10
DRUG DEVELOPMENT RESEARCH 39~279-288 (1 996) Venture Capital Enabling Technology Research Overview Multiple Roles of ATP and Adenosine in Somatosensory Processing: Therapeutic Implications Gary J. Keil II and Michael W. Salter* Division of Neuroscience, Hospital for Sick Children, Department of Physiology, University of Toronto, Toronto, Ontario, Canada Preclinical Development Clinical Development Toxicology, Formulation Phases 1-111 Postmarketing Drug Delivery, Pharmacokinetics Manufacturing Regulatory, Quality, Phase IV I and Health Policy INTRODUCTION The somatosensory system encodes, processes, and transmits peripheral sensory information into the central nervous system (CNS). The coding of information as nox- ious or innocuous depends not only on the activation of specific peripheral sensory receptors, but also on the ac- tions and interactions of numerous neurotrans,mitter/ neuroinodulator systems at peripheral and central sites. Multiple lines of evidence support roles for adenosine and adenosine 5 '-triphosphate (ATP) in the transinission of sensory information into the CNS [for review see Salter et al., 19931. In this article we outline evidence indicat- ing that adenosine and ATP participate in sensory trans- mission, and highlight recent developments in the understanding of purine nucleotides and nucleosides in nociceptive, or pain-related, and non-nociceptive sensory transmission at peripheral and spinal sites. Finally, we describe considerations related to possible therapeutic intervention in nociception by targeting compounds which act on or through purinergic receptors or down- stream signalling elements. ATP AND ADENOSINE IN SENSORY NEUROTRANSMISSION ATP as a Primary Afferent Transmitter The first evidence implicating ATP as a neurotrans- mitter in the somatosensory system came from studies in the mid- to late-1950s by Holton and Holton who dem- onstrated that ATP is released from peripheral endings of primary sensory neurons [Holton and Holton, 1954; Holton, 19591. They hypothesized that ATP may also he released from central terminals of primary afferents. Evidence for central release of ATP was provided by White and coworkers who showed that ATP was released in a calcium-dependent fashion from dorsal horn synap- tosomes by depolarizing concentrations of potassium [White et al., 19851. This release is greatly depressed by dorsal rhizotoiny suggesting that there is ATP released Contract grant sponsor: Medical Council of Canada *Correspondence to: Michael W. Salter, Division of Neuro- science, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5C 1 X8, Canada. 0 1997 Wiley-Liss, Inc.

Multiple roles of ATP and adenosine in somatosensory processing: Therapeutic implications

Embed Size (px)

Citation preview

DRUG DEVELOPMENT RESEARCH 39~279-288 (1 996)

Venture Capital Enabling Technology

Research Overview

Multiple Roles of ATP and Adenosine in Somatosensory Processing: Therapeutic Implications

Gary J. Keil II and Michael W. Salter* Division of Neuroscience, Hospital for Sick Children, Department of Physiology, University of Toronto,

Toronto, Ontario, Canada

Preclinical Development Clinical Development Toxicology, Formulation Phases 1 - 1 1 1 Postmarketing Drug Delivery, Pharmacokinetics Manufacturing

Regulatory, Quality, Phase IV

I and Health Policy

INTRODUCTION

The somatosensory system encodes, processes, and transmits peripheral sensory information into the central nervous system (CNS). The coding of information as nox- ious or innocuous depends not only on the activation of specific peripheral sensory receptors, but also on the ac- tions and interactions of numerous neurotrans,mitter/ neuroinodulator systems at peripheral and central sites. Multiple lines of evidence support roles for adenosine and adenosine 5 '-triphosphate (ATP) in the transinission of sensory information into the CNS [for review see Salter et al., 19931. In this article we outline evidence indicat- ing that adenosine and ATP participate in sensory trans- mission, and highlight recent developments in the understanding of purine nucleotides and nucleosides in nociceptive, or pain-related, and non-nociceptive sensory transmission at peripheral and spinal sites. Finally, we describe considerations related to possible therapeutic intervention in nociception by targeting compounds which act on or through purinergic receptors or down- stream signalling elements.

ATP AND ADENOSINE IN SENSORY NEUROTRANSMISSION

ATP as a Primary Afferent Transmitter

The first evidence implicating ATP as a neurotrans- mitter in the somatosensory system came from studies in the mid- to late-1950s by Holton and Holton who dem- onstrated that ATP is released from peripheral endings of primary sensory neurons [Holton and Holton, 1954; Holton, 19591. They hypothesized that ATP may also he released from central terminals of primary afferents. Evidence for central release of ATP was provided by White and coworkers who showed that ATP was released in a calcium-dependent fashion from dorsal horn synap- tosomes by depolarizing concentrations of potassium [White et al., 19851. This release is greatly depressed by dorsal rhizotoiny suggesting that there is ATP released

Contract grant sponsor: Medical Council of Canada

*Correspondence to: Michael W. Salter, Division of Neuro- science, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M 5 C 1 X8, Canada.

0 1997 Wiley-Liss, Inc.

280 KElL II A N D SALTER

from central terminals of primary afferent neurons [Sawynok et a]., 19931. Release of ATP from dorsal horn synaptosomes is riot affected by neonatal treatment with capsaicin, which destroys many nociceptive primary af- ferents. Thus, ATP released from primary afferents de- rives from a capsaicin-insensitive subpopulation.

Iontophoretic application of ATP in vivo demon- strated that it excites neurons in the cuneate nucleus [Galindo et al., 19671 and in the dorsal horn [Salt and Hill, 1983; Fyffe and Perl, 1984; Salter and Henry, 19851. These types of neurons receive direct input from primary afferents and it was found that the excitatory effects of ATP are correlated with input from non-nociceptivc pri- mary afferents. In vitro, ATP evokes an inward currcnt in dorsal horn neurons [Jahr and Jessell, 19831 by acti- vating a non-selective cation conductance [Jessell and Jahr, 19851. The effects of exogenously administered ATP would suggest that ifATP is released synaptically it should produce an excitatory post-synaptic response. Recently, evidence for an excitatory postsynaptic current (EPSC) mediated by KL'P in dorsal horn neurons has been ob- tained [Goldstein ct al., 19961. EPSCs resistant to block- ade by excitatory amino acid antagonists hut blocked by the P2-purinoccptor antagonists, suraniin or PPADS, have been recorded in a subpopulation of dorsal horn neurons in an in vitro slice preparation. These are fast EPSCs, which are evoked by dorsal root stimiilation, demonstrat- ing KIP-mediated synaptic responses at some priinaiy afferent-dorsal horn synapses. Taking the in vivo and in vitro electrophysiological evidence together with studies on A r P release from synaptosomes, suggests that ATP may be a mediator of non-nociceptive sen- sory transmission.

It is likely that the excitatory effect of ATP on dorsal horn neurons and the ATP-mediated EPSC are mediated via ligand-gated cation channels of the P2X-purinoceptor type [rcvicwcd by Surprenant et al., 19951. Moleciilar biological studies liavc revealed a family of P2X purinoceptors of which six (P2XI-P2Xfi) subunits have been identified [Collo et al., 19961. Messenger RNA (mRNA) for P2X1, P2X2, P2X4, P2Xs, and P2X(j, but not P2Xi3, purinoceptors is expressed in the spinal dorsal horn. Which of these P2X subunits mediate the synaptic ef- fects of ATP remains to be determined.

Adenosine as an Inhibitory Modulator in the Dorsal Horn

Considerahle evidence from physiological, bio- chemical, and pharmacological studies indicate that ad- enosine is an inhibitory mediator in many CNS areas [reviewed by Stone, 19811. In the spinal dorsal horn ad- enosinc has been shown to inhibit the firing discharge of neurons in somatosensory pathways in vivo [Salter and Henry, 19851. That this inhibition may be a postsynaptic

effect was suggested by the finding that discharge evoked by glutamate is inhibited by iontophoretic application of adenosine 5 '-monophosphate (AMP) and this inhibition is blocked by theophylline. Subsequently, adenosine was shown to cause hyperpolarization of dorsal horn neurones in vivo [DeKoninck and Henry, 19921 and in vitro [Li and Perl, 19941 due to an increase in po- tassium conductance.

Adenosine-mediated actions in the spinal cord may also involve presynaptic actions. Evidence that adenos- ine might have presynaptic effects on primary afferent terminals in the spinal cord was first shown by Phillis and Kirkpatrick [ 19781. Sn1)sequent studies have indi- cated that adenosine niay inhibit release of substancc P and CGRP from dorsal horn slices [Santicioli et al., 1992, 19931, but whether this was due to pre- or postsynaptic mechanisms was not determined. Decreases in the rate of spontaneously occurring niiniature EPSCs in dorsal horn neurons by adenosine [Li and Perl, 19943 suggest that adenosine inhibits glutamate-mediated synaptic transmission presynaptically in the dorsal horn. Presyn- aptic actions of adenosine in other CNS regions have consistently been shown to be mediated by A, receptors [see Fredholm and Dunwiddie, 19881, but the subtype of receptor, which may mediate the presynaptic action of spinal adenosine, has not been detcrniined.

A, and A2 receptor binding sites are demonstrated in the spinal cord [Goodman and Snyder, 1982; Murray and Cheney, 1982; Geiger et al., 1984; Choca et al., 1987, 19881 with binding occurring most heavily in the super- ficial laminae [Goodman and Snyder, 1982; Choca et al., 19881. Selective lesion studies show that adenosine rc- ccptor binding is partially decreased only following the destruction of spinal interneurons [Geiger et al., 1984; Choca ct al., 19881. Moreover, mRNA for A , and A2 re- ceptors is expressed in the spinal cord [Reppert et al., 1991; Rivkees and Reppert, 1992; Stehle et al., 1992; Rivkees, 19951, suggesting that intrinsic neurons express and contain adenosine rcceptors. That these receptors are found on dorsal horn neurons is consistent with post- synaptic effects of adenosine on dorsal horn neurons, and that Al or A2 receptors may participate in the physiologi- cal actions of adenosinc at spinal levels.

Extracellular Conversion of ATP to Adenosine in Dorsal Horn Signalling

ATP is rapidly hydrolyzed to adenosine [Zimmer- man, 19941, which is inactive at P2 purinoceptors [Burnstock, 19781. Thus, hydrolysis of ATP terminates its actions. Equally important niay he the generation of adenosine; ATP released from primaiy afferent terminals might he converted to adenosine at synaptic sites in the dorsal horn. An inhibitory postsynaptic response medi- ated by ATP-derived adenosine has been demonstrated

ATP AND ADENOSINE IN SOMATOSENSORY PROCESSING 281

in dorsal horn neurons [Salter and Henry, 1987; Li and Perl, 19951. The inhibition is a postsynaptic response me- diated hy an increase in potassium conductance [DeKoninck and Henry, 19921 due to activating KhTP channels [Salter et al., 19921. This adenosine-mediated inhibitory postsynap- tic potential (IPSP) is shown by nociceptive dorsal honi neurons and this is a physiological mechanism for decreas- ing nociceptive transmission in the dorsal horn.

Actions of ATP on Primary Sensory Neurons

In addition to releasing ATE primary afferent neu- rons are also sensitive to this nucleotide. ATP produces an excitatory effect on the cell bodies of primary afferent neiirons located within the dorsal root ganglia (DRG) [Jahr and Jessell, 1983; Krishtal et a]., 19831. The excita- tory effect of ATP results from activating a non-selective cation channel [Bean, 19901 and is blocked by P2- purinoceptor antagonists, indicating it is mediated via P2X-purinoceptors. mRNA for each of the six subunits of P2X receptors is expressed by DRG cells [Chen et al., 1995; Lewis et al., 1995; see Collo et al., 19961. The ef- fects of ATP on DRG cells raise the possibility that pc- ripheral endings of primary afferents may also be excited by ATE In this way ATP might be a sensory mediator in peripheral tissues.

Actions of Adenosine on Primary Sensory Neurons

Adenosine receptors are found on DRG cell bodies [Dolphin et al., 1986; Macdonald et a]., 19861, suggest- ing that these receptors might be located on the central and peripheral endings of primary sensory neurons. Ac- tivating adenosine receptors on neuronal DRG cell bod- ies inhibit high voltage-gated Ca2+ currents [Dolphin et al., 1986; Macdonald et al., 1986; Gross et al., 19881. These findings suggest adenosine might inhibit transmitter re- lease at nerve terminals of DRG neurons through inhibi- tion of calcium currents.

PURINES IN MODULATING NOCICEPTION

Adenosine Is a Mediator of Analgesia Evoked by Stimulating low Threshold Primary Afferents

The adenosine-mediated IPSP in nociceptive dor- sal horn is elicited by low-intensity peripheral stimula- tion, which activates non-nociceptive primary afferents. The IPSP is antagonized by 2-methylthiotheophylline, and is potentiated by the nucleoside transport inhibitor, dipyridamole [Salter et al., 19931. The inhibition of noci- ceptive dorsal horn neurons by activating non-nocicep- tive inputs may be the cellular basis for the analgesia produced by stimulation of these inputs by transcutane- ous electrical nerve stimulation (TENS) or by innocuous mechanical stimuli such as vibration [Woolf and Thomp- son, 19941. Therefore, it has been hypothesized that an-

algesia produced by these types of stimulation may be mediated by adenosine which causes activation of K,,rr channels [Salter et al., 19931. Recently, it has been re- ported that, under double blind conditions, TENS-in- duced analgesia is blocked in human subjects challenged with caffeine (200 mg, p a ) [Marchand et al., 19951. At this dose caffeine is expected to antagonize central ad- enosine receptors [Fredholm, 19801. Thus, the effect of caffeine provides strong support for the hypothesis that analgesia produced by TENS is mediated by adenosine.

Adenosine Induces Analgesia at Spinal Sites

As discussed above, the inhibitory actions of ad- enosine on dorsal horn neurons have been investigated in vivo using electrophysiological techniques. The ion- tophoretic application of AMP inhibits the ongoing ac- tivity of dorsal horn neurons and the responses to noxious peripheral stimuli [Salter and Henry, 19851. This inhibi- tory response is seen in approximately two-thirds of dor- sal horn neurons and is prevented by theophylline, suggesting that a large population of dorsal horn neu- rons are sensitive to the inhibitory effects of adenosine. In terms of presynaptic effects, Li and Perl [1994] dem- onstrated, in an in vitro slice preparation, that bath-ap- plied adenosine suppressed all polysynaptic EPSCs, and inhibited approximately 40% of the monosynaptic EPSCs, in dorsal horn neurons. Reeve and Dickenson [1995a] reported that selective Al adenosine receptor agonists inhibit the C-fiber evoked responses in dorsal horn neu- rons, hut that an A2A selective agonist was without sig- nificant effects. In addition, the excitatory responses of dorsal horn neurons following subcutaneous application of formalin were suppressed by centrally administered A, agonists. The inhibitory responses were blocked by prior treatment with adenosine receptor antagonists, sug- gesting that Al receptor activation might inhibit acute nociceptive transmission in the dorsal horn and responses to peripheral inflammation.

In animal models of acute and chronic pain, adenos- ine or adenosine analogs induce antinociception [re- viewedby Sawynok and Sweeney, 1989; Sawynok, 19911. Significant decreases in pain behavior are observed in the formalin test [Malmberg and Yaksh, 1993; Doak and Sawynok, 1995; Poon and Sawynok, 19951, in thermal hyperalgesic states following nerve injury [Yamamoto and Yaksh, 19931, and in strychnine- [Sosnowski and Yaksh, 1989; Sosnowski e t al., 19891 or prostaglandin I T S a -

[Minami et al., 1992a,h] induced allodynia (pain behav- iors to non-painful stimuli). Activation of spinal adenos- ine receptors, therefore, induces antinociception in a wide number of animal models of acute, chronic, and patho- logical pain states.

Studies using receptor-selective agonists indicate that activating A, adenosine receptors results in the

282 KElL I I AND SALTER

antinociception in behavioral models of pain [Karlsten ct al., 1991, 1992; DeLander et al., 1992; Malmberg and Yaksh, 1993; DeLander and Keil, 1994; Poon and Sawynok, 19951. As discussed previously, binding sites for both AI- and A2-type receptors are found in the spinal cord and mRNA for both receptor subtypes are found in intrinsic dorsal horn neurons, and spinally administered A2 receptor agonists have been found to induce antinociception [reviewed by Sawynok and Sweeney, 19897. Thus, it is possible that A2-type receptors may par- ticipate in antinociceptive effects.

The principal mechanism for terniiriatirig the ac- tion of extracellular adenosine is uptake and intracellu- lar metabolism. Pharmacological manipulations that decrease the clearance of adenosine in the dorsal horn would be expected to either induce antinociception or to enhance adenosine-mediated effects. Inhibition of ad- enosine uptake by inhibitors of nucleoside transporters has been shown to significantly potentiate and prolong adenosine-mediated antinociception [Keil and DeLander, 19951. The possibility that other inhibitors of adenosine clearance induce antinociception was examined by Keil and DeLander [ 19921 who found antinociceptive effects of an inhibitor of adenosine kinase. Other studies have also shown that inhibition of adenosine kinase results in antinociception [Keil and DeLander, 1994; Poon and Sawynok, 19951, and in vivo and in vitro studies iridicate that adenosine kinase inhibitors lead to increases in ex- tracellular adenosine levels [Golembiowska et al., 1995, 19961. Lastly, inhibition of adenosine deaminase did not induce demonstrable antinociception in the absence of exogenous adenosine [Kcil and DeLander, 1994; Poon and Sawynok, 19951, h i t antinociception induced h y exog- enous adenosine is potentiated following the inhibition of adenosine deaminase [Keil and DeLander, 19941. ‘Faken together, these studies indicate that pliarmacological manipulations which iricreasc endogenous adenosine lev- els may induce appreciable levels of antinociception, and potentiate physiological or pharmacological manipula- tions that increase extracellular adenosine levels.

Pronociceptive Effects of Spinal Adenosine Receptor Antagonists

In contrast to the antinociceptive effects that occur following spinal adenosine receptor agonist administra- tion, the spinal administration of adenosine receptor an- tagonists produces behaviors indicating hyperalgesia to noxious stimuli [Jurna, 1984; Sawynok et al., 19861, allodynia to innocuous stimuli [Keil and DeLander, 19961, or nociception without overt peripheral stimulation [Nagaoka et al., 1993; Keil and DeLander, 19961. That spinal adenosine receptor antagonists induce these be- haviors suggests adenosine might tonically inhibit noci- ccptive pathways, and processes that diminish spinal

adenosine activity might participate in or underlie cer- tain pain states.

Peripheral Actions of ATP Are Pro-Nociceptive

ATP has long been known to induce nociceptivc or algogenic responses following peripheral administration [Collier et al., 1966; Bleehen and Keele, 19773. Nocicep- tive responses induced b y ATP have been suggested to be due to direct activation of peripheral nerve terminals [reviewed by Illes and Niirenberg, 19931 or indirectly through actions on inflammatory cells [Dubyak et al., 1988; Handwerker and Reeh, 1991; Rang et al., 19911. A’L’P may be released into extracelliilar spaces as a con- sequence of tissue injury: it may be released from blood products such as platelets and mast cells [Dubyak arid El-Moatassini, 19931, or it may be released from post- ganglionic sympathetic nerve terminals [Burnstock, 19901, or from sensory afferents [Holton, 19591. As nien- tioned above, ATP depolarizes primary afferent cell bod- ies [Jahr and Jessel, 1983; Krishtal et al., 19831, and if ATP depolarizes the peripheral terminals of sensory neu- rons this could result in direct activation of primary afferent s .

Which type or types of P2-purinoceptors mediate the excitatoq or pronociceptive effects of ATP at periph- eral sites is presently unknown. mRNA for all six sub- types of P2X purinoceptors are expressed in sensory ganglia [see Collo et al., 19961, and two recent studies suggest a role of the P2X3 subiinit in nociceptive pro- cesses. mRNA for the P2X3 subunit is found only in sen- sory ganglia [Chen et al., 1995; Lewis et al., 19951, and cell body labelling for P2X3, in situ, occurs coincident with inimuriostaining to periphcrin [Chcn et al., 19951, a marker for C-fiber afferents. mRNA for P2X3 transcript is abolished following neonatal capsaicin treatment [Chen et al., 19951, indicating P2X3 is found exclusively in cap- saicin-sensitive, presumably nociceptive afferents. The peripheral nociceptive actions of ATP may, therefore, he mediated through activation of PeX:\-containing recep- tors located on nociceptive primary afferent neurons.

Peripheral Actions of Adenosine May Be Pro- or Anti-Nociceptive

In human subjects, the peripheral adniiriistr a t’ ion of adenosine has been shown to induce pain. Pain is evoked when adenosine is administered to blister bases [Bleechen and Keele, 19771 arid ischemic-like chest pain or discomfort is reported following peripheral [Sylvkn et al., 1986, 1988a,b; Biaggioni et a]., 19871 or intracoronary [Lagerqvist et al., 19901 adenosine administration. Such algogenic or pronociceptive effects are also seen in ani- mal models [‘Faiwo and Levine, 1990; Karlsten et al., 1992; Doak and Sawynok, 19951. Pharmacological studies in animal models indicate that the pronociceptive effects of

ATP AND ADENOSINE IN SOMATOSENSORY PROCESSING 283

peripherally administered adenosine are mediated by actions at A2 adenosine receptors [Taiwo and Levine, 1990,1991; Doak and Sawynok, 19951. Interestingly, the A2A-selective agonist, CGS21680, enhances, while the selective antagonist, DMPX, inhibits formalin-induced nociceptive behavior only during the latter phases of the test [Doak and Sawynok, 19951. Moreover, in this study the intradermal injection of adenosine did not induce nociceptive behavior in the absence of coadmiiiistered formalin. Such effects raise the possibility that adenos- ine may not activate sensory neurons directly, but may enhance the excitatory effects by other inflammatory mediators [Doak and Sawynok, 19951.

In contrast to the local algogenic effects of adenos- ine, the systemic administration of adenosine has been shown to induce analgesia [Belfrage et al., 1995; Segerdahl et al., 199.5; Sollevi et al., 19951. In thcse stud- ies patients occasionally experienced discomforting sen- sory phenomena, but these sensations were promptly diminished upon dosage reduction. The apparently con- trasting results from Sollevi and colleagues compared to the studies mentioned above may be explained by the multiplicative site- and receptor-dependent effects fol- lowing systemic administration of adenosine (see below). In animal models, the activation of peripheral Al adenos- ine receptors induces antinociceptive effects [Taiwo and Levine, 1990; Karlsten et al., 1992; Aley et a]., 199.5; Doak and Sawynok, 19951. Tissue adenosine levels are elevated in conditions of ischemia arid inflammation [reviewed in Cronstein, 19951. It has been suggested that this adenos- ine may activate peripheral Al receptors, which may par- ticipate in reducing postinflammatory pain since the peripheral administration of Al antagonists increase, but Al agonists decrease, pain behaviors in the formalin test [Doak and Sawynok, 19951.

Adenosine may also exert anti-inflammatory effects. Indeed, the anti-inflammatoiy actions of certain pharma- cological agents have also been attributed to the release of adenosine at inflamed sites [Cronstein, 1995; Gadangi et al., 19961. Similarly, the peripheral inhibition of ad- enosine kinase reduces inflammation [Firestein et al., 1994; Cronstein et a]., 1995; Rosengren et al., 19951. The anti-inflammatory effects have been attributed to periph- eral activation of A2-type receptors, which, amongst other actions, inhibits neutrophil adhesion and prevents the secretion of inflammatory cytokines [Gadangi et al., 19961. Thus, while A2-type receptor activation might be involved in pronociception, anti-inflammatory effects are also seen which may lead to diminished postinflammatory pain.

It is possible that adenosine-mediated actions at A-, adenosine receptors may participate in peripheral soma- tosensory processes. One function of An receptors is ad- enosine-mediated degranulation of mast cells [for review see Linden, 19941, which release multiple compounds

implicated in inflammation [rcviewed by Ilandwerker and Reeh, 19911. An additional involvement of A, rccep- tors in inflammation and pain may relate to adenosine- mediated inhibition of the release of the proinflammatory cytokine, tumor necrosis factor a (TNF-a) from macroph- ages and monocytes [Sajjadi et al., 19961.

THERAPEUTIC CONSIDERATIONS FOR PURlNERClC COMPOUNDS IN TREATING PAIN

Some potential roles for adenosine and ATP in modulating nociceptive signalling at peripheral and cen- tral sites are summarized in Figure 1. As discussed, these compounds have multiple functions in soma- tosensory processing, which are dependent upon the site and the receptor subtype that is activated. I’har- macological agents or physiological manipulations which specifically target the antinociceptive actions of adenosine and ATP may provide efficacious anal- gesia in a wide number of clinical pain states. How- ever, algogenic or pronociceptive results might occur if agents are used that are non-selective, either in their mechanism of action or in their primary site of action. Therefore, site- or receptor-targeting strategies are needed in order to maximize analgesic effects while minimizing any algogenic effects.

Targeting PI-Purinoceptors

Systemic administration of adenosine may induce analgesic effects through actions at spinal and periph- eral A1 receptors, and anti-inflammatory effects at peripheral A2 or A, receptors. A potential limiting ef- fect to using systemic adenosine in the treatment of pain and inflammation, however, may be the periph- eral AP type receptor mediated algogenic effects. The overall effect of systemically administered adenosine, therefore, may depend on the relative magnitude of opposing pro- and anti-nociceptive systems. The use of agents selective for a particular subtype of recep- tor would be considered a pharmacological targeting strategy, which could avoid possible pronociceptive effects seen with non-selective agents.

A,-selective agonists

Systemically administered Al-selective agonists would be expected to induce analgesia at both periph- eral and, if able to cross the blood brain barrier, central adenosine receptors. Moreover, localized peripheral ad- ministration of these compounds, for example intra- articularly, would also be expected to induce analgesia. Systemic or intra-articular administration could induce analgesia without concurrent pronociceptive effects. These agents could have actions as anti-inflammatory agents through possible effects on sensory nerve terminals, which inhibit the release of inflammatory mediators.

284 KElL II AND SALTER

Figure 1. Possible central and peripheral sites of adenosine- and ATP- mediated modulation of somatosensory processes. In the periphery, tis- sue damage could result in the release of ATP and adenosine. ATP and adenosine could induce nociception (lightening bolt) at peripheral P2X3- containing PJ-purinoceptor and Az-adenosine receptors, respectively. Adenosine-mediated actions at peripheral AT receptors induce antinociception. Not shown are the possible anti-inflammatory effects at A2 and/or A3 receptors. In the spinal cord, ATP released from large-diam-

A,-selective agents A,-selcctive agonists induce anti-inflammatory ac-

tions in the periphery, but concurrent algogenic effects might limit any clinical usage. In the spinal cord, A,-type agonists might induce analgesia; in animal models, how- ever, these agonists oftcn p r o d ~ ~ e profound motor-im- pairing effects which may also occur in human patients.

eter (AD) primary afferents following innocuous peripheral stimuli (feather) excites non-nociceptive dorsal horn neurons in deep laminae likely through P2X receptor increases in cation conductance (gc,3,,c,n). Metabolism of ATP to adenosine would induce inhibition through inward potas- sium conductance (gK) on nociceptive dorsal horn neurons. Adeno- sine might also inhibit presynaptic release from primary afferent terminals (not shown). In addition, ATP might affect dorsal horn as- trocytes through P2Y receptors.

Peripheral A2-type selective antagonists may inhibit ad- enosine-mediated pronociception, but these agents might facilitate inflammatory processes.

A,-selective agonists

The effects of compounds with agoiiist activity at A3 adenosine receptors might he expected to induce an-

ATP AND ADENOSINE IN SOMATOSENSORY PROCESSING 285

algesia via A3 receptor-mediated anti-inff aminatory ef- fects but additional studies are needed to clarify the ac- tivity of the receptors in somatosensory processes.

Targeting P,-Purinoceptors

As ATP may be involved in activating nociceptive sensory neurons through P2X3-containing receptors, pe- ripherally acting antagonists selective for these receptors might be analgesic. Potential actions of these compounds would not likely be associated with significant side ef- fects because P2X3 subunits are only expressed by cap- saicin-sensitive primary afferents.

The effect of P2X3-selective compounds adminis- tered spinally may lead to paradoxical effects. These re- ceptor subunits are not expressed by dorsal horn neurons so P2X3 agonists or antagonists would not be expected to have direct effects on these neurons. However, P2X3 ago- nists might have effects on central terminals of nocicep- tive primary afferents. Such agonists might induce primary afferent depolarization (PAD), which may in- hibit central release of transmitters [Shapiro et al., 19801 or which may antidromically activate these af- ferents, leading to release of pronociceptive media- tors in the periphery. Thus, the net effect of central agonists or antagonists at P2X3 receptors would be difficult to predict.

Therapeutic Implications of Adenosine-Mediated Antinociception in TENS Analgesia

From the observation that caffeine blocks analge- sia produced by TENS (vide supra), it is suggested that patients receiving TENS therapy for pain relief should refrain from consumption of caffeine or other niethyl- xanthines. Moreover, the analgesic effects of dorsaf col- umn stimulation or of vibration, two other treatments that activate low-threshold primary afferent neurons, may also be mediated by adenosine [Salter et al., 19931. Thus, simi- lar considerations concerning the consumption of methylxanthines would also apply to these treatments.

We have reported that the inhibition of nocicep- tive dorsal horn neurons by low-threshold primary affer- ent stimulation may be potentiated by inhibitors of adenosine uptake [Salter and Henry, 19871. This suggests that extracellular adenosine concentration produced by the stimulation may not be sufficient to fully activate the receptors or downstream signalling elcments. I f this is the case in humans, then several novel pharmacologi- cal means for increasing the efficacy of pain treatment with TENS, dorsal column stimulation, or vibration may be suggested. Thus, it may be possible to use as adjuvants in these treatments such as inhibitors of ad- enosine uptake or degradation, allosteric enhancers of adenosine receptors, or agents that enhance the activity of K,,, channels.

Therapeutic Implications of Adenosine-Mediated Antinociception in Opioid-Induced Analgesia

As previously described, the antinociceptivc cf'fccts of spinal adenosine may be mediated through the activa- tion of KATp channels. Opioid-induced antinociceptive effects have also been attributed in part to activity of these channels [Welch and Dunlow, 19931. Moreovei; spinal adenosine release has been shown to be involved in the antinociceptive actions of opioid compounds [reviewed by Sawynok and Sweeney, 1989; Sawynok et al., 1989; DeLander e t al., 1992; Cahill et al., 1993; Keil and DeLander, 1994; Cahill et al., 19951. Adenosine and opioid-mediated antinociception show overlap and this overlap might result in cross-tolerance in the spinal cord, similar to the cross-tolerance between opioids and ad- enosine analogs in the periphery [Aley et al., 19951. Not all opioid-mediated effects are thought to involve adeno- sine, however [reviewed by Sawynok and Sweeney, 1989; DeLander et al., 19921. In support ofindependent mecha- nisms of action, adenosine analogs can interact in a su- pra-additive manner with opioids when coadministered [DeLander and Keil, 1994; Keil and DcLander, 1995; Reeve and Dickenson, 1995b1. These results suggest su- pra-additive interactions between adenosine and opioid analgesics might occur in clinical settings, such that the overall efficacy to treat certain pains might be enhanced at lower total analgesic doses [DeLander and Keil, 19941.

Novel Therapeutic Target: Dorsal Horn Astrocytes

In the preceding sections we have focused on the efyects of ATP and adenosine on neurons. However, the effects of ATP in the dorsal horn are not restricted to neurons. In primary cultures prepared from the dorsal horn, ATP increases intracellular calcium levels in >99% of the astrocytes examined [Salter and Hicks, 19941, through a P2Y-purinoceptor-mediated activation of the phospholipase Cp/IP3 pathway [Salter and Hicks, 199SI. Astrocytes and other glial cells may have distinct roles in pathological processes that occur following peripheral nerve injury [Hajos et al., 1990; Gilmore et al., 1990; Garrison et al., 1991; Svensson et al., 19933 and these cells may contribute to abnormal nociceptive processing and pathological pain states [Meller et al., 19941. Although much work is needed in this area, it is conceivable that pharmacological agents with actions at P2Y receptors might represent a novel class of analgesic agents.

CONCLUSIONS

We have highlighted the multiple roles of adenos- ine and ATP in somatosensory processes. These multiple roles provide a rich number of potential therapeutic tar- gets and, thus, we see a great potential for purinergic compounds as analgesics. However, site- and receptor-se-

286 KElL II A N D SALTER

lective targeting strategies need to be employed to achieve maximum efficacy with minimal unwanted effects.

ACKNOWLEDGMENTS

This work was supported by the Medical Research Council of Canada.

REFERENCES

d e y KO, Grccn PG, Levine JD (199,5): Opioid and adenosine periph- eral antinociception are sul)ject to tolerance and withdrawal. J Neurosci 15:8031-8038.

Bean BP (1990): A'l'P-activated channels in rat and bullfrog sen- 7 ory neuron e s . <; on cc ii t ra t i on de pe II tle II cc an d kine t i cs . J Neurosci 10:1-10.

Belfrage M, Sollcvi A, Segt:rtlahl M, Sjiilurrd K-E Ilansson P (1995): Systenric aderiosine infiision alleviates sporrtarieoiis and stimiilus evoked pain iii patients with periplieral neiiropathic pain. Anestli Analg 8:713-717.

Biaggioni I, Olafsson B, Robertson R M , Hollister AS, Rollertson 11 (1987): Cardiovascular and respiratory effects of adenosincx in con- scious man. Evidence for carotid body clieniorcccptor activation. Circ Res 61:779-786.

Blcehen T, Kt:ele CA (1977): Observations on the algogeiiic actions of adcmosine compounds on the human Iilister bast preparation. Pain 3:367-377.

Burnstock G (1978): A basis for distinguishing hvo types of purincrgic receptor. In Straiih RW, Rolis L (etls): Celt Mein1)rane Receptors for Drugs and Hormones: A 1Lliiltidisciplin;iry Approach. New York: Rave Press, pp 107-118.

Burnstock C; (1990): Purinergic nic:clrariisnis. Ann NY Acad Sci 603:l-17.

Cahill CM, CVhite TD, Sawynok J (1 3 ) : Morphine activates W- conotoxin-sthnsitive Ca" channels to release atlcnosine froin spinal cord svnaptosomes. J Neurocheni 60:894-80 1 .

Cahill CM, Wlritc TU, Sawynok J (199Ei): Spinal opioid recqtors arid adenosine relcxse: Neurochernical and I)chavioural characterization of opioid siilitypes. J Pharniacol Exp Tlier 275234-93.

Chen C-C, Akoplan AN, Sivilotti L, Colqiihoun D, Rririistock G, Wood JN (1995): A P2X purinoceptor expressed Iiy ii w h e t of sensoiy neurons. Nature 377:428-431.

Choca JI, Proudfit HK, (:reen RD (1987): Identification of A1 and A2

adenosine rcccptors in the rat spinal cord. J Pharmacol Exp Ther 242:905-9 10.

Choca JI, Green RD, Proudfit H K (1988): Adwrosiiic A] and A2 recep- tors of the sulxtantia gelatinosa are located prrdorninantly on in- trinsic neurones: An audoradiographic study. J Pliarniacol Exp Ther 247:757-764.

Collicr HOJ, Jiinrcs GWL, Sclineider C (1966): Antagonism b y aspirin and fcmanates of I~rorichoconstriction aiid nociception by adenos- ine-5 '-ti-iphosphatc. Nature 21241 1412.

Cello G, North RA, Kawashima E , Merlo-Picli E, Ncidliart S, Snrprenant A, Bucll C (1996): Cloning of P2X5 and P2Xti receptors aiid the distri1)utioii and properties of an extended family of A'I'P- gated ion channels. J Neurosci 1632495-2507.

Cronstein BN (1995): A novc.1 approach to the devclopnicmt of anti- iirflanim:ctory agents: Atlrriosinc release at inf1;uned sites. J Invest Med 43:50-5i.

(:ronstein RN, Naimc I>, Firesteirr C;S (1995): The antiinfl~tnirnatorvitory

effects of an adenosine kinase inhil~itor arc nretliatcd by adenosine. Arthritis Rheinn 38:1040-1045.

DcKoninck Y, Henry J L (1992): Periphcwl vibration causes an adenos- ine-mediated postsynaptic inhibitory potential in dorsal horn nenrones in the cat spinal cord. Neuroscience 50:435-443.

DeLantler GE, Keil I1 GJ (1994): Antinociception induced b y i r i -

trathecal coadministration of selective adenosine receptor and selective opioid receptor agonists in mice. J Pharniacol Exp Ther 268343-951.

IleLandcr CE, Mossberg 111, Porreca F (1992): Involvement of atl- enosine in antinociception produced by spinal or supraspinal re- ceptor-selective opioid agonists: Dissociation from gastrointestinal effects in mice. J Pharniacol Ex11 Thcr 263:1097-1104.

Doak GJ, Sawynok J (1995): Complex role of peripheral adenosine in the genesis ofthe response to subcutaneous fommalin in the rat. Eur J Pharmacol 281:311-318.

Dolphin AC, Fortla SR, Scott RH (1986): Callcirnri-dependent currents in cultured rat dorsal root ganglion neurones arc inlrihited by an adenosine arialog. J Physiol (London) 373:47-61.

Dray A, Urban L, Dickensori A (1994): Pharmacology of chronic pain. Trends Pharmacol Sci 15:190-197.

Dricssen B, Reimann W, Selve N, Friderichs E , Biiltmann R (1994): Antinociceptive effect ofintratliccally administered Pz-piirinoccptor antagonists in rats. Brain Res 666:182-188.

Duhpak GR, El-Moatassim C (1993): Signal transduction via P1- piirincqic receptors from cxtracellular ATP and other nucleotides. Am J Physiol 265 (Cell Physiol 43):C577-606.

Dubyak GR, Cowen DS, Lazarus 11M (1988): Activation of the inosi- to1 phospholipid signaling system by receptors for extraccllular ATP in human neutrophils, nionocytes and nc:ritrophil/monocyte progen- tor cells. Ann NY Acad Sci 551218-237.

Firestein GS, Royle D, Bullough DA, Griiher HE, Sajjadi FC:, Montag A, Sanibol B, Mullare KM (1994): Protective effcct of an adenosine kinase inhihitor in septic shock. J Imniunol 152:5853-5859.

Fredholm BB (1980): Are mcthylxanthinr effwts due to antagonism of endogenous adenosine'? Trciids Pharniacol Sci 1:129-132.

Fredholm BB, Dunwiddie TV (1988): How does adenosine inhiliit trans- mitter relcase? Trends Pharmacol Sci 9:130-134.

Fredholm BB, A1)bracchio ME Rnrrrstock C:, Daly JW, IIarden TK, Jacobson KA, L,effP, Williams M (1994): Nonienclature and classifi- cation of purinoceptors. Pharrnacol Rev 46: 143-156.

Fyffe RE, Per1 ER (1984): Is ATP ii central synaptic mediator for cer- tain priinary afferent fibers froin mammalian skin? Proc Natl Acad Sci 81:6890-6893.

Gadangi F: Longaker M, Nainie L), Lcvin RI, Recht PA, Montesinos MC, Biickley M'r, Carlin C,, Cronstcin BN (1996): The anti-inflam- matory mechanism of sulfLsalazine is related to adenosine relcue at inflamed sitrs. ] Inimunol 156:1937-1911,

Galiirtlo A, Kriijevic K, Schwartz S (1967): 1Llicroiontophoretic studies on nenrorres in the cuneate nucleus. J Physiol (London) 192359-377

( :ailison _:

ing of CFAP in l u m h r spinal cord increases following a sciatic newc constriction injury. Brain Res 565:l-7.

Geiger JII, 1,aBella FS, Nagv J 1 (1984): Cheracterizatioii and lo- calization of adenosine receptors in rat spinal cord. J Ncurosci 1:2303-23 10.

Gilmore SA, Sims TJ, Leiting J E (1990): Astrocvtic reactions i n spinal

Coldstein PA, Lcc CJ, Bardoni R, Chi JG, SlacDcvmott AB (1996):

CJ, Doiigherty PM, Kajandcr KC, Carlton SM (1991): Stairi-

gray matter following sciatic axotorny. (%a 3:342-:349.

ATP AND ADENOSINE IN SOMATOSENSORY PROCESSING 28 7

ATP-receptor mediated synaptic transmission in the dorsal horn o f the rat spinal cord. Soc for Neurosci (Abs) Vol 1: 788.

Golenibiowska I<, White TD, Sawynok J (1995): Modulation of ad- enosine release from rat spinal cord by adenosine deaminase and ;&nosine kinase inhihitot-s. Brain Res 6993315-320.

Golernbiowska K, White TD, Sawynok J (1996): Adenosine kinase in- hibitors aiiginent release ofadenosine from spinal cord slices. Eur J Pharniacol307: 157-162.

Goodman RR, Snyder SH (1982): Autoradiographic localization of ad- enosine receptors in rat brain using (’€II)cyclohexvladeilosine. J Neiirosci 2:1230-1241.

Gross RA, Macdonald RL, Ryan-Jastrow T (1989): 2-Chloroadenosine rediices the N calcium current of cultured mouse sensory neiironcs in a pertussis toxin-serisitivc manner. J Physiol 411:585-595.

Hajos E Csillik B, Knyihar-Ckillik E (1990): Alterations in flial fibril- lary acidic protein imniunoreactivity in tlie upper dorsal horn ofthe rat spinal cord in the course of transganglionic degenerative atro- phy and regenerative proliferation. Neurosci Lett 117:8-13.

Handwerker HO, Reeh PW (1991) Pain and inflarnmation. In Bond MR, Charlton JE, Woolf CJ (eds): Pain Research arid Clinical Man- agement. New York: Elsevier Science, pp 59-70.

Holton P (1959): The liberation of adenosine triphosphate on an- tidromic stimulation of sensory nerves. J Physiol (],ondon) 145:494-504.

Holton FA, Holton P (1954): The capillary dilator suhtances in dry powders of spinal roots: a possible role of adenosine triphosphate in chemical transmission from nerve endings. J Physiol (London) 126: 124-140.

Tlles E: Niirenberg W (1993): Neuronal ATP receptors and their mecha-

Jahr CE, Jessel TM (1983): ATP excites a subpopulation of rat dorsal horn ncurones. Nature 304:730-733.

Jc~ssell TM, Jahr CE (1985): Fast and slow excitatory transmitters at primary afferent synapses in the dorsal horn of the spinal cord. In: Advdnces in Pain Research and Therapy. New York: Raven Press, pp 31-39.

Jiirna I (1984): Cyclic nucleotides and aniinophylline produce differ- ent effects on nociceptive motor and sensory responses in the rat spinal cord. Arch Pharniacol 327:2330.

Earlsten R, Gordh T, Hartviql, Post C (1990): Effects of intrathecal injection of the adenosine receptor agonist R-phenylisopropyl-ad- enosine and N-ethylcar1)oxamide-adenosine on nociception and motor function in the rat. Anesth Analg 71:60-64.

Karlsten R: Post C, Hide I, Daly JW (1991): Thc antinociceptive effect of intrathecally administered adenosine analogs in mice correlates with the affinity for the Al-adenosine receptor. Neurosci Lett 121:267-270.

nism of action. Trends Pharmacol Sci 14:50-54.

Karlstcin R, Gordh T, Post C (1992): Local antinociceptive arid hyper- algesic effects in the formalin test after peripheral administration of adenosine analogues in micc. Pharinacol Toxic01 70:434-4:38.

Keil I1 CJ, DeLdnder GI? (1992): Spinally-mediated antinociception is induced in inicc: by an adetiosiiie kinase-, but not hy an adenosine deaminase-, inhibitor. Life Sci 51:PL171-176.

Keil I1 CJ, DeLander GE (1994): Adenosine kinase and atlenosinc deaminase inhibition modulate spinal adenosine- and opioid ago- nist-induced antinociception in mice. Eur J Pharmacol271:3746.

Keil 11 CJ, DeLandcr GE (1995): Time-dependent antinociceptive interactions between opioids and nucleoside transport inlir1)itors. J Pharmacol Exp Ther 274:1387-1392.

Keil I1 GJ, DcbLander GE (1996): Altered srnsoty Ijchwiors i n nricc following manipulation ofendogenous spinal adenosine neurotrans- mission. Eur J Pharmacol 3127-14.

Krishtal OA, Marchcnko SM, Pidoplichko VI (1983): Receptor fol- ATP in tlie membrane of mammalian sensory neuronvs. Nerlrosci Lett 35: 4 1 4 5 .

Lagerqvist B, SylvGn C, Beermann B, Helniius C, Waldenstrh A (1990): Intracoronary adenosine causes angina pectoris like pain: An incpity into the nature of visceral pain. Cardiovasc Res 24:6O9-613.

Lewis C, Neldhart S, Holy C, North RA, Buell G, Surprenant A (1995): Coexpression of P2Xz and P2X3 receptor subunits can account for ATP-gated currents in sensory neiirons. Nature :377:432-43.5.

Li J, Per1 ER (1994): Adenosine inhibition of synaptic transnrission in the substantia gelatinosa. J Neurophysiol 72:1611-1621.

Li J, Per1 ER (1995): ATP rnodulation of synaptic transmission in thc spinal substantia gelatinosa. J Neurosci 15:33573365.

Lindcn J (1994): Cloned adenosine A1 receptors: Pharmacological prop- erties, species differences and receptor functiorrs. Trends Pharrnacol Sci 15:298-306.

Maedonald RL, Skerritt JH, Werz MA (1986): Adenosine agonists re- duce voltage-dependent calcium conductance of mouse sensory neiirones in cell culture. J Physiol (London) 370:75-90.

Malmherg AB, Yaksh TL (1993): Pharmacology of the spirial action of ketorolac, morphone, ST-91, U50,488H, and L-PIA on the forinalin test and an isobolographic analysis ofthe NSAID interaction. Anes- thesiology 79:270-281.

Marchand S, Li J, Charest J (1995): Effects of caffeine on analgesia from transcutaneous electrical nerve stimulation. NEJM 333:32%326.

Meller ST, Dykstra C, Grzyhycki D, Murphy S, Cebhart GF (1994): Thc possible role of glia in nociceptive processing and hyperalgc- sia in the spinal cord ofthe rat. Neuropharmacology 33:1471-1478.

Minanii T, Uda R, Horiguchi S, Ito S, Hyodo M, Hayaishi 0 (199221): EfFects of clonidine and helofen on andin Fz,-induccd

4. Minanri T, Uda R, Horiguchi S, Ito S, Hyodo M, Hayaishi 0 (19921)):

Allodynia evoked by intrathecal administration ofprostaglandin Fza to conscious mice. Pain 50:223-229.

Murray TE Chcney D L (1982): Neuronal location of N‘-cVclo- hexyl[”H]adenosine binding sites in rat and guinea pig hrain. Neu- ropharmacology 2 1:575-580.

Nagaoka €I, Sakurada S, SakuradaT, Takeda S, Nakagawa Y, Kisara K, Ami Y (1993): Theophylline-induced nociceptive behavioral response in mice: Possible indirect interaction with spinal N-methyl-I)-as- partate receptors. Neurochem Int 2269-74.

Phillis JW, Kirkpatrick JR (1978): The actions of adenosine and vari- ous nucleosides and nucleotides on the isolated toad spinal cord. Gen Pharniacol9:239-247.

Poon A, Sawynok J (1995): Antinociception by adenosine analogs and ana adenosine kinasc inhibitor: Dependence on fonnalin concen- tration. Enr J Pharniacol286:177-184.

Rang He Bevan S, Dray A (1991): Chemical activation of nociccptive peripheral nenroncs. Br Med Bull 47534-548.

Reeve AJ, Dickenson AH (1995): The roles of spinal adenosine re- ceptors in the control of acute and more persistent nociceptive re-

urones in the anaesthetized rat. Br J

Reeve AJ, Dickensori AH (19951)): Electrophysiological study on spi- nal antinociceptive intcmctions between adenosinc and morphine in the dorsal horn of the rat. Neurosci Lett 19423-84.

allodynia in conscious mice. Pain Res

288 KElL I I AND SALTER

Reppert SM, Weaver DR, Stehle JH, Rivkees SA (1991): Molecu- lar cloning and characterization of a rat A1-adenosine receptor that is widcly cxprcsscd in brain and spinal cord. Mol Endo- crinol 5:1037-1048.

Rivkees SA (lWC5): The ontogeiiy of cardiac and neural A1 adenosine receptor expression in rats. Dev Brain Res 89:202-213.

Rivkees SA, Reppert SM (1992): RFL9 cncodes an A ~ I ) adenosine re- ceptor. Mol Endocrinol 6: 1,598-1604.

Roscngrcri S, Bong CW, Fircstciii G S (199s): Anti-iriflainiiiatury c%f- fccts of an adcnosinc kinasc. inhibitor: Dccrci~s~d ncritrophil accu- mulation a i d vasciilar leakage. J Immunol 154:5444-5451.

Sajjadi FG, Takabayashi K, Foster AC, Dorningo RC, Firestein CS (1996): Inhibition of TNF-a by adenosine. Role of A3 adenosine receptors. J Iinmimol 156:3435-3442.

Salt TE, Hill RG (1983): Excitation of single sensory nenrones in the rat caudal trigeniinal nucleus by iontophoretically applied adenos- ine 5 '-triphosphate. Neurosci Lett 35:53-57.

Salter MW, Henry J L (1985): Effects ofadenosine 5'-rnonopliosphate and adenosiiw .5'-triphospliate on functionally itlentified units in the cat spinal dorsal 1m-n. Evidence for a differential effect of ad- enosine 5'-triphosphate on nociceptive vs noti-nocicrptivr units. Neuroscience 15815-825.

Salter M\V, IIenry JL (1987): Evidence that adenosine mediates the dcprcssion of spinal dorsal horn ncurones intluced b y peripheral vibration in the cat. Neurosciencc 221631-650.

Salter MW. Hicks JI, (1994): ATP-evoked increases in intracellular calcium in neurons and glia from the tlorsal spinal cord. J Neurosci

Salter MW, Hicks J L (1995); ATP causes release of intracellrilar Ca" via the pliospliolipase Cp/IP:j pathway i n astrocytes fi-on1 the tlorsal spinal cord. J Neiirosci 15:2961-2971.

Salter MW, DcKoninck Y, Hcriiy JL (1992): ATP-serisitive K t chaii- nels mediate an IPSP in dorsal horn neurones elicited by sensoiy stimiilation. Synapse 115214-220.

Salter MW, DeKoninck Y, Henry JL (1993): Physiological roles for adenosine and KrP in synaptic transmission in thc spinal dorsal horn. Prog Neurobiol 41: 125-156.

Santicioli E Del Bianco E, TrdinOntdlia M , Maggi CA (1992): Ad- enosine inhibits actions potential-dependelit rclease of calcitonin gene-related peptide- and su1)stancc P-like imniunoreactivitics froin p r i i i i a ry affereiits i n rat spinal cord. Neurosci Lett 14421 1-214.

Smticioli P, Del Biarrco E, Maggi C A (1993): Adenosine A1 reccp- tors mediate the presynaptic inhihition of calcitonin gene-re- lated peptide release in the rat spiiial c o r d Eiir J Pharmacol

Sawynok J (1091): Adenosine and pain. In Phillis JW (ed): Adenosine and Adenine Nucleotides as Regulators of C:ellular Function. Boca Raton: CXC Prcss, pp 391400.

Sawpnok J, Swecney MI (1989): The role of purines in nociception. Neuroscience 32557-569.

ney MI, White TD (1986): Classification of atknos- ine receptors mediating antinociception in the rat spinal cord. Br J Ph;irniacol 88:923-930.

Sawynok J, S w r ~ n e y M I , White TU (1989): Aderiosirie release niay nic:diate spinal analgesia h y niorpliine. Trends Pharmacol Sci 10: 186- 189.

14:1563-1575.

231:139-142.

release from dorsal spinal cord synaptosomes: Charactcrization and neural origin. Brain Res 610:32-38.

Segerdahl M, Ekhloni A, Sjolund K-E Belfrage M, Forslm-g C, Sollevi A (1995): Systemic adenosine attenuates touch evoked allosynia in- duced by niustard oil in humans. NeuroReport 6:753-756.

Shapiro E, Castchcci VF: Kandel ER (1980): Presynaptic inhibition i n aplysia involvcs a decrease i n the ~ a " + currcnt of the presynaptic neuron. PI-oc Natl Acad Sci USA 77:1185-1189.

Segc:i-dahl ?if, H;inhson P (1995): Systemic adenosine infcision: A new treatriierit iiiodality to alleviate neuropathic pain. Pain 61:155-158.

Sosnowski M, Yaks11 TL (1989): Role of spinal adenosine receptors in modulating the Iiyperesthesia produced by spinal glycine receptor antagonism. Anesth Analg 69~587-892.

Sosnowski M, Stevens CW, Yaksli 'I'L (1989): Assessment of the role of AliAz adenosine receptors mediating the purine antinociceptiori, motor, and autonomic function in the rat spinal cord. J Pliarrnacol Exp Ther 250:915-922.

Stehk JH, Rivkccs SA, Lee JJ, Weaver DR, Deeds JD. Reppert Sbl (1992): Molecular cloning and expression of the cDNA for a novel A2 aderiosirw rcccptor suhtypc!. Mol Endocrinol 6:384-393.

Stone TW (1981'): Physiological roles for adenosine and adenosine 5 '- triphosphate in the nervous system. Ncuroscicnce L523-555.

Suprenant A, Buell G, North R A (199.5): Pzx receptors bring new structure to ligantl-gated ion channels. Trends Neurosci 18:224-229.

Svensson hl, Eriksson P, Pesson JKE, Molarider C, Arvidsson Aldskogius 11 (1993): The resporise of central glia to pr:ripheral nerve injury. Brain I b s Bull 30:499-506.

Sy1vi.n C, Beermann 8, Jonzon B, Brandt R (1986): Angina pcctoris- like pain provokcd by intravenous adenosine in healthy volunteers. Br hled J 293227-230.

S y l v h C, Bccrniann B, Edlund A, Lewander R, Joiizoii B, hlogensen I, (1988a): Provocation of chest pain in paticnts with coronary insuf- ficiency using the vasodilator adenosine. Eur I leart J 9610.

Sylvt.n C:, Jonzon B, Fredholni AR, Kaijer I, (1988b): Adenosine in- jected into the brachial artery produces ischaernia-like pain or dis- comfort in tlit: forearm. Cardiovasc Rcs 22676678.

Taiwo YO, Leviric JD (1990): Direct cutaneous hyperalgesia induced b y adenosine. Neurosci 38:757-762.

'I:,iiwo YO, Lcvirie JD (1991): Further confirmation ofthe role ofade- iiyl cyclase arid of CAMP-dependent protein kin;ibc in primal? af- f r r rn t hyperalgesia. Neuroscierice 44:131-135.

Welch Sl: Dunlow LD (1993): Antinociccptivc~ activity of intrathecally administered potassium cliarrnel openers and opioid agonists: 11 c~miiion mechanism of action'? J Phamacol Exp Thcr 267:3'30-399.

White TD, Downie JW, Leslie RA (1985): Characteristics o f K t - and veratridine-induced release of ATP from synaptosomes prepared from dorsal and ventral spinal cord. Brain Res 334372-374.

Woolf CJ, Thompson JW (1994): Stirnulation-induced analgesia: Trun- scutancous electrical ncrvc stimulation (TENS) and viln-ation. I n Wall PD, Melzack R (eds): Textbook of Pain. Edinburgh: Churchill Livingstone, pp 1191-1208.

Ymnmioto '1; Yaksh 'TL (1993): Stereospecific effects of a nonpcptidic NK1 selective antagonist, CP-96,345: Aritinociccptiori iri the al~seiice of motor dysfiinction. Life Sci 4931955-1963.

Zimniernrann H (1994): Signalling via ATP i n the iiervous systc~m.

S o k v i A, Belfrase M, Lundeherg

Sawynok J, Downie Jn', Reid AR, (:ahill CM, White 'TD (1993): A'I'P 'Trends Ncurosci 17:420426