You are on page 1of 18

Acta Anuesthesiol Scnnd 1997; 41: 94-111 Copyright Q Actn Anaesthesiol Scnnd 1997

Printed in Denmark. All rights reserved ACTA ANAESTHESIOLOGICA SCANDINAVICA


ISSN 0001-5172

Pharmacology and mechanisms of opioid analgesic activity


T. L. YAKSH
Anesthesiology Research Laboratory, University of California, San Diego, U S A

Opiates by an action at specific receptors can induce a highly cord, this effect is mediated by an action presynaptic to the pri-
selective alteration in the response of humans and animals to mary afferent and by a post-synaptic effect to hyperpolarize pro-
strong and otherwise aversive chemical, mechanical or thermal jection neurons. In addition, it is now appreciated that p and K
stimuli. Specific investigations in a variety of species from ro- receptors in the periphery can modulate the sensitized state of
dent to primate using microinjection techniques to examine the the small afferent terminal innnervating inflamed tissue and ex-
pharmacology of local drug action have shown potent antino- ert an anti-hyperalgesic action. After systemic delivery of an
ciceptive actions to be mediated by a receptor specific action at opiate, it is thus clear that a wide array of central and peripheral
a number of sites within the brain, including the periaqueductal systems serve to explain the powerful analgesic effect exerted
gray (FAG: p receptor), the rostral ventral medulla (p/d recep- by this class of agents.
tor) and the substantia nigra (p receptor) and within the spinal
dorsal horn (p/a/K receptor). Mechanistic studies have shown
these actions in the different sites to be mediated by several Key words: periaqueductal gray; substantia nigra; bulbospinal
discrete mechanisms. For example, in the PAG, the local opiate pathway; rostral ventral medulla; spinal cord; opiate receptors;
effect is likely mediated by the indirect activation of bulbospinal mu receptors; delta receptors; kappa receptors; analgesia.
pathways, rostral projections to forebrain sites and by a local
alteration in afferent input into the brainstem core. In the spinal 0 Acta Anaesthesiologica Scandinavica 41 (1997)

mong the remedies which it has pleased almighty God


A to give to man to relieve his sufierings, none is so
universal and eficacious as opium (Sydenham 1680).
definition of the three principal receptor types. Thus,
work by Martin and colleagues (1) in spinally tran-
sected dogs led to the classification of agents as acting
at p and K sites. The subsequent work by Kostelitz
The potent effects of opiates in altering the animal’s and colleagues (2) using in vitro smooth muscle bioas-
or human’s response to strong and potentially tissue says with the then newly discovered enkephalin pep-
damaging stimulus has been long appreciated, as sug- tides led to the designation of the d receptor subclass.
gested by the quote attributed to Sydenham. The po- The profile of this activity is summarized in Table 1.
tency of the effect and the specificity of the action is, Receptor isolation and cloning have been accom-
moreover, indicative of a highly organized substrate plished for the p (3, 4), a (5) and K (6, 7, 8) receptor
which regulates the processing of and response to no-
ciceptive information. The following sections will re-
view some of the mechanisms by which this family
Table 2
of agents exert their actions as defined by preclinical
investigations. Opioid-receptor summary.

Receptor Bioassay Agonists Antagonists


Opioid receptors Mu Guinea pig Morphine Naloxone
ileum Sufentanil Naltrexone
Subtypes Meperidine beta-
At present, agents classified as opioids are believed to DAMGO funaltrexamine
exert their effects by a specific interaction with one or Delta Mouse vas DPDPE, DADLEl Naloxone
deferens Deltorphin, BNTX
more subclasses of three opiate receptors, designated DSLET Naltrindole
as: mu (p), delta (a) and kappa (K). Early in vivo phar-
Kappa Rabbit vas Butorphanol
macological work provided the initial definition of the deferens Bremazocine Naloxone
profile of actions that became associated with the Spiradoline Nor BNI

94
classes. Sequencing has revealed structures of 398 opening of voltage sensitive Ca++ channels (17, 18)
amino acids, 371 amino acids and 380 amino acids for which will subsequently depress the terminal release
the p, a and K sites, respectively. Cells made to express of neurotransmitters from the cell. These joint actions
the cloned sequences typically display the appropri- lead to a powerful, receptor-mediated inhibition that
ate ligand structure activity/affinity relationships an- is typically observed with these opiates. K receptors
ticipated for the respective sites. In general, they all appear to be distinctly coupled and have been shown
show considerable structural homology with each to depress Ca++ conductance in neurons. While the
other and to other G protein-coupled receptors (93. principal effect of the p receptor appears mediated
All, for example, display a characteristic 7 transmem- through an inhibitory effect, several lines of work
brane spanning domain in their structure. The cloned have suggested that under certain conditions, the p
receptors thus are in accord with the several general receptors may couple through Gs protein and exert a
classes of sites that have been identified in the phar- stimulatory effect (19). The significance of this action
macological literature. is less certain, however, than for the inhibitory effect,
A number of subclasses of the principal categories which appears to be broadly noted.
of receptors have been postulated based upon their
differential pharmacology: p1 and p2 (10); dl and d2
(11)and K~~ (lo), the characteristics of these subtype
Sites of opiate antinociceptive action
receptors continue to evolve as new agents are de- As noted, the systemic delivery of an opioid will pro-
veloped. It is important to note that in many cases duce a selective alteration in the animal’s response to
the clones have not yet demonstrated multiple types. a strong and otherwise aversive mechanical or ther-
However, the possibility of systematic changes in re- mal stimulus. This antinociceptive effect is character-
ceptor protein structure by post-translational pro- ized by a structure activity relationship and antagon-
cessing cannot be discounted. Moreover, separation of ist profile indicative of an action mediated by an
subclasses within the subtypes has frequently de- opioid receptor. The essential question is where are
pended upon a limited and structure activity relation- such receptors located? The methodological approach
ship. Thus, for example, the p1 and p2 receptor separ- to such an issue is to deliver the agent to specific sites
ation has been almost exclusively based on the ability in the organism and to define the effect and the phar-
of noncompetitive antagonists (such as naloxonazine) macology of the action if there is an effect after local
to antagonize one or another effect of an opioid exposure.
agonist (analgesia is reversed, respiratory depression For central delivery, such work has involved the
is not, (10). In the case of the p subclasses no selective placement of microinjection guide cannulae stereotax-
agonists have been described thus far. In contrast, in ically into specific brain sites and affixing them to the
the case of the a subclasses, selective competitive skull. Through the chronic guide, a yet smaller injec-
agonists (al: DPDPE; d2: deltorphin) have been iden- tion cannulae is inserted to deliver small volumes of
tified and a distinctive antagonist activity profile has injectate (0.2-0.5 pl). For the spinal cord, this local de-
been obtained for both sites (al: 7-brenzylidenenal- livery has entailed the placement of a spinal catheter
trexone; a2: naltriben) (11, 12, 13). into the intrathecal or epidural space (20). In each
For the purposes of the present review, emphasis case, the important issue has been to assess the effects
will largely remain with the primary classifications. of a local drug on the nociceptive behavior of the in-
tact and unanesthetized animal.
Membrane action
Agonist occupancy of opioid receptors typically leads Brain (supraspinal) sites
to several events which serve to inhibit the activation
of the neuron. Mu and a agonists have been shown to Mapping of brain sites in animals prepared with
depress cyclic AMP formation. Mu and 8 (14) and on mickoinjection cannulae has shown that opioid re-
occasion K (15) receptors can induce a membrane ceptors associated with systems which modulate the
hyperpolarization through the activation of an in- animal’s pain behavior are found in several surpris-
wardly rectifying K+channel. This effect of the p re- ingly restricted brain regions. Table 2 summarizes
ceptor is mediated by the activation of membrane Go several of the sites of actions which have been iden-
proteins, while the a effects are mediated by Gi/o pro- tified in the rat. The schematic localization of these
teins of a different nature (16). In addition to the sites is presented in Fig. 1 for the rat and Fig. 2 in
hyperpolarization induced by p and a agonist recep- the primate. The best characterization of these sites
tor occupancy, there is a concurrent inhibition of the so identified is the mesencephalic periaqueductal

95
T. L. Yaksh

Table 2
Summary of characteristicsof actions of opiates given into various sites in the unanesthetized rat.
Antinociceptive action
Microinjectionsites Reflex Escape Pharmacology References

Forebrairddiencephalon
amygdala (corticomedial) 0 ++ P? 1
N. accurnbens + P? 2
Mesencephalon
Periaqueductal grey +++ +++ p>>6=K=o 3
Mesencephalic reticular formation ++ ++ P 4
Substantia nigra ++ ++ P>>6=K=o 5
Lower brainstem
medial medulla ++ ++ p=6>K=0 6
Spinal cord +++ +++ p=&>K=O 7
Peripheral site' +++ p=K>6-0 8
~~

Reflex: tail flick/jaw jerk. Escape: Hot plate/paw pressure. Relative activity: ++ +>+ +>+>O.
*: Inflamed knee joint or foot pad.
References: 1. Rodgers RJ 1977 (42), Yaksh TL et al. 1976 (22); 2. Tseng LF, Wang Q 1992 (39). 3. Jensen TS, Yaksh TL 1986 (34), Smith
DJ et al. 1988 (155), Sanchez-Blazquez P, Garzon J 1989 (156); 4. Haigler HJ, Spring DD 1978 (33); 5. Baumeister AA et al. 1987 (36),
Baumeister AA et al. 1990 (37); 6. Jensen TS, Yaksh TL 1986 (34), Bodnar RJ et al. 1988 (182); 7. Drower EJ et al. 1991 (157), Malmberg
AB, Yaksh TL 1992 (791, Yaksh TL et al. 1986 (1581, Schmauss C, Yaksh TL 1984 (77); 8. Stein C et al. 1989 (104), NagasakaH, Yaksh TL
1995 (105).

gray (PAC). Microinjections of morphine into this re- injected agent emphasizes the possible activation of
gion will block nociceptive responses in the unanes- bulbospinal projections, as will be discussed below.
thetized rat, rabbit, cat, dog and primate (see below). Importantly, these effects are reversed by low doses
This local effect serves to block not only spinally-
mediated reflexes such as the tail flick, but supra-
spinally organized responses such as in the formalin
test (21), hot plate (see Fig. 1) or the shock titration
(22). This effect on a spinal reflex of a supraspinally

Fig. 1. Schematic representation of general regions in which the Fig. 2. Schematic drawings taken from sagittal histology (1-6
intracerebral microinjection of morphine will induce a signifi- mm from midline) of the rhesus monkey. Each dot presents the
cant increase in the response latency on thermal nociceptive effects of a single microinjection of morphine sulfate given
measures or increase the pressure applied to the hind paw through chronically implanted guide cannulae. 0: <50% increase
which is necessary to evoke withdrawal. A. Amygdala; B. Sub- in baseline shock titration threshold with 40 pg of morphine: 0:
stantial nigra; C. Mesecephalic reticular formation; Periaque- >50% increase in the baseline shock titration threshold with 40
ductal gray; D. Raphe magnus; nucleus gigantocellularis (rostra1 18 of morphine; V >50% increase in threshold with 10-20 pg of
ventral medulla); E. Spinal cord (dorsal horn). See text for details morphine. The stereotaxic axes are given for the most medial
and comments. (:LO) section (30).

96
of naloxone given either systemically or into the B-endorphin but not morphine displays significant ac-
microinjection site. tivity, suggesting two receptors (39).
In the ventral forebrain microinjections of p, a and
Mesencephalic periaqueductal gray K agonists into the region referred to as the area tem-
Tsou and Jiang discovered in 1963 that the local action pestas have been shown to attenuate thermal nocicep-
of morphine in the periventricular gray would block tion (41).
thermally-evoked hind limb reflexes and this has been
subsequently verified in a variety of species, includ- A mygdala
ing the rat (22,23), mouse (24), cat (25), dog (26), and Morphine given into the basolateral amygdala in-
primate (27,28) (Fig. 2). Within the PAG, a somatotop- creased escape but not spinal reflex latencies (22,42).
ic organization has been reported for opiate action (22, The pharmacology of the amygdala has not been
29). As indicated in Table 2, based on the relative ac- characterized but p opioid agonists are active (22, 30,
tivity of several receptor agonists and antagonists, the 42).
effects appear to be mediated by p but not a or K (30).
Binding studies focusing on the PAG have identified Thalamus and cortex
a single high affinity p site for which a and K agonists Several microinjection mapping studies of opiate ac-
have low affinity and which is coupled to a G protein tion have failed to observe activity following thalamic
(31, 32). or cortical injections (22, 27) but others (43) reported
that microinjection of morphine into the anterior pre-
Mesencephalic reticular formation. tectal region of the rat, but not adjacent nuclei, re-
Bilateral injection of opiates into the mesencephalic re- sulted in an inhibition of the tail flick.
ticular formation significantly increases escape la-
tency with only modest effects upon spinal reflexes
(33). The pharmacology of these systems through the
action of morphine implicates a p site. Supraspinal mechanisms of opiate
antinociception
Medulla The multiplicity of opiate sites virtually guarantees
Two distributions of opiate-sensitive sites have been that the systems with which the opiate receptors are
identified within the caudal medulla: i) medial sites coupled vary in the role each plays in altering no-
overlapping the nucleus raphe (34), and ii) lateral sites ciceptive processing. Accordingly, it is unlikely that
coincident with the nucleus gigantocellularis (34, 35). all of the mechanisms whereby opiates act within the
The pharmacology of these systems suggest both p brain to alter nociceptive transmission are identical. A
and a sites exist within the caudal medulla (34). brief overview of several classes of mechanisms will
be provided below.
Substantia nigra
Bilateral microinjection of opioids into the substantia
nigra will evoke an increase in the tail flick and hot
plate response latencies in rats without evidence of
significant motor impairment ,or change in the re-
sponse to non-noxious stimuli (36, 37). Examination
of the agonist’s and antagonist’s pharmacology of this
nigra action reveals the role of p, but not a or K recep-
50-
tors (38).

0-
Nucleus accumbens/ventral forebrain I -
Microinjections of opiates into the ventral forebrain, 0 10 30 60
Time after MlcroinJeclion(mln)
notably the nucleus accumbens, preoptic and arcuate
nuclei, will block spinal nociceptive reflexes (39, 40). Fig. 3. Figure presents the time course of the change in the tail
Tseng and colleagues observed that in the preoptic flick response latency after the microinjection of 5 p/0,5 p1 given
into the sites indicated. In this case, a single rat was prepared
and arcuate region, B-endorphin and morphine with a single microinjection guide and injections were made at
yielded a dose-dependent, naloxone-reversible in- progressively deeper sites (separated by 1 mm). Each injection
crease in tail flick latencies. In the nucleus accumbens, was separated by a 5 day interval (22).

97
T.L. Yaksh

Descending control
Mu and a opiates acting in the brainstem will inhibit
spinal nociceptive reflexes, reduce the spinal neuronal
activity evoked by noxious stimuli and alter supra-
spinally organized pain behavior. As discussed in de-
tail elsewhere (44, 45), bulbospinal pathways exert a
powerful regulatory influence over spinal cord affe-
rent processing. These effects are in accord with
studies in which:
i) activation of bulbospinal pathways which con-
tain noradrenaline or 5-HT will inhibit spinal no- MeduI lopet aI neu ron
ciceptive activity (46,47);
ii) enhancement of spinal monoamine receptor ac-
tivity by local delivery of a2/5-HT agonists will
inhibit pain behavior (48, 49, 50, 51), while the
4
(GABA-
Medulla
Fig. 4.Schematic of organization of opiate action within the peri-
spinal action of these agonists will inhibit the fir-
aqueductal gray. In this schema, 1 opiate actions block the re-
ing of dorsal horn neurons evoked by high inten- lease of GABA from tonically active systems that otherwise
sity stimuli (52, 53, 54); regulate the projections to the medulla leading to an activation
iii) opiate microinjection into brainstem sites in- of bulbospinal projections. See text for other considerations.
creases the spinal release or turnover of 5-HT
and/or noradrenaline (55,56);
iv) intrathecal injection of a2 or serotonergic anta-
gonists has minimal effect upon resting baseline ion within the periaqueductal gray can inhibit nucleus
response latencies, but reverses the effects of reticulo-gigantocellularis neurons (61). Fields et al.
brainstem opiates on spinal reflexes and anal- have shown powerful mesencephalic influences upon
gesia (57, 58). medullary cell populations (62). Given the role played
It is important to note that spinal inhibition and the by the medullary system in the ascending pain traffic,
increase in monoamine release both occur after the de- it seems probable that some of these cells may repre-
livery of the PAG opiates. This, in conjunction with sent projection neurons which contribute to the rostral
the absence of prominent effects on baseline responses movement of nociceptive information.
when the spinal monoamine antagonists are given
alone, argues that the effects of PAG opiates are as- Direct inhibition of brainstem afferent trufic
sociated with an increase in spinofugal outflow and Many regions in which opioids exert their effects,
this influence can regulate spinal nociceptive pro- such as within the mesencephalon and medulla, re-
cessing. The mechanisms whereby this increase in ac- ceive significant input from direct spinobulbar projec-
tivity in bulbospinal pathways occur after the local tions or collaterals of spinodiencephalic projections.
PAG action of 1.1 opioid agonists is of particular organ- Cervical hemisection results in a reduction in opiate
izational interest, given the membrane inhibitory ef- in medulla PAG and the mesencephalic reticular for-
fects of the p opioids. Current thinking emphasizes mation ipsilateral to the hemisection (63). This sug-
that the outflow of the periaqueductal gray is under gests that opiate binding may be presynaptic on spi-
modulatory control by GABAergic neurons. Thus, to nofugal terminals and that locally administered opi-
the extent that local opiates inhibit activity in this ates might alter nociceptive processing through a
population of tonically active neurons, there would reduction of the excitation otherwise evoked by the
be an inhibition that would permit the drive of an spinofugal projections.
excitation (Fig. 4). Indeed, opiates have been shown
to diminish a tonic GABA release from the PAG (59), Forebrain mechanism modulating the response
while GABA A antagonists enhance the activity of lo- Ample evidence suggests that opiates may interact
cal neurons (60). with brainstem mechanisms to alter input by a variety
of direct and indirect systems. The behavioral seque-
Brainstem-brainstem inhibition of afferent trafic lae of opioids possess a significant component that re-
Spinomedullary and spinal mesencephalic projections flects a change in the affective component of the re-
are thought to play a role in the generation of the sponse to the pain state. Numerous rostral projections
message evoked by high threshold stimuli. Stimulat- arise from the dorsal raphe nucleus (5-HT) and the

98
locus caeruleus (noradrenaline) that connect the peri- have been shown to be effective are presented in Table
aqueductal gray with forebrain systems that are 3.
known to influence motivational and affective com-
ponents of behavior, including the n. accumbens, Pharmacology
amygdala and lateral thalamus (40, 64).The locus ca- The pharmacology of spinal opioids has been exten-
eruleus has ample projections into the limbic fore- sively examined from the perspective of agonist struc-
brain and thalamus (65). Both monoaminergic sys- ture activity relationships, antagonist structure activ-
tems are strongly implicated in emotionality and ity relationships and antagonist potency. In general,
maintenance of consciousness. Lesions of raphe dor- on the tests outlined above, spinal opioids produce
salis diminish the effects of morphine (66, 67) and de- a monotonic dose-dependent increase in the latency
pletion of serotonin has been classically known to pro- response (tail flick, hot plate, skin twitch), the magni-
duce rats that are exceedingly irritable (68). tude of the response (writhing, number of paw licks,
change in blood pressure) or the threshold for escape
(paw pressure, shock titration) (see above for refer-
Spinal Action ences). Based on the consideration of the available
Behavioral Effects data, several points regarding the pharmacology of
Intrathecal administration of opioids reliably attenu- spinal opiates may be summarized.
ates the response of the animal to a variety of uncon- i) The structure activity relationship for these
ditioned somatic and visceral stimuli which otherwise opioids over a range of 2-3 orders of magnitude
evokes an organized escape behavior in a variety of are similar on the several endpoints and across
species including the mouse (69), rat (70), rabbit (70), species;
guinea pig (L. Crone, unpublished observations), cat ii) The rank order of potency in producing a block of
(71, 72), dog (73), and primate (macaque) (30, 74, 75). spinal reflexes after intrathecal delivery correlated
The antinociceptive endpoints in which spinal opiates with the potency of agents on the guinea pig

Table 3
Models of nociception in which spinal opiates have been examined.
General stimulus Animal Specific model References

Acute thermal Rat Hot plate (523) Yaksh TL, Rudy TA 1977 (20)
Tail flick
Rat Tail dip evoked hypertension and tachycar- Nagasaka H, Yaksh TL 1995 (105)
dia
Doglcat Skin twitch Tung AS, Yaksh TL 1982 (72)
Yaksh TI 1978 (85)
Sabbe MB et al. 1994 (73)
Primate Thermal escape Oliveras J-L et al. 1986 (97)
Yaksh TI ibid.
Acute mechanical Rat Paw pressure Taiwo YO et al. 1989 (160)
Rat Tail clamp/pinch Kuraishi Y et al 1985 (183)
Acute visceral Rat Colonic distension Ness TJ, Gebhart GF 1989 (161)
Harada Y et al. 1995 (162)
Acute electrical shock Rat Tail shocklvocalization Tang AH, Schoenfeld MJ 1978 (163)
Yaksh TL, Rudy TA 1977 (67)
Primate Shock titration Yaksh TL 1983 (75)
Acute chemical stimuli Rat Writhing Schmauss C, Yaksh TL 1984 (77)
Rat lntradermal formalin Yamamoto T, Yaksh TL 1992 (164)
Chronic neurogenic Rat Autonomy-neurectomy Weisenfeldt-HallinZ 1984 (165)
Rat Chronic nerve compression Yamamoto T, Yaksh TL 1991 (145)
(Bennett model)
Rat Root ligation (Chung model) Yaksh TI et al 1995 (76)
Lee Y-W et al. 1995 (146)
Fictive pain stimuli generated by intra- Rat lntrathecal strychnine Yaksh TL 1989 (166)
thecal drug treatment

99
T. L. Yaksh
Primate: 51C Tail Dip
ileum and on the mouse vas deferens (76), sug-
gesting a role for the p and d receptors in virtually INTRATHECAL DRUG
all endpoints (except certain neuropathic pain
models (Table 3). Kappa-preferring agonists thus
far studied typically have lower apparent efficacy
(77).
iii) Intrathecal opioids are subject to a competitive an-
tagonism by naloxone. For agents classified as p
preferring agonists (morphine, sufentanil, -20 40 loo 160
Time (min)
DAME), the pA2 (dose required to double the
EDs0 of the agonist) is similar across all tests /f T ITDRUG
examined (writhing, hot plate, tail flick in the rat)
and across species (rat and primate) and is about
0.1X that dose required to antagonize agonists
classified as d and K (78), while receptor preferring
antagonists block the agonists for the respective
receptors (e.g. 6 preferring opioids (DPDPE,
DADL) but not p preferring (morphine, DAG)
have been shown to be reversed bynaltrindole 1 10 100 1WO 1woo
Intrathecal Dose (pg)
(79). The pharmacology of these effects appear to
be similar across a number of species, including - 100

rat (12, 13, 79,80) and primate (74, 75,81) (Fig. 5). -z
w
00

6o NALOXONE (100 pg)


Based on the above overview, a case can be made that & 40
NALTRENDOLE (300 pg)
in the spinal cord of infrahuman species, there are at ae 20

least three distinguishable subpopulations of opioid 0


Mor (1000) DAMGO (30)DPDPE (3000)DADL (1000)
receptors which, according to the homology of their INTRATHECAL AGONISTS (pg)
agonist and antagonist structure activity profiles, re- Fig. 5. Top: Time effect curve for the effects of intrathecal p (mor-
semble the subclassifications designated as p, d and K. phine, sufentanil) and a (DPDPE) agonists on the response la-
tency to withdraw the tail of the cynomologous primate from
Mechanisms of spinal opiate action a 51°C waterbath. Each line presents the mean and SEM of 4
experiments. Middle: Intrathecal dose effect curve for intrathecal
Initial studies demonstrated that opiates applied by p (DAMGO, SUF: Sufentanil, MOR: morphine), 'pk (DADL: D-
iontophoresis (82,83) or systemically in spinally tran- ala2 d-leu 5 enkephalin: DPDPE: d-pen2-dpen5-enkephalin)or
sected animals (84, 85) would inhibit selectively the K (U50488) agonists. Bottom: Reversal of the effects of intrathecal
discharge of spinal dorsal horn neurons activated by Morphine (1000 pg); DAMGO (30 pg), DPDPE (3000 pg) and
small (high threshold) but not large (low threshold) DADL (1000 pg) after pretreatment with intrathecal saline (con-
trol), naloxone (100 pg) or Naltrindole (300 pg). * R 0 . 0 5 as com-
afferents. Fleetwood-Walker and colleagues (83) pared to control.
examined the pharmacology of the inhibition of dor-
sal horn neurons by iontophoretically applying recep-
tor preferring agonists for the p (DAG), d (DPDPE)
and K (U50488).In Lamina I, with nociceptive specific revealed a number of important characteristics of
neurons or with multireceptive (WDR)neurons, p and spinal opiate binding. These revealed that binding
d agonists exerted a suppressive effect upon the no- was limited for the most part to the upper laminae of
ciceptive component. Kappa agonists had no effect. In Rexed, particularly in the substantia gelatinosa (87,
laminae 11-V, p and d agonists were with minimal ef- 88, 89, 90), the region in which small afferents show
fect, while K agonists caused a selective inhibition of their principal termination. The proportions of the p,
the nociceptive component. Recording evoked activity d and K opioid binding sites, assessed by autoradi-
in lamina I1 neurons in spinal cord slices revealed that ography in laminae I and 11, were found to be ap-
p (DAMGO) and d l (DPDPE) agonists produced a proximately 70%, 20% and lo%, respectively. Dorsal
dose-dependent reduction in the excitatory post-syn- rhizotomies resulted in a significant reduction in dor-
aptic potentials. In contrast, the d2 agonists (D-ala2- sal horn binding, suggesting that the binding corre-
glu 4-deltorphin) had less than a 50% effect at the sponding to p, a and K sites were all reduced by 50-
highest dose examined (86). 75% in the dorsal horn ipsilateral to the lesions, sug-
Receptor autoradiography with opiate ligands have gesting that a significant proportion of all three bind-

100
ing sites were associated with the degenerating pri-
mary afferents (91,92). This association was consistent
with the observation that opiate bindkg was found
in the dorsal root ganglia (93). This organization sug-
\ C Fiber terminal

gested that opiates might thus exert a portion of their Receptors


activity by a presynaptic effect upon primary afferents
and a postsynaptic effect upon dorsal horn projection a
neurons. Confirmation of the presynaptic action was C I K
provided by the observation that opiates in vitro and
in vivo would reduce the release of primary afferent K + I
peptide transmitters such as substance P, which were
2nd Order neuron
contained in small primary afferents. Thus, it has been
reported that morphine, DAMGO, DPDPE and
DADL, but not U50488 (94,95,96,97) would block sP
and/or CGRP release in viva These interactions are,
however, complex as such interactions can influence Fig. 6. Schematic presents a summary of the anticipated organ-
multiple spinal terminal systems and there may be ization of opiate receptors in the dorsal horn regulating nocicep-
tive processing. As indicated, p/a and K binding is high in the
predictable differences on the effects assessed under
dorsal, particularly in the region associated with the termination
resting and stimulated conditions (98). of small unmyelinated afferents (C fibers). A significant pro-
A post-synapticaction was demonstrated by the abil- portion of these sites are located on the terminals of the small
ity of opiates to block the excitation of dorsal horn neu- afferent as suggested by the loss of such binding after rhi-
rons evoked by glutamate, presumably reflecting a di- zotomy. In addition, there is a post-afferent terminal localization
of these sites that are apparently coupled through Gi protein to
rect activation of the dorsal horn neuron (99). The pre- K channels leading to a hyperpolarization of the neuron. Occu-
synaptic action corresponded to the ability of opiates to pancy of the presynaptic p and a sites reduce the release of sP
prevent the opening of voltage-sensitive Ca+ chan-
+
and/or CGRP in part by an inhibition of the opening of voltage-
nels, thereby preventing release. The activation of pot- sensitive Calcium channels.
assium channels leading to a hyperpolarization was
consistent with the direct postsynaptic inhibition. The
joint ability of p and d opiates to reduce the release of Alternately, studies employing the direct injection
excitatory neurotransmitters from C fibers as well as of these agents into peripheral sites have demon-
decrease the excitability of dorsal horn neurons is be- strated that under conditions of inflammation where
lieved to account for the powerful and selective effects there is a “hyperalgesia”, the local action of opiates
upon spinal nociceptive processing (Fig. 6). can be demonstrated to exert a normalizing effect
There is evolving data that suggests that spinal opi- upon the exaggerated thresholds. This has been dem-
ates may exert a portion of their action by a local re- onstrated for the response to mechanical stimulation
lease of adenosine (100). However, it should be noted applied to the inflamed paw (104) or inflamed knee
that spinally delivered adenosine agonists are, unlike joints (105). As indicated in Fig. 7, examination of the
spinal p and a opiates, relatively poorly effective pharmacology of this articular action has emphasized
against acute high threshold stimuli (101) and do not the likely role of p and K, but perhaps less importantly
block spinal afferent peptide release (102). a sites (105, 106).

Mechanisms of peripheral action


Peripheral While there are opiate ‘%inding” sites being trans-
Behavioral efects ported in the peripheral sensory axon (107), there is
It has been a principal tenet of opiate action that these no evidence that these sites are coupled to mechan-
agents are “centrally” acting. Direct application of isms governing the excitability of the membrane, thus
opiates to the peripheral nerve can in fact produce a the effects are not naloxone reversible (103). High
local anesthetic-like action at high concentrations, but doses of agents such as sufentanil can block the com-
this is not naloxone reversible and is believed to re- pound action potential, but this is not naloxone re-
flect a “non-specific’’ action (103). Moreover, in pain versible and is thought to reflect upon a “local anes-
models examining normal animals, it can be readily thetic” action of the lipid soluble agent. It is certain
demonstrated that if the agent does not readily pen- that opiate receptors exist on the distant peripheral
etrate the brain, its opiate actions are limited. terminals. It has been shown that opiate receptors are

101
T. L. Yaksh

. Intra-articular T stem will produce a powerful alteration in nociceptive


processing. After systemic delivery, the net result will
likely reflect upon the effects of concurrent interaction
at the sites. Ample evidence indicates that the effects
of concurrent opiate receptor occupancy result in a
powerful functional facilitation. Several specific in-
stances are outlined.

Brainstem-spinal cord
Early studies revealed that delivery of an opioid an-
tagonist into the cerebral ventricles (111, 112) or into
the lumbar intrathecal space (67) can produce a com-
plete antagonism of the effects of a systemic opioid
2 agonist. Given the ability of opiate agonists to pro-
DRUG DOSE (mg)
duce a maximum effect with an action limited to
Fig. 7. Figure presents the dose response of the suppressive ef- either site alone, this significant reversal of the sys-
fects of maximum suppression of the evoked change in blood
temic agonist after local antagonist delivery was
pressure (expressed as the mean?SEM of the compression
evoked change in blood pressure) after injection, intra-articu- hypothesized to indicate that the spinal and supraspi-
lady or intramuscularly of SUF (sufentanil), PD: (PD117302), nal opiate systems were interacting in a synergistic
MOR: (morphine),SPIR (spiradoline),DADL (dala2 dleu5 enke- fashion. In other words, levels of local receptor occu-
phalin), DPDPE (d-pen2-d-pen5 enkephalin) and U-50,488H. pancy which were sufficient to produce a behavioral
Each point presents the mean2SEM of 4-8 rats. Because the IA
injection alone of the vehicle produced an augmentation re-
analgesia when both spinal and supraspinal sites
sponse, all IM drug injections were given concurrently with an were occupied were inadequate when either contri-
IA injection of saline. The horizontal lines indicate the mean bution was removed (113). The specific test of this
(so1id)eSEM (dotted lines) for rats receiving IA saline and no hypothesis was provided by Yeung and Rudy (114),
other drug at time zero. who demonstrated that the concurrent administration
of morphine spinally and supraspinally would lead to
on the distal terminals of C fibers and that agonist a prominent synergy, as indicated by hyperbolic iso-
occupancy of these sites can block antidromic release bolograms. Similar results have been observed in mice
of C fiber transmitters e.g., sP; "axon reflex" (108).Im- (115,116).
portantly, the models in which peripheral opiates ap- Consistent with the powerful nonlinear interaction
pear to work are those that possess a significant de- between spinal and supraspinal opiates, and given the
gree of inflammation and are characterized by a considered role of bulbospinal systems in mediating
hyperalgesic component. This raises the possibility some of the supraspinal actions of opiates, it has been
that these peripheral actions normalize a process lead- shown that the concurrent spinal delivery of a2and p
ing to an increased sensitivity to the local stimulus and 3 opioid agonists as well as concurrent p and d
environment, but does not alter normal transduction. agonists can interact synergistically (44).
Previous work has indeed demonstrated that local
opiates in the knee joint (109) and in the skin (110) Supraspinal-supraspinal
can reduce the firing of spontaneously active afferents In the brain, multiple sites exist that mediate an opiate
observed when these tissues are inflamed. The mech- antinociceptive effect. Co-injection of opiates into the
anism of the antihyperalgesic effects of opiates ap- periaqueductal gray and locus caeruleus (117) re-
plied to the inflamed regions (as in the knee joint) is at sulted in a synergic interaction, while co-injection into
present unexplained. It is possible, for example, that it the periaqueductal gray and nucleus reticularis gigan-
may act upon inflammatory cells that are present and to-cellularis (118) appeared to have only a simple ad-
are releasing cytokines and products that activate or ditive interaction.
sensitize the nerve terminal (106).
Spinal-spinal cord
It is apparent that within the spinal cord there are
Interactions between opiate modulated multiple points of interaction for opiates, including
systems presynaptic on C fibers and a concurrent postsynaptic
As outlined above, opioids with an action limited to interaction that leads to a hyperpolarization of dorsal
several sites, notably the spinal cord, and to the brain- horn projection neurons. It is likely that the net effect

102
Table 4
Summary of activity of several opiates delivered epidurally on human clinical pain.

Drug Receptor Approximate dose Reference

Lofentanil Van den Abele G, Camu F 1983 (167)


Buprenorphine Simpson KH et al. 1988 (168)
Fentanyl Welchew EA 1983 (169)
Sufentanil Geller E et al. 1993 (170)
Hydromorphone Brose WG et al. 1991 (171)
Alfentanil Chauvin M et al. 1985 (172)
Butorphanol Camann WR et al. 1992 (173)
DADL Onofrio BM, Yaksh TL 1983 (137)
P-endorphin Oyama T et al. 1982 (175)
Morphine Yaksh TL 1987 (132)
Methadone Torda TA, Pybus DA 1982 (176)
Evron S et al. 1985 (177)
Meperidine P Torda TA, Pvbus DA 1982 (176)

of spinally delivered agents is mediated by this con- pain relief in cancer patients (124,125,126,127,128). An
current action which reduces the excitatory input and important characteristicof this action is that the time of
the excitability of the dorsal horn neuron. While the onset is relatively rapid for even the water-soluble
importance of this concurrent interaction on the net agent morphine. Gamma scans of human brain after
analgesic action of spinal opiates has not been system- 1123-morphine have shown that the agent, even at one
atically examined, a comparison can be made between hour after injection, remains close to the ventricular lu-
spinal receptor systems that have and do not have this men (129). This distribution is consistent with the slow
property. Thus, p, d and a2 agonists and neuropeptide permeation of the water soluble agent from the ven-
Y have pre-synaptic effects upon C fiber transmitter tricular lumen (130).Given that the rate of diffusion for
release and appear to hyperpolarize neurons through such a drug and the size of the human brain, it seems
a G protein coupling (119, 120). These agents are probable to hypothesize that the site of opiate action in
known to have a significant therapeutic ratio in ani- humans must lie close to the ventricular lumen. In this
mals and, where examined, in humans. In contrast, regard, the preclinical studies in species such as the pri-
agents such as adenosine A1 agonists and GABA B mate have emphasized the importance of the periaque-
(baclofen) agonists have binding in the dorsal horn, ductal sites. Such a site of action would in fact lie in
can increase K conductance through G protein link- close proximity to the ventricles and permit a relatively
ages, but do not significantly decrease spinal sub- rapid access of a slowly moving drug from the ven-
stance P release (44,45). These agents after spinal de- tricular lumen. While an extensive activity relationship
livery have a comparatively poor therapeutic ratio, has not been achieved with human intracerebroventri-
e.g., they produce significant motor dysfunction at cular delivery, morphine and &endorphin have been
doses that alter nociceptive endpoints (121,122). It has delivered and analgesia produced with B-endorphin
been hypothesized that this concurrent pre-terminal appears to show a greater activity (131).
effect on small afferents and the post-synaptic effects
enhance the therapeutic ratio (123). Thus, these two Spinal
classes of agents argue for a potent functional contri- There is an extensive literature indicating that opiates
bution of the two concurrent sites of action leading to delivered spinally can induce a powerful analgesia in
antinociception. humans. The pharmacology of this action has been
relatively widely studied and it appears certain that
p, 8 and to a lesser degree K agonists are effective after
Similarity of mechanisms in human and intrathecal or epidural delivery (132). The effects of
animal models spinal opiates are reversed by low doses of systemic
Supraspinal naloxone (133, 134, 135, 136, 137). Importantly, in the
In humans it is not feasible to routinely assess the site activity of spinally-delivered agents in modulating
of action within the brain where opiates may act to alter acute nociception in animal models, such as for the
nociceptive transmission. However, intracerebroven- rodent, hot plate reveals an ordering of activity that
tricular opioids have been employed in humans for closely resembles that observed in humans for con-

103
T. L. Yaksh
1 OOOOl 1 tized by a local change in the chemical milieu that is

5
A

L
1oooj

100-
r-0.872 7 generated by local injury and inflammation. Given
these properties, it is not surprising that the clinically
employed opiates (largely p and to a lesser extent K
agonists) are noted to be particularly effective in the
postinjury/-operative pain state, as they are in the
3. Hydromorphone
L acute nociceptive models (Fig. 8). In each case, it is
in believed that the principal input generated by mech-
0
L 6. Morphine
w 7. Alfentanil anical distortion of the tissue or by the elaboration of
8. Methadone active factors (141, 142) from the injury similarly re-
9. Butorphanol
10. Buprenorphine flects input that arises from activity in small afferents.
11. Meperidine In neuropathic pain models, the normal processing
.1 1 10 100 1000 of sensory input is altered to yield a hyperalgesic com-
RAT (HOT PLATE 52.5.C) ponent, as indicated in the thermal escape latency in
Fig. 8. The relative ordering of activity for clinically effective the model of Bennett and Xie (143), or in the tactile allo-
doses of spinal opiates in the postoperative pain state expressed dynia in the nerve ligation model of Kim and Chung
relative to the activity of morphine (e.g. morphine=l) in (144) or the intrathecal strychnine model of Yaksh (108).
humans is plotted versus the ordering of activity relative to mor- As summarized in Table 5, in these models, opiates
phine after spinal delivery in the rat model on the 52°C hot plate
test. * indicates that buprenorphine on the hot plate is a partial
have been shown to have distinguishable effects. Thus,
agonist. in the Bennett and Xie model (143), systemic and intra-
thecal morphine has been shown to be effective and the
dose response curves have been shown to be shifted
modestly to the right (145).In contrast, on the tactile al-
trolling clinical pain states. This similarity is indicated lodynia models, intrathecal morphine has been shown
by the close regression observed when the approxi- to have no effect in altering the allodynia, while sys-
mate relative ordering of activity observed in rats and temic and intraventricular opiates have been shown to
after spinal delivery in humans is plotted (Fig. 8). be effective in reversing the tactile threshold (76, 108,
146). These observations, suggesting that at least some
Peripheral actions preclinical neuropathic states may not be sensitive to
The observation that opiates injected in the periphery spinal opiates are consistent with previous retrospec-
led to the clinical studies showing that observation of tive analyses in human causalgic pain states (147). As
injection of morphine into the knee joint of humans the tactile allodynia is believed to be mediated by large,
after knee surgery (138, 139, 140) showed that intra- low threshold mechanoreceptors, the modest, if any, ef-
articular morphine injections have a powerful sparing fect upon the tactile allodynia by spinal opiates is con-
effect upon subsequent analgesics. The appropriate sistent with the current thinking regarding their pre-
controls emphasize that the effects are indeed med- sumed mechanisms of action at the spinal level (see
iated by a local action and not by a CNS redistri- above). In this regard, the apparent efficacy of systemic
bution. These results are in close accord with the ob- or supraspinal opiates in these models may point to an
servation reported in the animal models of hyperalge- alteration in the supraspinal systems activated by the
sia induced by peripheral inflammation. anomalous ascending message generated by the tactile
stimulus in the post nerve injury pain state. On the
other hand, the efficacy of spinal and systemic opiates
Preclinical versus clinical pain states
in some pain models such as the Bennett model and in
and opiate action other models by a systemic or supraspinal action as in
Mechanistic variables the Chung model (Table 5) would suggest that in
A variety of nociceptive models employed in animals humans at least some components of the post nerve in-
have been shown to be sensitive to the supraspinal jury pain state should be affected by spinal opiates (148,
and spinal actions of opiates (see above and Table 3). 149).
A common property of these states are that they in-
volve: i) activity in small primary afferents, as acti- Stimulus intensity
vated by high threshold thermal, mechanical and Opiate agonists generate their effect by an interaction
chemical stimuli; or ii) in the case of the peripheral with a specific opiate site for which it has affinity and
action, a high threshold afferent that has been sensi- the magnitude of effect produced by a given degree

2 04
Table 5
Comparable sensitivity of preclinical and clinical pain states to morphine'.

Rat Human
Spinal ICVT Systemic Spinal Systemic
Somatic (small afferent)
Acute thermaVmechanical (rat') Yes Yes Yes
Post injury (human') Yes Yes
Neuropathic
Bennett (thermal hyperalgesia: rat) Yes2 - Yes3
Chung (tactile allydynia: rat) NO^ Yes5 Yes5
IT strychnine (tactile allodynia: rat) Nos - -
Spinal ischaemia (tactile allodynia: rat) - - No7
Nerve section (autotomy) Yese - NO'
Causalgia (human) Nolo YedNoll
Visceral
Colonic distension (rat) Yes12 - -
Uteral colic (human) Yes13 --
' See text and Table 3 for references; Yamamoto T, Yaksh TL 1991 (145); Yaksh TL, unpublishedobservations; Yaksh TL et al. 1995 (76);
Lee Y-W et al. 1995 (146); Yaksh TI 1989 (166); Xu XJ et al. 1992 (178); Wiesenfeld-Hallin2 1984 (165), Puke MJ, Wiesenfeld-Hallin
'
Z 1993 (179) (Pretreatmenffextended treatment); Yamamoto T, Mizuguchi T 1992 (180) (Prolonged oral); lo Arner S,Meyerson BA 1988
(147); 'l Cherny NI et al. 1994 (149); l2Ness TJ, Gebhart GF 1989 (161); l3Olshwang D et al. 1984 (181).

of receptor occupancy is defined by the intrinsic effi-


IT SUF, LOW
cacy of the agonist. Thus, if two p agonists produce a IT SUF, Mod
given effect with different degrees of occupancy, the IT SUF, High
agent requiring the lowest occupancy is said to have
higher intrinsic efficacy. Agents with low efficacy may
be unable to achieve a maximum effect even when
the fraction of receptor occupancy approaches 1. In
contrast, other agents may produce maximum effects
when only a small fraction of the available receptors
are occupied. The relative efficacy of two agonists
may be compared by measuring the shifts in their $ 1 -20, . .......I . '..'...I . ..'.... .
I . ' ' ~ ' ' '' ~.- ..-
dose response curves in the face of receptor inacti- - 0 ITMOR,LOW
100
vation with noncompetitive antagonists. Thus, for
opiates, in models of nociception using non B-funal-
5 8 0 -- ' ITMOR,Mld
ITMOR,Hlgh
trexamine, it has been possible to demonstrate after 0
2

/&
60
spinal and/or systemic delivery that sufentanil= 8
DAMGO>morphine (150, 151). An important conse- 2 40

-1
quence of differences in efficacy arise in the face of
20
changing stimulus intensities. It is known that an in- X
crease in stimulus intensity will result in a right shift $ 0
in the dose response curve for an analgesic and if the E
intensity is sufficient, the agonists may be unable to -20, . . ....-. . .
I *'."'I ' . ...'.. . .
I .."..1 ' ' '.--I

.001 .01 .1 1 10 100


produce a full effect, e.g., it will appear as a partial lntrathecal Dose (pg)
agonist. It has been hypothesized that the degree of Fig. 9. Dose response curves for sufentanil (SUF, top panel) and
right shift is defined by the intrinsic efficacy of the morphine (MOR, bottom panel) at progressively increasing ther-
agonist. This speculation has been supported for p mal stimdus intensities (Low, open circles; Medium, closed
opiate agonists (152) and generalized to a2 adrenergic circles; and High, closed squares). All results expressed as per-
agonists (153). An example of this is presented in Fig. cent maximum possible effect (%WE) versus Logarithm of opi-
ate dose (pg) with the mean of 4-6 animals2SEM represented
9. Here, the spinal delivery of sufentanil and mor- at each data point. As indicated, for a given increase in stimulus
phine results in a dose-dependent increase in the ther- intensity, sufentanil shows a lesser right shift than morphine and
mal escape latency. By increasing the intensity of the no decreasing maximum (154).

105
T. L. Yaksh

stimulus, the doses of sufentanil and morphine are effects on function at levels of receptor occupancy
shifted to the right, with the greatest shift and the de- at which other systems (e.g. respiratory or auto-
pression of maximum being greatest for morphine. In nomic) are not sufficiently affected to produce a
humans, some evidence for this has been obtained physiologically relevant action. This multitude of
from the ratio of potencies for sufentanil and mor- mechanisms and their synergistic interaction
phine defined in mild to moderate pain states (low likely accounts for the most part for how opiates
stimulus) versus that required for intraoperative anal- can exert a relatively potent, yet selective, effect
gesia (strong stimulus) (152,154). In this regard, the p upon nociceptive processing.
opioid agonist buprenorphine is appreciated to be a iv) Finally, our appreciation of the actions of the opi-
clinically useful analgesic, whereas in animal models ate systems provide important insights into the
it is frequently seen as having a plateau effect. This mechanisms by which nociceptive information is
plateau effect reflects upon the fact that at the maxi- processed. Thus, the characteristics of the antino-
mum achievable receptor occupancy, the agent has in- ciception will be defined by the systems with
sufficient efficacy to achieve a maximum effect. This which the opiates interact. In the case of neuro-
is manifested in Fig. 8, where intrathecally in the rat, pathic pain, for example, it might be speculated
on the 52°C hotplate, buprenorphine displays a pla- that in pain states in which the afferent drive is
teau effect and its estimated relative potency (as com- mediated by low threshold myelinated afferents
pared to morphine) is less than that observed in the (as in allodynia), the agent may be relatively less
clinical setting. Thus, the efficacy of the opiate ap- effective than in a pain state in which the pain
pears to have an impact upon the relative potency in state is mediated by a small afferent input (with
the preclinical model and in its clinical use. which opiate receptors are believed to be associ-
ated in the spinal cord).
v) Finally, while the data for human action of these
agents is relatively less exhaustive than the pre-
Conclusions clinical work, there is a prominent comparability
This overview of opioid receptor activity raises a across specifies when site of action and pharma-
number of important issues that are relevant to the cology are considered. Thus, there is little doubt
understanding of pain and pain processing. that in humans, as in the animal model, it is pos-
i) The ability of opiates to produce a powerful effect sible to demonstrate comparable supraspinal,
upon the organized response of the animal to a spinal and peripheral actions. This similarity in
strong and potentially tissue damaging stimulus pharmacology and function provides validating
reflects first upon the action mediated by a speci- support for the conclusion that animal models re-
fic family of receptors. veal mechanisms of processing that are present in
ii) The antinociceptive effects associated with such the human.
receptor occupancy reflects the coupling of these
receptors to specific neuronal systems that are
part of a circuitry that either regulates the excitab- Acknowledgement
ility of a neuronal pathway (as in the bulbospinal
projections that control spinal activity) or serves Work reported herein was supported in part by NIDA 02110. I
to directly impede the release of a transmitter link would like to dedicate this paper to Dr.Florella Magora who
contributed to the early clinical literature on spinal opiates, in
(as in the prevention of the release of a neuro- honor of her retirement.
transmitter from the primary afferent or by hyper-
polarizing the projection neuron).
iii) The complexity of each of these systems alone is References
amplified by the appreciation that the organized
processing is impacted at many levels by even a 1. Martin WR, Eades CG, Thompson WO, Thompson JA,
locally acting agent (as in the PAG or in the spinal Flanary HG. Morphine physical dependence in the dog.
Pharmacol Exp Ther 1974:189:759-771.
cord where a single site reflects multiple effect). 2. Lord JA, Waterfield AA, Hughes J, Kosterlitz HW. Endo-
Importantly, in many cases, it appears certain that genous opioid peptides: multiple agonists and receptors.
the concurrent involvement of the several separ- Nature 1977: 267 495-499.
ate components yields a physiological or biochem- 3. Chen Y,Mestek A, Liu J, Hurley JA, Yu L. Molecular clon-
ing and functional expression of a mu-opioid receptor from
ical consequence that is greater than an additive rat brain. Mol Pharmacol 1993:44:8-12.
(e.g. synergistic). The synergy permits significant 4. Thompson RC, Mansour A, Akil H, Watson SJ.Cloning and

2 06
pharmacological characterization of a rat mu opioid recep- 24. Criswell HD. Analgesia and hypereactivity following mor-
tor. Neuron 1993: 11: 903-913. phine microinjections into mouse brain. Phurmucol Biochem
5. Kieffer BL, Befort K, Gaveriaux-Ruff C, Hirth CG. The Behuv 1976 4 23-26.
delta-opioid receptor: isolation of a cDNA by expression 25. Ossipov MH, Goldstein FJ, Malseed RT. Feline analgesia
cloning and pharmacological characterization. Proc Nutl following central administration of opioids. Neurophurmu-
Acud Sci U S A 1992: 89: 12048-12052. cology 19% 23: 925-929.
6. Chen Y, Mestek A, Liu J, Yu L. Molecular cloning of a rat 26. Wettstein JG, Kamerling SG,Martin WR. Effects of microin-
kappa opioid receptor reveals sequence similarities to the jections of opioids into and electrical stimulation (ES) of the
mu and delta opioid receptors. Biochem J 1993: 295 625- canine periaqueductal gray (PAG) on electrogenesis (EEG),
628. heart rate (HR), pupil diameter OPD), behavior and anal-
7. Li S, Zhu J, Chen C, Chen YW, Deriel JK, Ashby B, Liu- gesia. Neurosci Abstr 1982 8: 229.
Chen LY. Molecular cloning and expression of a rat kappa 27. Pert A, Yaksh TL. Sites of morphine induced analgesia in
opioid receptor. Biochem J 1993 295: 629-633. the primate brain: relation to pain pathways. Bruin Res
8. Meng F, Xie GX, Thompson RC, Mansour A, Goldstein A, 1974 8 0 135-140.
Watson SJ, Akil H. Cloning and pharmacological character- 28. Pert A, Yaksh TL. Localization of the antinociceptive action
ization of a rat kappa opioid receptor. Proc Nutl Acud Sci of morphine in primate brain. Phurmacol Biochem Behuv
U S A 1993 90: 9954-9958. 1975: 3: 133-138.
9. Uhl GR, Childers S, Pasternak G. An opiate-receptor gene 29. Kasman GS, Rosenfeld JP. Opiate microinjections into mid-
family reunion. Trends Neurosci 1994: 1 7 89-93. brain do not affect the aversiveness of caudal trigeminal
10. Pasternak GW. Pharmacological mechanisms of opioid an- stimulation but produce somatotopically organized peri-
algesics. Clin Neurophurmacol 1993: 1 6 1-18. pheral hypoalgesia. Brain Res 1986 383: 271-278.
11. Jiang Q, Takemori AE, Sultana M, Portoghese PS, Bowen 30. Yaksh TL, Al-Rodhan NRF, Jensen TS. Sites of action of
WD, Mosberg HI, Porreca F. Differential antagonism of opiates in production of analgesia. Prog Bruin Res 1988: 77:
opioid delta antinociception by [D-ala2,Leu5, Cys61 - enke- 371-394.
phalin and naltrindole 5’-isothiocyanate: evidence for delta 31. Fedynyshyn JP,Kwiat G, Lee N. Characterization of high
receptor subtypes. J Phurmucol E x p Ther 1991: 257 1069- affinity opioid binding sites in rat periaqueductal gray P2
1075. membrane. Eur J Pharmacol 1989: 159: 83-88.
12. Hammond DL, Stewart PE, Littell L. Antinociception and 32. Fedynyshyn JE’,Lee NM. Mu type opioid receptors in rat
delta-1 opioid receptors in the rat spinal cord: studies with periaqueductal gray-enriched P2 membrane are coupled to
intrathecal 7-benzylidenenaltrexone.J Phurmacol Exp Ther G-protein-mediated inhibition of adenylyl cyclase. FEBS
1995: 274 1317-1324. Letters 1989: 253: 23-27.
13. Stewart PE; Hammond DL. Activation of spinal delta-1 or 33. Haighler HJ, Spring DD. A comparison of the analgesic
delta-2 opioid receptors reduces carrageenan-induced and behavioral effects of [D-Ala2] metenkephalinamide
hyperalgesia in the rat. J Phurmucol E x p Ther 1994 268: 701- and morphine in the mesencephalic reticular formation of
708. rats. Life Sci 1978: 2 3 1229-1240.
14. Yoshimura M, North RA. Substantia gelatinosa neurones in 34. Jensen TS, Yaksh TL. Comparison of the antinociceptive ac-
vitro hyperpolarised by enkephalin. Nature 1983: 305 529- tion of mu and delta opioid receptor ligands in the peri-
530. aqueductal gray matter, medial and paramedial ventral
15. Grudt TJ, Williams JT. Kappa-Opioid receptors also in- medulla in the rat as studied by the microinjection
crease potassium conductance. Proc Nutl Acud Sci U S A technique. Bruin Res 1986 372: 301-312.
1993: 9 0 11429-11432. 35. Satoh M, Oku R, Akaike A. Analgesia produced by micro-
16. Carter BD, Medzihradsky F. Go mediates the coupling of injection of L-glutamate into the rostra1 ventromedial
the mu opioid receptor to adenylyl cyclase in cloned neural bulbar nuclei of the rat and its inhibition by intrathecal U-
cells and brain. Proc Nutl Acud Sci U S A 1993: 9 0 4062-4066. adrenergic blocking agents. Bruin Res 1983: 261: 361-364.
17. Kaneko S, Fukuda K, Yada N, Akaike A, Mori Y, Satoh M. 36. Baumeister AA, Hawkins MF, Anticich TG, Moore LL, Hig-
Ca2+ channel inhibition by kappa opioid receptors ex- gins TD, Vaughn A, Chatellier MO. Bilateral intranigral
pressed in Xenopus oocytes. Neuroreport 1994: 5 2506-2508. microinjection of morphine and opioid peptides produces
18. Piros ET, Prather PL, Loh HH, Law PY, Evans CJ, Hales antinociception in rats. Bruin Res 1987 411: 183-186.
TG. Ca2+ channel and adenylyl cyclase modulation by 37. Baumeister AA, Nagy M, Hebert G, Hawkins MF, Vaughn
cloned mu-opioid receptors in GH3 cells. Mol Phurmucol A, Chatellier MO. Further studies of the effects of intra ni-
1995: 4 7 1041-1049. gral morphine on behavioral responses to noxious stimuli.
19. Shen K-F, Crain SM. Dual opioid modulation of the action Brain Res 1990: 525 115-125.
potential duration of mouse dorsal root ganglion neurons 38. Baurneister AA. The effects of bilateral intranigral microin-
in culture. Brain Res 1989: 491: 227-242. jection of selective opioid agonists on behavioral responses
20. Yaksh TL, Rudy TA Chronic catheterization of the spinal to noxious thermal stimuli. Brain Res 1991: 557 136-145.
subarachnoid space. Phys Behuv 1976 17: 1031-1036. 39. Tseng LP, Wang Q. Forebrain sites differentially sensitive
21. Manning BH, Morgan MJ, Franklin KB. Morphine anal- to beta-endorphin and morphine for analgesia and release
gesia in the formalin test: evidence for forebrain and mid- of Met-enkephalin in the pentobarbital-anesthetized rat. J
brain sites of action. Neuroscience 1994: 63289-294. Phurmacol E x p Ther 1992 261: 1028-1036.
22. Yaksh TL, Yeung JC, Rudy TA. Systematic examination in 40.Ma QP, Han JS. Neurochemical and morphological evi-
the rat of brain sites sensitive to the direct application of dence of an antinociceptive neural pathway from nucleus
morphine: Observation of differential effect within the pe- raphe dorsalis to nucleus accumbens in the rabbit. Bruin
riaqueductal gray. Bruin Res 1976 114 83-103. Res Bull 1992 2 8 931-936.
23. Sharpe LG, Gamett JE, and Cicero TJ. Analgesia and hyper- 41. d’Amore A, Lorenzini P, Massotti M. Antinociceptive ac-
reactivity produced by intracranial microinjections of mor- tion of opiates and opioid peptides after unilateral microin-
phine into the periaqueductal gray matter of the rat. Behuv jection into area tempestas in rats. J P h u m c o l Exp Ther
Biol 1974: 11: 303-313. 1991: 259 1308-1315.

2 07
T. L. Yaksh
42. Rodgers RJ. Elevation of aversive threshold in rats by intra 60. Ehbehani MM, Jiang MR, Chandler SD, Ennis M. The effect
amygdaloid injection of morphine sulfate. Pharmacol Bio- of GABA and its antagonists on midbrain periaqueductal
chem Behau 1977 6:385-390. gray neurons in the rat. Pain 1990: 40: 195-204.
43. Prado WA. Antinociceptive effect of agonists microinejcted 61. Mohrland S, Gebhart G. Effects of focal electrical stimula-
into the anterior pretectal nucleus of the rat. Brain Res 1989: tion and morphine microinjection in the periaqueductal
493:145-154. gray of the rat mesencephalon on neuronal activity in the
44. Yaksh TL, and Malmberg AB. Central pharmacology of no- medullary reticular formation. Brain Res 1980: 201: 23-37.
ciceptive transmission. In:Wall P, Melzack R,eds. Textbook 62. Fields HL, Heinricher MM, Mason P. Neurotransmitters in
of Pain 3. ed. Edinburgh Churchil Livingstone, 1994: 165- nociceptive modulatory circuits. Ann Rev Neurosci 1991: 14:
200. 219-245.
45. Yaksh TL, and Malmberg AB. Interaction of spinal modula- 63. Ramberg DA, Yaksh TL. Effects of cervical spinal hemisec-
tory receptor systems. In. Fields HL, Liebeskind JC, eds. tion of dihydromorphine binding in brainstem and spinal
Progress in Pain Research and Management, Vol. 1. Seattle: cord in cat. Brain Res 1989: 483: 6 1 4 7 .
IASP Press, 1994: 151-171 64. Ma QP, Ym GF, Ai MK, Han JS. Serotonergic projections
46. Gebhart GF, Sandkuhler J, Thalhammer J and Zimmerman from the nucleus raphe dorsalis to the amygdala in the rat.
M. 1984. Inhibition in spinal cord of nociceptive infor- Neurosci Lett 1991: 134:21-24.
mation by electrical stimulation and morphine microinjec- 65. Amaral DG, Sinnamon HM. The locus ceruleus: Neurobiol-
tions at identical sites in midbrain of the cat. J Neurophysiol ogy of a central noradrenergic nucleus. Prog Neurobioll977:
1984: 51: 75-89. 9: 147-196.
47. Gebhart GF, Jones SL. Effects of morphine given in the 66. Samanin R, Gumulka W, Valzelli L. Reduced effect of mor-
brainstem on the activity of dorsal horn nociceptive neu- phine in midbrain raphe lesioned rats. Eur J Pharmacoll970:
rons. Prog Brain Res 1988: 77 229-243. 1 0 339.
48. Reddy SVR, Maderdrut JL, Yaksh TL. Spinal cord pharma- 67. Yaksh TL, Rudy TA. Studies on the direct spinal action of
cology of adrenergic agonist-mediated antinociception. narcotics in the production of analgesia in the rat. J Pharma-
Pharmacol Exp Ther 1980: 213:525-533. col Exp Ther 1977 202: 411-428.
49. Takano Y, and Yaksh TL. Characterization of the pharma- 68. Tenen SS. Antagonism of the analgesic effect of morphine
cology of intrathecally administered alpha-2 agonists and and other drugs by p-chlorophenylalanine, a serotonin de-
antagonists in rats. J Pharmacol Exp Ther 1992: 261: 764-772. pletor. Psychopharmacology 1968: 12: 278.
50. Yaksh TL, Wilson PR. Spinal serotonin terminal system me- 69. Hylden JLK, Wilcox GL. Pharmacological, characterization
diates antinociception. J Pharmacol Exp Ther 1979: 208: 446- of substance P induced nociception in mice: Modulation
453. by opioid and noradrenergic agonists at the spinal level. J
51. Danzebrink RM, Gebhart GF. Evidence that spinal 5-HT1, Pharmacol E x p Ther 1983: 226: 398404.
5-HT2 and 5-HT3 receptor subtypes modulate responses to 70. Yaksh TL, Rudy TA. Analgesia mediated by a direct spinal
noxious colorectal distention in the rat. Brain Res 1991: 538: action of narcotics. Science 1976 192:1357-1358.
64-75. 71. Yaksh TL. Analgetic actions of intrathecal opiates in cat and
52. El-Yassir N, Fleetwood-Walker SM, Mitchell R. Heterogen- primate. Brain Res 1978: 153:205-210
eous effects of serotonin in the dorsal horn of rat: the in- 72. Tmg AS, Yaksh TL. The antinociceptive effects of epidural
volvement of 5-HT1 receptor subtypes. Brain Res 1988: 456 opiates in the cat: studies on the pharmacology and the
147-58. effects of lipophilicity in spinal analgesia. Pain 1982: 12:
53. Nakagawa I, Omote K, Kitahata LM, Collins JG, Murata K. 343-356.
Serotonergic mediation of spinal analgesia and its interac- 73. Sabbe MB, Grafe MR, Mjanger E, Tiseo PJ, Hill HF, Yaksh
tion with noradrenergic systems. Anesthesiology 1990: 73: TL. Spinal delivery of sufentanil, alfentanil and morphine
474-478. in dogs. Physiologic and toxicologic investigations. Anes-
54. Fleetwood-Walker SM, Mitchell R, Hope PJ, Molony V, thesiology 1994: 81: 899-920.
Iggo A. An alpha-2 receptor mediates the selective inhi- 74. Yaksh TL, Reddy SVR. Studies in the primate on the
bition by noradrenaline of nociceptive responses of iden- analgesic effects associated with intrathecal actions of opi-
tified dorsal horn neurones. Brain Res 1985: 334: 243-254. ates, a-adrenergic agonists and baclofen. Anesthesiology
55. Yaksh TL, and Tyce GM. Microinjection of morphine into 1981: 54: 451-467.
the periaqueductal gray evokes the release of serotonin 75. Yaksh TL. In vivo studies on spinal opiate receptor systems
from spinal cord. Brain Res 1979: 171: 176-181. mediating antinociception. Mu and delta receptor profiles
56. Takagi H, Shiomi H, Kuraishi Y, Fukui K, Ueda H. Pain in the primate. J Pharmacol E x p Ther 1983: 226: 303-316.
and the bulbospinal noradrenergic system: Pain-induced 76. Yaksh TL, Pogrel JW, Lee YW, Chaplan SR. Reversal of
increase in normetanephrine content in the spinal cord and nerve ligation-induced allodynia by spinal alpha-2 adreno-
its modification by morphine. Eur J Pharmacoll979: 54: 99- receptor agonists. J Pharmacol Exp Ther 1995: 272:207-214.
107. 77. Schmauss C, Yaksh TL. In vivo studies on spinal opiate
57. Yaksh TL. Direct evidence that spinal serotonin and nor- receptor systems mediating antinociception. 11. Pharmaco-
adrenaline terminals mediate the spinal antinociceptive ef- logical profiles suggesting a differential association of mu,
fects of morphine in the periaqueductal gray. Brain Res delta and kappa receptors with visceral chemical and cu-
1979: 160: 180-185. taneous thermal stimuli in the rat. J Pharmacol E x p Ther
58. Camarata PJ, Yaksh TL. Characterization of the spinal adre- 1984: 228 1-12.
nergic receptors mediating the spinal effects produced by 78. Yaksh TL, Noueihed R. The physiology and pharmacology
the microinjection of morphine into the periaqueductal of spinal opiates. Ann Rev Pharmacol Toxic01 1985: 25 433-
gray. Brain Res 1985: 336:133-142. 462.
59. Renno Wh4, Mullett MA, Beitz AJ. Systemic morphine re- 79. Malmberg AB, and Yaksh TL. Isobolographic and dose-re-
duces GABA release in the lateral but not the medial por- sponse analyses of the interaction between intrathecal mu
tion of the midbrain periaqueductal gray of the rat. Brain and delta agonists: Effects of naltrindole and its benzofuran
Res 1992: 594 221-232. analog (NTB). ] Pharmacol Exp Ther 1992: 263: 264-275.

108
80. Stewart PE, Holper EM, Hammond DL. Delta antagonist 99. Zieglghberger W, Bayerl H. The mechanism of inhibition
and kappa agonist activity of Naltriben: evidence for differ- of neuronal activity by opiates in the spinal cord of cat.
ential kappa interaction with the delta-1 and delta-2 opioid Brain Res 1976 115: 111-128.
receptor subtypes. Life Sci 1994: 55: 79-84. 100. Cahill CM, White TD, Sawynok J. Spinal opioid receptors
81. Yaksh TL, Gross KE, Li CH. Studies on the intrathecal effect and adenosine release: neurochemical and behavioral char-
of P-endorphin in primate. Brain Res 1982 241: 261-269. acterization of opioid subtypes. J Pharmacol Exp Ther 1995:
82. Calvillo 0, Henry JL, Neuman RS.Effects of morphine and 275: 8493.
naloxone on dorsal horn neurones in the cat. Can J Physiol 101. Sosnowski M, Stevens CW and Yaksh TL. Assessment of
Pharmacol 1974: 52: 1207-1211. the role of AI/A2 adenosine receptors mediating the purine
83. Fleetwood-Walker SM, Mitchell R, Hope PJ, El-Yassir N, antinociception, motor and autonomic function in the rat
Molony V, Bladon CM. The involvement of neurokinin re- spinal cord. j Pharmacol Exp Ther 1989: 250 915-922.
ceptor subtypes in somatosensory processing in the super- 102. Vasko MR, Cartwright S, On0 H. Adenosine agonists do
ficial dorsal horn of the cat. Brain Res 1990: 519: 169-182. not inhibit the K+ stimulated release of substance P from
84. Le Bars D, Menetrey D, Conseiller C, Besson JM. Depres- rat spinal cord slices. Society Neurosci Abstr 1986 12: 799.
sive effects of morphine upon lamina V cells activities in 103. Gissen AJ, Gugino LD, Datta S, Miller J, Covino BG. Effects
the dorsal horn of the spinal cat. Brain Res 1975: 98: 261- of fentanyl and sufentanil on peripheral mammalian
277. nerves. Anesth Analg 1987 66: 1272-1276.
85. Yaksh TL. Inhibition of etorphine of the discharge of dorsal 104. Stein C, Millan MJ, Shippenberg TS, Peter K, Herz A
horn neurons: Effects upon the neuronal response to the Peripheral opioid receptors mediating antinociception in
both high- and low-threshold sensory input in the decere- inflammation. Evidence for involvement of mu, delta and
brate spinal cat. Exp Neuroll978: 60: 23-40. kappa receptors. J Pharmacol Exp Ther 1989 248: 1269-1275.
86. Glaum SR, Miller RJ, Hammond DL. Inhibitory actions of 105. Nagasaka H, Yaksh TL. Peripheral and spinal actions of
delta 1-,delta 2-, and mu-opioid receptor agonists on excit- opioids in the blockade of the autonomic response evoked
atory transmission in lamina I1 neurons of adult rat spinal by compression of the inflamed knee joint. Anesthesiology
cord. J Neurosci 1994: 14: 49654971, 1995, in press.
87. Gouarderes C, Cros J, Quirion R. Autoradiographic localiz- 106. Stein C S Peripheral mechanism of opioid analgesia. Anesth
ation of p, 8 and K opioid receptor binding sites in rat and Analg 1993: 76:182-191.
guinea pig spinal cord. Neuropeptides 1985: 5: 331-342. 107. Laduron PM. AX OM^ transport of opiate receptors in cap-
88. Morris BJ, Herz A. Distinct distribution of opioid receptor saicin-sensitive neurones. Brain Res 19M 294: 157-160.
types in rat lumbar spinal cord. Naunyn-Schmiedeberg's Arch 108. Yaksh TL.:Substance P release from knee joint afferent ter-
Pharmacol 1987 336: 240-243. minals: modulation by opioids. Brain Res 1988: 458 319-
89. Bouchenafa 0, Livingston A. Distribution of immunoreac- 324.
tive metenkephalin and autoradiographic [SHIDAGO and 109. Russell NJW,Schaible H-G, Schmidt RF. Opiates inhibit the
[3H]DPDPEin the spinal cord of sheep. Advances Biosciences discharges of fine afferent units from inflamed knee joint
1989: 75: 289. of the cat. Neurosci Lett 1987: 7 6 107-112.
90. Stevens CW, Lacey CB, Miller KE, Elde RP, Seybold VS. 110. Andreev N, Urban L, Dray A. Opioids suppress spon-
Biochemical characterization and regional quantification of taneous activity of polymodal nociceptors in rat paw skin
mu, delta and kappa opioid binding sites in rat spinal cord. induced by ultraviolet irradiation. Neuroscience 1994 58:
Brain Res 1991: 550 77-85. 793-798.
91. Besse D, Lombard M-C, Zajac J-M, Roques BP, Besson J-M. 111. Tsou K. Antagonism of morphine analgesia by the intracer-
Pre- and postsynaptic location of mu, delta and kappa ebral microinjection of nalorphine. Acta Physiologica Sinica
opioid receptors in the superficial layers of the dorsal horn
1963 2 6 332-337.
of the rat spinal cord. In: International Narcotics Research
112. Vigouret J, Tesechemacher H, Albus K, Herz A. Differen-
Conference (INRC). Alan R. Liss, Inc. 1990 182.
tiation between spinal and supraspinal sites of action of
92. Stevens CW, Seybold VS. Changes of opioid binding den-
morphine when inhibiting the hind limb flexor reflex in
sity in the rat spinal cord following unilateral dorsal rhi-
rabbits. Neuropharmacology 1973 1 2 111-121.
zotomy. Brain Res 1995: 687 53-62.
113. Yaksh TL, Rudy TA. Narcotic analgesics: CNS sites and
93. Fields HL, Emson PC, Leigh BK, Gilbert W, Iverses LL.
Multiple opiate receptor sites on primary afferent fibres. mechanisms of action as revealed by intracerebral injection
Nature 1980: 284. 351-353. techniques. Pain 1978: 4: 299-359.
94. Yaksh TL, Jessell TM, Gamse R, Mudge AW, Leeman SE. 114. Yeung JC, Rudy TA. Multiplicative interaction between
Intrathecal morphine inhibits substance P release from narcotic agonism expressed at spinal and supraspinal sites
mammalian spinal cord in vivo. Nature 1980: 286: 155-156. of antinociceptive action as revealed by concurrent intra-
95. Go VL, Yaksh TL. Release of substance P from the cat spinal thecal and intracerebroventriuclar injections of morphine.
cord. j Physiol Lond 1987: 391: 141-167. Pharmacol Exp Ther 1980 215: 633-642.
96. Hirota N, Kuraishi Y, Hino Y, Sato Y, Satoh M, Takagi H. 115. Roerig SC,Fujimoto JM. Multiplicative interaction between
Met-enkephalin and morphine but not dynorphin inhibit intrathecally and intracerebroventricularlyadministered
the noxious stimuli-induced release of substance P from mu opioid agonists but limited interactions between delta
rabbit dorsal horn in situ. Neuropharmacoll985:24: 567-570. and kappa agonists for antinociception in mice. J Pharmacol
97. Aimone LD, Yaksh TL. Opioid modulation of capsaicin- Exp Ther 1989: 249: 762-768.
evoked release of substance P from rat spinal cord in vivo. 116. Tallarida RJ, Porreca F, Cowan A. Statistical analysis of
Peptides 1989: 1 0 1127-1131. drug-drug and site-site interactions with isobolograms. Life
98. Bourgoin S, Benoliel JJ, Collin E, Mauborgne A, Pohl M, Sci 1989: 45: 947-961.
Hamon M, Cesselin F. Opioidergic control of the spinal re- 117. Bodnar R, Paul D, Pastemak GW. Synergistic analgesic in-
lease of neuropeptides. Possible significance for the anal- teractions between the periaqueductal gray and the locus
gesic effects of opioids. Fund Clin Pharmacol 1994 8: 307- coeruleus. Brain Res 1991: 5%. 224-230.
321. 118. Xia LY, Huang KH, Rosenfeld Jl? Behavioral and trigeminal

109
T. L. Yaksh
neuronal effects of rat brainstem-nanoinjected opiates. Phy- 135. Bromage PR, Camporesi E, Leslie J. Epidural narcotics in
siol Behav 1992: 52 65-73. volunteers: sensitivity to pain and to carbon dioxide. Pain
119. Yaksh TL, Jage J, and Takano Y. Pharmacokinetics and 1980: 9:145-160.
pharmacodynamics of medullar agents. The spinal actions 136. Rawal N, Mollefors K, Axelsson K, Lingardh G, Widman
of a2-adrenergic agonists as analgesics. In: Aitkenhead AR, 8. An experimental study of urodynamic effects of epidural
Benad G, Brown BR, Cousins MC, Jones JG, Strunin L, morphine and of naloxone reversal. Anesth Analg 1983 62:
Thomson D, Van Aken H, eds. Bailliere's Clinical Anaesthe- 641-647.
siology, vol. 7, no. 3, London: BailliereTindall, 1993:597-614. 137. Onofrio BM, Yaksh TL. Intrathecal delta-receptor ligand
120. Hua X-Y, Boublik JH,Spicer MA, Rivier JE,Brown MR, and produces analgesia in man. Lancef 1983: 1: 386-1387.
Yaksh TL. The antinociceptive effects of spinally adminis- 138. Stein C, Cornisel K, Haimerl E, Yassouridis A, Lehrberger
tered neuropeptide Y in the rat: Systematicstudies on struc- K, Herz A, Peter K. Analgesic effect of intra-articular mor-
ture-activity relationship. \ Pharmacol E x p Ther 1991: 258: phine after arthroscopic knee surgery. N Engl \ Med 1991:
243-258 325: 1123-1126.
121. Wilson PR, Yaksh TL. Baclofen is antinociceptive in the 139. Khoury GF, Chen ACN, Garland DE, Stein C. Intra-articu-
spinal intrathecal space of animals. Eur J Pharmacol 1978: lar morphine, bupivacaine, and morphine/bupivacaine for
51: 323-330. pain control after knee videoarthroscopy. Anesthesiology
122. Sosnowski M, and Yaksh TL. The role of spinal and brain- 1992 7 263-266.
stem adenosine receptors in the modulation of the volume- 140. Haynes TK, Appadurai IR, Power I, Rosen M, Grant A.
evoked micturition reflex in the unanesthetized rat. Brain Intra-articular morphine and bupivacaine analgesia after
Res 1990: 515:207-213. arthroscopic knee surgery. Anaesthesia 1994: 49:54-56.
123. Yaksh TL. Tolerance: Factors involved in changes in the 141. Handwerker HO, Reeh PW Pain and inflammation, Pro-
dose-effect relationship with chronic drug exposure. In: ceedings of the VIth World Congress on Pain. Bond MR,
Basbaum AI, Besson J-M, eds. Towards a New Pharmaco- Harlton JE, Woolf CJ, eds. Elsevier Science Publishers BV
therapy of Pain, Dahlem Workshop Reports, John Wiley & 1991: 59-70.
Sons Ltd. 1991: 157-179. 142. Schaible H-G, Grubb BD. Afferent and spinal mechanism
124. Lobato RD, Madrid JL, Fatela LV, Sarabia R, Rivas JJ, Goza- of joint pain. Pain 1993: 55:5-54.
lo A. Intraventricular morphine for intractable cancer pain: 143. Bennett GJ, Xie YK. A peripheral mononeuropathy in rat
rationale, methods, clinical results. Acta Anaesthesiol Scand, that produces disorders of pain sensation like those seen in
Suppl 1987 85:68-74. man. Pain 1988: 33:87-107.
125. Lazorthes Y. Intracerebroventricular administration of mor- 144. Kim SH, Chung JM. An experimental model for peripheral
phine for control of irreducible cancer pain. Ann N Y Acad neuropathy produced by segmental spinal nerve ligation
Sci 1988: 531: 123-132. in the rat. Pain 1992: 50: 355-363.
126. Lenzi A, Galli G, Gandolfini M, Marini G. Intraventricular 145. Yamamoto T, and Yaksh TL. Spinal pharmacology of ther-
morphine in paraneoplastic painful syndrome of the cervi- mal hyperesthesia induced by incomplete ligation of sciatic
cofacial region: experience in thirty-eight cases. Neurosur- nerve. I. Opioid and nonopioid receptors. Anesthesiology
gery 1985: 1 7 6-11. 1991: 75 817-826.
127. Lazorthes Y, Verdie JC, Caute B, Maranhao R, Tafani M. 146. Lee Y-W, Chaplan S, Yaksh TL. Systemic and supraspinal,
Intracerebroventricular morphinotherapy for control of but not spinal opiates suppress allodynia in a rat neuro-
chronic cancer pain. Prog Brain Res 1988: 77 395405. pathic pain model. Neurosci Lett 1995: 186:111-114.
128. Sandouk P, Serrie A, Urtizberea M, Debray M, Got P, 147. Arner S, Meyerson BA. Lack of analgesic effect of opioids
Scherrmann JM. Morphine pharmacokinetics and pain as- on neuropathic and idiopathic forms of pain. Pain 1988: 33:
sessment after intracerebroventricular administration in 11-23.
patients with terminal cancer. Clin Pharmacol Ther 1991: 49: 148. Kenner DJ. Pain forum. Part 2. Neuropathic pain. Austral
442-448. Fam Physician 1994 23:1279-1283.
129. Tafani JA, Lazorthes Y, Danet 8, Verdie JC, Esquerre JF, 149. Cherny NI, Thaler HT, Friedlander-Klar H, Lapin J, Foley
Simon J, Guiraud R. Human brain and spinal cord scan KM,Houde R, Portenoy RK. Opioid responsiveness of can-
after intracerebroventricular administration of iodine-123 cer pain syndromes caused by neuropathic or nociceptive
morphine. International Journal of Radiation Applications mechanisms: a combined analysis of controlled, single-
and Instrumentation. Part B. Nucl Med Biol 1989: 16: 505- dose studies. Neurology 1994: 44: 857-861.
509. 150. Mjanger E, and Yaksh TL. Characteristics of dose-depend-
130. Herz A, Tescehmacher H. Activities and sites of antino- ent antagonism by iJ-funaltrexamine of the antinociceptive
ciceptive action of morphine-like analgesics and kinetics effects of intrathecal mu agonists. \ Pharmacol E x p Ther
of redistribution following intravenous, intracerebral and 1991: 258: 544-550.
intraventricular application. A d v Drug Res 1971: 6:79-119. 151. Adams JU, Paronis, CA and Holtzman SG. Assessment of
131. Foley KM, Kourides IA,Inturrisi CE, Kaiko RF, Zaroulis relative intrinsic activity of mu-opioid analgesics in vivo
CG, Posner JB, Houde RW, Li CH. Beta-endorphin: anal- by using beta-funaltrexamine. I Pharmacol E x p Ther 1990:
gesic and hormonal effects in humans. Proc Nutl Acud Sci 255:1027-1032.
USA: 1979: 76: 5377-5381. 152. Saeki S, Yaksh TL. Suppression of nociceptive responses by
132. Yaksh TL. Spinal opiates: a review of their effect on spinal spinal mu opioid agonists: Effects of stimulus intensity and
function with emphasis on pain processing. Acta Anaes- agonist efficacy. Anesth Analg 1993: 7: 265274.
thesiol Scand 1987 31: 25-37. 153. Saeki S, and Yaksh TL. Suppression by spinal alpha-2
133. Behar M, Magora F, Olshwang D, Davidson JT. Epidural agonists of motor and autonomic responses evoked by low-
morphine in treatment of pain. Lancet 1979: 1: 527-529. and high-intensity thermal stimuli. J Pharmacol E x p Ther
134. Magora F, Olshwang D, Eimerl D, Shorr J, Katzenelson R, 1992: 260: 795-802.
Cotev S, Davidson JT. Observations on extradural mor- 154. Dirig DM, Yaksh TL. Differential right shifts in the dose
phine analgesia in various pain conditions. Br J Anaesth response curves for intrathecal morphine and sufentanil as
1980: 52: 247-252. a function of stimulus intensity. Pain 1995: 62: 321-328.

110
155. Smith DJ, Perrotti JM, Crisp T, Cabral ME, Long JT, Scalzitti sufentanil versus intravenous sufentanil or epidural fen-
JM. The mu opiate receptor is responsible for descending tanyl analgesia after major abdominal surgery. Anesth
pain inhibition originating in the periaqueductal gray re- Analg 1993 76: 1243-250.
gion of the rat brain. Eur J Pharmacol 1988: 156:47-54. 171. Brose WG, Tanelian DL, Brodsky JB, Mark JB, Cousins MJ.
156. Sanchez-Blazquez P, Garzon J. Evaluation of delta receptor CSF and blood pharmakinetics of hydromorphone and
mediation of supraspinal opioid analgesia by in vivo pro- morphine following lumbar epidural administration. Pain
tection against the beta-funaltrexamine antagonist effect. 1991: 45 11-15.
Eur J Pharmacoll989: 159:9-23. 172. Chauvin M, Salbaing J, Perrin D, Levron JC, Viars P. Clin-
157. Drower EJ, Stapelfeld A, Rafferty MF, de Costa BR, Rice ical assessment and plasma pharmacokinetics associated
KC, Hammond DL. Selective antagonism by naltrindole of with intramuscular or extradural alfentanil. Brit J Anaesth
the antinociceptive effects of the delta opioid agonist cyclic 1985: 57 886-891.
[D-penicillamine2-D-penicillamine51enkephalin in the rat. 173. Camann WR, Loferski BL, Fanciullo GJ, Stone ML, Datta
J Pharmacol Exp Ther 1991: 259725-731. S. Does epidural administration of butorphanol offer any
158. Yaksh TL, Noueihed RY, Durant PAC. Studies of the phar- clinical advantage over the intravenous route? A double-
macology and pathology of intrathecally administered 4- blind, placebo-controlled trial. Anesthesiology 1992 76:216-
anilinopiperidine analogues and morphine in rat and cat. 220.
Anesthesiology 1986: 64: 54-66. 174. Moulin DE, Max MB, Kaiko RF, Inturrisi CE, Maggard J,
159. Oliveras J-L, Maixner W, Dubner R, Bushel1 MC, Duncan Yaksh TL., Foley KM. The analgesic efficacy of intrathecal
G, Thomas DA, Bates R. Dorsal horn opiate administration D-Ala2-DLeu5-enkephalin in cancer patients with chronic
attenuates the perceived intensity of noxious heat stimula- pain. Pain 1985: 23: 213-221.
tion in behaving monkey. Brain Res 1986: 371:368-371. 175. Oyama T, Fukushi S, Jin T. Epidural P-endorphin in treat-
160. Taiwo YO, Coderre TJ, Levine JD. The contribution of train- ment of pain. Can Anaesth Soc J 1982: 29: 24-26.
ing to sensitivity in the nociceptive paw-withdrawal test. 176. Torda TA, Pybus DA. Comparison of four narcotic anal-
Brain Res 1989: 487 148-151. gesics for extradural analgesia. Br J Anaesth 1982: 54: 291-
161. Ness TJ, Gebhart GF. Differential effects of morphine and 295.
clonidine on visceral and cutaneous spinal nociceptive 177.Evron S, Samueloff A, Simon A, Drenger B, Magora F. Uri-
transmission in the rat. J Neurophysiol 1989: 6 2 220-230. n a y function during epidural analgesia with methadone
162. Harada Y, Nishioka K, Kitahata LM, Nakatani K, Collins and morphine in postcesarean section patients. Pain 1985:
JG. Contrasting actions of intrathecal U50,488H, morphine, 23:135-144.
or [D-Pen2, D-Pen51 enkephalin or intravenous U50,488H 178. Xu XJ, Hao JX, Aldskogius H, Seiger A,Wiesenfeld-Hallin
on the visceromotor response to colorectal distension in the Z. Chronic pain-related syndrome in rats after ischemic
rat. Anesthesiology 1995: 83 336-343. spinal cord lesion: a possible animal model for pain in pa-
163. Tang AH, Schoenfeld MJ. Comparison of subcutaneous and tients with spinal cord injury. Pain 1992 48:279-290.
spinal subarachnoid injections of morphine and naloxone 179. Puke MJ, Wiesenfeld-Hallin Z. The differential effects of
of analgesic tests in the rat. Eur J Pharmacol 1978: 52 215- morphine and the alpha-2-adrenoceptor agonists clonidine
223. and dexmedetomidine on the prevention and treatment of
164. Yamamoto T, and Yaksh TL. Comparison of the antino- experimental neuropathic pain. Anesth Analg 1993: 77 104-
ciceptive effects of pre- and posttreatment with intrathecal 109.
morphine and MK801, an NMDA antagonist, on the for- 180.Yamamoto T, Mizuguchi T. Time-dependent effects of oral
malin test in the rat. Anesthesiology 1992: 77: 757-763. morphine on autotomy following brachial nerve section in
165. Wiesenfeld-Hallin Z. The effects of intrathecal morphine the rat. Neurosci Lett 1992: 141: 166-168.
and naltrexone on autotomy in sciatic nerve sectioned rats. 181. Olshwang D, Shapiro A, Perlberg S, Magora F. The effect
Pain 1984: 18:267-278. of epidural morphine on ureteral colic and spasm of the
166. Yaksh TL. Behavioral and autonomic correlates of the tac- bladder. Pain 1984: 18:97-101.
tile evoked allodynia produced by spinal glycine inhi- 182. Bodnar RJ, Williams CL, Lee SJ, Pastemak GW. Role of mu
bition: Effects of modulatory receptor systems and excit- I-opiate receptors in supraspinal opiate analgesia: a micro-
atory amino acid antagonists. Pain 1989: 37 111-123. injection study. Brain Res 1988: 447 25-34.
167. Van den Abeele G, Camu F, Clinical evaluation of the anal- 183. Kuraishi Y, Hirota N, Satoh M, Takagi H. Antinociceptive
gesic potency of lofentanil in postoperative pain. Acta effects of intrathecal opioids, noradrenaline and serotonin
Anaesthesiol Belg 1983 34: 4147. in rats: mechanical and thermal algesic tests. Brain Res 1985:
168. Simpson KH, Madej TH, McDowell JM, MacDonald R, 326: 168-171.
Lyons G. Comparison of extradural buprenorphine and
extradural morphine after caesarean section. Br J Anaesth
1988: 60:627-31. Address:
169. Welchew EA. The optimum concentration for epidural fen- Tony L. Yaksh, PhD
tanyl. A randomised, double-blind comparison with and Department of Anesthesiology
without 1200 000 adrenaline. Anaesthesia 1983: 338: 1037- University of California, San Diego
1041. 9500 Gilman Drive
170. Geller E, Chrubasik J, Graf R, Chrubasik S, Schulte-Mon- La Jolla, California 92093-0818
ting J.A. Randomized double-blind comparison of epidural USA

111

You might also like