Mechanisms of in nonmalignant disease Victoria L. Harvey and Anthony H. Dickenson

Department of Pharmacology, University College Purpose of review London, London, UK To review key mechanisms underlying the transmission of nociceptive information from Correspondence to Victoria L. Harvey, Department of the periphery to the central nervous system implicated in different acute pain states. Pharmacology, University College London, Gower St, London, WC1E 6BT, UK Recent findings E-mail: [email protected] Advances in molecular and transgenic approaches have helped to identify novel therapeutic targets for the treatment of pain from tissue and nerve damage such as acid-

Current Opinion in Supportive and Palliative sensing ion channels, transient receptor potential and NaV channels. The subsequent Care 2008, 2:133–139 development of selective pharmacological ligands has also strengthened the role of other receptors such as hyperpolarization-activated cyclic nucleotide-gated channels and the further development of subunit specific antagonists, such as those available for NR2B, will further advance our understanding of the mechanisms involved in nociceptive transmission. Summary Inflammatory and differ considerably in their peripheral mechanisms but certain central spinal and brain mechanisms are common to both. The mechanisms of pain are not fully established but are thought to be underpinned by changes in the expression of receptors (nociceptive plasticity), central spinal hyperexcitability (central sensitization) and alterations in descending control from the midbrain. This review considers these mechanisms and highlights recent advances in the understanding of pain perception.

Keywords dorsal horn, facilitation, inflammatory pain, neuropathic pain, plasticity

Curr Opin Support Palliat Care 2:133–139 ß 2008 Wolters Kluwer Health | Lippincott Williams & Wilkins 1751-4258

Characteristic symptoms experienced with pain, resulting Introduction from these various causes, include an increased amplitude Pain, although generally initiated by damaging events of response to a given stimulus (), pain elicited within the body and subsequently transmitted into the by normally innocuous stimuli () and spon- central nervous system, can also be generated within taneous pain in the absence of external stimuli. Sensory central nervous system circuits such as post-stroke pain. deficits can also exist in neuropathic pain. In addition, as This account will consider various peripherally gener- pain persists, even over short periods there are the affective ated acute , a series of heterogeneous conditions and emotional responses that have to be considered along involving a cohort of neuronal mechanisms. These pain with the sensory aspects of the stimulus. The sensory and states can be broadly categorized into those accompany- psychological aspects of pain are separable in terms of ing tissue damage (trauma, surgery, tumour growth and pathways within the central nervous system but there is an arthritic conditions) and those following nerve damage interplay between the neural pathways that contribute to caused again by trauma, surgery, tumours and also these components. It is clear from these symptoms that at viruses (postherpetic , HIV) and metabolic both peripheral and central sites, there are mechanisms disorderssuchasdiabetes.The inflammatory and neuro- that can amplify and prolong the experience of a painful pathic pains differ considerably in their peripheral stimulus and a miscoding of normally innocuous stimuli – mechanisms but share more common central spinal this can result in severe pain in the presence of a relatively and brain mechanisms. Current research emphasis minor peripheral disorder. envelops the nociceptive plasticity of ion channels at peripheral (e.g. sodium channels) and central sites (e.g. calcium channels), central spinal hyperexcitability Tissue damage (central sensitization) and alterations in descending con- Peripheral sensory nerve fibres are divided into A-fibres in trol from the midbrain. which A-b fibres normally convey innocuous information

1751-4258 ß 2008 Wolters Kluwer Health | Lippincott Williams & Wilkins

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. 134 Pain: nonmalignant disease

and A-d fibres that can respond to noxious mechanical play a major role in the maintenance of pain arising from stimuli and a population of C-fibres that respond to a wide tissue or nerve inflammation [11–13]. Although 5-HT has range of noxious stimuli. The latter groups respond to been identified as playing a key role in the pain associated acute noxious mechanical and thermal stimuli and are with , its mechanism of action is less clear and sensitized by tissue damage. Tissue damage elicits the is not known whether changes in this mediator are per- release of a number of endogenous chemical mediators at ipherally or centrally driven [14]. 5-HT3 receptors, when both peripheral and central sites. Of these, prostaglandins activated, cause pain [15] and antagonists (e.g. ondanse- (in particular PGE2) represent an important component of tron) have been shown to possess peripheral analgesic inflammation, sensitizing primary afferent fibres to other actions in human studies. ATP is present in all cells so key mediators and stimuli and reducing their thresholds tissue damage rapidly causes a marked rise in extracellular so that they now respond to lower intensity stimuli levels of this purine that is a potent algogen. Of the 7 P2X (peripheral sensitization). Steroids and nonsteroidal anti- receptors for ATP cloned to date, P2X3 homomeric and inflammatory drugs (NSAIDs) are able to block cyclo- PX2X2/3 heteromeric receptors are expressed in unmye- oxygenase (COX), an enzyme upstream of the produc- linated sensory wherein they sense ATP release tion of prostaglandins [1]. Classically, NSAIDs inhibit following injury to the surrounding tissues [16]. Following the constitutive isoform (COX-1) and subsequently lead inflammation, P2X3 receptors enhance neurotransmitter to gastrointestinal complications. More recently, a second release either through an upregulation of their expression inducible form (COX-2) with a different pattern of distri- or through receptor sensitization [17,18]. Further support bution had been described [2]. Although COX-2 inhibitors for the importance of these channels comes from antisense have avoided the gastropathy associated with COX-1 studies in which P2X3 receptor knockdown with siRNAs inhibition, increased cardiovascular complications have attenuated nocifensive behaviour in rats following an been described and their therapeutic gain is now unas- inflammatory or neuropathic insult [19,20]. sured [3]. Central targets downstream of PGE2 have been identified to be important in inflammatory pain. COX-1 , the hot ingredient in chilli peppers, evokes a and COX-2 are present and upregulated in the spinal cord sensation of burning pain thought to occur from the and prostaglandins can be released at this level, indicative activation of TRPV1 (VR1), a receptor thought originally of a role of centrally penetrating NSAIDs in pain control. to represent the sole molecular identity of our heat sensor Interestingly, spinal GlyRa3 receptors are thought to be [21]. Since then 27 transient receptor potential (TRP) selectively inhibited through the phosphorylation of a channels have been cloned in humans [22], and a subset key serine (S346), which is not present in the other major of these (thermoTRPs) are thought responsible for ther- adult spinal isoform (a1) [4]. The distinct expression pat- mal signalling [23]. Of these, transgenic studies [21,24] tern of the GlyRa3 receptors in lamina II of the dorsal horn have revealed that TRPV1 is required for the thermal suggests that such a disinhibition could lead to facilitated hypersensitivity observed in inflammation, and TRPM8 nociceptive transmission in response to inflammation. is important in cold hypersensitivity in neuropathy and inflammation [25]. In addition to thermal signalling, Bradykinin, hydrogen ions, ATP and 5-HT also accumu- TRP channels may also serve as receptor-operated chan- late in damaged tissue, and cause acidosis, vasodilation and nels, such as TRPA1 that depolarizes nociceptors in plasma extravasation causing the oedema and vasodilata- response to proalgesic and proinflammatory agents that tion that occur in inflammation. Acid-sensing ion channels activate phospholipase C (PLC)-signalling pathways (ASICs), due to their location and properties are likely [26,27]. candidates for Hþ-gated nociceptors, sensing tissue acido- sis in response to inflammation [5]. Upregulation of ASIC The interplay between these numerous pain transducers subunit expression has been observed experimentally at C-fibre endings explains the commonly observed per- during inflammation [6] and inhibitors have displayed ipheral sensitization that occurs after tissue damage. efficacy in acid-induced pain in humans [7]. Furthermore, Although these multiple targets at peripheral levels are as inflammatory mediators such as nerve growth factor potential targets for novel analgesic therapies, the issue of (NGF), 5-hydroxytryptamine (5-HT), interleukin-1, bra- redundancy with multiple pain sensors is an important dykinin and brain-derived neurotrophic factor (BDNF) factor in decisions on which receptor to target. can upregulate ASIC transcription, these channels may contribute to the pronociceptive effects of these mediators [8]. Interestingly, increased expression of the proinflam- Nerve damage matory cytokine, tumour necrosis factor-a (TNF), in both Sodium channels generate the action potential. Follow- the dorsal root ganglia and the spinal cord has been ing damage to peripheral nerves, primary afferent fibres reported following several types of nerve injury [9,10]. often display aberrant ‘ectopic’ activity altering their The mechanisms involved are unclear but the downstream pattern of neuronal excitability and conduction causing activation of p38 mitogen-activated kinase is thought to spontaneous pain and hyperalgesia. These changes are

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Mechanisms of pain in nonmalignant disease Harvey and Dickenson 135

thought to arise namely through the accumulation of pain from varying origins [42–44]. Following nerve sodium channels at or around the site of injury. Although injury, a2d subunits are slowly upregulated in the central sodium channels have long been associated with terminals of peripheral nerves [45] possibly allowing GBP the efficacy of local anaesthetics such as lignocaine and and PGB to selectively inhibit abnormal neuronal the carbemazepine they have, through the function associated with neuropathy [46–49]. A further generation of mutant mice, received renewed interest. In permissive factor in the actions of these drugs is particular, tetrodotoxin (TTX)-resistant Nav 1.8 and 1.9 activity in spinal 5-HT3 receptors, themselves driven channels are expressed almost exclusively in small, by activity in descending pathways from the brain – unmyelinated fibres, whereas TTX-sensitive Nav 1.7 this rapidly switching circuit may not only explain the channels are expressed in both sensory and sympathetic actions of a2d ligands after nerve injury but their acute neurons. Although it is likely that these ion channels are effects such as those seen after surgery. regulated by nerve and tissue damage, transgenic animals lacking 1.7 or 1.8 show marked reductions in neuronal Hyperpolarization-activated cyclic nucleotide-gated responses to acute noxious mechanical stimuli. Inherited (HCN) channels are predominantly located within the ‘gain of function’ mutations of SCN9A, the gene encod- cardiovascular system and DRG, wherein they are thought ing Nav 1.7, results in the rare autosomal dominant to generate spontaneous rhythmic activity as well as con- disease primary erythermalgia [28], whereas a ‘loss of tributing to neuronal excitability and plasticity [50]. Nerve function’ channelopathy produces analgesia [29]. damage elicits a marked increase in the expression of particularly HCN1 channels in large diameter afferents The biophysical properties of Nav 1.8 currents permits evoking spontaneous pacemaker-driven action potentials them to participate in action potential electrogenesis in the damaged nerve [51]. Further studies [52] with an even at potentials close to 40 mV, which can occur HCN antagonist (ZD7288) reversed behavioural mecha- following nerve injury [30] and more recently have been nical hypersensitivities in animals with experimentally identified in signalling cold-induced pain [31]. Nav 1.3, a induced neuropathic (spinal nerve ligation, SNL) and mild TTX-sensitive channel, is developmentally regulated thermal injury (MTI) pain. wherein expression is primarily associated with embryo- nic development and, under normal conditions, declines to low levels following birth. Following peripheral nerve Central excitatory mechanisms damage Nav 1.3 levels in dorsal root ganglia increase, and Ad and C fibres terminate primarily in the superficial the biophysical properties of these channels suggests that laminae of the dorsal horn, namely lamina I, wherein they are capable of supporting ectopic neuronal firing the large majority of neurones are nociceptive specific [32–36]. Recently, Jarvis et al. [37] have demonstrated with small receptive fields, responding to only noxious the efficacy of the first Nav 1.8 selective antagonist pinch and/or heat stimulation and with major outputs to (A-803467) in a variety of animal models of inflammatory the brain. A smaller population of polymodal (HPC) and neuropathic pain. neurones are found that are also cold responsive. Lamina I neurones have been shown to project to areas such as the Although sodium channels act as the accelerator, potas- periaqueductal grey (PAG), lateral parabrachial nucleus, sium channels serve as the molecular brakes of the system, thalamus, nucleus tractus solitarius (NTS) and the medul- playing an important role in modulating neuronal lary reticular formation. A large number of projection hyperexcitability. In particular, spinally applied retiga- neurones from lamina I express the receptor for substance bine, a KCNQ channel (KV7, M-current) opener, inhibits P, NK1 [53]. This group of neurones is the origin from C-mediated and Ad-mediated responses in dorsal horn which a spinobulbospinal loop originates that drives des- neurones in both naı¨ve and neuropathic rats [38], and cending controls and can set dorsal horn excitability from diminishes behavioural hypersensitivity in neuropathic higher centres. Within deeper lamina V, neurones are rats [39]. More recently, TREK-1, a member of the two- wide-dynamic range, responding to both innocuous and þ pore-domain K channel family (K2P), has been found in noxious stimuli, exhibiting wind-up and projecting to small sensory neurones and colocalizes with TRPV1 [40]. sensory areas of the brain such as the thalamus. Both Knockout mouse studies [40] have revealed a role for this the lamina I and V neurones therefore provide parallel receptor in polymodal pain, and studies [41] in transfected outputs to the sensory and affective areas of the brain. cell lines have suggested a role in anaesthesia as it is There is clear evidence from both preclinical and clinical activated by volatile anaesthetics. studies that apart from peripheral processes, central hyper- excitability plays important roles in determining the level (GBP) and (PGB), which attenu- of pain perceived. ate neurotransmitter release through binding to the a2d subunit of voltage-gated Ca channels, have demonstrated Neurones of the dorsal horn of the spinal cord represent a their value in the treatment of peripheral neuropathic key modulatory site for the processing of nociceptive

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. 136 Pain: nonmalignant disease

information. The major excitatory neurotransmitter glu- istration of ondansetron, a 5-HT3 antagonist, attenuates tamate is found in both primary afferent and projection 5-HT-mediated pronociception [65,66]. Furthermore, this neurones. Glutamate acts at both the metabotropic descending hyperalgesic serotinergic (5-HT3) pathway (mGlu) receptors G-protein coupled) and the ionotropic from the brainstem is permissive for the actions of GBP AMPA, Kainate and N-methyl-D-aspartic acid (NMDA) [67–69]. receptors (coupled directly to ion channels). During high frequency firing of C fibres, glutamate and its coreleased transmitter substance P cooperate to allow NMDA recep- Central inhibitory mechanisms tor activation, resulting in the wind-up of dorsal horn and fentanyl, mu (m) receptor agonists, neurones. Neurones rapidly switch from a constant low- remain the gold standard for the treatment of moderate level response to a higher prolonged and amplified level and severe pain. These receptors represent the of activity. This has been reported in a wide range of endogenous system for the suppression of pain. Expres- human acute and persistent pain conditions. This sion studies have shown that opioid receptors (m, d, k process is thought to underpin central sensitization, and ORL1) are predominantly expressed in the super- whereby nociceptive pathways display increased ficial laminae (I and II) of the spinal cord wherein synaptic efficacy (LTP) and is thought to represent nociceptive primary afferents terminate [70], and their the mechanism responsible for hyperalgesia and allody- expression is differentially regulated following neuro- nia [54]. First generation NMDA anatagonists such as pathy and inflammation [71–74]. Their principal actions ketamine, memantine and dextromethorphan are still involve the presynaptic attenuation on neurotransmitter well used today in a wide variety of pain states [55–58], release by inhibition of voltage-gated calcium channels although their long-term use is more limited due to and postsynaptic inhibition of projection neurones their cognitive contraindications. Alternative subunit through the activation of G protein-regulated inwardly specific regimes are being investigated such as NR2B rectifying Kþ (GIRK) channels [75,76]. The supraspinal antagonism with ifenprodil, as these subunits are activity of , in areas such as the rostroventral specifically upregulated following nerve injury in medulla (RVM), PAG matter and the locus coeruleus animals [59]. disinhibit spinally projecting noradrenergic and seroto- nergic fibres possibly through the inhibition of presyn- Genes are rapidly switched on in spinal neurones follow- aptic g-aminobutyric acid (GABA) receptors [77,78]. A ing intense acute noxious stimuli including genes role for nociceptin, the endogenous ligand for mor- thought to be important in forebrain memory processes phine-resistant ORL1 receptors, has been demonstrated such as zif 268. These more enduring changes in synaptic in animal models of neuropathic and inflammatory pain events may be a process that could lead to the transition [79–81]. Furthermore, ORL1 receptors are thought to from acute to chronic pain. form unique complexes with CaV2.2 channels such that prolonged exposure to nociceptin triggers a protein Alterations in both the transmission of nociceptive infor- kinase C-dependent internalization of the ORL1/ 2þ mation within the spinal cord and the changes in the CaV2.2 complex, thus downregulating Ca channel descending controls, which relay information from higher activity [82]. brain centres to the spinal cord, are frequently observed in pain states. These pathways are highly complex in nature and also involve supraspinal centres, such as those Conclusion associated with affective-cognitive information process- It is evident from the literature that the mechanisms ing. Increased activity within the spinal NK1-receptor- underlying pain perception are highly complex in nature driven spino-bulbo-spinal loop is associated with patho- involving the plasticity and functional dysregulation of physiology [60]. NK1-receptor expressing lamina 1 numerous ion channels and receptors within both the neurones are predominantly projection neurones that peripheral and the central nervous system (Fig. 1). These terminate in a number of brainstem areas involved in changes underlie alterations in the transmission and both the sensory and the affective components of processing of nociceptive information resulting in pain nociception [53]. These areas are the key sites for the related phenomena such as ectopic neuronal discharge, descending noradrenergic and serotonergic controls that peripheral and central hyperexcitability and the facili- regulate spinal nociceptive processing and underlie the tation of descending algogenic pathways. efficacy of antidepressants for the treatment of pain [61,62]. Modulation of descending serotonergic pathways Advances in molecular and genomic techniques continue is, however, complex as the release of 5-HT can lead to to identify novel therapeutic targets; the subsequent both antinociception and pronociception [63,64] and most development of more selective and subtype specific likely depends on the activation of specific receptor sub- ligands will greater advance our understanding of the types. The depletion of spinal 5-HT or intrathecal admin- mechanisms of pain.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Mechanisms of pain in nonmalignant disease Harvey and Dickenson 137

Figure 1 Pathophysiological changes in nociceptive transmission

Thalamus cortex

Limbic Peripheral insult PAG system Inflammatory mediators Na+, HCN channel plasticity Parabrachial RVM area Transcription/ function: ASIC 5-HT3-R P2X3 + TRPV1 + Prostaglandin Spontaneous Spinal activity EP neurone 2 -- Ca2+ Inhibitory channel GlyRα3 interneurone function function NMDA-R function

Damage and/or inflammation in the periphery evoke changes in the transcription and function of channels and receptors in both the peripheral and the central nervous systems. This leads to an increase in the output of spinal neurones that project to higher centres of the brain that, in turn, engage descending pathways from the periaqueductal grey (PAG) and the rostroventral medulla (RVM). These synapse on dorsal horn neurones and further facilitate nociceptive transmission. ASIC, acid sensing ion channels; EP2, prostaglandin E2 receptor; GlyRa3, glycine a 3 receptor; HCN, cyclic nucleotide-gated ion channel; NMDA-R, N-methyl-D-aspartic acid receptor; TRPV, transient receptor potential ion channel.

8 Mamet J, Baron A, Lazdunski M, Voilley N. Proinflammatory mediators, Acknowledgement stimulators of sensory excitability via the expression of acid-sensing The present research is funded by the Wellcome Trust, the London ion channels. J Neurosci 2002; 22:10662–10670. Pain consortium, BBSRC and MRC. 9 Schafers M, Geis C, Brors D, et al. Anterograde transport of tumor necrosis factor-alpha in the intact and injured rat sciatic nerve. J Neurosci 2002; 22:536–545. 10 Schafers M, Geis C, Svensson CI, et al. Selective increase of tumour necrosis References and recommended reading factor-alpha in injured and spared myelinated primary afferents after chronic Papers of particular interest, published within the annual period of review, have constrictive injury of rat sciatic nerve. Eur J Neurosci 2003; 17:791–804. been highlighted as: 11 of special interest Ji RR, Samad TA, Jin SX, et al. p38 MAPK activation by NGF in primary of outstanding interest sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 2002; 36:57–68. Additional references related to this topic can also be found in the Current World Literature section in this issue (pp. 146–148). 12 Milligan ED, Twining C, Chacur M, et al. Spinal glia and proinflammatory cytokines mediate mirror-image neuropathic pain in rats. J Neurosci 2003; 1 Bingham S, Beswick PJ, Blum DE, et al. The role of the cylooxygenase 23:1026–1040. pathway in nociception and pain. Semin Cell Dev Biol 2006; 17:544– 13 Xu L, Huang Y, Yu X, et al. The influence of p38 mitogen-activated protein 554. kinase inhibitor on synthesis of inflammatory cytokine tumor necrosis factor 2 Burian M, Geisslinger G. COX-dependent mechanisms involved in the anti- alpha in spinal cord of rats with chronic constriction injury. Anesth Analg nociceptive action of NSAIDs at central and peripheral sites. Pharmacol Ther 2007; 105:1838–1844. (table of contents). 2005; 107:139–154. 14 Ramadan NM, Buchanan TM. New and future migraine therapy. Pharmacol 3 FDA. Food and Drug Administration announces series of changes to the class Ther 2006; 112:199–212. of marketed nonsteroidal anti-inflammatory drugs (NSAIDs). Rockville: FDA; 15 Oatway MA, Chen Y, Weaver LC. The 5-HT3 receptor facilitates at-level 2005. mechanical allodynia following spinal cord injury. Pain 2004; 110:259– 4 Harvey RJ, Depner UB, Wassle H, et al. GlyR alpha3: an essential target for 268. spinal PGE2-mediated inflammatory pain sensitization. Science 2004; 16 Khakh BS, North RA. P2X receptors as cell-surface ATP sensors in health and 304:884–887. disease. Nature 2006; 442:527–532. 5 Wemmie JA, Price MP, Welsh MJ. Acid-sensing ion channels: advances, questions and therapeutic opportunities. Trends Neurosci 2006; 29:578–586. 17 Ramer MS, Bradbury EJ, McMahon SB. Nerve growth factor induces P2X(3) expression in sensory neurons. J Neurochem 2001; 77:864–875. 6 Voilley N, de Weille J, Mamet J, Lazdunski M. Nonsteroid anti-inflammatory drugs inhibit both the activity and the inflammation-induced expression of 18 Xu GY, Huang LY. Peripheral inflammation sensitizes P2X receptor-mediated acid-sensing ion channels in nociceptors. J Neurosci 2001; 21:8026–8033. responses in rat neurons. J Neurosci 2002; 22:93–102. 7 Jones NG, Slater R, Cadiou H, et al. Acid-induced pain and its modulation in 19 Dorn G, Patel S, Wotherspoon G, et al. siRNA relieves chronic neuropathic humans. J Neurosci 2004; 24:10974–10979. pain. Nucleic Acids Res 2004; 32:e49.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. 138 Pain: nonmalignant disease

20 Honore P, Kage K, Mikusa J, et al. Analgesic profile of intrathecal P2X(3) 43 Freynhagen R, Strojek K, Griesing T, et al. Efficacy of pregabalin in neuro- antisense oligonucleotide treatment in chronic inflammatory and neuropathic pathic pain evaluated in a 12-week, randomised, double-blind, multicentre, pain states in rats. Pain 2002; 99:11–19. -controlled trial of flexible- and fixed-dose regimens. Pain 2005; 115: 254–263. 21 Caterina MJ, Schumacher MA, Tominaga M, et al. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 1997; 389:816– 44 Rowbotham M, Harden N, Stacey B, et al. Gabapentin for the treatment of 824. postherpetic neuralgia: a randomized controlled trial. JAMA 1998; 280: 22 Venkatachalam K, Montell C. TRP channels. Annu Rev Biochem 2007; 1837–1842. 76:387–417. 45 Li CY, Zhang XL, Matthews EA, et al. Calcium channel alpha(2)delta(1) 23 Dhaka A, Viswanath V, Patapoutian A. TRP ion channels and temperature subunit mediates spinal hyperexcitability in pain modulation. Pain 2006; sensation. Annu Rev Neurosci 2006; 29:135–161. 125:20–34. 24 Davis JB, Gray J, Gunthorpe MJ, et al. Vanilloid receptor-1 is essential for 46 Chapman V, Suzuki R, Chamarette HL, et al. Effects of systemic carbama- inflammatory thermal hyperalgesia. Nature 2000; 405:183–187. zepine and gabapentin on spinal neuronal responses in spinal nerve ligated rats. Pain 1998; 75:261–272. 25 Colburn RW, Lubin ML, Stone DJ Jr, et al. Attenuated cold sensitivity in TRPM8 null mice. Neuron 2007; 54:379–386. 47 Field MJ, Oles RJ, Lewis AS, et al. Gabapentin (neurontin) and S-(þ)-3- An exciting study reporting that mice lacking the cold-gated and menthol-gated ion isobutylgaba represent a novel class of selective antihyperalgesic agents. Br J channel TRPM8 exhibit deficiencies in their behavioral responses to cold Pharmacol 1997; 121:1513–1522. temperatures. 48 Stanfa LC, Singh L, Williams RG, Dickenson AH. Gabapentin, ineffective in 26 Bandell M, Story GM, Hwang SW, et al. Noxious cold ion channel TRPA1 is normal rats, markedly reduces C-fibre evoked responses after inflammation. activated by pungent compounds and bradykinin. Neuron 2004; 41:849– Neuroreport 1997; 8:587–590. 857. 49 Tanabe M, Takasu K, Kasuya N, et al. Role of descending noradrenergic 27 Bautista DM, Jordt SE, Nikai T, et al. TRPA1 mediates the inflammatory system and spinal alpha2-adrenergic receptors in the effects of gabapentin on actions of environmental irritants and proalgesic agents. Cell 2006; 124: thermal and mechanical nociception after partial nerve injury in the mouse. Br J 1269–1282. Pharmacol 2005; 144:703–714. 28 Yang Y, Wang Y, Li S, et al. Mutations in SCN9A, encoding a sodium channel 50 Robinson RB, Siegelbaum SA. Hyperpolarization-activated cation currents: alpha subunit, in patients with primary erythermalgia. J Med Genet 2004; from molecules to physiological function. Annu Rev Physiol 2003; 65:453– 41:171–174. 480. 29 Cox JJ, Reimann F, Nicholas AK, et al. An SCN9A channelopathy causes 51 Chaplan SR, Guo HQ, Lee DH, et al. Neuronal hyperpolarization-activated congenital inability to experience pain. Nature 2006; 444:894–898. pacemaker channels drive neuropathic pain. J Neurosci 2003; 23:1169– 30 Blair NT, Bean BP. Roles of tetrodotoxin (TTX)-sensitive Naþ current, 1178. þ 2þ TTX-resistant Na current, and Ca current in the action potentials of 52 Luo L, Chang L, Brown SM, et al. Role of peripheral hyperpolarization- nociceptive sensory neurons. J Neurosci 2002; 22:10277–10290. activated cyclic nucleotide-modulated channel pacemaker channels in 31 Zimmermann K, Leffler A, Babes A, et al. Sensory neuron sodium channel acute and chronic pain models in the rat. Neuroscience 2007; 144: Nav1.8 is essential for pain at low temperatures. Nature 2007; 447:855– 1477–1485. 858. 53 Todd AJ, McGill MM. Shehab SA. Neurokinin 1 receptor expression by This elegant electrophysiological study of knock out mice posits Nav1.8 as an neurons in laminae I, III and IV of the rat spinal dorsal horn that project to essential molecule for cold nociception. the brainstem. Eur J Neurosci 2000; 12:689–700. 32 Cummins TR, Waxman SG. Downregulation of tetrodotoxin-resistant sodium 54 Dickenson AH. Mechanisms of central hypersensitivity: excitatory amino acid currents and upregulation of a rapidly repriming tetrodotoxin-sensitive sodium mechanisms and their control. In: The pharmacology of pain. Handbook of current in small spinal sensory neurons after nerve injury. J Neurosci 1997; experimental pharmacology. Berlin: Springer-Verlag; 1997. 17:3503–3514. 55 Bell RF, Dahl JB, Moore RA, Kalso E. Perioperative ketamine for acute 33 Black JA, Liu S, Tanaka M, et al. Changes in the expression of tetrodotoxin- postoperative pain. Cochrane Database Syst Rev 2006:CD004603. sensitive sodium channels within dorsal root ganglia neurons in inflammatory pain. Pain 2004; 108:237–247. 56 Eide PK, Jorum E, Stubhaug A, et al. Relief of postherpetic neuralgia with the N-methyl-D-aspartic acid receptor antagonist ketamine: a double-blind, 34 Dib-Hajj SD, Fjell J, Cummins TR, et al. Plasticity of sodium channel expres- cross-over comparison with morphine and placebo. Pain 1994; 58:347– sion in DRG neurons in the chronic constriction injury model of neuropathic 354. pain. Pain 1999; 83:591–600. 57 Max MB, Byas-Smith MG, Gracely RH, Bennett GJ. Intravenous infusion of the 35 Garry EM, Delaney A, Anderson HA, et al. induces NMDA antagonist, ketamine, in chronic posttraumatic pain with allodynia: a neuropathic changes in rat dorsal root ganglia and behavioral reflex sensitisa- double-blind comparison to alfentanil and placebo. Clin Neuropharmacol tion that is attenuated by gabapentin or sodium channel blocking drugs. Pain 1995; 18:360–368. 2005; 118:97–111. 58 Weinbroum AA. A single small dose of postoperative ketamine provides 36 Hains BC, Klein JP, Saab CY, et al. Upregulation of sodium channel Nav1.3 rapid and sustained improvement in morphine analgesia in the presence and functional involvement in neuronal hyperexcitability associated with of morphine-resistant pain. Anesth Analg 2003; 96:789–795. (table of central neuropathic pain after spinal cord injury. J Neurosci 2003; 23: contents). 8881–8892. 59 Iwata H, Takasusuki T, Yamaguchi S, Hori Y. NMDA receptor 2B subunit- 37 Jarvis MF, Honore P, Shieh CC, et al. A-803467, a potent and selective mediated synaptic transmission in the superficial dorsal horn of peripheral Nav1.8 sodium channel blocker, attenuates neuropathic and inflammatory nerve-injured neuropathic mice. Brain Res 2007; 1135:92–101. pain in the rat. Proc Natl Acad Sci U S A 2007; 104:8520–8525. This exciting study discovers a selective Nav 1.8 channel blocker that attenuates 60 Hunt SP, Mantyh PW. The molecular dynamics of pain control. Nat Rev 2001; pain behaviours in a variety of animal models of neuropathic and inflammatory 2:83–91. pain. 61 Gidal BE. New and emerging treatment options for neuropathic pain. Am J Manag Care 2006; 12 (9 Suppl):S269–S278. 38 Passmore GM, Selyanko AA, Mistry M, et al. KCNQ/M currents in sensory neurons: significance for pain therapy. J Neurosci 2003; 23:7227– 62 Watson CP, Evans RJ, Reed K, et al. Amitriptyline versus placebo in post- 7236. herpetic neuralgia. 1982; 32:671–673. 39 Blackburn-Munro G, Jensen BS. The anticonvulsant attenuates 63 Hylden JL, Wilcox GL. Intrathecal serotonin in mice: analgesia and inhibition of nociceptive behaviours in rat models of persistent and neuropathic pain. Eur J a spinal action of substance P. Life Sci 1983; 33:789–795. Pharmacol 2003; 460:109–116. 64 Yaksh TL, Wilson PR. Spinal serotonin terminal system mediates antinoci- 40 Alloui A, Zimmermann K, Mamet J, et al. TREK-1, a Kþ channel involved in ception. J Pharmacol Exp Ther 1979; 208:446–453. polymodal pain perception. EMBO J 2006; 25:2368–2376. 65 Rahman W, Suzuki R, Webber M, et al. Depletion of endogenous spinal 5-HT 41 Patel AJ, Honore E, Lesage F, et al. Inhalational activate two-pore- attenuates the behavioural hypersensitivity to mechanical and cooling stimuli domain background Kþ channels. Nat Neurosci 1999; 2:422–426. induced by spinal nerve ligation. Pain 2006; 123:264–274. 42 Backonja M, Beydoun A, Edwards KR, et al. Gabapentin for the symptomatic 66 Suzuki R, Morcuende S, Webber M, et al. Superficial NK1-expressing neurons treatment of painful neuropathy in patients with mellitus: a rando- control spinal excitability through activation of descending pathways. Nat mized controlled trial. JAMA 1998; 280:1831–1836. Neurosci 2002; 5:1319–1326.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Mechanisms of pain in nonmalignant disease Harvey and Dickenson 139

67 Suzuki R, Rahman W, Hunt SP, Dickenson AH. Descending facilitatory 75 Moises HC, Rusin KI, Macdonald RL. Mu- and kappa-opioid receptors control of mechanically evoked responses is enhanced in deep dorsal selectively reduce the same transient components of high-threshold calcium horn neurones following peripheral nerve injury. Brain Res 2004; 1019: current in rat dorsal root ganglion sensory neurons. J Neurosci 1994; 14: 68–76. 5903–5916. 68 Dickenson AH, Bee LA, Suzuki R. Pains, gains, and midbrains. Proc Natl Acad 76 Marker CL, Lujan R, Colon J, Wickman K. Distinct populations of spinal cord Sci U S A 2005; 102:17885–17886. lamina II interneurons expressing G-protein-gated potassium channels. J Neurosci 2006; 26:12251–12259. 69 Iannetti GD, Zambreanu L, Wise RG, et al. Pharmacological modulation of pain-related brain activity during normal and central sensitization states in 77 Finnegan TF, Li DP, Chen SR, Pan HL. Activation of mu-opioid receptors humans. Proc Natl Acad Sci U S A 2005; 102:18195–18200. inhibits synaptic inputs to spinally projecting rostral ventromedial medulla neurons. J Pharmacol Exp Ther 2004; 309:476–483. 70 Maekawa K, Minami M, Yabuuchi K, et al. In situ hybridization study of mu- and kappa-opioid receptor mRNAs in the rat spinal cord and dorsal root ganglia. 78 Vaughan CW, Ingram SL, Connor MA, Christie MJ. How opioids inhibit Neurosci Lett 1994; 168:97–100. GABA-mediated neurotransmission. Nature 1997; 390:611–614. 71 Porreca F, Tang QB, Bian D, et al. Spinal opioid mu receptor expression in 79 Carpenter KJ, Vithlani M, Dickenson AH. Unaltered peripheral excitatory lumbar spinal cord of rats following nerve injury. Brain Res 1998; 795:197– actions of nociceptin contrast with enhanced spinal inhibitory effects after 203. carrageenan inflammation: an electrophysiological study in the rat. Pain 2000; 85:433–441. 72 Chen SR, Sweigart KL, Lakoski JM, Pan HL. Functional mu opioid receptors are reduced in the spinal cord dorsal horn of diabetic rats. Anesthesiology 80 Maie IA, Dickenson AH. Cholecystokinin fails to block the spinal inhibitory 2002; 97:1602–1608. effects of nociceptin in sham operated and neuropathic rats. Eur J Pharmacol 2004; 484:235–240. 73 Sung B, Loh HH, Wei L. Association of kappa opioid receptor mRNA upregulation in dorsal root ganglia with mechanical allodynia in mice following 81 Stanfa LC, Chapman V, Kerr N, Dickenson AH. Inhibitory action of nociceptin nerve injury. Neurosci Lett 2000; 291:163–166. on spinal dorsal horn neurones of the rat, in vivo. Br J Pharmacol 1996; 118:1875–1877. 74 Gendron L, Lucido AL, Mennicken F, et al. Morphine and pain-related stimuli enhance cell surface availability of somatic delta-opioid receptors in rat dorsal 82 Altier C, Khosravani H, Evans RM, et al. ORL1 receptor-mediated internaliza- root ganglia. J Neurosci 2006; 26:953–962. tion of N-type calcium channels. Nat Neurosci 2006; 9:31–40.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.