105:97–107.

Pogatzki E.M., Zahn P.K., Brennan T.J., et al. Effect of pretreatment with intrathecal excitatory amino acid receptor antagonists on the development of pain behavior caused by plantar incision. Anesthesiology. 2000;93:489–496.

Polgar E., Al-Khater K.M., et al. Large projection neurons in lamina I of the rat spinal cord that lack the neurokinin 1 receptor are densely innervated by VGLUT2-containing axons and possess GluR4-containing AMPA receptors. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2008;28(49):13150–13160.

Polgár E., Fowler J.H., McGill M.M., et al. The types of neuron which contain protein kinase C gamma in rat spinal cord. Brain Research. 1999;833:71–80.

Poon A., Sawynok J. Antinociception by adenosine analogs and inhibitors of adenosine metabolism in an inflammatory thermal hyperalgesia model in the rat. Pain. 1998;74:235–245.

Popescu G., Auerbach A. The NMDA receptor gating machine: lessons from single channels. Neuroscientist. 2004;10:192–198.

Porreca F., Burgess S.E., Gardell L.R., et al. Inhibition of neuropathic pain by selective ablation of brainstem medullary cells expressing the mu-opioid receptor. Journal of Neuroscience. 2001;21:5281–5288.

Potrebic S., Ahn A.H., Skinner K., et al. Peptidergic nociceptors of both trigeminal and dorsal root ganglia express serotonin 1D receptors: implications for the selective antimigraine action of triptans. Journal of Neuroscience. 2003;23:10988–10997.

Prast H., Philippu A. Nitric oxide as modulator of neuronal function. Progress in Neurobiology. 2001;64:51–68.

Price G.W., Kelly J.S., Bowery N.G. The location of GABAB receptor binding sites in mammalian spinal cord. Synapse. 1987;1:530–538.

Price T.J., Cervero F., de Koninck Y. Role of cation-chloride-cotransporters (CCC) in pain and hyperalgesia. Current Topics in Medicinal Chemistry. 2005;5:547–555.

Proudlock F., Spike R.C., Todd A.J. Immunocytochemical study of somatostatin, neurotensin, GABA, and glycine in rat spinal dorsal horn. Journal of Comparative Neurology. 1993;327:289–297.

Puig S., Sorkin L.S. Formalin-evoked activity in identified primary afferent fibers: systemic lidocaine suppresses phase-2 activity. Pain. 1996;64:345–355.

Purcell A.L., Carew T.J. Modulation of excitability in Aplysia tail sensory neurons by tyrosine kinases. Journal of neurophysiology. 2001;85(6):2398–2411.

Radhakrishnan V., Henry J.L. Novel substance P antagonist, CP-96,345, blocks responses of cat spinal dorsal horn neurons to noxious cutaneous stimulation and to substance P. Neuroscience Letters. 1991;132:39–43.

Rady J.J., Fujimoto J.M., Tseng L.F. Dynorphins other than dynorphin A(1–17) lack spinal antianalgesic activity but do act on dynorphin A(1–17) receptors. Journal of Pharmacology and Experimental Therapeutics. 1991;259:1073–1080.

Rahman O.I., Terayama R., Ikeda T., et al. Differential effects of NMDA and AMPA/KA receptor antagonists on c-Fos or Zif/268 expression in the rat spinal dorsal horn induced by noxious thermal or mechanical stimulation, or formalin injection. Neuroscience Research. 2002;43:389–399.

Rahman W., Suzuki R., Webber M., et al. Depletion of endogenous spinal 5-HT attenuates the behavioural hypersensitivity to mechanical and cooling stimuli induced by spinal nerve ligation. Pain. 2006;123:264–274.

Rajaofetra N., Ridet J.L., Poulat P., et al. Immunocytochemical mapping of noradrenergic projections to the rat spinal cord with an antiserum against noradrenaline. Journal of Neurocytology. 1992;21:481–494.

Ramos K.M., Lewis M.T., Morgan K.N., et al. Spinal upregulation of glutamate transporter GLT-1 by ceftriaxone: therapeutic efficacy in a range of experimental nervous system disorders. Neuroscience. 2010;169:1888–1900.

Ramwell P.W., Shaw J.E., Jessup R. Spontaneous and evoked release of prostaglandins from frog spinal cord. American Journal of Physiology. 1966;211:998–1004.

Randic M., Hecimovic H., Ryu P.D. Substance P modulates glutamate-induced currents in acutely isolated rat spinal dorsal horn neurones. Neuroscience Letters. 1990;117:74–80.

Randic M., Jiang M.C., Cerne R. Long-term potentiation and long-term depression of primary afferent neurotransmission in the rat spinal cord. Journal of Neuroscience. 1993;13:5228–5241.

Randic M., Miletic V. Depressant actions of methionine–enkephalin and somatostatin in cat dorsal horn neurones activated by noxious stimuli. Brain Research. 1978;152:196–202.

Reddy S.V., Maderdrut J.L., Yaksh T.L. Spinal cord pharmacology of adrenergic agonist–mediated antinociception. Journal of Pharmacology and Experimental Therapeutics. 1980;213:525–533.

Reddy S.V., Yaksh T.L. Spinal noradrenergic terminal system mediates antinociception. Brain Research. 1980;189:391–401.

Reeve A.J., Dickenson A.H., Kerr N.C., et al. Spinal effects of bicuculline: modulation of an allodynia-like state by an A1-receptor agonist, morphine, and an NMDA-receptor antagonist. Journal of Neurophysiology. 1998;79:1494–1507.

Ren K., Hylden J.L., Williams G.M., 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;50:331–344.

Ren K., Williams G.M., Hylden J.L., et al. The intrathecal administration of excitatory amino acid receptor antagonists selectively attenuated carrageenan-induced behavioral hyperalgesia in rats. European Journal of Pharmacology. 1992;219:235–243.

Rhee J.S., Wang Z.M., Nabekura J., et al. ATP facilitates spontaneous glycinergic IPSC frequency at dissociated rat dorsal horn interneuron synapses. Journal of Physiology. 2000;524:471–483.

Roberts R.G., Stevenson J.E., Westerman R.A., et al. Nicotinic acetylcholine receptors on capsaic-insensitive nerves. Neuroreport. 1995;6:1578–1582.

Ross C.A., Ruggiero D.A., Joh T.H., et al. Rostral ventrolateral medulla: selective projections to the thoracic autonomic cell column from the region containing C1 adrenaline neurons. Journal of Comparative Neurology. 1984;228:168–185.

Ross R.A., Coutts A.A., et al. Actions of cannabinoid receptor ligands on rat cultured sensory neurones: implications for antinociception. Neuropharmacology. 2001;40(2):221–232.

Ruda M.A., Iadarola M.J., Cohen L.V., et al. In situ hybridization histochemistry and immunocytochemistry reveal an increase in spinal dynorphin biosynthesis in a rat model of peripheral inflammation and hyperalgesia. Proceedings of the National Academy of Sciences of the United States of America. 1988;85:622–626.

Rudomin P. Selectivity of the central control of sensory information in the mammalian spinal cord. Advances in Experimental Medicine and Biology. 2002;508:157–170.

Russell F.A., McDougall J.J. Proteinase activated receptor (PAR) involvement in mediating arthritis pain and inflammation. Inflammation Research. 2009;58:119–126.

Ryu P.D., Gerber G., Murase K., et al. Actions of calcitonin gene–related peptide on rat spinal dorsal horn neurons. Brain Research. 1988;441:357–361.

Sabbe M.B., Penning J.P., Ozaki G.T., et al. Spinal and systemic action of the alpha2 receptor agonist dexmedetomidine in dogs. Antinociception and carbon dioxide response. Anesthesiology (Laboratory Investigations). 1994;80:1057–1072.

Sagen J., Proudfit H.K. Effect of intrathecally administered noradrenergic antagonists on nociception in the rat. Brain Research. 1984;310:295–301.

Salt T.E. Mediation of thalamic sensory input by both NMDA receptors and non-NMDA receptors. Nature. 1986;322:263–265.

Salter M.W., Henry J.L. Responses of functionally identified neurones in the dorsal horn of the cat spinal cord to substance P, neurokinin A and physalaemin. Neuroscience. 1991;43:601–610.

Sandkuhler J., Fu Q.G., Helmchen C. Spinal somatostatin superfusion in vivo affects activity of cat nociceptive dorsal horn neurons: comparison with spinal morphine. Neuroscience. 1990;34:565–576.

Sang C.N., Hostetter M.P., Gracely R.H., et al. AMPA/kainate antagonist LY293558 reduces capsaicin-evoked hyperalgesia but not pain in normal skin in humans. Anesthesiology. 1998;89:1060–1067.

Sang C.N., Ramadan N.M., Wallihan R.G., et al. LY293558, a novel AMPA/GluR5 antagonist, is efficacious and well-tolerated in acute migraine. Cephalalgia: An International Journal of Headache. 2004;24:596–602.

Santicioli P., Del Bianco E., Maggi C.A. Adenosine A1 receptors mediate the presynaptic inhibition of calcitonin gene–related peptide release by adenosine in the rat spinal cord. European Journal of Pharmacology. 1993;231:139–142.

Santicioli P., Del Bianco E., Tramontana M., et al. Release of calcitonin gene–related peptide like–immunoreactivity induced by electrical field stimulation from rat spinal afferents is mediated by conotoxin-sensitive calcium channels. Neuroscience Letters. 1992;136:161–164.

Sanudo-Pena M.C., Strangman N.M., et al. CB1 receptor localization in rat spinal cord and roots, dorsal root ganglion, and peripheral nerve. Zhongguo Yao Li Xue Bao. 1999;20(12):1115–1120.

Saria A., Gamse R., Petermann J., et al. Simultaneous release of several tachykinins and calcitonin gene–related peptide from rat spinal cord slices. Neuroscience Letters. 1986;63:310–314.

Saxen M.A., Smith F.L., Dunlow L.D., et al. The hypothermic and antinociceptive effects of intrathecal injection of CGRP (8–37) in mice. Life Sciences. 1994;55:1665–1674.

Scemes E., Giaume C. Astrocyte calcium waves: what they are and what they do. Glia. 2006;54:716–725.

Schmidlin F., Loeffler S., Bertrand C., et al. PLA2 phosphorylation and cyclooxygenase-2 induction, through p38 MAP kinase pathway, is involved in the IL-1beta–induced bradykinin B2 receptor gene transcription. Naunyn-Schmiedeberg`s Archives of Pharmacology. 2000;361:247–254.

Schmitt P., Eclancher F., Karli P. Etudes des systèmes de renforcement négatif et de renforcement positiv au niveau de la substance grise centrale chez le rat. Physiology and Behavior. 1974;12:271–279.

Schneider S.P., Perl E.R. Comparison of primary afferent and glutamate excitation of neurons in the mammalian spinal dorsal horn. Journal of Neuroscience. 1988;8:2062–2073.

Schoepp D.D., Jane D.E., Monn J.A., et al. Pharmacological agents acting at subtypes of metabotropic glutamate receptors. Neuropharmacology. 1999;38:1431–1476.

Schousboe A. Role of astrocytes in the maintenance and modulation of glutamatergic and GABAergic neurotransmission. Neurochemical Research. 2003;28:347–352.

Schulte G., Fredholm B.B. Signalling from adenosine receptors to mitogen-activated protein kinases. Cell Signalling. 2003;15:813–827.

Schuman E.M., Madison D.V. A requirement for the intercellular messenger nitric oxide in long-term potentiation. Science. 1991;254:1503–1506.

Seagrove L.C., Suzuki R., Dickenson A.H. Electrophysiological characterisations of rat lamina I dorsal horn neurones and the involvement of excitatory amino acid receptors. Pain. 2004;108:76–87.

Segond von Banchet G., Schindler M., Hervieu G.J., et al. Distribution of somatostatin receptor subtypes in rat lumbar spinal cord examined with gold-labelled somatostatin and anti-receptor antibodies. Brain Research. 1999;816:254–257.

Sekiguchi F., Mita Y., Kamanaka Y., et al. The potent inducible nitric oxide synthase inhibitor ONO-1714 inhibits neuronal NOS and exerts antinociception in rats. Neuroscience Letters. 2004;365:111–115.

Senaris R.M., Schindler M., Humphrey P.P., et al. Expression of somatostatin receptor 3 mRNA in the motor neurones of the rat spinal cord, and the sensory neurones of the spinal ganglia. Brain Research. Molecular Brain Research. 1995;29:185–190.

Seybold V.S. The role of peptides in central sensitization. Handbook of Experimental Pharmacology. 2009;194:451–491.

Seybold V.S., Hylden J.L., Wilcox G.L. Intrathecal substance P and somatostatin in rats: behaviors indicative of sensation. Peptides. 1982;3:49–54.

Seybold V.S., Jia Y.P., Abrahams L.G. Cyclooxygenase-2 contributes to central sensitization in rats with peripheral inflammation. Pain. 2003;105:47–55.

Sher G.D., Mitchell D. N-methyl-D-aspartate receptors mediate responses of rat dorsal horn neurones to hindlimb ischemia. Brain Research. 1990;522:55–62.

Sherman S.E., Luo L., Dostrovsky J.O., et al. Spinal strychnine alters response properties of nociceptive-specific neurons in rat medial thalamus. Journal of Neurophysiology. 1997;78:628–637.

Sherman S.E., Luo L., Dostrovsky J.O., et al. Altered receptive fields and sensory modalities of rat VPL thalamic neurons during spinal strychnine-induced allodynia. Journal of Neurophysiology. 1997;78:2296–2308.

Sherriff F.E., Henderson Z. A cholinergic propriospinal innervation of the rat spinal cord. Brain Research. 1994;634:150–154.

Sherrington C.S. The integrative action of the nervous system. New Haven, CT: University Press; 1906.

Shi T.S., Winzer-Serhan U., Leslie F., et al. Distribution and regulation of alpha(2)-adrenoceptors in rat dorsal root ganglia. Pain. 2000;84:319–330.

Sim L.J., Joseph S.A. Serotonin and substance P afferents to parafascicular and central medial nuclei. Peptides. 1992;13:171–176.

Simmons D.R., Spike R.C., Todd A.J., et al. Galanin is contained in GABAergic neurons in the rat spinal dorsal horn. Neuroscience Letters. 1995;187:119–122.

Sinclair R.J., Sathian K., Burton H. Neuronal responses in ventroposterolateral nucleus of thalamus in monkeys (Macaca mulatta) during active touch of gratings. Somatosensory and Motor Research. 1991;8:293–300.

Siuciak J.A., Wong V., Pearsall D., et al. BDNF produces analgesia in the formalin test and modifies neuropeptide levels in rat brain and spinal cord areas associated with nociception. European Journal of Neuroscience. 1995;7:663–670.

Sivilotti L., Woolf C.J. The contribution of GABA and glycine receptors to central sensitization: disinhibition and touch-evoked allodynia in the spinal cord. Journal of Neurophysiology. 1994;72:169–179.

Skilling S.R., Smullin D.H., Beitz A.J., et al. Extracellular amino acid concentrations in the dorsal spinal cord of freely moving rats following veratridine and nociceptive stimulation. Journal of Neurochemistry. 1988;51:127–132.

Sluka K.A., Westlund K.N. An experimental arthritis in rats: dorsal horn aspartate and glutamate increases. Neuroscience Letters. 1992;145:141–144.

Smith P.B., Martin B.R. Spinal mechanisms of delta 9-tetrahydrocannabinol-induced analgesia. Brain Res. 1992;578(1-2):8–12.

Snider W.D., McMahon S.B. Tackling pain at the source: new ideas about nociceptors. Neuron. 1998;20:629–632.

Sokal D.M., Chapman V. Inhibitory effects of spinal baclofen on spinal dorsal horn neurones in inflamed and neuropathic rats in vivo. Brain Research. 2003;987:67–75.

Solomon R.E., Gebhart G.F. Mechanisms of effects of intrathecal serotonin on nociception and blood pressure in rats. Journal of Pharmacology and Experimental Therapeutics. 1988;245:905–912.

Sorkin L.S. NMDA evokes an L-NAME sensitive spinal release of glutamate and citrulline. Neuroreport. 1993;4:479–482.

Sorkin L.S., Doom C.M., et al. Secondary hyperalgesia in the rat first degree burn model is independent of spinal cyclooxygenase and nitric oxide synthase. European journal of pharmacology. 2008;587(1-3):118–123.

Sorkin L.S., Maruyama K., Boyle D.L., et al. Spinal adenosine agonist reduces c-fos and astrocyte activation in dorsal horn of rats with adjuvant-induced arthritis. Neuroscience Letters. 2003;340:119–122.

Sorkin L.S., McAdoo D.J., Willis W.D. Raphe magnus stimulation–induced antinociception in the cat is associated with release of amino acids as well as serotonin in the lumbar dorsal horn. Brain Research. 1993;618:95–108.

Sorkin L.S., Westlund K.N., Sluka K.A., et al. Neural changes in acute arthritis in monkeys. IV. Time-course of amino acid release into the lumbar dorsal horn. Brain Research Reviews. 1992;17:39–50.

Sorkin L.S., Yaksh T.L., Doom C.M., et al. Pain models display differential sensitivity to Ca2+-permeable non-NMDA glutamate receptor antagonists. Anesthesiology. 2001;95:965–973.

Sosnowski M., Stevens C.W., Yaksh T.L. Assessment of the role of A1/A2 adenosine receptors mediating the purine antinociception, motor and autonomic function in the rat spinal cord. Journal of Pharmacology and Experimental Therapeutics. 1989;250:915–922.

Sosnowski M., Yaksh T.L. Role of spinal adenosine receptors in modulating the hyperesthesia produced by spinal glycine receptor antagonism. Anesthesia and Analgesia. 1989;69:587–592.

Spampinato S., Ferri S. Pharmacology of spinal peptides affecting sensory and motor functions: dynorphins, somatostatins and tachykinins. Pharmacological Research. 1991;23:113–127.

Spike R.C., Puskar Z., Andrew D., et al. A quantitative and morphological study of projection neurons in lamina I of the rat lumbar spinal cord. European Journal of Neuroscience. 2003;18:2433–2448.

Standaert D.G., Watson S.J., Houghten R.A., et al. Opioid peptide immunoreactivity in spinal and trigeminal dorsal horn neurons projecting to the parabrachial nucleus in the rat. Journal of Neuroscience. 1986;6:1220–1226.

Stanfa L.C., Dickenson A.H. Inflammation alters the effects of mGlu receptor agonists on spinal nociceptive neurones. European Journal of Pharmacology. 1998;347:165–172.

Stanfa L.C., Hampton D.W., Dickenson A.H. Role of Ca2+-permeable non-NMDA glutamate receptors in spinal nociceptive transmission. Neuroreport. 2000;11:3199–3202.

Stanzione P., Zieglgansberger W. Action of neurotensin on spinal cord neurons in the rat. Brain Research. 1983;268:111–118.

Steiger J.L., Russek S.J. GABAA receptors: building the bridge between subunit mRNAs, their promoters, and cognate transcription factors. Pharmacology and Therapeutics. 2004;101:259–281.

Steinhoff M., Vergnolle N., Young S.H., et al. Agonists of proteinase-activated receptor 2 induce inflammation by a neurogenic mechanism. Nature Medicine. 2000;6:151–158.

Stevens B., Fields R.D. Response of Schwann cells to action potentials in development. Science. 2000;287:2267–2271.

Stevens C.W. Opioid research in amphibians: an alternative pain model yielding insights on the evolution of opioid receptors. Brain Research. Brain Research Reviews. 2004;46:204–215.

Stevens C.W., Yaksh T.L. Dynorphin A and related peptides administered intrathecally in the rat: a search for putative kappa opiate receptor activity. Journal of Pharmacology and Experimental Therapeutics. 1986;238:833–838.

Stewart W., Maxwell D.J. Distribution of and organisation of dorsal horn neuronal cell bodies that possess the muscarinic m2 acetylcholine receptor. Neuroscience. 2003;119:121–135.

Stone L.S., Broberger C., Vulchanova L., et al. Differential distribution of alpha2A and alpha2C adrenergic receptor immunoreactivity in the rat spinal cord. Journal of Neuroscience. 1998;18:5928–5937.

Storer R.J., Goadsby P.J. Microiontophoretic application of serotonin (5HT)1B/1D agonists inhibits trigeminal cell firing in the cat. Brain. 1997;120:2171–2177.

Stucky C.L., Galeazza M.T., Seybold V.S. Time-dependent changes in Bolton-Hunter–labeled 125I-substance P binding in rat spinal cord following unilateral adjuvant-induced peripheral inflammation. Neuroscience. 1993;57:397–409.

Sugiura T., Kondo S., Sukagawa A., et al. 2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain. Biochemical and Biophysical Research Communication. 1995;215:89–97.

Sun R.Q., Lawand N.B., Willis W.D. The role of calcitonin gene–related peptide (CGRP) in the generation and maintenance of mechanical allodynia and hyperalgesia in rats after intradermal injection of capsaicin. Pain. 2003;104:201–208.

Sun X.F., Larson A.A. Behavioral sensitization to kainic acid and quisqualic acid in mice: comparison to NMDA and substance P responses. Journal of Neuroscience. 1991;11:3111–3123.

Supowit S.C., Hallman D.M., Zhao H., et al. Alpha 2-adrenergic receptor activation inhibits calcitonin gene–related peptide expression in cultured dorsal root ganglia neurons. Brain Research. 1998;782:184–193.

Suzuki R., Morcuende S., Webber M., et al. Superficial NK1-expressing neurons control spinal excitability through activation of descending pathways. Nature Neuroscience. 2002;5:1319–1326.

Svensson C.I., Hua X.Y., Protter A.A., et al. Spinal p38 MAP kinase is necessary for NMDA-induced spinal PGE2 release and thermal hyperalgesia. Neuroreport. 2003;14:1153–1157.

Svensson C.I., Yaksh T.L. The spinal phospholipase-cyclooxygenase-prostanoid cascade in nociceptive processing. Annual Review of Pharmacology and Toxicology. 2002;42:553–583.

Sweeney M.I., White T.D., Sawynok J. Morphine, capsaicin and K+ release purines from capsaicin-sensitive primary afferent nerve terminals in the spinal cord. Journal of Pharmacology and Experimental Therapeutics. 1989;248:447–454.

Taal W., Holstege J.C. GABA and glycine frequently colocalize in terminals on cat spinal motoneurons. Neuroreport. 1994;5:2225–2228.

Tachibana M., Wenthold R.J., Morioka H., et al. Light and electron microscopic immunocytochemical localization of AMPA-selective glutamate receptors in the rat spinal cord. Journal of Comparative Neurology. 1994;344:431–454.

Taiwo Y.O., Levine J.D. Indomethacin blocks central nociceptive effects of PGF2 alpha. Brain Research. 1986;373:81–84.

Takano M., Takano Y., Yaksh T.L. Release of calcitonin gene–related peptide (CGRP), substance P (SP), and vasoactive intestinal polypeptide (VIP) from rat spinal cord: modulation by alpha 2 agonists. Peptides. 1993;14:371–378.

Takasusuki T., Yaksh T.L. Regulation of spinal substance p release by intrathecal calcium channel blockade. Anesthesiology. 2011;115(1):153–164.

Takeda D., Nakatsuka T., Papke R., et al. Modulation of inhibitory synaptic activity by a non-alpha4beta2, non-alpha7 subtype of nicotinic receptors in the substantia gelatinosa of adult rat spinal cord. Pain. 2003;101:13–23.

Tao Y.X. Dorsal horn alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor trafficking in inflammatory pain. Anesthesiology. 2010;112(5):1259–1265.

Tang Q., Lynch R.M., Porreca F., et al. Dynorphin A elicits an increase in intracellular calcium in cultured neurons via a non-opioid, non-NMDA mechanism. Journal of Neurophysiology. 2000;83:2610–2615.

Taniguchi K., Shinjo K., Mizutani M., et al. Antinociceptive activity of CP-101,606, an NMDA receptor NR2B subunit antagonist. British Journal of Pharmacology. 1997;122:809–812.

Tao F., Tao Y.X., Mao P., et al. Intact carrageenan-induced thermal hyperalgesia in mice lacking inducible nitric oxide synthase. Neuroscience. 2003;120:847–854.

Tao F., Tao Y.X., Zhao C., et al. Differential roles of neuronal and endothelial nitric oxide synthases during carrageenan-induced inflammatory hyperalgesia. Neuroscience. 2004;128:421–430.

Tata A.M., Vilaro M.T., Mengod G. Muscarinic receptor subtypes expression in rat and chick dorsal root ganglia. Brain Research. Molecular Brain Research. 2000;82:1–10.

Tessler A., Himes B.T., Gruber-Bollinger J., et al. Characterization of forms of immunoreactive somatostatin in sensory neuron and normal and deafferented spinal cord. Brain Research. 1986;370:232–240.

Testa C.M., Friberg I.K., Weiss S.W., et al. Immunohistochemical localization of metabotropic glutamate receptors mGluR1a and mGluR2/3 in the rat basal ganglia. Journal of Comparative Neurology. 1998;390:5–19.

Thomas N.K., Wright R.A., et al. (S)-3,4-DCPG, a potent and selective mGlu8a receptor agonist, activates metabotropic glutamate receptors on primary afferent terminals in the neonatal rat spinal cord. Neuropharmacology. 2001;40(3):311–318.

Tiseo P.J., Adler M.W., Liu-Chen L.Y. Differential release of substance P and somatostatin in the rat spinal cord in response to noxious cold and heat; effect of dynorphin A(1–17). Journal of Pharmacology and Experimental Therapeutics. 1990;252:539–545.

Todd A.J. An electron microscope study of glycine-like immunoreactivity in laminae I-III of the spinal dorsal horn of the rat. Neuroscience. 1990;39:387–394.

Todd A.J. GABA and glycine in synaptic glomeruli of the rat spinal dorsal horn. European Journal of Neuroscience. 1996;8:2492–2498.

Todd A.J. Anatomy of primary afferents and projection neurones in the rat spinal dorsal horn with particular emphasis on substance P and the neurokinin 1 receptor. Experimental Physiology. 2002;87:245–249.

Todd A.J., Hughes D.I., Polgár E., et al. The expression of vesicular glutamate transporters VGLUT1 and VGLUT2 in neurochemically defined axonal populations in the rat spinal cord with emphasis on the dorsal horn. European Journal of Neuroscience. 2003;17:13–27.

Todd A.J., McGill M.M., Shehab S.A. Neurokinin 1 receptor expression by neurons in laminae I, III and IV of the rat spinal dorsal horn that project to the brainstem. European Journal of Neuroscience. 2000;12:689–700.

Todd A.J., Polgár E., Watt C., et al. Neurokinin 1 receptor–expressing projection neurons in laminae III and IV of the rat spinal cord have synaptic AMPA receptors that contain GluR2, GluR3 and GluR4 subunits. European Journal of Neuroscience. 2009;29:718–726.

Todd A.J., Sullivan A.C. Light microscope study of the coexistence of GABA-like and glycine-like immunoreactivities in the spinal cord of the rat. Journal of Comparative Neurology. 1990;296:496–505.

Todd A.J., Watt C., Spike R.C., et al. Colocalization of GABA, glycine, and their receptors at synapses in the rat spinal cord. Journal of Neuroscience. 1996;16:974–982.

Todorovic S., Anderson E.G. 5-HT2 and 5-HT3 receptors mediate two distinct depolarizing responses in rat dorsal root ganglion neurons. Brain Research. 1990;511:71–79.

Todorovic S., Anderson E.G. Serotonin preferentially hyperpolarizes capsaicin-sensitive C type sensory neurons by activating 5-HT1A receptors. Brain Research. 1992;585:212–218.

Todorovic S.M., Jevtovic-Todorovic V. T-type voltage-gated calcium channels as targets for the development of novel pain therapies. British Journal of Pharmacology. 2011;163:484–495.

Tracey D.J., De Biasi S., Phend K., et al. Aspartate-like immunoreactivity in primary afferent neurons. Neuroscience. 1991;40:673–686.

Tseng L.F., Collins K.A. Cholecystokinin administered intrathecally selectively antagonizes intracerebroventricular beta-endorphin–induced tail-flick inhibition in the mouse. Journal of Pharmacology and Experimental Therapeutics. 1992;260:1086–1092.

Tsou K., Lowitz K.A., et al. Suppression of noxious stimulus-evoked expression of Fos protein-like immunoreactivity in rat spinal cord by a selective cannabinoid agonist. Neuroscience. 1996;70(3):791–798.

Tsumoto T. Excitatory amino acid transmitters and their receptors in neural circuits of the cerebral neocortex. Neuroscience Research. 1990;9:79–102.

Tyce G.M., Yaksh T.L. Monoamine release from cat spinal cord by somatic stimuli: an intrinsic modulatory system. Journal of Physiology. 1981;314:513–529.

Uda R., Horiguchi S., Ito S., et al. Nociceptive effects induced by intrathecal administration of prostaglandin D2, E2, or F2 alpha to conscious mice. Brain Research. 1990;510:26–32.

Urban L., Randic M. Slow excitatory transmission in rat dorsal horn: possible mediation by peptides. Brain Research. 1984;290:336–341.

van den Pol A.N., Gorcs T. Glycine and glycine receptor immunoreactivity in brain and spinal cord. Journal of Neuroscience. 1988;8:472–492.

Vanderah T.W., Gardell L.R., Burgess S.E., et al. Dynorphin promotes abnormal pain and spinal opioid antinociceptive tolerance. Journal of Neuroscience. 2000;20:7074–7079.

Vanderah T.W., Laughlin T., Lashbrook J.M., et al. Single intrathecal injections of dynorphin A or des-Tyr-dynorphins produce long-lasting allodynia in rats: blockade by MK-801 but not naloxone. Pain. 1996;68:275–281.

Vane J.R., Bakhle Y.S., Botting R.M. Cyclooxygenases 1 and 2. Annual Review of Pharmacology and Toxicology. 1998;38:97–120.

Verge V.M., Wiesenfeld-Hallin Z., Hokfelt T. Cholecystokinin in mammalian primary sensory neurons and spinal cord: in situ hybridization studies in rat and monkey. European Journal of Neuroscience. 1993;5:240–250.

Vergnolle N., Bunnett N.W., Sharkey K.A., et al. Proteinase-activated receptor-2 and hyperalgesia: a novel pain pathway. Nature Medicine. 2001;7:821–826.

Verhage M., McMahon H.T., Ghijsen W.E., Boomsma F., Scholten G., Wiegant V.M., Nicholls D. Differential release of amino acids, neuropeptides, and catecholamines from isolated nerve terminals. Neuron. 1991;6:517–524.

Versteeg H.H., van Bergen en Henegouwen P.M., van Deventer S.J., et al. Cyclooxygenase-dependent signalling: molecular events and consequences. FEBS Letters. 1999;445:1–5.

Vidnyanszky Z., Hamori J., Negyessy L., et al. Cellular and subcellular localization of the mGluR5a metabotropic glutamate receptor in rat spinal cord. Neuroreport. 1994;6:209–213.

Vikman K.S., Rycroft B.K., Christie M.J. Switch to Ca2+-permeable AMPA and reduced NR2B NMDA receptor–mediated neurotransmission at dorsal horn nociceptive synapses during inflammatory pain in the rat. Journal of Physiology. 2008;586:515–527.

Vincent J.P., Mazella J., Kitabgi P. Neurotensin and neurotensin receptors. Trends in Pharmacological Sciences. 1999;20:302–309.

Vulchanova L., Arvidsson U., Riedl M., et al. Differential distribution of two ATP-gated channels (P2X receptors) determined by immunocytochemistry. Proceedings of the National Academy of Sciences of the United States of America. 1996;93:8063–8067.

Vulchanova L., Olson T.H., Stone L.S., et al. Cytotoxic targeting of isolectin IB4–binding sensory neurons. Neuroscience. 2001;108:143–155.

Vulchanova L., Riedl M.S., Shuster S.J., et al. Immunohistochemical study of the P2X2 and P2X3 receptor subunits in rat and monkey sensory neurons and their central terminals. Neuropharmacology. 1997;36:1229–1242.

Walker G.E., Yaksh T.L. Studies on the effects of intrathalamically injected DADL and morphine on nociceptive thresholds and electroencephalographic activity: a thalamic delta receptor syndrome. Brain Res. 1986;383(1-2):1–14.

Walker J.M., Huang S.M., Strangman N.M., et al. Pain modulation by release of the endogenous cannabinoid anandamide. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:12198–12203.

Wang H., Ubl J.J., Reiser G. Four subtypes of protease-activated receptors, co-expressed in rat astrocytes, evoke different physiological signaling. Glia. 2002;37:53–63.

Wang J.K. Antinociceptive effect of intrathecally administered serotonin. Anesthesiology. 1977;47:269–271.

Wang Y.T., Salter M.W. Regulation of NMDA receptors by tyrosine kinases and phosphatases. Nature. 1994;369:233–235.

Wang Y.T., Wu J.J., Wu Z., et al. Regulation of AMPA receptors in spinal nociception. Molecular Pain. 2010;6:5.

Waterman A., Livingston A., Bouchenafa O. Analgesic effects of intrathecally-applied alpha 2-adrenoceptor agonists in conscious, unrestrained sheep. Neuropharmacology. 1988;27:213–216.

Waters S.M., Krause J.E. Distribution of galanin-1, -2 and -3 receptor messenger RNAs in central and peripheral rat tissues. Neuroscience. 2000;95:265–271.

Wajima Z., Hua X.Y., et al. Inhibition of spinal protein kinase C blocks substance P-mediated hyperalgesia. Brain research. 2000;877(2):314–321.

Weng H.R., Aravindan N., et al. Spinal glial glutamate transporters downregulate in rats with taxol-induced hyperalgesia. Neuroscience letters. 2005;386(1):18–22.

Westlund K.N., Bowker R.M., Ziegler M.G., et al. Noradrenergic projections to the spinal cord of the rat. Brain Research. 1983;263:15–31.

White W.F., Heller A.H. Glycine receptor alteration in the mutant mouse spastic. Nature. 1982;298:655–657.

Whiting P.J. GABA-A receptor subtypes in the brain: a paradigm for CNS drug discovery? Drug Discovery Today. 2003;8:445–450.

Wiertelak E.P., Maier S.F., Watkins L.R. Cholecystokinin antianalgesia: safety cues abolish morphine analgesia. Science. 1992;256:830–833.

Wiesenfeld-Hallin Z. Intrathecal somatostatin modulates spinal sensory and reflex mechanisms: behavioral and electrophysiological studies in the rat. Neuroscience Letters. 1985;62:69–74.

Wiesenfeld-Hallin Z. Substance P and somatostatin modulate spinal cord excitability via physiologically different sensory pathways. Brain Research. 1986;372:172–175.

Wiesenfeld-Hallin Z. Nerve section alters the interaction between C-fibre activity and intrathecal neuropeptides on the flexor reflex in rat. Brain Research. 1989;489:129–136.

Wiesenfeld-Hallin Z., Villar M.J., Hokfelt T. Intrathecal galanin at low doses increases spinal reflex excitability in rats more to thermal than mechanical stimuli. Experimental Brain Research. 1988;71:663–666.

Wilding T.J., Huettner J.E. Functional diversity and developmental changes in rat neuronal kainate receptors. Journal of Physiology. 2001;532:411–421.

Willcockson H., Valtschanoff J. AMPA and NMDA glutamate receptors are found in both peptidergic and non-peptidergic primary afferent neurons in the rat. Cell and Tissue Research. 2008;334:17–23.

Willcockson W.S., Chung J.M., Hori Y., et al. Effects of iontophoretically released amino acids and amines on primate spinothalamic tract cells. Journal of Neuroscience. 1984;4:732–740.

Willis W.D., Jr. Dorsal horn neurophysiology of pain. Annals of the New York Academy of Sciences. 1988;531:76–89.

Willis W.D. Role of neurotransmitters in sensitization of pain responses. Annals of the New York Academy of Sciences. 2001;933:142–156.

Willis W.D. Long-term potentiation in spinothalamic neurons. Brain Research. Brain Research Reviews. 2002;40:202–214.

Willis W.D., Westlund K.N. Neuroanatomy of the pain system and of the pathways that modulate pain. Journal of Clinical Neurophysiology. 1997;14:2–31.

Wilson P.R., Yaksh T.L. Baclofen is antinociceptive in the spinal intrathecal space of animals. European Journal of Pharmacology. 1978;51:323–330.

Wisden W., Gundlach A.L., Barnard E.A., et al. Distribution of GABAA receptor subunit mRNAs in rat lumbar spinal cord. Brain Research. Molecular Brain Research. 1991;10:179–183.

Woolf C.J., King A.E. Physiology and morphology of multireceptive neurons with C-afferent fiber inputs in the deep dorsal horn of the rat lumbar spinal cord. Journal of Neurophysiology. 1987;58:460–479.

Woolf C.J., Thompson S.W. The induction and maintenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation; implications for the treatment of post-injury pain hypersensitivity states. Pain. 1991;44:293–299.

Woolf C.J., Wall P.D. Relative effectiveness of C primary afferent fibers of different origins in evoking a prolonged facilitation of the flexor reflex in the rat. Journal of Neuroscience. 1986;6:1433–1442.

Worsley M.A., Todd A.J., King A.E. Serotoninergic-mediated inhibition of substance P sensitive deep dorsal horn neurons: a combined electrophysiological and morphological study in vitro. Experimental Brain Research. 2005;160:360–367.

Wright D.H., Nantel F., Metters K.M., et al. A novel biological role for prostaglandin D2 is suggested by distribution studies of the rat DP prostanoid receptor. European Journal of Pharmacology. 1999;377:101–115.

Xi Z.X., Shen H., et al. Inhibition of non-vesicular glutamate release by group III metabotropic glutamate receptors in the nucleus accumbens. Journal of neurochemistry. 2003;87(5):1204–1212.

Xie J., Ho Lee Y., Wang C., et al. Differential expression of alpha1-adrenoceptor subtype mRNAs in the dorsal root ganglion after spinal nerve ligation. Brain Research. Molecular Brain Research. 2001;93:164–172.

Xie Y., Zhang J., Petersen M., et al. Functional changes in dorsal root ganglion cells after chronic nerve constriction in the rat. Journal of Neurophysiology. 1995;73:1811–1820.

Xin W.J., Weng H.R., Dougherty P.M. Plasticity in expression of the glutamate transporters GLT-1 and GLAST in spinal dorsal horn glial cells following partial sciatic nerve ligation. Molecular Pain. 2009;5:15.

Xu X.J., Wiesenfeld-Hallin Z. An analogue of growth hormone releasing factor (GRF), (Ac-Try1, D-Phe2)-GRF-(1-29), specifically antagonizes the facilitation of the flexor reflex induced by intrathecal vasoactive intestinal peptide in rat spinal cord. Neuropeptides. 1991;18:129–135.

Xu X.J., Wiesenfeld-Hallin Z. Intrathecal pituitary adenylate cyclase activating polypeptide facilitates the spinal nociceptive flexor reflex in the rat. Neuroscience. 1996;72:801–804.

Yaksh T.L. Inhibition by etorphine of the discharge of dorsal horn neurons: effects on the neuronal response to both high- and low-threshold sensory input in the decerebrate spinal cat. Experimental Neurology. 1978;60:23–40.

Yaksh T.L. Direct evidence that spinal serotonin and noradrenaline terminals mediate the spinal antinociceptive effects of morphine in the periaqueductal gray. Brain Research. 1979;160:180–185.

Yaksh T.L. Central and peripheral mechanisms for the antianalgesic action of acetylsalicylic acid. In: Barnet J.M., Hirsh J., Mustard J.F., eds. Acetylsalicylic acid: new uses for an old drug. New York: Raven Press; 1982:137–152.

Yaksh T.L., Elde R.P. Factors governing release of methionine enkephalin-like immunoreactivity from mesencephalon and spinal cord of the cat in vivo. J Neurophysiol. 1981;46(5):1056–1075.

Yaksh T.L., Reddy S.V. Studies in the primate on the analgetic effects associated with intrathecal actions of opiates, alpha-adrenergic agonists and baclofen. Anesthesiology. 1981;54(6):451–467.

Yaksh T.L. Pharmacology of spinal adrenergic systems which modulate spinal nociceptive processing. Pharmacology, Biochemistry, and Behavior. 1985;22:845–858.

Yaksh T.L. Spinal opiates: a review of their effect on spinal function with emphasis on pain processing. Acta Anaesthesiologica Scandinavica. Supplementum. 1987;85:25–37.

Yaksh T.L. Behavioral and autonomic correlates of the tactile evoked allodynia produced by spinal glycine inhibition: effects of modulatory receptor systems and excitatory amino acid antagonists. Pain. 1989;37:111–123.

Yaksh T.L. Pharmacology and mechanisms of opioid analgesic activity. Acta Anaesthesiologica Scandinavica. 1997;41:94–111.

Yaksh T.L. Regulation of spinal nociceptive processing: where we went when we wandered onto the path marked by the gate. Pain. 1999;6(Suppl):S149–S152.

Yaksh T.L. Calcium channels as therapeutic targets in neuropathic pain. Journal of Pain. 2006;7:S13–S30.

Yaksh T.L., Abay E.O., 2nd., Go V.L. Studies on the location and release of cholecystokinin and vasoactive intestinal peptide in rat and cat spinal cord. Brain Research. 1982;242:279–290.

Yaksh T.L., Chaplan S.R., Malmberg A.B., et al. Future directions in the pharmacological management of hyperalgesic and allodynic pain states: the NMDA receptor. NIDA Research Monograph. 1995;147:84–103.

Yaksh T.L., Dirig D.M., Conway C.M., et al. The acute antihyperalgesic action of NSAIDs and release of spinal PGE2 is mediated by the inhibition of constitutive spinal COX-2 but not COX-1. Journal of Neuroscience. 2001;21:5847–5853.

Yaksh T.L., Elde R.P. Factors governing release of methionine enkephalin–like immunoreactivity from mesencephalon and spinal cord of the cat in vivo. Journal of Neurophysiology. 1981;46:1056–1075.

Yaksh T.L., Jessell T.M., Gamse R., et al. Intrathecal morphine inhibits substance P release from mammalian spinal cord in vivo. Nature. 1980;286:155–157.

Yaksh T.L., Malkmus S.A. Animal models of intrathecal and epidural drug delivery. In: Yaksh T.L., ed. Spinal drug delivery. Amsterdam: Elsevier; 1999:317–344.

Yaksh T.L., Michener S.R., Bailie J.E., et al. Survey of distribution of substance P, vasoactive intestinal polypeptide, cholecystokinin, neurotensin, Met-enkephalin, bombesin and PHI in the spinal cord of cat, dog, sloth and monkey. Peptides. 1988;9:357–372.

Yaksh T.L., Ozaki G., McCumber D., et al. An automated flinch detecting system for use in the formalin nociceptive bioassay. Journal of Applied Physiology. 2001;90:2386–2402.

Yaksh T.L., Reddy S.V. Studies in the primate on the analgetic effects associated with intrathecal actions of opiates, alpha-adrenergic agonists and baclofen. Anesthesiology. 1981;54:451–467.

Yaksh T.L., Rudy T.A. Analgesia mediated by a direct spinal action of narcotics. Science. 1976;192:1357–1358.

Yaksh T.L., Rudy T.A. Narcotic analgesics: CNS sites and mechanisms of action as revealed by intracerebral injection techniques. Pain. 1978;4:299–359.

Yaksh T.L., Schmauss C., Micevych P.E., et al. Pharmacological studies on the application, disposition, and release of neurotensin in the spinal cord. Annals of the New York Academy of Sciences. 1982;400:228–243.

Yaksh T.L., Wilson P.R. Spinal serotonin terminal system mediates antinociception. Journal of Pharmacology and Experimental Therapeutics. 1979;208:446–453.

Yaksh T.L., Yamamoto T., Myers R.R., et al. Pharmacology of nerve compression–evoked hyperesthesia. In: Willis Jr W.D., ed. Hyperalgesia and allodynia. New York: Raven Press; 1992:245–258.

Yamamoto T., Nozaki-Taguchi N. The role of cyclooxygenase-1 and -2 in the rat formalin test. Anesthesia and Analgesia. 2002;94:962–967.

Yamamoto T., Shimoyama N., Mizuguchi T., et al. The effects of morphine, MK-801, an NMDA antagonist, and CP-96,345, an NK1 antagonist, on the hyperesthesia evoked by carrageenan injection in the rat paw. Anesthesiology. 1993;78:124–133.

Yamamoto T., Yaksh T.L. Stereospecific effects of a nonpeptidic NK1 selective antagonist, CP-96,345: antinociception in the absence of motor dysfunction. Life Sciences. 1991;49:1955–1963.

Yamamoto T., Yaksh T.L. Comparison of the antinociceptive effects of pre- and posttreatment with intrathecal morphine and MK801, an NMDA antagonist, on the formalin test in the rat. Anesthesiology. 1992;77:757–763.

Yashpal K., Radhakrishnan V., Coderre T.J., et al. CP-96,345, but not its stereoisomer, CP-96,344, blocks the nociceptive responses to intrathecally administered substance P and to noxious thermal and chemical stimuli in the rat. Neuroscience. 1993;52:1039–1047.

Yashpal K., Sarrieau A., Quirion R., et al. [125I]vasoactive intestinal polypeptide binding sites: quantitative autoradiographic distribution in the rat spinal cord. Journal of Chemical Neuroanatomy. 1991;4:439–446.

Yasphal K., Wright D.M., Henry J.L., et al. Substance P reduces tail-flick latency: implications for chronic pain syndromes. Pain. 1982;14:155–167.

Yaster M., Guan X., Petralia R.S., et al. Effect of inhibition of spinal cord glutamate transporters on inflammatory pain induced by formalin and complete Freund’s adjuvant. Anesthesiology. 2011;114:412–423.

Yermakova A.V., Rollins J., Callahan L.M., et al. Cyclooxygenase-1 in human Alzheimer and control brain: quantitative analysis of expression by microglia and CA3 hippocampal neurons. Journal of Neuropathology and Experimental Neurology. 1999;58:1135–1146.

Yokota T., Nishikawa N., Nishikawa Y., et al. Effects of strychnine upon different classes of trigeminal subnucleus caudalis neurons. Brain Research. 1979;168:430–434.

Yoshimura M., Jessell T. Amino acid–mediated EPSPs at primary afferent synapses with substantia gelatinosa neurones in the rat spinal cord. Journal of Physiology. 1990;430:315–335.

Yoshimura M., Jessell T.M. Primary afferent–evoked synaptic responses and slow potential generation in rat substantia gelatinosa neurons in vitro. Journal of Neurophysiology. 1989;62:96–108.

Yoshimura M., Nishi S. Blind patch-clamp recordings from substantia gelatinosa neurons in adult rat spinal cord slices: pharmacological properties of synaptic currents. Neuroscience. 1993;53:519–526.

Yoshimura M., Nishi S. Primary afferent–evoked glycine- and GABA-mediated IPSPs in substantia gelatinosa neurones in the rat spinal cord in vitro. Journal of Physiology. 1995;482:29–38.

Young M.R., Blackburn-Munro G., Dickenson T., et al. Antisense ablation of type I metabotropic glutamate receptor mGluR1 inhibits spinal nociceptive transmission. Journal of Neuroscience. 1998;18:10180–10188.

Young M.R., Fleetwood-Walker S.M., Mitchell R., et al. The involvement of metabotropic glutamate receptors and their intracellular signalling pathways in sustained nociceptive transmission in rat dorsal horn neurons. Neuropharmacology. 1995;34:1033–1041.

Young M.R., Fleetwood-Walker S.M., Mitchell R., et al. Evidence for a role of metabotropic glutamate receptors in sustained nociceptive inputs to rat dorsal horn neurons. Neuropharmacology. 1994;33:141–144.

Yousufzai S.Y., Chen A.L., Abdel-Latif A.A., et al. Species differences in the effects of prostaglandins on inositol trisphosphate accumulation, phosphatidic acid formation, myosin light chain phosphorylation and contraction in iris sphincter of the mammalian eye: interaction with the cyclic AMP system. Journal of Pharmacology and Experimental Therapeutics. 1988;247:1064–1072.

Yu L.C., Hansson P., Brodda-Jansen G., et al. Intrathecal CGRP8-37–induced bilateral increase in hindpaw withdrawal latency in rats with unilateral inflammation. British Journal of Pharmacology. 1996;117:43–50.

Yu X.M., Askalan R., Keil G.J., 2nd., et al. NMDA channel regulation by channel-associated protein tyrosine kinase Src. Science. 1997;275:674–678.

Yung K.K. Localization of glutamate receptors in dorsal horn of rat spinal cord. Neuroreport. 1998;9:1639–1644.

Zahn P.K., Brennan T.J. Lack of effect of intrathecally administered N-methyl-D-aspartate receptor antagonists in a rat model for postoperative pain. Anesthesiology. 1998;88:143–156.

Zahn P.K., Umali E., Brennan T.J., et al. Intrathecal non-NMDA excitatory amino acid receptor antagonists inhibit pain behaviors in a rat model of postoperative pain. Pain. 1998;74:213–223.

Zamponi G.W., Lewis R.J., Todorovic S.M., et al. Role of voltage-gated calcium channels in ascending pain pathways. Brain Research Reviews. 2009;60:84–89.

Zaratin P., Angelici O., Clarke G.D., et al. NK3 receptor blockade prevents hyperalgesia and the associated spinal cord substance P release in monoarthritic rats. Neuropharmacology. 2000;39:141–149.

Zeitz K.P., Guy N., Malmberg A.B., et al. The 5-HT3 subtype of serotonin receptor contributes to nociceptive processing via a novel subset of myelinated and unmyelinated nociceptors. Journal of Neuroscience. 2002;22:1010–1019.

Zemlan F.P., Murphy A.Z., Behbehani M.M., et al. 5-HT1A receptors mediate the effect of the bulbospinal serotonin system on spinal dorsal horn nociceptive neurons. Pharmacology. 1994;48:1–10.

Zhang H., Nei H., Dougherty P.M. A p38 mitogen–activated protein kinase–dependent mechanism of disinhibition in spinal synaptic transmission induced by tumor necrosis factor-alpha. Journal of Neuroscience. 2010;30:12844–12855.

Zhang J.M., Li H., Liu B., et al. Acute topical application of tumor necrosis factor alpha evokes protein kinase A–dependent responses in rat sensory neurons. Journal of Neurophysiology. 2002;88:1387–1392.

Zhang L., Berta T., Xu Z.Z., et al. TNF-alpha contributes to spinal cord synaptic plasticity and inflammatory pain: distinct role of TNF receptor subtypes 1 and 2. Pain. 2011;152:419–427.

Zhang L., Lu Y., Chen Y., et al. Group I metabotropic glutamate receptor antagonists block secondary thermal hyperalgesia in rats with knee joint inflammation. Journal of Pharmacology and Experimental Therapeutics. 2002;300:149–156.

Zhang W., Gardell S., Zhang D., et al. Neuropathic pain is maintained by brainstem neurons co-expressing opioid and cholecystokinin receptors. Brain: A Journal of Neurology. 2009;132:778–787.

Zhang X., Nicholas A.P., Hokfelt T., et al. Ultrastructural studies on peptides in the dorsal horn of the spinal cord—I. Co-existence of galanin with other peptides in primary afferents in normal rats. Neuroscience. 1993;57:365–384.

Zhang Y., Malmberg A.B., Yaksh T.L., et al. Capsaicin-evoked release of pituitary adenylate cyclase activating peptide (PACAP) and calcitonin gene–related peptide (CGRP) from rat spinal cord in vivo. Regulatory Peptides. 1997;69:83–87.

Zhang Y.Z., Sjolund B., Moller K., et al. Pituitary adenylate cyclase activating peptide produces a marked and long-lasting depression of a C-fibre–evoked flexion reflex. Neuroscience. 1993;57:733–737.

Zheng J.H., Chen J. Modulatory roles of the adenosine triphosphate P2x-purinoceptor in generation of the persistent nociception induced by subcutaneous bee venom injection in the conscious rat. Neuroscience Letters. 2000;278:41–44.

Zhou Z., Peng X., Hagshenas J., et al. A novel cell-cell signaling by microglial transmembrane TNFalpha with implications for neuropathic pain. Pain. 2010;151:296–306.

Zhuo M., Gebhart G.F. Tonic cholinergic inhibition of spinal mechanical transmission. Pain. 1991;46:21–222.

Zieglgansberger W., Herz A. Changes of cutaneous receptive fields of spino-cervical-tract neurones and other dorsal horn neurones by microelectrophoretically administered amino acids. Experimental Brain Research. 1971;13:111–126.

Zimmer A., Zimmer A.M., Hohmann A.G., et al. Increased mortality, hypoactivity, and hypoalgesia in cannabinoid CB1 receptor knockout mice. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:5780–5785.

Zimmermann H., Braun N., Kegel B., et al. New insights into molecular structure and function of ectonucleotidases in the nervous system. Neurochemistry International. 1998;32:421–425.

Zygmunt P.M., Petersson J., Andersson D.A., et al. Vanilloid receptors on sensory nerves mediate the vasodilator action of anandamide. Nature. 1999;400:452–457.

Suggested Readings

Boyce S., Wyatt A., Webb J.K., 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.

Buczynski M.W., Svensson C.I., Dumlao D.S., et al. Inflammatory hyperalgesia induces essential bioactive lipid production in the spinal cord. Journal of Neurochemistry. 2010;114:981–993.

Choi J.I., Svensson C.I., Koehrn F.J., et al. Peripheral inflammation induces tumor necrosis factor dependent AMPA receptor trafficking and Akt phosphorylation in spinal cord in addition to pain behavior. Pain. 2010;149:243–253.

Damaj M.I., Meyer E.M., Martin B.R., et al. The antinociceptive effects of alpha7 nicotinic agonists in an acute pain model. Neuropharmacology. 2000;39:2785–2791.

De Koninck Y. Altered chloride homeostasis in neurological disorders: a new target. Current Opinion in Pharmacology. 2007;7:93–99.

Deuchars S.A., Brooke R.E., Deuchars J., et al. Adenosine A1 receptors reduce release from excitatory but not inhibitory synaptic inputs onto lateral horn neurons. Journal of Neuroscience. 2001;21:6308–6320.

Djouhri L., Koutsikou S., Fang X., et al. Spontaneous pain, both neuropathic and inflammatory, is related to frequency of spontaneous firing in intact C-fiber nociceptors. Journal of Neuroscience. 2006;26:1281–1292.

Dougherty P.M., 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, chemical, thermal, and electrical stimuli. Journal of Neuroscience. 1992;12:3025–3041.

Du J., Zhou S., Carlton S.M. Group II metabotropic glutamate receptor activation attenuates peripheral sensitization in inflammatory states. Neuroscience. 2008;154:754–766.

Gold M.S., Levine J.D., Correa A.M. Modulation of TTX-R INa by PKC and PKA and their role in PGE2-induced sensitization of rat sensory neurons in vitro. Journal of Neuroscience. 1998;18:10345–10355.

Guindon J., Hohmann A.G. The endocannabinoid system and pain. CNS & Neurological Disorders Drug Targets. 2009;8:403–421.

Hartmann B., Ahmadi S., Heppenstall P.A., et al. The AMPA receptor subunits GluR-A and GluR-B reciprocally modulate spinal synaptic plasticity and inflammatory pain. Neuron. 2004;44:637–650.

Harvey R.J., Depner U.B., Wassle H., et al. GlyR alpha3: an essential target for spinal PGE2-mediated inflammatory pain sensitization. Science. 2004;304:884–887.

Hokfelt T., Arvidsson U., Cullheim S., et al. Multiple messengers in descending serotonin neurons: localization and functional implications. Journal of Chemical Neuroanatomy. 2000;18:75–86.

Iadarola M.J., Brady L.S., Draisci G., et al. Enhancement of dynorphin gene expression in spinal cord following experimental inflammation: stimulus specificity, behavioral parameters and opioid receptor binding. Pain. 1988;35:313–326.

Ji R.R., Gereau R.W., 4th., Malcangio M., et al. MAP kinase and pain. Brain Research Reviews. 2009;60:135–148.

Jin X., Gereau R.W., 4th. Acute p38-mediated modulation of tetrodotoxin-resistant sodium channels in mouse sensory neurons by tumor necrosis factor-alpha. Journal of Neuroscience. 2006;26:246–255.

Karim F., Wang C.C., Gereau R.W., 4th. Metabotropic glutamate receptor subtypes 1 and 5 are activators of extracellular signal–regulated kinase signaling required for inflammatory pain in mice. Journal of Neuroscience. 2001;21:3771–3779.

Kumar N., Laferriere A., Yu J.S., et al. Metabotropic glutamate receptors (mGluRs) regulate noxious stimulus–induced glutamate release in the spinal cord dorsal horn of rats with neuropathic and inflammatory pain. Journal of Neurochemistry. 2010;114:281–290.

Leong M.L., Gu M., Speltz-Paiz R., et al. Neuronal loss in the rostral ventromedial medulla in a rat model of neuropathic pain. Journal of Neuroscience. 2011;31:17028–17039.

Liu H., Mantyh P.W., Basbaum A.I., et al. NMDA-receptor regulation of substance P release from primary afferent nociceptors. Nature. 1997;386:721–724.

Liu J., Zhao X., Cao J., et al. Differential roles of PKA and epac on the production of cytokines in the endotoxin-stimulated primary cultured microglia. Journal of Molecular Neuroscience. 2011;45:186–193.

Liu X.J., Salter M.W. Glutamate receptor phosphorylation and trafficking in pain plasticity in spinal cord dorsal horn. European Journal of Neuroscience. 2010;32:278–289.

Malmberg A.B., Yaksh T.L. Hyperalgesia mediated by spinal glutamate or substance P receptor blocked by spinal cyclooxygenase inhibition. Science. 1992;257:1276–1279.

Martin W.J., Malmberg A.B., Basbaum A.I. PKCgamma contributes to a subset of the NMDA-dependent spinal circuits that underlie injury-induced persistent pain. Journal of Neuroscience. 2001;21:5321–5327.

McGeehan G.M., Becherer J.D., Bast R.C., Jr., et al. Regulation of tumour necrosis factor-alpha processing by a metalloproteinase inhibitor. Nature. 1994;370:558–561.

Merighi A., Salio C., Ghirri A., et al. BDNF as a pain modulator. Progress in Neurobiology. 2008;85:297–317.

Milligan E.D., Sloane E.M., Watkins L.R. Glia in pathological pain: a role for fractalkine. Journal of Neuroimmunology. 2008;198:113–120.

Moss A., Blackburn-Munro G., Garry E.M., et al. A role of the ubiquitin-proteasome system in neuropathic pain. Journal of Neuroscience. 2002;22:1363–1372.

Neugebauer V., Chen P.S., Willis W.D., et al. Groups II and III metabotropic glutamate receptors differentially modulate brief and prolonged nociception in primate STT cells. Journal of Neurophysiology. 2000;84:2998–3009.

Niclou S.P., Suidan H.S., Pavlik A., et al. Changes in the expression of protease-activated receptor 1 and protease nexin-1 mRNA during rat nervous system development and after nerve lesion. European Journal of Neuroscience. 1998;10:159–1607.

Ossipov M.H., Bazov I., Gardell L.R., et al. Control of chronic pain by he ubiquitin proteasome system in the spinal cord. Journal of Neuroscience. 2007;27:8226–8237.

Palecek J., Paleckova V., Dougherty P.M., et al. The effect of phorbol esters on the responses of primate spinothalamic neurons to mechanical and thermal stimuli. Journal of Neurophysiology. 1994;71:529–537.

Pogatzki E.M., Niemeier J.S., Sorkin L.S., et al. Spinal glutamate receptor antagonists differentiate primary and secondary mechanical hyperalgesia caused by incision. Pain. 2003;105:97–107.

Porreca F., Burgess S.E., Gardell L.R., et al. Inhibition of neuropathic pain by selective ablation of brainstem medullary cells expressing the mu-opioid receptor. Journal of Neuroscience. 2001;21:5281–5288.

Price T.J., Cervero F., de Koninck Y. Role of cation-chloride-cotransporters (CCC) in pain and hyperalgesia. Current Topics in Medicinal Chemistry. 2005;5:547–555.

Ramos K.M., Lewis M.T., Morgan K.N., et al. Spinal upregulation of glutamate transporter GLT-1 by ceftriaxone: therapeutic efficacy in a range of experimental nervous system disorders. Neuroscience. 2010;169:1888–1900.

Rudomin P. Selectivity of the central control of sensory information in the mammalian spinal cord. Advances in Experimental Medicine and Biology. 2002;508:157–170.

Seybold V.S. The role of peptides in central sensitization. Handbook of Experimental Pharmacology. 2009;194:451–491.

Steinhoff M., Vergnolle N., Young S.H., et al. Agonists of proteinase-activated receptor 2 induce inflammation by a neurogenic mechanism. Nature Medicine. 2000;6:151–158.

Suzuki R., Morcuende S., Webber M., et al. Superficial NK1-expressing neurons control spinal excitability through activation of descending pathways. Nature Neuroscience. 2002;5:1319–1326.

Svensson C.I., Hua X.Y., Protter A.A., et al. Spinal p38 MAP kinase is necessary for NMDA-induced spinal PGE2 release and thermal hyperalgesia. Neuroreport. 2003;14:1153–1157.

Todd A.J. Anatomy of primary afferents and projection neurones in the rat spinal dorsal horn with particular emphasis on substance P and the neurokinin 1 receptor. Experimental Physiology. 2002;87:245–249.

Tyce G.M., Yaksh T.L. Monoamine release from cat spinal cord by somatic stimuli: an intrinsic modulatory system. Journal of Physiology. 1981;314:513–529.

Willis W.D. Role of neurotransmitters in sensitization of pain responses. Annals of the New York Academy of Sciences. 2001;933:142–156.

Woolf C.J., Thompson S.W. The induction and maintenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation; implications for the treatment of post-injury pain hypersensitivity states. Pain. 1991;44:293–299.

Yaksh T.L. Behavioral and autonomic correlates of the tactile evoked allodynia produced by spinal glycine inhibition: effects of modulatory receptor systems and excitatory amino acid antagonists. Pain. 1989;37:111–123.

Yaksh T.L., Jessell T.M., Gamse R., et al. Intrathecal morphine inhibits substance P release from mammalian spinal cord in vivo. Nature. 1980;286:155–157.

Zamponi G.W., Lewis R.J., Todorovic S.M., et al. Role of voltage-gated calcium channels in ascending pain pathways. Brain Research Reviews. 2009;60:84–89.

Zieglgansberger W., Herz A. Changes of cutaneous receptive fields of spino-cervical-tract neurones and other dorsal horn neurones by microelectrophoretically administered amino acids. Experimental Brain Research. 1971;13:111–126.