25
Fundam Clin Pharrnacol(1995) 9 , 4 0 9 4 3 3 Q Elsevier, Paris 409 Review Opioid and anti-opioid peptides F Cesselin Inserm U 288 and Service de Biochimie Mkdicale, Faculte‘ de Mkdecine, Pitik-Salpt?trit?re, 91 Boulevard de 1’Hbpital. 75634 Paris cedex 13, France (Received 12 April 1995; accepted 9 June 1995) Summary - The numerous endogenous opioid peptides (pendorphin, enkephalins, dynorphins ...) and the exogenous opioids (such as morphine) exert their effects through the activation of receptors belonging to four main types: p, 6, rand E. Opioidergic neurones and opioid receptors are largely distributed centrally and peripherally. It is thus not surprising that opioids have numerous pharmacological effects and that endogenous opioids are thought to be involved in the physiological control of various functions, among which nociception is particularly emphasized. Some opioid targets may be components of homeostatic systems tending to reduce the effects of opioids. “Anti-opioid” properties have been attributed to various peptides, especially cholecystokinin (CCK). neuropeptide FF (NPFF) and melanocyte inhibiting factor (MIF)-related peptides. In addition, a particular place should be attributed. paradoxically, to opioid peptides themselves among the anti-opioid peptides. These peptides can oppose some of the acute effects of opioids. and a hyperactivation of anti-opioid peptidergic neurones due to the chronic administration of opioids may be involved in the development of opioid tolerance andor dependence. In fact, CCK, NPFF and the MIF family of peptides have complex properties and can act as opioid-like as well as anti-opioid peptides. Thus, “opioid modulating peptides” would be a better term to designate these peptides, which probably participate, together with the opioid systems, in multiple feed-back loops for the maintenance of homeostasis. “Opioid modulating peptides” have generally been shown to act through the activation of their own receptors. For example, CCK appears to exert its anti-opioid actions mainly through the activation of CCK-B receptors, whereas its opioid-like effects seem to result from the stimulation of CCK-A receptors. However, the partial agonistic properties at opioid receptors of some MIF-related peptides very likely contribute to their ability to modulate the effects of opioids. CCK- and NPFF-related drugs have potential therapeutic interest as adjuncts to opioids for alleviating pain andor for the treatment of opioid abuse. opioid peptides I morphine 1 anti-opioid peptides 1 cholecystokinin 1 neuropeptide FF I MIF I analgesia 1 opioid tolerance I opioid dependence INTRODUCTION In addition to pain inhibitory pathways, which include endogenous opioid systems, several lines of evidence suggest that there are “anti-analgesic’’ neuronal peptide systems which act in an opposite manner by blocking pain inhibition. As these systems can oppose not only the analgesic actions of opioids but also certain others of their pharmac- ological or physiological effects, they can be more generally referred as to “anti-opioid” systems. Besides reducing the acute effects of opioids, the triggering of these systems by opioids themselves could explain, at least partly, the development of tolerance and dependance to opioids. Currently available data, which will be summarized below, show that the three most studied “anti-opioid” pep- tides (cholecystokinin [CCK], neuropeptide FF [NPFF] and peptides of the melanocyte-inhibiting factor [MIF] family) in fact have complex proper- ties: they can act as anti-opioid as well as opioid- like peptides. This suggests that “opioid modulat- ing peptides” would be a better term to designate these peptides, and that the systems of multiple feed-back loops they actually form with the opioid systems take place within the framework of the classical and general concept of homeostasis. THE OPIOID PEPTIDES AND THEIR RECEPTORS Introduction To date, numerous endogenous opioid peptides (“endomorphins”) have been identified which are synthesized by neurones widely distributed in both

Opioid and anti-opioid peptides

Embed Size (px)

Citation preview

Page 1: Opioid and anti-opioid peptides

Fundam Clin Pharrnacol(1995) 9 , 4 0 9 4 3 3 Q Elsevier, Paris

409

Review

Opioid and anti-opioid peptides

F Cesselin

Inserm U 288 and Service de Biochimie Mkdicale, Faculte‘ de Mkdecine, Pitik-Salpt?trit?re, 91 Boulevard de 1’Hbpital. 75634 Paris cedex 13, France

(Received 12 April 1995; accepted 9 June 1995)

Summary - The numerous endogenous opioid peptides (pendorphin, enkephalins, dynorphins ...) and the exogenous opioids (such as morphine) exert their effects through the activation of receptors belonging to four main types: p, 6, rand E. Opioidergic neurones and opioid receptors are largely distributed centrally and peripherally. It is thus not surprising that opioids have numerous pharmacological effects and that endogenous opioids are thought to be involved in the physiological control of various functions, among which nociception is particularly emphasized. Some opioid targets may be components of homeostatic systems tending to reduce the effects of opioids. “Anti-opioid” properties have been attributed to various peptides, especially cholecystokinin (CCK). neuropeptide FF (NPFF) and melanocyte inhibiting factor (MIF)-related peptides. In addition, a particular place should be attributed. paradoxically, to opioid peptides themselves among the anti-opioid peptides. These peptides can oppose some of the acute effects of opioids. and a hyperactivation of anti-opioid peptidergic neurones due to the chronic administration of opioids may be involved in the development of opioid tolerance andor dependence. In fact, CCK, NPFF and the MIF family of peptides have complex properties and can act as opioid-like as well as anti-opioid peptides. Thus, “opioid modulating peptides” would be a better term to designate these peptides, which probably participate, together with the opioid systems, in multiple feed-back loops for the maintenance of homeostasis. “Opioid modulating peptides” have generally been shown to act through the activation of their own receptors. For example, CCK appears to exert its anti-opioid actions mainly through the activation of CCK-B receptors, whereas its opioid-like effects seem to result from the stimulation of CCK-A receptors. However, the partial agonistic properties at opioid receptors of some MIF-related peptides very likely contribute to their ability to modulate the effects of opioids. CCK- and NPFF-related drugs have potential therapeutic interest as adjuncts to opioids for alleviating pain andor for the treatment of opioid abuse.

opioid peptides I morphine 1 anti-opioid peptides 1 cholecystokinin 1 neuropeptide FF I MIF I analgesia 1 opioid tolerance I opioid dependence

INTRODUCTION

In addition to pain inhibitory pathways, which include endogenous opioid systems, several lines of evidence suggest that there are “anti-analgesic’’ neuronal peptide systems which act in an opposite manner by blocking pain inhibition. As these systems can oppose not only the analgesic actions of opioids but also certain others of their pharmac- ological or physiological effects, they can be more generally referred as to “anti-opioid” systems. Besides reducing the acute effects of opioids, the triggering of these systems by opioids themselves could explain, at least partly, the development of tolerance and dependance to opioids. Currently available data, which will be summarized below, show that the three most studied “anti-opioid” pep- tides (cholecystokinin [CCK], neuropeptide FF

[NPFF] and peptides of the melanocyte-inhibiting factor [MIF] family) in fact have complex proper- ties: they can act as anti-opioid as well as opioid- like peptides. This suggests that “opioid modulat- ing peptides” would be a better term to designate these peptides, and that the systems of multiple feed-back loops they actually form with the opioid systems take place within the framework of the classical and general concept of homeostasis.

THE OPIOID PEPTIDES AND THEIR RECEPTORS

Introduction

To date, numerous endogenous opioid peptides (“endomorphins”) have been identified which are synthesized by neurones widely distributed in both

Page 2: Opioid and anti-opioid peptides

410 F Cesselin

ACTH 5LPH

PC1 PROOPlOMEUNOCORTlN (POMC)

I COOH

I PC1, Pc2 a-MSH

0 i B-ENDORPHIN

11-13,

I metenkephalin sequence : Tyr-Gly-Gly-Phe-Met 1 0 2 7

8-ENDORPHIN-1-26 n26 Fig 1. Schematic representation of proopiomelanocortin and its main derivatives. In addition to the peptides represented in this figure, others also generated from POMC have been identified. In the anterior lobe of the pituitary, POMC is mainly cleaved to produce ACTH and PLPH by the proprotein convertase PC1. Due to the presence of proprotein convertase PC2, together with PCI, the main products of POMC in the intermediate lobe of the pituitary and the CNS are Pendorphin and a-MSH. Pendorphin (in its initial form, 1-31) and its shorter forms (1-27 and 1-26) lose their opioid activity when they are acetylated (not shown). Pendorphin-1-31 and P endorphin- 1-27 act as agonist and antagonist, respectively, at opioid receptors.

the central and the peripheral nervous systems. These peptides act at specific receptors, which belong to several types also largely distributed centrally and peripherally. It is thus not surprising that opioids have numerous pharmacological effects and that endogenous opioids are thought to be involved in the physiological control of various functions, among which nociception is particularly emphasized (see Millan, 1986).

At least twenty peptides form the group of endo- morphins. All of them have in common the amino acid sequence of their N-terminal moiety: Tyr- Gly-Gly-Phe-Met, or Tyr-Gly-Gly-Phe-Leu. These pentapeptide sequences are by themselves the two shortest endomorphins, ie met-enkephalin and leu- enkephalin, respectively. The endogenous opioid peptides originate from three distinct precursors: proopiomelanocortin (POMC), proenkephalin A (PA), and prodynorphin (PD), also called proen- kephalin B.

The proopiomelanocortin (POMC) family of peptides (fig 1)

The POMC gene is mainly expressed in the ante- rior pituitary and the central nervous system

(CNS). It can also be expressed in immune cells of inflamed tissue (Przewlocki et al, 1992). As indi- cated by its name, POMC is the precursor of opi- oid, melanotropic (MSH) and corticotropic (ACTH) peptides. Depending on the proprotein convertases (PC1, PC2 ...) which act upon it, POMC is cleaved mainly to form ACTH and p lipotropin (P-LPH) (in the anterior lobe of the pituitary), or in Pendorphin (a 31 amino acid pep- tide acting as an agonist at opioid receptors) and a-MSH (in the intermediate lobe and the CNS) (see Loh, 1992). pendorphin-1-31 can be further cleaved into pendorphin- 1-27 (&endorphin) and pendorphin-1-26 (fig 1). Endorphins can be acety- lated, a modification which leads to the loss of their opioid activity (see Loh, 1992). Such trans- formations probably have important physiological significance. For instance, pendorphin- 1-27 (in its non acetylated form) blocks the analgesic effect of pendorphin-1-3 1 with a potency five-fold higher than that of the most commonly used opioid antag- onist, naloxone (Hammonds et al, 1984). In other words, the same precursor, POMC, can generate peptides with opposite biological activities.

POMC is synthesized by neurones located in the hypothalamus (essentially in the arcuate nucleus)

Page 3: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 41 1

I I I 0 7

O I I 0 Proenkephalln A

II I I I COOH , Synenkephaiin , , PeptideF , Amidorphln

0 I metenkephalin sequence (TyrGly-Gly-Phe-Met)

leu-enkephalln sequence (Tyr-Gly-Gly-Phe-Leu) P

Adrenomhln

8 0 octapeptide

7 - heptapeptide

Fig 2. Schematic representation of proenkephalin A and its main derivatives. When all potential sites are cleaved (a frequent situation in the CNS), each precursor molecule yields 7 endomorphin molecules:4 of met-enkephalin, one of leu-enkephalin, one of an octapep- tide (8) and one of a heptapeptide (7). However, proenkephalin A can be processed differently to generate longer peptides (peptide E, adrenorphin etc).

and in the brain stem (principally the nucleus trac- tus solitarius). Hypothalamic POMC neurones have very diffuse projections in the brain, notably in hypothalamic and limbic areas. Several struc- tures (such as the thalamus, the periaqueductal gray [PAG] and the reticular formation) involved in nociception contain POMC terminals.

The proenkephalin A (PA) family of peptides (fig 2)

PA is mainly synthesized in the adrenal medulla (of some species) and the CNS. Like POMC, PA can also be expressed in immune cells (Przewlocki et al, 1992). Each molecule of PA contains seven sequences of endomorphins. There is also a vari- ability in the processing of PA, and a large number of different opioid peptides can derive from h s pre- cursor. In the CNS, the major ones are met- and leu-enkephalins, a heptapeptide: met-enkephalin- Arg6-Phe7, an octapeptide: met-enkephalin-Arg6- Gly7-Leu8, and a C-terminal-amidified 8 amino acid peptide (adrenorphin or metorphamide), the latter being present in substantial amounts mainly in the hypothalamus, striaturn and spinal cord (fig 2).

PA synthesizing neurones are numerous and largely distributed. They are found mainly in the striatum, cerebral cortex, olfactory tubercle, hippo- campus, septum, several hypothalamic and tha- lamic nuclei, PAG and the dorsal horn of the spi- nal cord. They are mainly interneurones, although some of them send long projections such as, for instance, those which form an enkephalinergic pathway between the posterior raphe nuclei and the dorsal horn of the spinal cord (see Khachatu- rian et al, 1985). Enkephalins are also present in peripheral neurones, notably in some primary afferent fibres which convey sensory information from the periphery to the dorsal horn (Pohl et al, 1994).

The prodynorphin (PD) family of peptides (fig 3)

The gene encoding of the third precursor of endo- morphins, PD, is mainly expressed in the CNS, the adrenals and the anterior pituitary. Each molecule of PD contains three leu-enkephalin sequences. Indeed, in some areas, like the substantia nigra, leu-enkephalin can actually arise from PD. How- ever, the main products of PD are in fact longer

Page 4: Opioid and anti-opioid peptides

412 F Cesselin

Prodynorphin (Proenkephalin B)

Dynorphln 1-32 a-Neo-endorphin

leuenkephalin sequence (Tyr-GlyGly-Phe-Leu) I Dynorphin B P-Nwndorphin 1-17 1-29

Fig 3. Schematic representation of prodynorphin (or proenkephalin B) and its main derivatives. Each prodynorphin molecule is poten- tially processed into 3 opioid peptide molecules. Processing enzymes can generate leu-enkephalin, but the main derivatives of prody- norphin are peptides of higher molecular weight (neo-endorphins. dynorphins A and B).

peptides, ie neoendorphins and dynorphins A and B (fig 3). In the CNS, the distribution of neurones synthesizing PD is as large as (and apparently very close to) that of PA neurones (see Khachaturian el al, 1985). The two peptide families even coexist in some neurones of the raphe nuclei and the dorsal horn of the spinal cord. However, in a given struc- ture, the distributions of PA and PD neurones are most often distinct. As enkephalins, PD derived peptides, especially dynorphins, are expressed in at least some sensory neurones (Basbaum et al, 1986; Sweetnam et al, 1986; Weihe et al, 1985).

The opioid receptors

Endomorphins and exogenous opioids exert their physiological and pharmacological effects, respec- tively, by interacting with opioid receptors belong- ing to four main types, which can be divided into subtypes: y Wll 4). a (a,, a,), K(K,, %, K3) and E. In addition, opioid receptors can be associated in complexes with mixed p, a andor ~properties, in which 'allosteric interactions can occur, such that the binding of a given opioid agonist (a selective for instance) can non-competitively affect the binding of another ligand (p selective for example) (see Holaday et al, 1991).

Although they were discovered more than 20 years ago, representatives of the first three types, with pharmacological properties close to those of the y,, a2 and K, receptors, respectively, have only recently been cloned (see Chen et al,

1993; Reisine and Bell, 1993; Uhl ef al, 1994). By contrast, E opioid receptors remain poorly known. Cloning of opioid receptors has confirmed that they belong to the superfamily of G protein-cou- pled receptors. Their effectors can be enzymes (like adenylate cyclase) or ion channels (K+, Ca2+).

The various opioid receptors can be distin- guished from each other owing to agonist and antagonist ligands, discovered andor synthesized during the last years, which exhibit a much higher affinity for a given type of receptor than for the others. When they are used at concentrations or doses such that their binding to the receptors with which they interact with a low affinity remains negligible, they act as selective ligands of a given opioid receptor type. However, except in a few cases, endogenous ligands, endomorphins, have a relatively weak selectivity for the various opioid receptors types. Opioid receptors are widely dis- tributed in the CNS and the peripheral nervous system (see Collin and Cesselin, 1993 and refs therein).

Possible functions of opioid systems

The wide distributions of the opioidergic neurones and opioid receptors suggest that the endomorphi- nergic systems play various physiological roles. Indeed, evidence has been presented for their implication in the control of respiratory, cardiovas- cular and gastrointestinal functions, pituitary hor-

Page 5: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 413

monal secretion, motor activity, and the etiology of some pathological states such as epilepsy and psychosis (see Olson et al, 1994).

One of the better established functional roles of the opioid systems is the control of nociception. Thus, morphine, or more generally opioids, could exert their analgesic effects by mimicking a phys- iological mechanism normally involving endo- morphins (see Millan, 1986). The neuroanatomi- cal substrate for this action exists, since the spinal and cerebral areas involved in nociception (dorsal horn of the spinal cord, PAG, some thalamic nuclei etc) all contain endomorphinergic neurones and opioid receptors. Opioid receptors are also present on the peripheral terminals of the primary afferent fibres which convey nociceptive mes- sages. These receptors could be activated by opi- oid peptides locally released from immunocytes (see above). Thus, although the details of their involvement in physiological or pathological situ- ations are far from being clearly elucidated, it is very likely that endogenous (and exogenous) opi- oids decrease the transmission of nociceptive mes- sages by acting at numerous strategical points on the pain pathways, from the periphery to the cere- bral cortex, including the spinal, bulbo-mesence- phalic and thalamic relays etc (see Cesselin, 1991 and refs therein).

ANTI-OPIOID PEPTIDES

Introduction

Endogenous and exogenous opioids affect the activity of the neurones which express opioid receptors, and numerous neuronal networks, involving various neurotransmitters, are implicated in the diverse physiological or pharmacological effects of opioids. Some opioid targets may partic- ipate in a homeostatic system tending to reduce the effects of opioids. Thus, opioids could activate neurones that would release neuropeptides with “anti-opioid” properties. Such peptides would be able to reduce some of the acute (particularly anti- nociceptive) effects of endogenous or exogenous opioids, while the blockade of their receptors would enhance the effects of opioids. According to this concept, the effects of opioids should be per- manently lowered due to the triggering of anti-opi- oid systems (fig 4).

It is well known that chronic administration of opioids can lead to the development of tolerance to some of their pharmacological effects, as well as to a state of dependence. Blocking the action of some anti-opioid peptides can reduce tolerance and

+ - EFFECTS

Fig 4. General concept of anti-opioid peptides. In this figure, as in the following ones, the term “effects” is used for desig- nating both pharmacological and physiological effects, what- ever the level at which they are appreciated (from the molecu- lar to the behavioural level). However, most of the data obtained relate to nociception and analgesia. Opioids, either exogenous or endogenous, activate neurones which release neuropeptides acting as endogenous “antagonists” of opioids, by reducing their pharmacological or physiological effects. These “anti-opioids” can exert an excitatory influence on opi- oidergic neurones, leading to an enhancement of the release of endogenous opioid peptides.

attenuate the withdrawal syndrome. These obser- vations have led to the idea that hyperactivation of anti-opioid peptidergic neurones due to the chronic administration of opioids may be responsible, at least partly, for the development of opioid toler- ance andor dependence.

“Anti-opioid” properties have been attributed to various peptides including TRH (Bhargava et al, 1983; Holaday et al, 1978; Tache et al, 1977), somatostatin (Terenius, 1976), calcitonin (Giusti et al, 1985), ACTH (Bertolini et al, 1979; Galina and Amit, 1985; Smock and Fields, 1981; Terenius, 1976; Terenius et al, 1975), a-MSH (Contreras and Takemori, 1984a, b; Sandman and Kastin, 1981), and arginine vasopressin (Xu et al, 1992), but the most studied anti-opioid peptides have been cholecystokinin (CCK), neuropeptide FF (NPFF) and melanocyte inhibiting factor (MIF) and its derivatives (table I). Excellent reviews have already been devoted to these peptides (see Baber et al, 1989; Galina and Kastin, 1986; Reed et al, 1994; Rothman 1992) and the purpose of the present review is to take stock of the present knowledge of the possible implication of these three peptides in opioid tolerance andor depen- dence.

In addition to CCK, NPFF and MIFs, a particu- lar place should be attributed, paradoxically, to opioid peptides themselves among the anti-opioid peptides. We will begin with some brief considera- tions on this issue before summarizing in greater detail the anti-opioid properties of CCK, NPFF and MIFs.

Page 6: Opioid and anti-opioid peptides

414 F Cesselin

Table I. Amino acid sequences of cholecystokinin-8 (CCK), neuropeptide FF (NPFF) and rnelanocyte inhibiting factor (M1F)-related peptides.

Cholecystokinin-8 (CCK): Asp-Tyr-Met-Gly-Trp-Met-Asp-Phe-NH, Neuropeptide FF (NPFF): Phe-Leu-Phe-Gln-Pro-Gln-Arg-Phe-Nh, Melanocyte inhibiting factor (MIF)-related peptides: Pro-Leu-Gly-NH, (MIF- 1 )

Tyr-Pro-Leu-Gly-NH, (Tyr-MIF- 1 ) Tyr-Pro-Trp-Gly-NH, (Tyr-W-MIF- 1 ) Tyr-Pro-Lys-Gly-NH, (Tyr-K-MIF- 1)

Opioid peptides as anti-opioids

Some opioids can reduce or block the effects of other opioids. Thus, as noted above, one of the derivatives of POMC, P-endorphin- 1-27, can inhibit the antinociceptive activity of pendorphin- 1-31 (Hammonds et al, 1984; Suh et al, 1988; Takemori and Portoghese, 1993). When both com- pounds are administered intracerebroventricularly (icv) to mice, the antagonistic activity of pendor- phin-1-27 appears to be mediated through 3, opi- oid receptors (Takemori and Portoghese, 1993). Alterations in the processing of POMC leading to an enhanced production of this antagonist have been proposed to be responsible, in part, for the development of tolerance to the analgesic effect of opioids after chronic administration of morphine (Bronstein et al, 1990).

In line with the possible existence of opioid receptor complexes (see above), or of interactions between various neuronal systems beyond the receptors, dynorphins and synthetic opioid ago- nists acting at K receptors have been shown to modulate the effects due to the stimulation of p or 3 receptors. In particular, endogenous and exoge- nous K agonists have been shown to block the effects of morphine and other opioid agonists on various parameters on which they are devoid of effects by themselves (Dickenson and Knox, 1987; Friedman et al, 1981; Fujimoto and Arts, 1990; Fujimoto and Holmes, 1990; Fujimoto et al, 1990; Hong et al, 1988; Janiri et al, 1988; Katoh et al, 1990; Lee and Smith, 1984; Miaskowski et al, 1992; Petrillo et al, 1984; Ramabadran, 1983; Ramarao et al, 1988; Schmauss and Herz, 1987; Sheldon et al, 1987, 1988, 1989; Tulunay et at, 1981; Wood et al, 1983). For instance, although icv administered dynorphin exerts no effect on its own, it significantly reduces morphine- and P- endorphin- 1-3 l-induced antinociception (Fried- man et al, 1981; Tulunay et al, 1981). In addition, electrophysiological studies revealed that the inhi- bition of spinal neurones by p agonists is less pro- nounced upon IC opioid receptor stimulation (Dick- enson and Knox, 1987). In our laboratory, we

demonstrated that the stimulation of K receptors, without any effect by itself, can modulate the influence of p receptor stimulation on the release of met-enkephalin-, CCK-, substance P- and CGRP-like materials from the rat spinal cord, in vivo as well as in vitro (Benoliel et al, 1991, 1994; Collin et al, 1992a, b, 1993, 1994).

Although the stimulation of opioid receptors (including those which have been cloned) is classi- cally known to trigger inhibitory effects (decrease in intracellular CAMP levels, hyperpolarization, reduction in the release of neurotransmitters etc), through the activation of pertussis toxin-sensitive G proteins (ie Gi/Go), increasing evidence sup- ports the existence of opioid receptors, with a high affinity for agonists, which are coupled to cholera toxin-sensitive G proteins (ie Gs) and whose stim- ulation leads to excitatory effects (see Crain and Shen, 1990; Gintzler and Xu, 1991; Lin and Car- penter, 1994; Wang and Gintzler, 1994). Further- more, opioid receptors in a given cellular type can interact simultaneously with both GdGo and Gs proteins (Cruciani et al, 1993). In other words, the net effects of opioids will depend on their relative excitatory influence through the activation of receptors (with high affinity for the agonists) cou- pled to Gs proteins and inhibitory influence medi- ated by receptors (with a lower affinity) coupled to GdGo proteins. Thus, Crain and Shen have shown that chronic administration of an opioid agonist leads to a reduction in its inhibitory influence together with an increase in its excitatory action (Crain and Shen, 1992; Shen and Crain, 1992). Such alterations have been proposed to be impli- cated in the development of the tolerance to the inhibitory effects of opioids and in the opioid with- drawal syndrome (Shen and Crain, 1994).

Cholecystokinin

Introduction Cholecystokinin was first identified as a hormone in the gastrointestinal system. It was subsequently shown to be present in the CNS (Vanderhaeghen e f al, 1975), essentially in its sulfated octapeptide

Page 7: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 415

Table 11. Main antagonists of CCK receptors.

Nonselective Proglumide

CCK-A Devazepide (MK-329, L-364,7 18) Benzotript

Lorglumide (CR-1409) L-365,03 1

CCK-B L-365,260 Cl-988 (PD-134,308) PD-135,158

form (CCK, Rehfeld, 1985; table I). At least two types of CCK receptors (A and B) have been iden- tified (Moran et al , 1986). Although CCK-A “alimentary” receptors are found primarily in the periphery, they are also present in some brain areas. CCK-B “brain” receptors have a widespread distribution in the CNS. Selective agonists and antagonists at these two types of CCK receptors, which have been recently cloned, are available (see refs in Crawley and Corwin, 1994) (table 11).

There is a striking overlap in the anatomical dis- tributions of CCK, opioid peptides and their respective receptors in the CNS (Stengaard-Peder- sen and Larsson, 1981; Zabin et al, 1983). Both CCK and opioids are localized within principal nociceptive centres, including layers I and I1 of the spinal cord, the PAG, and intralaminar nuclei of the thalamus. They coexist in some neurones of the thalamus, PAG, and allocortex (Gall et al, 1987).

CCK and opioids have been shown to exert opposite effects in several experimental paradigms and physiological functions. For instance, in rats, food intake increases following administration of opioid agonists, but decreases following adminis- tration of CCK (see Baile e t al, 1986, for a review). At the cellular level, CCK and opioids are also known to act frequently in an opposite way, as shown for instance on ion transport in the intestine (Kachur et al, 1980, 1991). Indeed, as summarized below, CCK frequently acts as an anti-opioid com- pound, especially in pain processing and control.

CCK as an anti-opioid peptide The results of numerous studies support this asser- tion. Thus, it has been shown that CCK antago- nizes butorphanol-induced feeding (Morley et al, 1983), morphine’s effects on locomotor activity (Schnur et al, 1986), and pendorphin-induced cat- alepsy (Itoh and Katsuura, 1981) and disruption of maternal behaviour (Felicio et al, 1991). Similarly, CCK reduces the hypothermic (Kapas et al, 1989) and hypotensive effects of opioids (Mei and Han,

1993), as well as the excitation of pyramidal neu- rones due to opioid receptor activation in the rat hippocampus (Miller and Lupica, 1994). In addi- tion, CCK blocks the increase in brain tryptophan hydroxylase activity elicited by morphine (Johannessen et al, 1989) and prevents &endor- phin from inhibiting brain and liver DNA synthe- sis in rat pups (Bartolome et af, 1994). However, as summarized below, the majority of experiments have been devoted to the anti-analgesic effects of CCK.

CCK as an anti-analgesic peptide

Effects of exogenous CCK

CCK reduces the antinociceptive effects of exogenous opioids Numerous reports have indicated that CCK antag- onizes opioid antinociceptive effects. Indeed, administered by peripheral or central (icv, intrathe- cal) routes, CCK can reduce morphine-, PL017- or ohmefentanyl- (two selective p agonists), and p endorphin-induced analgesia, as well as the mor- phine-induced depression of the nociceptive flex- ion reflex in rats (Faris et al, 1983; Han et al, 1985; Itoh et al, 1982; Li and Han, 1989; Suh et al, 1992; Wang et al, 1990; Wiesenfeld-Hallin and Duranti, 1987; Wiesenfeld-Hallin and Xu, 1993; Zhang et at, 1994). In the mouse tail flick test, CCK attenuates the analgesic effects of morphine (Barbaz et al, 1989). In addition, in the same spe- cies, CCK administered intrathecally antagonizes the inhibition of tail-flick by icv j3-endorphin (Tseng and Collins, 1992). Furthermore, the inhib- itory effect of morphine and DAGO on C fibre- evoked activity of rat spinal nociceptive neurones can be reduced by CCK (Kellstein et al, 1991; Magnuson et al, 1990; Suberg et al, 1985a, b).

Thus, from these data, it can be concluded that CCK has the pharmacological property of reducing the antinociceptive effects of exogenous opioids.

CCK reduces the antinociceptive effects of endogenous opioids Several observations suggest that exogenous CCK can also reduce the analgesia due to endogenous opioids. Indeed, CCK blocks the naloxone-sensitive analgesia in rats subjected to forepaw shocks or in rats conditioned to these shocks (Faris, et al, 1983). In addition, in the same species, CCK attenuates opioid analgesia induced by restraint stress (Dourish et al, 1988a). Finally, CCK antagonizes electroacu- puncture-induced analgesia, which is thought to be mediated through the release of endomorphins

Page 8: Opioid and anti-opioid peptides

41 6 F Cesselin

(Chen et al, 1994; Han and Terenius, 1982; Han et al, 1986), as well as the acupuncture-induced sup- pression of the electrical activity of thalamic pain- related neurones in the rat (Bian et al, 1993).

Studies with CCK receptor antagonists as tools for establishing the role of endogenous CCK CCK antagonists potentiate the antinociceptive effects of exogenous opioids A large amount of data strongly suggests that the above summarized effects of exogenous CCK in fact mimic the action of the endogenous peptide. Thus, active immunization against cholecystokinin or administration of an anti-CCK antiserum poten- tiate morphine and Pendorphin analgesia in rats (Faris et al, 1984; Itoh et al, 1985; see also Ding et al, 1986).

Proglumide, a non-selective CCK receptor antagonist, and the selective CCK-A antagonists, devazepide and lorglumide, enhance opioid anal- gesia in rodents and baboons (Bodnar et al, 1990; Dourish et al, 1988b, 1990b; Hendrie et af, 1989; Katsuura and Itoh, 1985; Kellstein and Mayer, 1990; Kellstein et al, 1991; Klein et al, 1992, Lavigne et al, 1992; Li and Han, 1989; O'Neill et al, 1989; Panerai et al, 1987; Poggioli et al, 1991; Rattray et al, 1988; Rovati et al, 1985; Suh and Tseng, 1990; Takeshige et al, 1991; Tang et al, 1984a; Watkins et al, 1984, 1985a, b, c; Zhou et al, 1993b). Proglumide also potentiates the inhibi- tory effect of intrathecal morphine on the electrical activity of spinal nociceptive neurones (Suberg et al, 1985b).

In addition to enhancing opioid antinociception in rodents and squirrel monkeys, the CCK-B antagonist, L-365,260, produces by itself an anal- gesic effect in the latter species (Dourish et al, 1990a; Lavigne et al , 1992; Maldonado et al, 1993; O'Neill et al, 1990; Ossipov et al, 1994; Zhou et ul , 1993b). In the rat, another CCK-B antagonist, CI-988, also enhances the analgesic effect of morphine on the flexor reflex, and exerts a naloxone-reversible antinociceptive effect per se (Wiesenfield-Hallin and Xu, 1993; Wiesenfield- Hallin e t al , 1990, 1991). Moreover, it was recently shown that treatment with antisense oligo- nucleotides targeting the cloned CCK-B receptor produces a 5-fold leftward displacement of the morphine analgesic dose-effect curve (Vanderah et al, 1994).

Interestingly, an induction of the expression of the proCCK gene has been observed in rat dorsal root ganglia after peripheral axotomy, a model of neuropathic pain, which induces autotomy behavi- our (Xu et al, 1993). Furthermore, this behaviour

can be inhibited by combined treatment with mor- phine and the CCK-B antagonist CI-988 (Xu et al, 1993). In axotomized rats, the depressive effect of CI-988 on the flexor reflex is more pronounced than in normal animals, and the (weak) inhibitory influence of morphine on this reflex is enhanced by CCK-B receptor blockade by this antagonist (Xu et al, 1994b). These observations led the authors to propose that an increased release of CCK from terminals of (lesioned) primary afferent fibres could antagonize the actions of opioid anal- gesics either released endogenously or applied exogenously, resulting in the development of a neuropathic pain syndrome and in the ineffective- ness of opioids. Similarly, in another model of neuropathic pain due to photochemically provoked spinal cord ischemia, CI-988 (but not CAM 1481, a selective CCK-A antagonist) relieves the lesion- induced allodynia-like symptoms (Xu et al , 1994a).

The importance of the pain state to reveal the possible anti-analgesic action of endogenous CCK is further demonstrated by the observations of Stanfa and Dickenson (1993). These authors showed that CCK-B receptor blockade by L- 365,260 or PD 135158 potentiates the inhibitory effect of intrathecal morphine or DAGO on the C- fibre-evoked responses of dorsal horn nociceptive neurones in normal rats (Stanfa and Dickenson, 1993; Sullivan et al, 1994) but not in animals with carrageenin-induced inflammation (Stanfa and Dickenson, 1993). Conversely, CCK attenuates the effects of morphine only in rats with carragee- nin inflammation (Stanfa and Dickenson, 1993). The effects of both CCK and its antagonists L- 365,260 and PD 135138 are therefore dependent on the inflammatory state of the animal, with the agonist and the antagonists being active in oppo- site situations. This led to the proposal that in nor- mal rats morphine may produce a maximal stimu- lation of CCK release, such that exogenous CCK is unable to further reduce the opioid analgesia, whereas in rats with inflammation there is a reduction in the spinal release of CCK, which could explain the enhanced potency of morphine in these animals (Stanfa and Dickenson, 1993; Stanfa et al, 1994).

More generally, the experimental context seems to be critical with regard to the anti-anal- gesic action of CCK. In line with this view, Lavigne et al. (1992) showed that the CCK-A and CCK-B receptor antagonists, devazepide and L-365,260, enhance morphine antinociception only in non-acclimated rats exposed to a novel environment.

Page 9: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 417

CCK antagonists enhance the antinociceptive effects of endogenous opioids In agreement with the idea that electroacupuncture not only triggers the release of endogenous opioids (Han and Terenius, 1982), but also that of CCK, which may limit the analgesic effects of endomor- phins, CCK-B receptor blockade by L-365,260 (intrathecal injection) has been shown to enhance electroacupuncture analgesia in the rat (Zhou et al, 1993a). Similarly, restraint stress-induced analge- sia can be increased by CCK antagonists (Dourish et al, 1988a). Furthermore, the antinociceptive responses induced by endogenous enkephalins (fully protected from their catabolism by a com- plete inhibitor of the enkephalin-catabolizing enzymes) are strongly potentiated by the CCK-B receptor antagonists L-365,260, CI-988 and RB 21 1 in both rats and mice (Maldonado et al, 1993; Valverde et al, 1994).

Whereas neural circuitry that produces analgesia can be activated by a variety of environmental events (conditioned analgesia), this is also true for anti-analgesia systems: safety signals could become inhibitors of analgesia, either conditioned (and due to the triggering of endogenous opioid systems) or induced by morphine (Wiertelak et al, 1992). CCK appears to be involved in the media- tion of conditioned anti-analgesia at the spinal cord level. Indeed, proglumide abolishes the abil- ity of a safety signal to block intrathecal morphine analgesia (Wiertelak et al, 1994).

Conclusion (fig 5 )

That the consequences of proglumide administra- tion may in fact be due to its direct interaction with opioid receptors has been suggested due to its ago- nistic properties at ? opioid sites (Rezvani et al, 1987). Similarly, benzotript and lorglumide have high affinity for both CCK and opioid receptors (Gaudreau et al, 1990). Such properties do not apply to the other CCK antagonists, including devazepide, L-365,260 and CI-988, which do not bind to opioid receptors (Gaudreau et al, 1990; Hughes et al, 1990). Thus, altogether, these data indicate that endogenous CCK tends to reduce the antinociceptive action of exogenous as well as endogenous opioids by acting primarily at CCK receptors.

It would seem that the anti-opioid effects of CCK arise as a result of the stimulation by the pep- tide of CCK-B rather than CCK-A receptors. Indeed, the rank order of potency of the CCK antagonists (L-365,03 1, devazepide and L- 365,260) in potentiating morphine analgesia in the

DELTA

OPlOlDS CCK

EFFECTS Fig 5. Schematic representation of CCWopioid interactions. Opioids, either exogenous or endogenous, could activate (through 6 opioid receptors) CCKergic systems, and CCK, through CCK-B receptors, could reduce the opioid effects. In turn, CCK, or exogenous agonists, through an action at CCK-A receptors, could increase the release of endogenous opioid pep- tides, and facilitate opioid-mediated effects.

rat correlates with their affinity for CCK-B recep- tors (Dourish et al , 1990a). For instance, L- 365,260 appears to be 5 to 20 times more potent than devazepide both in potentiating morphine analgesia in rodents and in blocking CCK-B recep- tor activation (Dourish et a1, 1990a). Moreover, in contrast to selective CCK-B antagonists, neither lorglumide nor devazepide, two CCK-A antago- nists, are able to potentiate antinociception medi- ated by endogenous enkephalins (Maldonado et al, 1993).

The data summarized above suggest that exoge- nous and endogenous opioids very likely activate CCKergic systems, leading to increased extracellu- lar levels of CCK which counteract the antinoci- ceptive actions of opioids. Surprisingly, the ability of opioids to actually enhance the release of CCK has been investigated in relatively few studies. However, it was shown, in both in vivo and in vitro experiments, that, although p opioid agonists reduce the release of CCK, morphine and 6 opioid agonists increase the outflow of this peptide from various central areas (Benoliel et al, 1991, 1992, 1994; Rattray and de Belleroche, 1987; Rodriguez and Sacristan, 1989; Tang et al, 1984a; Zhou et al, 1993b). Furthermore, in rat brain, the in vivo bind- ing of a selective tritiated CCK-B agonist (BC 264) is reduced by a selective 6 agonist or by pro- tecting enkephalins from degradation, strongly suggesting that endogenous enkephalins increase

Page 10: Opioid and anti-opioid peptides

418 F Cesselin

the extracellular levels of CCK through activation of 6 receptors (Ruiz-Gay0 et al, 1992).

Involvement of CCKergic systems in the development of opioid tolerance Chronic use of various types of drugs, including opioids, results in adaptive changes that lead to the development of a loss of responsiveness to the drug used (tolerance) andor of a state in which the drug is required for the maintenance of normal physiological functions (dependence). Neurobio- logical mechanisms of opioid tolerance and depen- dence are still poorly understood. Various changes (down- and/or up-regulation) affecting opioid receptors and the postreceptor mechanisms (func- tional uncoupling from G protein) have been sug- gested to be responsible for these phenomena (see Collin and Cesselin, 199 1, for a review). However, cessation of stimulation of opioid receptors, by disruption of opioid administration or administra- tion of an opioid antagonist, leads to a withdrawal syndrome with various behavioural manifestations. Moreover, as an animal becomes more tolerant to morphine, less naloxone is required to trigger the withdrawal syndrome (see Rothman, 1992 for review and discussion). This suggests that during a chronic opioid treatment, opioid receptors remain stimulated, stimulatable, and can be blocked, prop- erties which are not easily reconcilable with the tolerance mechanisms proposed above, particu- larly those implicating a desensitization (whatever its cause) of opioid receptors. Without excluding the role of adaptive changes at the level of the receptors, the hypothesis of an opioid-induced compensatory hyperactivity of anti-opioid systems could offer an additional explanation for the devel- opment of opioid tolerance and dependence.

A large amount of data supports the idea that CCKergic systems might play a major role in the development of opioid tolerance. Thus, in condi- tions where chronic morphine administration results in the development of tolerance to its anal- gesic effects, daily administration of proglumide or of selective CCK-A or CCK-B receptor antago- nists, during the whole period of morphine treat- ment, inhibits the development of tolerance to the drug (Dourish et al, 1988b, 1990a; Kellstein and Mayer, 1991; Panerai et al, 1987; Rovati et al, 1985; Watkins et at, 1984; Wiesenfeld Hallin and Xu, 1993; Xu et al, 1992). Similarly, the acute desensitization to morphine (induced in 14 hours by repeated morphine injections) can be reduced by proglumide (Tang et al, 1984a). Furthermore, although active immunization against CCK does not affect morphine tolerance (Faris et al, 1984),

icv or intrathecal injection of an anti-CCK anti- serum reverses tolerance to morphine in rats (Ding et al, 1986). Not surprisingly, since the same anti- opioid activity of CCK as that implicated in its anti-analgesic effects is assumed to be. involved, the CCK receptors more likely implicated in the prevention of morphine tolerance appear to be those of the B type (Dourish et al, 1990a; Wood- ruff and Hughes, 1991; Xu et al, 1992). Moreover, it was recently shown that the CCK-B receptor antagonist CI 988 is not only able to block the development of morphine tolerance but also able to reverse tolerance to the analgesic effects of mor- phine in the hot plate test: in rats rendered tolerant, a significant degree of antinociception is observed when CI 988 is administered together with mor- phine (Hoffmann and Wiesenfeld-Hallin, 1994).

Interestingly, the naloxone-precipitated with- drawal syndrome is not affected by daily CCK antagonist administration (Dourish et al, 1988b, 1990a; Panerai et al, 1987; Rovati et at, 1985; Xu et al, 1992). In addition, administration of CCK itself, or of a selective CCK-B receptor agonist, instead of naloxone, does not precipitate with- drawal in morphine-dependent animals (Maldo- nado et al, 1994; Pournaghash and Riley, 1991). Incidentally, these observations are consistent with the suggestion that independent mechanisms medi- ate opioid tolerance and naloxone-precipitated withdrawal (Ling et al, 1984; Shen and Crain, 1992; see also Collin and Cesselin, 1991 and refs therein).

As for the question of the possible direct acute effects of opioids on CCKergic systems (see above), data concerning the effects of long-term opioid receptor stimulation on CCKergic neuro- transmission are still scarce. Indeed, it remains to be unambiguously demonstrated that there is actu- ally a compensatory increase in the activity of some CCKergic neurones which would contribute to tolerance following chronic morphine adminis- tration. Thus, in rats, Pohl et a1 (1992) found that neither acute nor repeated morphine treatment affected the levels of CCK and of its encoding &A in various brain areas. With regard to acute morphine treatment, similar results have been reported by Rosen and Brodin (1989). At least in the cerebral cortex, CCK receptor binding charac- teristics also did not differ from control values in the phases of analgesia, tolerance, abstinence or withdrawal in rats which had been implanted with a miniosmotic pump to deliver morphine (Welin et at, 1994). However, contradictory data have been published. Thus, other groups have reported that morphine, especially following repeated adminis-

Page 11: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 419

tration, stimulates CCK biosynthesis in selective CNS regions, including the spinal cord, the brain stem and the hypothalamus (Ding and Bayer, 1993; Faris et al, 1986; Pu et a1, 1994a; Zhou et al, 1992). In addition, it was shown that cerebrospinal fluid levels of radioimmunoassayable CCK increased significantly in rats receiving morphine under conditions which induce tolerance (Watkins et al, 198.5~). The latter observations are congruent with the idea that repeated morphine administra- tion elicits a compensatory enhancement in the synthesis andor release of CCK that may underlie the development of morphine tolerance.

CCK as an opioid-like peptide If, as discussed above, opioids can modulate CCKergic systems, the reverse is also true: the stimulation of CCK receptors is able in turn to affect opioid systems. In other words, there could be reciprocal modulations between CCK and opi- oid systems. Numerous observations support the idea that CCK can activate opioid systems. For instance, Clarke et a1 (1992) concluded that the effects of proglumide on the sural-gastrocnemius medialis reflex in the rabbit, which is tonically inhibited by endogenous opioid peptides, can be explained through a stimulatory influence of CCK on the release of opioids. CCK was also found to activate an opioid mechanism in the guinea pig ileum (Garzon et al, 1987). Perhaps more impor- tantly, it was shown that subcutaneously or icv administered CCK or CCK receptor agonists exert naloxone-reversible antinociceptive effects in the mouse (Barbaz et al, 1986; Derrien et al, 1993; Hong and Takemori, 1989; Kubota et al, 1985; Noble et al, 1993; Zetler, 1980). In the rat, by the same routes of administration or by the intrathecal route, CCK proved to be an antinociceptive com- pound in the paw pressure and writhing tests, but not in the tail flick test (Gericke et al, 1990; Hill et al, 1987; Pittaway et al, 1987). However, when administered into the periaqueductal gray matter, CCK had an analgesic action in the tail flick test in this species (Jurna and Zetler, 1981). In addition, CCK has been reported to potentiate morphine antinociception (Pittaway and Hill, 1987). Finally, in apparent contradiction with the above-men- tioned hypothesis of a pronociceptive effect of CCK in animal models of neuropathic pain, Ros- sitch et a1 (1987) found that intraventricular administration of CCK attenuates scratching and biting due to dorsal root ganglionectomy in rats.

Variable results have been obtained with CCK receptor antagonists. Thus, whereas proglumide has been reported to have no effect on morphine

antinociception in the mouse’ hot plate test (Pitt- away and Hill, 1987), devazepide was shown to be able to antagonize morphine antinociception when administered icv to mice (Marrama et al, 1989, Poggioli et al, 1991). The latter effect was attrib- uted to alterations in opioid receptors. Indeed, a significant reduction in the number of [3H]nalox- one binding sites has been found in the brain stem of devazepide-treated mice (Marrama et al, 1989; Poggioli et al, 1991).

It should be emphasized that the analgesic effects of CCK generally require higher doses than those which reverse the antinociceptive effects of opioids. Since low doses of CCK are “physiologi- cal” whereas high doses are “pharmacological”, it is generally accepted that analgesia due to a large dose of CCK is a pharmacological phenomenon whereas the blockade of opioid analgesia by small doses of the peptide may be physiologically rele- vant (Baber et al, 1989). In fact, dose-dependent biphasic effects of CCK have been reported for many experimental models (see eg Dourish et al, 1988b; Hendrie et al, 1989; Noble et al, 1993; O‘Neil et al, 1989; Watkins et al, 1984, 1985a, b, and discussion in Baber et al, 1989), and the bell- shaped dose-response curves obtained with CCK- related compounds may also be explained through the differential effects of CCK-A and CCK-B receptor activation on endogenous opioid systems. In particular, Derrien et a1 (1993) showed that CCK-A but not CCK-B receptor stimulation induces endogenous opioid-dependent antinoci- ceptive effects. This observation and others (Noble et al, 1994; Ossipov et al, 1994) have led to the proposal that activation of CCK-A receptors enhances the release of endogenous enkephalins, while the reverse situation occurs upon stimulation of CCK-B receptors.

The recent observation that CCK receptor ago- nists can block the jumping behaviour precipitated in mice by naloxone following a single dose of morphine can also be interpreted by an activation of endogenous opioid systems through CCK recep- tor stimulation (Malinge et al, in press). Interest- ingly, it has been established that exogenous CCK can actually increase the spinal release of met- enkephalin (Cesselin et al, 1984), and that endoge- nous CCK appears to exert a tonic stimulatory influence on spinal enkephalinergic neurones through the activation of CCK-A receptors (Beno- lie1 et al, unpublished data).

Congruent with the hypothesis that CCK can modulate the opioid systems through activation of (at least) two receptor types with opposite conse- quences (CCK-A agonists having excitatory

Page 12: Opioid and anti-opioid peptides

420 F Cesselin

effects), it was recently shown that CCK can pre- vent tolerance to the antinociceptive effects of morphine (Rezayat et al, 1994), via a probable action at CCK-A receptors, since the unsulfated form of the octapeptide does not affect the devel- opment of tolerance. This result was not unex- pected, if one considers that CCK and opioid systems are mutually antagonistic. Indeed, the resulting effect of the stimulation of one of them always depends on the state of the other. Thus, activating endogenous opioid systems through CCK-A receptors would alter the balance between the two systems in disfavour of the anti-opioid action of CCK via CCK-B receptors, resulting in less tolerance to the opioid effects. Evidence for the existence of subtypes of CCK-B receptors has been provided (Derrien et al, 1994; Durieux et al, 1992; Jagerschmidt et al, 1994; Kaufmann et al, 1994), but their respective (possibly opposite) roles in the modulation of opioid systems remain to be investigated.

Sites and mechanisms of CCWopioid interactions The localization of CCK neurones and receptors in the sensory cortex, PAG, thalamus, limbic struc- tures and spinal cord (Beinfeld et al, 1981; Hill et al, 1988) (where opioid systems are also present [see above] and where CCK and endomorphins can even coexist in the same neurones [Gall et al, 1987; Liu et al, 1994]), suggests that the phannac- ological and physiological effects of CCK in noci- ception can be due, at least in part, to its influence on both sensory and affective components of pain.

The results of numerous studies favour the idea that the site of interaction between CCK and opi- oids may lie in the spinal cord. At this level, the anti-opioid effect of CCK could be selective for p- and K-, but not 6, opioid analgesia (Magnuson et al, 1990; Wang et al, 1990). However, Wiertelak et a1 (1994) recently showed that conditioned anti- analgesia, which is very likely mediated at the spi- nal level by CCKergic systems (see above), abol- ishes conditioned analgesia triggered through not only p but also 6 opioid receptor activation.

If the dorsal horn of the spinal cord is very likeky one of the major sites of CCWopioid inter- actions in rodents, other areas involved in nocicep- tion must also be considered. Thus, CCK adminis- tered into the nucleus accumbens antagonizes the analgesic effect of systemic morphine in rats. Con- versely, morphine analgesia is markedly poten- tiated by intra-nucleus accumbens infusion of a CCK-B receptor antagonist (Pu et al, 1994b). Also in the PAG, infusion of CCK blocks morphine- and electroacupuncture-induced analgesia in the

rat (Chen et al, 1994; Li and Han, 1989), while local infusion of the non-selective CCK antagonist proglumide enhances the antinociceptive action of morphine (Watkins et al, 1985b).

The hypothesis of CCWopioid interactions in the context of a feedback loop composed of two reciprocal antagonist systems (fig 5 ) does not exclude that these interactions may involve events beyond receptor stimulation or blockade, provided both opioid and CCK receptors, which belong to the superfamily of G protein-coupled receptors, are located on the same targets. Such a colocalization occurs notably in the spinal cord of several spe- cies, at least on the terminals of primary afferent fibres (Ghilardi et al, 1992). In addition, CCK actually appeared to be able to affect opioid bind- ing to brain membranes. For instance, by acting at its own receptors, CCK reduces [3H]etorphine binding to opioid receptors (Wang et al, 1989), and more specifically to p and K, but not 6, bind- ing sites in the rat brain (Wang and Han, 1990). In vitro experiments with hippocampal slices suggest that an allosteric interaction occurs between the opioid and CCK-B receptors, in which the confor- mational state of the opioid receptor could be mod- ified (Miller and Lupica, 1994). It has even been suggested that CCK and opioid receptors in fact belong to the same complex (Hudson et a1, 1992).

Interaction between opioid and CCK receptors might also involve some intermediate component of a signal transduction cascade that is coupled to different effector mechanisms. For instance CCK and opioid receptors might act through the same G protein(s). It was suggested that CCK and opioids decrease and increase, respectively, K+ conduc- tance, via separate K+ channels located on the same neurone (Dourish, 1991). Others have pro- posed that, while opioid ligands reduce free intra- cellular calcium by blocking voltage-dependent Ca2+ influx, the anti-opioid effect of CCK could be associated with the mobilization of Ca2+ from intracellular stores (Wang et al, 1992).

The potential therapeutic use of CCK-related drugs for pain treatment Systemic proglumide has been found to potentiate morphine-induced analgesia in humans (Lavigne et al, 1989; Price et al, 1985). In line with the experimental findings showing that the effects of devazepide and L-365,260 on morphine antinoci- ception depend on the rat’s response to a new environment or to the stress induced by an unac- customed experimental paradigm (Lavigne et al, 1992), proglumide appeared to enhance the anal- gesic effect of morphine for the postoperative

Page 13: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 42 1

period only in non-sedated patients (Lavigne et al, 1989), but not in sedated patients using a con- trolled analgesia paradigm to manage their postop- erative pain (Lehmann et al, 1989). With regard to the possible long-term clinical use of CCK recep- tor antagonists as an adjunct to opioid therapy, it is important to note that the CCK-A receptor antago- nist devazepide does not enhance morphine- induced respiratory depression or constipation in the squirrel monkey (Dourish et al, 1990b). Fur- thermore, this compound has been reported to block the acquisition of morphine conditioned place preference in rats (Higgins et al, 1992). By contrast, the CCK-B antagonist L-365,260, in addition to potentiating the analgesic properties of morphine and reducing tolerance to the alkaloid, was shown to enhance the rewarding properties of the drug in the place preference test (Higgins et ul, 1992). This was considered as an important limita- tion in the possible therapeutic use of the associa- tion of CCK-B antagonist and exogenous opioids. Fortunately, this fear was allayed by subsequent studies which demonstrated that the potentiation of morphine-induced behaviour by L-365,260 does not extend to other paradigms. Indeed, neither this compound nor other CCK-A or CCK-B antago- nists potentiate the subjective effects of opioids in a morphine drug discrimination paradigm and a model of heroin self-administration (Higgins et ~1,1994; Jackson et al, 1994). However, another caveat could emerge, which suggests that CCK antagonists should nevertheless be used with cau- tion in the management of chronic pain in man. Indeed, a chronic treatment with proglumide or lorglumide may no longer facilitate, and may even attenuate, opioid-induced antinociception in rats (Kellstein and Mayer, 1990).

Neuropeptide FF

Introduction The peptide Phe-Met-Arg-Phe-NH, JFMRFamide) was originally isolated from ganglia of the clam Macrocallista nimbosa (Price and Greenberg, 1977). FMRFamide-like immunoreactive material was subsequently detected in the nervous system of various invertebrates and vertebrates (Boer et at , 1980; Galabov, 1991). In particular, two FMRFamide-like peptides were isolated and sequenced from the bovine brain: an octapeptide, FLFQPQFWamide (F8Fa, or morphine-modulating peptide, now called neuropeptide FF [NPFF, table I]), and an octadecapeptide (A18Fa, NPAF) (Yang et al, 1985b). The structure of the precursor pro- tein of these peptides is not yet known.

The presence of NPFF- and NPAF-related pep- tides in the brain and spinal cord of rat, mouse, guinea pig, pig and man has been established by several groups (Allard et al, 1991; Holmes and Dockray, 1989; Kivipelto and Panula, 1991; Kivi- pelto et al, 1989; Majane et al, 1988, 1989). NPFF immunoreactivity has notably been described in the rat CNS, with the highest concentrations in the spinal cord, hypothalamus, pons-medulla and neu- ral lobe of the pituitary (Allard et al, 1991; Kivi- pelto and Panula, 1991; Majane et al, 1989). NPFF-containing cells are located within two main areas in the rat brain: the medial hypothalamus and the nucleus tractus solitarius (Lee et al, 1993).

Functional NPFF receptors were first observed in electrophysiological studies using cultured mouse spinal cord neurones (Guzman et al, 1989). Subsequently, specific binding sites for NPFF were characterized in the rat CNS, with a distribu- tion different from that of opioid receptors. The highest densities of NPFF specific binding sites are found in the spinal cord, PAG, thalamus, amygdala and lateral septum (Allard et al, 1989, 1992). NPFF binding sites have also been identi- fied in the human spinal cord (Allard et ul, 1994).

Initially, NPFF receptors were labelled with a tyrosyl derivative of NPFF ([*25I]YLFQPQRFa- mide, Allard et al, 1989, 1992), but new analogues of NPFF or FMRFamide, resistant to degradation, and with high affinity for NPFF receptors, are now available for binding and behavioural studies (Devillers et at, 1994; Gicquel et at, 1992; Malin et al, 1993b, c). Opioid ligands do not affect the specific binding of iodinated NPFF analogues to rat spinal cord membranes (Allard et al, 1989; Devillers et al, 1994) and, reciprocally, FRMFa- mide and NPFF-related peptides do not display significant affinity at opioid receptors in rat or guinea-pig brain membranes (Raffa et al, 1994). NPFF receptors are coupled to G proteins (Devill- ers et al, 1994; Payza and Yang, 1993; Payza et al, 1993).

Anti-opioid effects of FMRFamide, NPFF and related peptides Behavioural studies have shown that NPFF and NPFA decrease tail-flick latency, as expected from a hyperalgesic action, in rats (Yang el at, 1985b). In addition, FMRFamide, NPFF and their ana- logues can attenuate exogenous opioid-induced analgesia as well as stress- and defeat-induced analgesia (which is thought to be due, in part, to the triggering of endogenous opioid systems) in rodents (Brussaard et al, 1989; Kavaliers, 1987a, 1990; Kavaliers and Hirst, 1985b, 1986b; Kavali-

Page 14: Opioid and anti-opioid peptides

422 F Cesselin

ers and Innes, 1992a; Kavaliers and Yang, 1991; Million et al, 1993; Oberling et al, 1993; Raffa, 1988; Tang et al, 1984b; Yang et al, 1985b). In line with these behavioural data, in vivo electro- physiological studies indicate that NPFF can atten- uate the inhibitory effects of p opioid agonists (but not those of 6 agonists) on C-fibre-evoked responses in nociceptive neurones in the rat dorsal horn (Magnuson et al, 1990).

In addition to reducing opioid-induced analge- sia, FMRFamide- and NPFF-related peptides were found to attenuate other opioid responses. For instance, these peptides suppress opioid-induced feeding (Kavaliers and Hirst, 1985a, 1986a), mor- phine-induced inhibition of electrically provoked contractions of the guinea pig ileum (Demichel et al, 1993), and morphine-induced increases in loco- motor activity (Marco et al, 1995; Raffa, 1989).

Intraventricular or intrathecal injection of immu- noglobulin Gs raised against FMRFamide or NPFF produces, by itself, antinociception, and augments morphine- and stress-induced analgesia (Kavaliers and Innes, 1992a; Kavaliers and Yang, 1989, 1991; Tang et al, 1984b; Yang et al, 1985b).

There is also evidence that NPFF is involved in opioid tolerance and dependence. When injected into the third ventricle of morphine-tolerant and -dependent rats, anti-NPFF immunoglobulin Gs or a putative NPFF antagonist peptide reverse toler- ance and attenuate the naloxone-induced with- drawal syndrome (Lake et al, 1991, 1992; Malin et at, 1990b, 1991). In contrast, NPFF or ana- logues injected into the third ventricle or subcuta- neously precipitate the opioid abstinence syn- drome in morphine-dependent rats (Malin et al, 1990a, 1993a, b, c). This is not due to alterations in NPFF binding characteristics since chronic mor- phine treatment alters neither the affinity, nor the number or G protein coupling of NPFF receptors in the rat CNS (Payza ef al, 1993). Interestingly, at higher doses, NPFF can cause a quasi-abstinence syndrome in normal (non-dependent) rats (Malin et al, 1990a).

From these data, it appears that NPFF not only counteracts exogenous and endogenous opioid analgesia (like CCK) but, in itself, can also exert a hyperalgesic action. In fact, this peptide seems to be involved not only in opioid tolerance (like CCK), but also in dependence. At odds with what was observed for spinal CCKergic systems, spinal NPFF systems do not seem to be involved in con- ditioned anti-analgesia (see above), since intrathe- cal administration of an anti-NPFF antiserum does not affect this process (Wiertelak et al, 1994).

The general scheme of such anti-opioid actions

OPlOlDS 7 NPFF

Receptors 2 ?

Receptors 1 ?

EFFECTS Fig 6. Schematic representation of NF'FF/opioid interactions. Opioids, either exogenous or endogenous, could activate NPFFergic systems, and the resulting enhanced release of NPFF could trigger anti-opioid actions. In turn, NF'FF, or exog- enous agonists, could increase the release of endogenous opioid peptides and facilitate opioid-mediated effects. By analogy with CCK (see fig 5). one could hypothesize that NPFF exerts its anti-opioid effects through type 1 receptors, and its excitatory influence on opioidergic systems through type 2 receptors.

implies that exogenous and endogenous opioids should be able to activate NPFF systems (fig 6). Indeed, the spinal release of NPFF or FMRFa- mide-like material (see Devillers and Simonnet, 1994; Devillers et al, 1995; Simonnet e f al, 1994; Tang et al , 1984b; Zhu et al , 1992) can be enhanced by morphine in rats (Simonnet et al, 1994; Tang et al, 1984b). In the same species, administration of heroin has been shown to reduce NPFF levels in the spinal cord, as expected from an increased release of the peptide (Simonnet et al, 1994). This idea is supported by the observation that naloxone not only reverses heroin-induced analgesia but also induces a clear hyperalgesia, which can be attributed to the enhanced extracellu- lar levels of the pronociceptive peptide NPFF pro- voked by heroin (Simonnet et al, 1994). Finally, Malin et a1 (1990b) reported that NPFF levels in the cerebrospinal fluid are markedly increased in opioid-tolerant and -dependent rats, further sup- porting the hypothesis that opioid/NPFF equilib- rium may be involved in opioid tolerance and dependence.

Opioid-like effects of FMRFamide, NPFF and related peptides Among the NPFF analogues with high affinity for NPFF receptors synthesized by Gicquel et a1 (1992), some behave as NPFF, ie have hyperal- gesic effects and inhibit morphine analgesia, whereas others exhibit significant analgesic prop- erties and increase morphine analgesia (Gicquel et

Page 15: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 423

al, 1992). Although the latter compounds could act as antagonists at NPFF receptors, several observa- tions suggest that their opioid-like properties are due to other mechanisms. Indeed, the analgesic effects of these NPFF-related compounds can be blocked by naloxone (Gicquel et al, 1992). Fur- thermore, NPFF itself, FMRFamide, and related peptides, when administered intrathecally or supraspinally, can exert naloxone-sensitive anti- nociceptive effects against thermal and mechanical stimuli in the rat and the mouse (Gouardbres et al, 1993; Hill et al, 1987; Raffa and Connelly, 1992). In addition, by the intrathecal route, NPFF, at a sub-analgesic dose, has been shown to potentiate the analgesic effect of morphine (Gouardbres et al, 1993).

In fact, careful examination of the influence of NPFF on nociception revealed that the peptide's action is biphasic. Thus, in rats, icv administration of NPFF induces a rapid and short-lasting hyperes- thesic effect, followed, during the night but not the day, by a long-lasting analgesic effect, the magni- tude of which being related to the magnitude of the hyperesthesic effect (Oberling et al, 1993). The analgesic rebound could result from the triggering of endogenous opioid systems, which are known to be more active during the night in rodents (Naber et al, 1981; Wesche and Frederickson, 1979). This is congruent with the observations that NPFF effects on nociception and opioid withdrawal are more easily observed during the dark phase (Kava- liers, 1990; Malin et at, 1990a). However, the abil- ity of NPFF-related peptides to enhance the release of endogenous opioids remains to be directly established.

Whatever their direction, the effects of NPFF are obtained when the peptide is injected in the third ventricle, but not in the lateral ventricles. This sug- gests that one site of action of NPFF for modulating nociception could be the PAC, which is rich i n NPFF receptors (AUard et al, 1992) and is immedi- ately adjacent to the third ventricle. Similarly, NPFF produces withdrawal signs only when administered in the third ventricle, but not in the lateral ventricle or intrathecally (Rothman et al, 1993). Obviously, these observations do not exclude that NPFF and opioids can also interact at the level of the spinal cord (see eg Tang el al, 1984b).

Other naloxone-reversible opioid-like effects of Fh4RFamide or NPFF have been reported. These include actions of these peptides on colonic bead expulsion in mice, intestinal myoelectrical activity (Million et al, 1993), heart rate and blood pressure in rats (Raffa, 1988; Raffa and Jacoby, 1989; Raffa et al, 1992; Wong et al, 1985).

The fact that the opioid antagonistic activity of an analogue of FMRFamide is displayed at lower doses than its opioid-like activity (which is remi- niscent of what is observed for CCK, see above), led Raffa and Connelly (1992) to suggest that the analgesic effects of this compound may be due to a weak agonistic activity at opioid receptors. How- ever, this hypothesis is not applicable to the opi- oid-like activity of the other FMRFamide- and NPFF-related peptides, since they have a very low affinity for opioid receptors (see above). In fact, as established for CCK, there could be two types of NPFF receptors, the stimulation of which would lead to opposite effects on opioid systems (see fig 6). This hypothesis is substantiated by the observation that NPFF has a multiphasic effect on the electrical activity of cultured mouse spinal cord neurones (Guzman et al, 1989). Furthermore, two receptors with different affinities for Fh4RFa- mide have already been described in the snail Helix aspersa (Payza, 1987).

Conclusion (fig 6) Thus, as with CCK and opioid systems, NPFF and opioid systems could constitute two physiologi- cally coupled systems, each controlling the activity of the other, with NPFF exhibiting anti-opioid effects via the activation of one type of NPFF receptor, and an opioid-like action due to the trig- gering of endogenous opioid systems through the activation of another NPFF receptor type.

There is presently a lack of potent and selective NPFF receptor antagonists, which could be poten- tial adjuncts to morphine for alleviating pain in humans or as a treatment medication for opioid abuse. However, it must be taken into account that chronic manipulations of NPFF systems could alter the characteristics of the opioid systems. In particular, chronic icv administration of anti-NPFF IgG has been shown to upregulate y opioid recep- tors (Rothman et al, 1991), and, conversely, chronic icv infusion of NPFF downregulates these receptors in the rat brain (but this latter change does not seem to affect the acute antinociceptive effects of icv injected morphine) (Rothman et al, 1993).

MIF and related peptides (table I)

Introduction The C-terminal tripeptide of oxytocin (Pro-Leu- Gly-NH,) is a melanocyte inhibiting factor (MIF- 1) (Celis et al, 1971; Nair et al, 1971) with anti- opioid properties (Galina and Kastin, 1986, see below). The hypothalamus contains an exopepti-

Page 16: Opioid and anti-opioid peptides

424 F Cesselin

dase which can produce MIF-1 from oxytocin (Walter et al, 1979). Other peptides, structurally related to MIF-1, but which do not derive from oxytocin, also have anti-opioid properties. These peptides are Tyr-MIF- 1 (Tyr-Pro-Leu-Gly-NH2), and Tyr-W-MIF-1 (Tyr-Pro-Trp-Gly-NH,). Tyr- MIF- 1 has been isolated from bovine hypothala- mus and human parietal cortex (Horvath and Kas- tin, 1989, 1990), and Tyr-W-MIF-1 from bovine hypothalamic tissue and human frontal cortex (Erchegyi et al, 1992; Hackler et al, 1993).

In contrast to CCK and NPFF, Tyr-MIF-1 and Tyr-W-MIF-1 (but not MIF-1 itself, Lucian0 et al, 1981) are able to interact with opioid receptors with a higher potency at p sites than at 6 and K sites (Zadina and Kastin, 1986; Zadina et al, 1992). Thus, Tyr-W-MIF-1 and Tyr-MIF-1 inhibit 50% of the specific binding of tritiated DAGO (a selective p agonist) to rat brain membranes at con- centrations of 0.3 and 6.0 jM, respectively (Erche- gyi et al, 1992; Zadina and Kastin, 1986). How- ever, non-opioid specific binding sites for Tyr-MIF- 1 have been identified in brain (Zadina and Kastin, 1985; Zadina et al, 1982, 1989, 1990; Zhang et at, 1992). The affhity of Tyr-MIF-1 for these sites is about 50-fold higher than for p opioid receptors. However, MIF-1 does not bind to these Tyr-MIF-1 specific sites (Zadina and Kastin, 1985; Zhang et al, 1992).

A fourth member of the MIF family has recently been isolated from human brain cortex, and named Tyr-K-MIF- 1 (Tyr-Pro-Ly s-Gl y -NH2) (Hackler et al, 1994, table I). Tyr-K-MIF-1 appears to bind to Tyr-MIF-1 specific sites and to its own sites. This peptide exhibits only a low activity on opioid bind- ing (see Reed et al, 1994).

Anti-opioid properties of MIFs MIF- 1 and Tyr-MIF- 1 have been shown to reduce exogenous opioid-induced analgesia in rodents (in the tail flick and scratching tests, Datta et al, 1982; Kastin et al, 1979, 1980, 1984, 1985), goldfish (Ehrensing et al, 1982), and man (in the cold pres- sor test, Ehrensing et a l , 1984). MIF-1 also decreases opioid-induced ingestive behaviour (Kavaliers and Hirst, 1985c; Olson et al, 1980) and hypothermia (Yehuda et al, 1980).

Data also indicate that MIF-1 and Tyr-MIF-1 can reduce the effects of endogenous opioids. In particular, both peptides are able to inhibit the expression of some forms of stress-induced anal- gesia in various species (Galina and Kastin, 1985, 1987, 1988; Kavaliers, 1987b; Kavaliers and Hirst, 1986b, c; Kavaliers and Innes, 1992b; Krowicki, 1991a, b; Lipa et al, 1989).

That Tyr-MIF-1 activity might increase in response to chronic opioid administration has been proposed by Zadina et a1 (1989) to explain why prolonged, but not acute, exposure to morphine decreases the specific binding of Tyr-MIF- 1 to brain tissue. Such an increase in Tyr-MIF-1 activ- ity could be involved in the behavioural expression of opioid withdrawal, since an icv injection of exogenous MIF- 1 has been reported to precipitate withdrawal signs in levorphanol dependent mice (Contreras and Takemori, 1984b). Similarly, Tyr- MIF-1 (protected from degradation by the pepti- dase inhibitor bestatin) triggers an abstinence syn- drome in morphine-dependent rats, but not in opioid-naive animals when injected into the third venticle (Malin et al, 1993d). However, rather than favourizing tolerance, icv administered MIF- 1 can antagonize the development of tolerance to levor- phanol in mice (Contreras and Takemori, 1984b).

Opioid-like actions of MIFs Similarly to CCK and NPFF, MIFs also exert some opioid-like actions. Thus, icv administration of Tyr-W-MIF- 1 induces a long-lasting naloxone- reversible analgesia in the tail-flick test in rats (Zadina et al, 1993). In addition, central or oral administration of Tyr-MIF- 1 modulates gastric emptying and gastrointestinal motility in a nalox- one-reversible manner in rodents (Million et al, 1992, 1994; Zadina et al , 1992). Tyr-MIF-1- induced inhibition of contractions of the guinea pig ileum can be reversed by CTOP, a selective p opioid receptor antagonist, but not by a selective K antagonist, nor-binaltorphimine (Erchegyi et al, 1993).

Are MIFs partial agonists at opioid receptors? In the guinea-pig ileum, Tyr-MIF-1 and Tyr-W- MIF- 1 have both opioid agonistic and antagonistic activities. Thus, Tyr-MIF- 1 and Tyr-W-MIF- 1 exhibit agonist-like activity as shown by a dose- dependent inhibition of electrically induced con- tractions through a selective action at the p (but not 6 or K) opioid receptor (Erchegyi et al, 1993; Zadina and Kastin, 1986; Zadina et al, 1992). In contrast, the tetrapeptides can also attenuate the inhibition of contractions due to morphine and DAGO, especially in morphine-tolerant animals and in preparations of ileum pretreated with the opioid alkylating agent, P-chlornaltrexamine (Erchegyi et al, 1992; Zadina et al, 1992). Without excluding the possibility that MIFs (especially Tyr-MIF- 1) may exert their anti-opioid properties through their own high affinity binding sites, alto- gether these data suggest that the antagonistic

Page 17: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 425

OPlOlDS MIFs

Opioid receptors I

P EFFECTS

Fig 7. Schematic representation of the interactions between the peptides of the MIF family and the opioids. Although specific binding sites exist for Tyr-h4IF-1 in the brain, through which this peptide could exert an anti-opioid effect, thepeptides of the MLF family could also act directly at opioid receptors where they behave as antagonists at low concentrations and as ago- nists at high concentrations.

action of these compounds would rather result from their “partial agonistic” properties at p opioid receptors (fig 7). The fact that antagonistic proper- ties are best observed under conditions of reduced receptor reserve would appear to be consistent with an efficacy of MIFs at the p receptors low enough to block the action of other opioid ligands. Indeed, agonists with relatively low efficacy are known to become antagonists when the efficiency of the stimulus-response coupling is reduced (see Kenakin, 1987).

CONCLUSION (fig 8)

The data summarized in this review initially led to the concept of “anti-opioid” peptides on the basis of extensive clinical and fundamental studies, which were carried out in order to elucidate the mechanisms of tolerance and dependence to opi- oids. In fact, the three peptides which have been reviewed in this article, CCK, NPFF and MIFs, have both anti-opioid and opioid-like properties, such that it would be more appropriate to refer to them as “opioid modulating” peptides (the label initially given to NPFF) than “anti-opioid” pep- tides. With regard to the models which tend to explain opioid tolerance and dependence only by

- * + 1

\/ + - RESULTING EFFECT

Fig 8. Homeostatic mechanism implying two mutually “antago- nistic” substances, able to reciprocally stimulate their produc- tion andor release. “A” enhances the release and reduces the effects of “B”, and reciprocally “B” modulates the release and effects of “A”, such that the value of a given parameter affected in opposite directions by A and B is the result of the dynamic equilibrium between the two systems, A and B.

alterations affecting opioid receptors themselves, and/or the transducers and effectors to which they are coupled (see Collin and Cesselin, 1991), the concept of anti-opioid peptides has the advantage of explaining why opioid receptors remain func- tional following chronic exposure to opioids, since their blockade with an antagonist results in a with- drawal syndrome (whereas this blockade is with- out effect in normal man or animal). In fact, the multiple feed-back loops within a system or between systems, as they appear in the figures illustrated in this paper (see figs 4-7), do not con- stitute a peculiar originality. They have to be con- sidered in the general context of homeostasis, ie the coordinated reactions which maintain most of the biological dynamic equilibria (fig 8).

ACKNOWLEDGMENTS

The author is indebted to Drs M Hamon and I Nevo for their critical reading of the manuscript.

REFERENCES

Allard M, Geoffre S, Legendre P, Vincent JD, Simonnet G . Characterization of rat spinal cord receptors to FLFQPQRFa- mide, a mammalian morphine modulating peptide: a binding study. Brain Res 1989;500: 169-76

Allard M, Jordan D, Zajac JM et al. Autoradiographic localiza- tion of receptors for neuropeptide FF, FLFQPQFWamide, in human spinal sensory system. Brain Res 1994;633: 127-32

Allard M, Theodosis DT, Rousselot P, Lombard MC, Simonnet G. Characterization and localization of a putative morphine- modulating peptide, FLFQPQRFamide, in the rat spinal cord: biochemical and immunocytochemical studies. Neu- roscience I99 I ;40:8 1-92

Page 18: Opioid and anti-opioid peptides

426 F Cesselin

Allard M, Zajac JM, Simonnet G. Autoradiographic distribu- tion of receptors to FLFQPQRFamide. a morphine-mod- ulating peptide, in rat central nervous system. Neuroscience 199249: I0 1-1 6

Baber NS, Dourish CT, Hill DR. The role of CCK, caerulein, and CCK antagonists in nociception. Pain 1989;39:307-28

Baile CA, McLaughlin CL, Della-Fera MA. Role of cholecys- tokinin and opioid peptides in control of food intake. Phys- iol Rev 1986;66: 172-234

Barbaz BS, Autry WL, Ambrose FG, Hall NR, Liebman JM. Antinociceptive profile of sulfated CCK-8: comparison with CCK-4, unsulfated CCK-8 and other neuropeptides. Neuropharmacology 1986;25:823-29

Barbaz BS, Hall NR, Liebman JM. Antagonism of morphine analgesia by CCK-8-S does not extend to all assays nor all opiate analgesics. Peptides 1989;9: 1295-300

Bartolome JV, Lorber BA, Bartolome MB. Brain cholecystoki- nin and Pendorphin systems may antagonistically interact to regulate tissue DNA synthesis in rat pups. Brain Res

Basbaum AI, Cruz L, Weber E. Immunoreactive dynorphin B in sacral primary afferent fibers of the cat. J Neurosci

Beinfeld MC, Meyer DK, Eskay RL, Jensen RT, Brownstein MJ. The distribution of cholecystokinin immunoreactivity in the central nervous system of the rat as determined by radioimmunoassay. Brain Res 1981 ;212:51-7

Benoliel JJ, Bourgoin S, Mauborgne A, Legrand JC, Hamon M, Cesselin F. Differential inhibitorylstimulatory modulation of spinal CCK release by p and a opioid agonists, and selective blockade of p-dependent inhibition by K receptor stimulation. Neurosci Lett 199 I ; 124:204-07

Benoliel JJ, Collin E, Mauborgne A et al. Mu and delta opioid receptors mediate opposite modulations by morphine of the spinal release of cholecystokinin-like material. Brain Res

Benoliel JJ, Mauborgne A, Bourgoin S. Legrand JC, Hamon M, Cesselin F. Opioid control of the in witro release of chole- cystokinin-like material from the rat substantia nigra. J Neurochem 1992;58:91&22

Bertolini A, Poggioli R, Ferrari W. ACTH-induced hyperalge- sia in rats. Experientia 1979;35:121&17

Bhargava HN, Yousif DJ, Matwyshyn GA. Interactions of thy- rotropin releasing hormone, its metabolites and analogues with endogenous and exogenous opiates. Gen Pharmacol

Bian JT, Sun MZ, Xu MY, Han JS. Antagonism by CCK-8 of the antinociceptive effect of electroacupuncture on pain- related neurons in nucleus-parafascicularis of the rat. Asia

Bodnar RJ, Paul D, Pasternak GW. Proglumide selectively pot- entiates supraspinal p, opioid analgesia in mice. Neuro- pharmacology 1990;29:507-10

Boer HH, Schot LP, Veenstra JA, Reichelt D. Immunocyto- chemical identification of neuronal elements in the central nervous system of a snail, some insects, a fish and a mam- mal with an antiserum to the molluscan cardio-excitatory tetrapeptide FMFWamide. Cell Tissue Res 1980;213:21-7

Bronstein DM, Przewlocki R, Akil H. Effects of morphine treatment on proopiomelanocortin systems in rat brain. Brain Res 1990;519:102-1 I

Brussaard AB, Kits KS, Maat AT, Mulder AH, Schoffelmeer ANM. Peripheral injection of DNS-Wa, a FMWa agonist. suppresses morphine-induced analgesia in rats. P eptides

Celis ME, Taleisnik S, Walter R. Regulation of formation and proposed structure of the factor inhibiting the release of

1994;66 1 : 19-24

1986;6: 127-33

I994;653:8 1-91

1983; 14565-70

PaC J Phatmacol 1993;8:89-97

1989; 10:735-39

melanocyte-stimulating hormone. Proc Natl Acad Sci USA 1971;68:1428-33

Cesselin F. Endomorphines, rkepteurs des opioydes et sites d'action des morphinomimttiques. Agressologie 1991 ;32:3 10-17

Cesselin F, Bourgoin S . Artaud F, Hamon M. Basic and regula- tory mechanisms of in vitro release of met-enkephalin from the dorsal zone of the rat spinal cord. J Neurochem 1984;43:763-73

Chen XH, Geller EB, Adler MW. Cholecystokinin-octapeptide microinjected into the periaqueductal grey antagonizes the analgesic effect induced by peripheral electroacupuncture stimulation in rats. Regul Pep? 1994;54:554

Chen Y , Mestek A, Liu J, Yu L. Molecular cloning of a rat K opioid receptor reveals sequence similarities to the p and a opioid receptors. Biochem J 1993;295:625-28

Clarke RW, Harris J, Ford Tw. Seeking a role for CCK in con- trol of spinal reflexes In: Dourish CT, Cooper SJ, Iversen SD, Iversen LL, eds, Multiple cholecystokinin receptors in the CNS. New York: Oxford University Press, 1992322-28

Collin E, Bourgoin S, Ferhat L, Hamon M, Cesselin F. K-opioid receptor stimulation abolishes p- but not &mediated inhibi- tory control of spinal met-enkephalin release. Neurosci Lett 1992a; 134: 134:23842

Collin E, Cesselin F. Neurobiological mechanisms of opioid tolerance and dependence. Clin Neuropharmacol 199 1 ; 14:465-88

Collin E, Cesselin F. OntogenBse des rkepteurs des opioides. In: Gauvain-Piquard A, Murat I, Pons G, eds. La douleur chez 1 'enfmt: dchelles d'6waluation. traitements m'dicamenteux. Paris:Springer-Verlag France, 1993;37-5 I

Collin E, Frechilla D, Pohl M et a l . Opioid control of the release of calcitonin gene-related peptide-like material from the rat spinal cord in wivo. Brain Res 1993;609:2 I 1-22

Collin E, Mauborgne A, Bourgoin S, Benoliel JJ, Hamon M, Cesselin F. Morphine reduces the release of met-enkeph- ah-like material from the rat spinal cord in wivo by acting at a opioid receptors. Newopeprides I994;27:75-83

Collin E, Mauborgne A, Bourgoin S et al. Kappa-/mu-receptor interactions in the opioid control of the in viwo release of sub- stance P-lie material from the rat spinal cord. Neuroscience

Contreras PC, Takemori AE. Antagonism of morphine-induced analgesia, tolerance and dependence by alpha-melanocyte- stimulating hormone. J Pharmacol Exp Ther 1984%229:214

Contreras PC, Takemori AE. Effect of prolyl-leucyl-glycina- mide and alpha-melanocyte-stimulating hormone on levor- phanol-induced analgesia, tolerance and dependence. Life Sci 1984b;34:255946

Crain SM, Shen KF. After chronic opioid exposure sensory neurons become supersensitive to the excitatory effects of opioid agonists and antagonists as occurs after elevation of GMI ganglioside. Brain Res 1992575: 13-24

Crain SM, Shen KF. Opioids can evoke direct receptor-medi- ated excitatory effects in sensory neurons. Trends Pharma- col Sci 1990; 1 1 :77-8 1

Crawley JN, Convin RL. Biological actions of cholecystokinin. Peptides 1994; 15:73 1-55

Cruciani RA, Dvorkin B, Moms SA, Crain SM, Makman MH. Direct coupling of opioid receptors to both Gs and Gi pro- teins in F-l 1 neuroblastoma X sensory neuron hybrid cells. P roc Nut1 Acad Sci USA 1993;90:30 I 9 4 2 3

Datta PC, Sandman CA, Hoehler FK. Attenuation of morphine analgesia by a-MSH, MIF-I, melatonin and naloxone in the rat. Peptides 1982;3:433-37

Demichel P, Rodriguez JC, Roquebert J, Simonnet G. Neuro- peptide FF, a Fh4RF-NH, like peptide, blocks opioid effects on ileum contractions. Peptides 1993;14: 1005-9

1992b;51:347-55

Page 19: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 427

Derrien M, McCort-Tranchepain I, Ducos B, Roques BP, Durieux C. Heterogeneity of CCK-B receptors involved in animal models of anxiety. Pharmacol Biochem Behav 1994;49: 133-4 1

Derrien M, Noble F. Maldonado R, Roques BP. Cholecystokinin- A but not cholecystokinin-B receptor stimulation induces endogenous opioid-dependent antinociceptive effects in the hot plate test in mice. Neurosci Len 1993; 160: 193-6

Devillers JP, Labrouche SA, Castes E, Simonnet G. Release of neuropeptide FF, an anti-opioid peptide, in rat spinal cord slices is voltage- and Ca*+-sensitive. Possible involvement of P-type Ca2+ channels. J Neurochem, I995;64: 1567-75

Devillers JP, Mazarguil H, Allard M, Dickenson AH, Zajac JM, Simonnet G. Characterization of a potent agonist for NPFF receptors: binding study on rat spinal cord membranes. Neuropharmacology 1994;33:661-69

Devillers JP, Simonnet G. Modulation of neuropeptide FF release from rat spinal cord slices by glutamate. Involvement of NMDA receptors. Eur J Phunnucof, 1994;271: 185-92

Dickenson AH, Knox RJ. Effects of selective and non-selective K-opioid receptor agonists on cutaneous C-fibre-evoked responses of rat dorsal horn neurones. Brain Res 1987; 41 521-9

Ding X Z , Bayer BM. Increases of CCK messenger RNA and peptide in different brain areas following acute and chronic administration of morphine. Brain Res 1993;625: 139-44

Ding XZ, Fan SG, Zhou JP, Han JS. Reversal of tolerance to morphine but not potentiation of morphine induced analge- sia by antiserum against cholecystokinin octapeptide. Neu- ropharmucology 1986;25:1155-160

Dourish CT, Clark ML, Iversen SD. Analgesia induced by restraint stress is attenuated by CCK and enhanced by the CCK antagonists MK-329, L-365-031 and CR 1409. SOC Neurosci Abstr 1988a;14:290

Dourish CT, Hawley D, Iversen SD. Enhancement of morphine analgesia and prevention of morphine tolerance in the rat by the cholecystokinin antagonist L-364,7 18. Eur J Pharmucol 1988b; 147:469-72

Dourish CT, O’Neill MF, Coughlan J, Kitchener SJ, Hawley D, Iversen SD. The selective CCK-B receptor antagonist L- 365,260 enhances morphine analgesia and prevents mor- phine tolerance in the rat. Eur J Pharmacol 1990a; 1 7 6 : 3 5 4

Dourish CT, O’Neill MF, Schaffer LW, Siegl PKS, Iversen SD. The cholecystokinin receptor antagonist devazepide enhances morphine-induced analgesia but not morphine- induced respiratory depression in the squirrel monkey. J Phunnucol Exp Ther 1990b;255: 1 158-65

Dourish CT. The role of CCK, and CCK, receptors in mediat- ing the inhibitory effect of CCK on opiate analgesia. In: Dourish CT, ed. Multiple cholecystokinin receptors in the CNS. New York: Oxford University Press, 1991;455-79

Durieux C, Ruiz-Gayo M, Corringer PJ, Bergeron F, Ducos B, Roques BP. [3H]pBC 264, a suitable probe for studying cholecystokinin-B receptors: binding characteristics in rodent brains and comparison with [3H]SNF 8702. Mol Pharmucof 1992;4 I : I 089-095

Ehrensing RJ, Kastin AJ, Michell GF. Antagonism of morphine analgesia by prolylleucyl-glycinamide (MIF-I) in humans. Phurmucol Biochem Behuv 1984;21:975-8

Ehrensing RJ, Michell GF, Kastin AJ. Similar antagonism of morphine analgesia by MIF-I and naloxone in Carussius auratus. Pharmucol Biochem Behuv 1982; 17:757-61

Erchegyi J, Kastin AJ, Zadina JE. Isolation of a novel tetrapep- tide with opiate and antiopiate activity from human cortex: Tyr-Pro-Trp-Gly-NH2 (Tyr-W-MIF- I ) . Peptides 1992; 13~623-31

Erchegyi J, Zadina JE, Qiu XD er al. Structure-activity relation- ships of analogs of the endogenous brain peptides Tyr-MIF- 1 and Tyr-W-MIF-I. Peptide Res 1993;6:31-8

Faris PL, Beinfeld MC, Scallet AC, Johannessen JN, Olney JW. Increase in hypothalamic cholecystokinin following acute and chronic morphine. Brain Res 1986;367:405-07

Fans PL, Komisaruk BR, Watkins LR, Mayer DJ. Evidence for the neuropeptide cholecystokinin as an antagonist of opiate analgesia. Science I983;219:3 10-1 2

Faris PL, McLaughlin CL, Baile CA, Olney JW, Komisaruk BR. Morphine analgesia potentiated but tolerance not affected by active immunization against cholecystokinin. Science 1984;226: 1215-17

Felicio LF, Mann PE, Bridges RS. Intracerebroventricular chol- ecystokinin infusions block beta-endorphin-induced disrup- tion of maternal behavior. Pharmacol Biochem Behav 199 I ;39:20144

Friedman HJ, Jen MF, Chang JK, Lee NM, Loh HH. Dynor- phin: a possible modulatory peptide on morphine or beta- endorphin analgesia in mouse. Eur J Pharmacol 1981;69: 3 5 7 4 0

Fujimoto J, Arts KS. Clonidine, administered intracerebroven- tricularly in mice, produces an anti-analgesic effect which may be mediated spinally by dynorphin A (1-17). Neuro- pharmacology 1990;29:35 1 - 8

Fujimoto JM, Arts KS, Rady JJ, Tseng LF. Spinal dynorphin A (1-17): possible mediator of antianalgesic action. Neuro- pharmacology 1990;29:609-17

Fujimoto JM, Holmes B. Systemic single dose morphine pre- treatment desensitizes mice to the spinal antianalgesic action of dynorphin A ( 1 - 17). J Phurmacol Exp Ther 1990; 254: 1-7

Galabov PG. FMRF-amide-like immunoreactivity (FMRF- amide-LIR) in the vegetative areas and dorsal horn of the spinal cord of some rodents. J Hirnforsch 1991 ;32:263-75

Galina ZH, Amit Z. Interactions between ACTH, morphine, and naloxone and their effects on locomotor behavior. frog Neuropsychophnnnucol Biol Psychiatry 1985;9:691-5

Galina W, Kastin AJ. Differential activity of the endogenous antiopiate Tyr-MIF- 1 after various intensities of stress. Neurosci Lett 1988;84:312-16

Galina ZH, Kastin AJ. Existence of antiopiate systems as illus- trated by MIF-lmyr-MIF-I. Life Sci 1986;39:2153-159

Galina ZH, Kastin AJ. MIF-1 antagonizes warm-, but not cold- water stress-induced analgesia. Peptides 1985;6: 1 109-12

Galina ZH, Kastin AJ. Tyr-MIF-1 attenuates antinociceptive response induced by three models of stress-analgesia. Br J

Gall C. Lauterborn J, Burks D, Seroogy K. Co-localisation of enkephalin and cholecystokinin in discrete areas of rat brain. Brain Res 1987;403:403-8

Garzon J, Hollt V, Herz A. Cholecystokinin octapeptide acti- vates an opioid mechanism in the guinea-pig ileum: a pos- sible role of substance P. Eur J Pharmacol 1987;136:

Gaudreau P, Lavigne GJ, Quirion R. Cholecystokinin antago- nists proglumide, lorglumide, and benzotript, but not L- 364.7 18, interact with brain opioid binding sites. Neuropep- tides 1990;16:51-5

Gericke M, Morgenstern R, Ott T. The influence of tifluadon on cholecystokinin-induced antinociception. Eur J Phanna-

Ghilardi JR, Allen CJ, Vigna SR, McVey DC, Manty PW. Tri- geminal and dorsal root ganglion neurons express CCK receptor binding sites in the rat, rabbit, and monkey: pos- sible site of opiate-CCK analgesic interactions. J Neurosci 1992;12:4854-66

P hurmucol 1987;90:669-74

36 1-70

C O ~ 1990 180 1 87-90

Page 20: Opioid and anti-opioid peptides

428 F Cesselin

Gicquel S, Mazarguil H, Allard M, Simonnet G, Zajac JM. Ana- logues of F8Famide resistant to degradation, with high affin- ity and in vivo effects. Eur J Pharmacol 1992;222:61-7

Gintzler AR, Xu H. Different G proteins mediate the opioid inhi- bition or enhancement of evoked [5-methionine]-enkephalin release. Proc Nut1 AcudSci USA 1991;88:474145

Giusti P, Carrara M, Zampiron S, Cima L, Borin G. Are calcit- onins analgesic and/or hyperalgesic? Peptides 1985;6

Gouardtres C, Sutak M, Zajac JM, Jhamandas K. Antinocicep- tive effects of intrathecally administered FBFamide and FMRFamide in the rat. Eur J Pharmucol 1993;237:73-8 1

Guzman A, Legendre P, Allard M, Geoffre S, Vincent JD, Simonnet G. Electrophysiological effects of FTFQPQRF amide, an endogenous brain morphine modulating peptide, on cultured mouse spinal-cord neurons. Neuropeprides 1989; 14:253-61

Hackler L, Kastin AJ, Erchegyi J, Zadina JE. Isolation of Tyr- W-MIF- 1 from bovine hypothalami. Neuropeptides 1993; 24: 159-64

Hackler L, Kastin AJ, Zadina JE. Isolation of Tyr-K-MIF-1 (Tyr- Pro-Lys-NH,) from human brain cortex. Peptides 1994; 15:945-50

Hammonds RC, Nicolas P, Li CH. Beta-endorphin (1-27) is an antagonist of Pendorphin analgesia. Proc Nut1 Acad Sci

Han JS, Ding XZ, Fan SG. Cholecystokinin octapeptide (CCK-8): antagonism to electroacupuncture analgesia and a possible role in electroacupuncture tolerance. Pain 198627: 101-1 5

Han JS, Ding XZ, Fan SG. Is cholecystokinin octapeptide (CCK-8) a candidate for endogenous anti-opioid substrates? Newopeprides 1 985 ;5: 399402

Han JS, Terenius L. Neurochemical basis of acupuncture anal- gesia. Annu Rev Phurmucol Toxic01 1982;22: 193-220

Hendrie CA, Shepherd JK, Rodgers RJ. Differential effects of the CCK antagonist, MK-329, on analgesia induced by mor- phine, social conflict (opioid) and defeat experience (non- opioid) in male mice. Neurophumcology 1989;28: 1025-32

Higgins GA, Joharchi N, Wang Y, Conigall WA, Sellers EM. The CCK(A) receptor antagonist devazepide does not mod- ify opioid self-administration or drug discrimination. Com- parison with the dopamine antagonist haloperidol. Brain Res 1994;64024&54

Higgins GA, Nguyen P, Sellers EM. Morphine place condition- ing is differentially affected by CCK, and CCK, receptor antagonists. Brain Res 1992;572:208-15

Hill DR, Shaw T M , Woodruff GN. Binding sites for 125I-chole- cystokinin in primate spinal cord are of the CCK-A sub- class. Neurosci Len 1988;89: 133-9

Hill RG, Hughes J, Pittaway KM. Antinociceptive action of cholecystokinin octapeptide (CCK-8) and related peptides in rats and mice: effects of naloxone and peptidase inhibi- tors. Neurophurmacology 1987;26:289-300

Hoffmann 0, Wiesenfeld-Hallin Z. The CCK-B receptor antag- onist CI 988 reverses tolerance to morphine in rats. Neuro- report 1994;5:2565-8

Holaday JW. Porreca F, Rothman RB. Functional coupling among opioid receptor types: implication for anaesthesio- logy. In: Estefanous FG, ed. Opioids in anaesthesia II. Lon- don: Butterworth, 1991;5040

Holaday JW, Tseng LF, Loh HH. Thyrotropin releasing hor- mone antagonizes beta endorphin hypothermia and cata- lepsy. Life Sci 197822: 1 5 3 7 4

Holmes S, Dockray GJ. Identification by radioimmunoassay and HPLC of morphine modulatory peptide-immunoreac- tivity in rat spinal cord and brain. Neurochern Int 1989; 14:477-82

(SUPPI 3):277-82

USA 1984;81:1389-90

Hong EK, Takemori AE. Indirect involvement of delta opioid receptors in cholecystokinin octapeptide-induced analgesia in mice. J Pharmacol Exp Ther 1989;25 1 :594-8

Hong 0, Young GA, Khazan N. Modulation of morphine- induced EEG and behavioral effects by dynorphin A (1 - 13) in non tolerant and morphine tolerant rats. Neuropharma- cology 1988;27:807-12

Horvath A, Kastin AJ. Evidence for presence of Tyr-MIF-I (Tyr-Pro-Leu-Gly-NH2) in human brain cortex Int J Protein Res 1990;36:28 1 4

Horvath A, Kastin AJ. Isolation of tyrosine-melanocyte-stimu- lating hormone release-inhibiting factor I from bovine brain tissue. J Biol Chem 1989;264:2175-179

Hudson GM, Marquis LK, Stamidis H, Young GA. Cholecys- tokinin octapeptide alters morphine-induced effects on EEG power spectra both quantitatively and qualitatively. Eur J Pharmacol 1992;221:217-22

Hughes J, Boden P, Costall B, Domeney A. Kelly E. Horwell DC, Hunter JC, Pinnock RD, Woodruff GN. Development of a class of selective cholecystokinin type B receptor antagonists having potent anxiolytic activity. P roc Nut1 Acod Sci USA 1990;87:6728-32

Itoh S , Katsuura G. Suppresive effect of cholecystokinin and its related peptides on /%endorphin-induced catalepsy in rats. EurJ Phannacol1981;74:3814

Itoh S, Katsuura G, Maeda Y. Caerulein and cholecystokinin suppress pendorphin-induced analgesia in the rat. Eur J P harmacol 1982;80:42 1-25

Itoh S, Katsuura G, Yoshikawa K, Rehfeld JF. Potentiation of /%endorphin effects by cholecystokinin antiserum in rats. Can J Physiol Phurmacol 1985;63:81-3

Jackson A, Tattersall D. Bentley G et al. An investigation into the discriminative stimulus and reinforcing properties of the CCK,-receptor antagonist, L-365,260 in rats. Neuropep- tides 1994;26:343-53

Jagerschmidt A, Popovici T, O'Donohue M, Roques BP. Iden- tification and characterization of various cholecystokinin B receptor mRNA forms in rat brain tissue and partial deter- mination of the cholecystokinin B receptor gene structure. J Neurochem 1994;63: I 199-206

Janiri L, D'Amato R, Zieglgansberger W. Dynorphin (1-17) reduces the inhibitory actions of p- and &selective opioid agonists in cortical neurons of the rat in vivo. Neurosci Lett 1988;84:79-83

Johannessen JN, Stuckey JA, Phan TH, Boadle-Biber MC. Blockade of morphine-induced increases in brain tryptophan hydroxylase activity by systemic pretreatment with CCK-8: no reversal by vagotomy. Brain Res 1989;499:357-62

Jurna I, Zetler G. Antinociceptive effect of centrally adminis- tered caerulein and cholecystokinin octapeptide (CCK-8). EurJ Pharmucol 1981;73:323-31

Kachur JF, Miller RJ, Field M. Control of guinea pig intestinal electrolyte secretion by a delta-opioid receptor. P roc Nut1 AcadSci USA 1980;77:2753-56

Kachur JF, Wang SX, Gullikson GW, Gaginella TS. Cholecys- tokinin-mediated ileal electrolyte transport in the guinea pig. Characterization of receptor subtypes. Gustroenrerol- ogy 199 1; 10 1 : 1428-3 1

Kapas L, Benedek G, Penke B. Cholecystokinin interferes with the thermoregulatory effect of exogenous and endogenous opioids. Neuropeptides 1989; 14:85-92

Kastin AJ, Nissen C, Zadina JE, Schally AV, Ehrensing RH. Naloxone-like actions of MIF-1 do not require the presence of the pituitary. fharmacol Biochem Behav 198013:907-12

Kastin AJ, Olson RD, Ehrensing RH, Berzas MC, Schally AV, Coy DH. MIF-1's differential actions as an opiate antago- nist. Phurmacol Biochem Behav 1979;11:721-23

Page 21: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 429

Kastin AJ, Stephens E, Ehrensing RH, Fischman AJ. Tyr-MIF- 1 acts as an opiate antagonist in the tail-flick test. Phurmu- col Biochem Behav 1984;21:937-41

Kastin AJ, Stephens E, Zadina JE, Coy DH, Fischman AJ. Tyr- MIF-I, identified in brain tissue, and its analogs are active in two models of antinociception. Pharmacol Biochem Behuv 1985;23:1045-49

Katoh A, Nabeshima T, Kameyama T. Behavioral changes induced by stressful situati0ns:effects of enkephalins, dynorphin, and their interactions. J Pharmacol Exp Ther 1990;253 :600-7

Katsuura G, Itoh S. Potentiation of /%endorphin effects by pro- glumide in rats. Eur J Pharmucol 1985;107:363-6

Kaufmann R, Schoneberg T, Henklein P, Boomgaarden M, Sohr D, Ott T. CCK,-type receptors in guinea-pig cortex and continuous cell lines. Comparative [3H]CCK-8S bind- ing studies. Neuropeptides 1994;26 (suppl 1):4

Kavaliers M. Calcium channel blockers inhibit the antagonistic effects of Phe-Met-Arg-Phe-amide (FMRFamide) on mor- phine- and stress-induced analgesia in mice. Bruin Res 1987a;4 15:38&4

Kavaliers M. Inhibitory influences of mammalian FMRF-amide (Phe-Met-Arg-Phe-amide)-related peptides on nociception and morphine- and stress-induced analgesia in mice. Neuro-

Kavaliers M. MIF-1 and Tyr-MIF-I antagonize morphine and opioid but not non-opioid stress-induced analgesia in the snail, Cupaea nemoralis. Peptides l987b;g: 1-5

Kavaliers M, Hirst M. FMRFamide: an endogenous peptide with marked inhibitory effects on opioid-induced feeding behavior. Brain Res Bull 1986a;17:403-8

Kavaliers M, Hirst M. FMRFamide suppresses kappa-opiate induced feeding in the mouse. Peptides 1985a;6:847-49

Kavaliers M, Hirst M. FMRFamide, a putative endogenous opiate antagonist: evidence for suppression of defeat-induced anal- gesia and feeding in mice. Neuropeptides 1985b;6:485-94

Kavaliers M, Hirst M. Inhibitory influences of FMRFamide and PLG on stress-induced opioid analgesia and activity. Brain Res 1986b;372:370-4

Kavaliers M, Hirst M. Inhibitory influences of MIF-l (PLG) and Tyr-MIF-1 (YPLG) on agression and defeat-induced analgesia in mice. Peptides 1986c;7:1007-10

Kavaliers M, Hirst M. PLG and FMRFamide: endogenous pep- tides with marked inhibitory effects on opioid-induced feeding. f rog Neuropsychopharmacol Biol Psychiatry 1985c;9:709-12

Kavaliers M, Innes D. Sex differences in the effects of neuro- peptide FF and IgG from neuropeptide FF on morphine- and stress-induced analgesia. Peptides 1992a;13:603-7

Kavaliers M, Innes DGL. Sex differences in the effects of Tyr- MIF-I on morphine- and stress-induced analgesia. Peptides I992b; 13: 1295-7

Kavaliers M, Yang HY. IgG from antiserum against endoge- nous mammalian FMRF-NH,-related peptides augments morphine- and stress-induced analgesia in mice. Peptides 1989;10:741-45

Kavaliers M, Yang HYT. Effects of mammalian FMRF-NH,- related peptides and IgG from antiserum against them on aggression and defeat-induced analgesia in mice. P eptides 1991;12:235-9

Kellstein DE, Mayer DJ. Chronic administration of cholecys- tokinin antagonists reverses the enhancement of spinal mor- phine analgesia induced by acute pretreatment. Brain Res

Kellstein DE, Mayer DJ. Spinal co-administration of cholecys- tokinin antagonists with morphine prevents the develop- ment of opioid tolerance. Pain 1991;47:221-29

S C ~ Lett 1990;115:307-12

19905 16~263-70.

Kellstein DE, Price DD, Mayer DJ. Cholecystokinin and its antagonist lorglumide respectively attenuates and facilitates morphine-induced inhibition of C-fiber evoked discharges of dorsal horn nociceptive neurons. Brain Res 1991 ; 540:302-6

Kenakin T. Agonists, partial agonists, antagonists, inverse ago- nists and agonistlantagonists? Trends Phurmucol Sci 1987;

Khachaturian H. Lewis ME, Schafer MKH, Watson SJ. Anat- omy of the CNS opioid systems. Trends Neurosci 1985; 8: 1 11-19

Kivipelto L, Majane EA, Yang HYT, Panula P. Immunohisto- chemical distribution and partial characterization of FLFQPQRFamidelike peptides in the central nervous system of rats. J Comp Neurol 1989;286:269-87

Kivipelto L, Panula P. Comparative distribution of neurons containing FLFQPQRFamide-like (morphine-modulating) peptide and related neuropeptides in the rat brain. Eur J Neurosci 1991;3:175-85

Klein H, Jackson R, McCormick G et al. Enhancement of opi- ate analgesia by devazepide in a baboon dolorimetry model. In: Dourish CT, Cooper SJ, Iversen SD, Iversen LL, eds. Multiple cholecystokinin receptors in the CNS. Oxford: Oxford University Press, 1992;529-36

Krowicki ZK. Cimetidine does not change the effect of Tyr- MIF-1 (Tyr-Pro-Leu-Gly-NHJ on the opiate form of foot- shock-induced analgesia. Life Sci 199 1 a;49: 1 163-68

Krowicki ZK. Dopamine receptor antagonists block the effect of Tyr-MIF 1 (Tyr-Pro-Leu-Gly-NH,) on the opiate form of footshock-induced analgesia. Neuropep t ides 199 1 b;

Kubota K, Sugaya K, Matsuda I, Matsuoka Y, Terawaki Y. Reversal of the antinociceptive effect of cholecystokinin by benzodiazepines and a benzodiazepine antagonist RO 15-

Lake JR, Hammond MV, Shaddox RC, Hunsicker LM, Yang HY, Malin DH. IgG from neuropeptide FF antiserum reverses morphine tolerance in the rat. Neurosci Lett 199 1 ; 132:2%32

Lake JR, Hebert KM. Payza K et al. Analog of neuropeptide FF attenuates morphine tolerance. Neurosc i Lert 1992; 146:203-6

Lavigne GJ, Hargreaves Khf, Schmidt EA, Dionne RA. Proglu- mide potentiates morphine analgesia for acute postsurgical pain. Clin Phannacol Ther 1989;45:666-73

Lavigne GJ, Millington WR, Mueller GP. The CCK-A and CCK-B receptor antagonists, devazepide and L-365,260. enhance morphine antinociception only in nonacclimated rats exposed to a novel environment. Neuropeptides 1992;

Lee CH, Wasowicz K, Brown R, Majane EA, Yang HYT, Panula P. Distribution and characterization of neuropeptide FF-like immunoreactivity in the rat nervous system with a monoclonal antibody. Eur J Neurosci 1993;5: 1339-48

Lee NM, Smith AF'. Possible regulatory function of dynorphin and its clinical implications. Trends Pharmuco l Sci

Lehmann KA, Schliisener M, Arabatsis P. Failure of proglu- mide, a cholecystokinin antagonist, to potentiate clinical morphine analgesia. Anesth Anulg 1989;68:5 1-6

Li Y, Han JS. Cholecystokinin octapeptide antagonizes mor- phine analgesia in periaqueductal gray of the rat. Brain Res 1989;480:105-10

Lin Y, Carpenter DO. Direct excitatory opiate effects mediated by non-synaptic actions on rat medial vestibular neurons. Eur J P h n m c o l I994;262:99-106

Ling GSF, MacLeod JM, Lee S, Lockhart SH, Pasternak GW.

81423-26

19:281-5

1788. JUP J P h ~ m c 0 1 1985;37: 101-5

21 : 119-29

1984;5: 108-10

Page 22: Opioid and anti-opioid peptides

430 F Cesselin

Separation of morphine analgesia from physical depen- dence. Science 1984;226:4624

Lipa SM, Kavaliers M, Ossenkopp KP. Differential inhibitory effects of MIF-I, Tyr-MIF-I, naloxone and pfunaltrexa- mine on body rotation-induced analgesia in the meadow vote, Microtus pennsyfvanicus. Peprides 1989; 10:493-7

Liu HY, Chandler S, Beitz AJ, Shipley MT, Behbehani MM. Characterization of the effects of cholecystokinin (CCK) on neurons in the periaqueductal gray of the rat - Immunocy- tochemical and in vivo and in virro electrophysiological studies. Brain Res 1994;642:83-94

Loh YP. Molecular mechanisms of kndorph in biosynthesis. Biochem Pharmucol 1992;44:84349

Lucian0 MG, Zadina JE, Kastin AJ, Coy DH. Mu and delta opi- ate receptors in rat brain are affected by GTP but not by MIF-I . Brain Res Bull 198 1 ;7:677-82

Magnuson DSK, Sullivan AF, Simonnet G, Roques BP, Dick- enson AH. Differential interactions of cholecystokinin and FLFQPQRF-NH, with j i and a opioid antinociception in the rat spinal cord. Neuropeptides 1990;16:213-18

Majane EA, Casanova MF, Yang HYT. Biochemical character- ization of W - N H , - l i k e peptides in spinal cords of vari- ous mammalian species using specific radioimmunoassays. Peptides 1988;8:1137-144

Majane EA, Panula P, Yang HYT. Rat brain regional distribu- tion and spinal cord neuronal pathway of FLFQPQRF-NH,, a mammalian FMRF-NH,-like peptide. Brain Res 1989; 494: 1-1 2

Maldonado R, Derrien M, Noble F, Roques BP. Association of a peptidase inhibitor and a CCK-B antagonist strongly pot- entiates antinociception mediated by endogenous enkepha- lins. Neuroreport 1993;7:947-50

Maldonado R, Valverde 0, Denien M, Tejedorreal P, Roques BP. Effects induced by BC 264, a selective agonist of CCK- B receptors, on morphine-dependent rats. Pharmacol Bio- chem Behav 1994;48:363-9

Malin DH, Lake JR, Arcangelli KR. Subcutaneous injection of an analog of neuropeptide FF precipitates morphine absti- nence syndrome. Life Sci 1993a;53:PL 2 6 1 4

Malin DH, Lake JR, Fowler DE et al. FMRF-NH2-like mam- malian peptide precipitates opiate-withdrawal syndrome in the rat. Peptides 1990a;l1:277-80

Malin DH, Lake JR, Hammond MV. FMRF-NH,-like mam- malian octapeptide: possible role in opiate dependence and abstinence. Peptides 1990b; 1 1 :969-72

Malin DH, Lake JR, Leyva JE el al. Analog of neuropeptide FF attenuates morphine abstinence syndrome. P e p t i d e s 1991;12:1011-14

Malin DH, Lake JR, Payza K er al. Enhanced antiopiate activity and enzyme resistance in peptidomimetics of FMRFamide containing (E)-2,3-methanomethionine. Peprides 1993b;

Malin DH, Payza K, Lake JR et al. Enhanced antiopiate activity in peptidomimetics of FMRFamide containing Z-2.3-meth- anomethionine. Peptides 1993c;14:47-51

Malin DH, Zadina JE, Lake JR. Tyr-MIF-1 precipitates absti- nence syndrome in morphine-dependent rats. Brain Res

Malinge M, Colombel MC, Bourin M. Cholecystokinin recep- tor antagonists block the jumping behavior precipitated in morphine-dependent mice by naloxone. 1995; in press

Marco N, Stinus L, Allard M, Le Moal M, Simonnet G. Neuro- peptide FF receptors within the ventral mesencephalon and dopaminergic terminal areas: localization and functional antiopioid involvement. Neuroscience 1995;64: 1035-44

Marrama D, Poggioli R, Vergoni AV, Sandrini M, Bertolini A. Influence of L-364,718, a specific CCK-A antagonist, on

14:731-34

1993d;610: 169-71

pain threshold, morphine analgesia and opioid receptors. Pharmacol Res 1989;21:4734

Mei L, Han JS. The antagonistic effect of cholecystokinin octa- peptide (CCK-8) on opioid effects in cardiovascular activ- ities was mediated by CCK-B receptor. Sci China Ser B 1993;36:8 17-23

Miaskowski C, Sutters KA, Taiwo YO, Levine JD. Antinoci- ceptive and motor effects of delta/mu and kappdmu combi- nations of intrathecal opioid agonists. Pain 1992;49: I 3 7 4 4

Millan MJ. Multiple opioid systems and pain. Pain 1986; 27:30347

Miller KK, Lupica CR. Morphine-induced excitation of pyram- idal neurons is inhibited by cholecystokinin in the CAI region of the rat hippocampal slice J Pharmucol Exp Ther 1994;268:7534 1

Million M, Fioramenti J, Bueno L. Central administration of Tyr-MIF-I stimulates gastrointestinal motility in rats:evi- dence for the involvement of dopamine, sigma and CCK receptors. Neuropeptides 1994;26:77-85

Million M, Fioramonti J, Bueno L. Oral administration of Tyr- MIF-I stimulates gastric emptying and gastrointestinal motility in rodents. Peptides 1992; 13:469-74

Million M, Fioramonti J, Gicquel S, Zajac JM, Bueno L. Com- parative action of Phe-Leu-Phe-Gln-Pro-Gln-Arg-Phe-NH, analogs on intestinal motility and nociception in rats. J Pharmacol Exp Ther 1993;265:96- 102

Moran TH, Robinson PH, Goldrich MS, McHugh PR. Two brain cholecystokinin receptors: implications for behavorial actions. Brain Res 1986;362: 175-9

Morley JE, Levine AS, Kneip J, Grace M. Billington CJ. The effects of peripherally administered satiety substances on feeding induced by butorphanol tartrate. Pharmacol Bio- chem Behav 1983;19:577-82

Naber D, Wirz-Justice A, Kafka MS. Circadian rhythm in rat brain opiate receptor. Neurosci Lett 198 1 ;21:45-50

Nair RMG, Kastin AJ, Schally AV. Isolation and structure of hypothalamic MSH release-inhibiting hormone. Biochem Biophys Res Commun 197 1 ;43: 1376-8 1

Noble F, Derrien M, Roques BP. Modulation of opioid antinoc- iception by CCK at the supraspinal 1evel:evidence of regu- latory mechanisms between CCK and enkephalin systems in the control of pain. BrJ Pharmucol 1993;109:1064-70

Noble F, Smadja C, Roques BP. Role of endogenous cholecys- tokinin in the facilitation of mu-mediated antinociception by delta-opioid agonists. J Pharmacol Exp Ther 1994; 271:1127-l34

O’Neill MF, Dourish CT, Iversen SD. Morphine-induced anal- gesia in the rat paw pressure test is blocked by CCK and enhanced by the CCK antagonist MK-329. Neurophurma- cology 1989;28:24>7

O’Neill MF, Dourish CT, Tye SJ, Iversen SD. Blockade of CCK-B receptors by L-365.260 induces analgesia in the squirrel monkey. Brain Res 1990;534:287-90

Oberling P, Stinus L, Le Moal M, Simonnet G. Biphasic effect on nociception and antiopiate activity of the neuropeptide FF (FLFQPQRFamide) in the rat. Peptides 1993;14:919-24

Olson GA, Olson RD, Kastin AJ. Endogenous opiates:1993. P eprides 1994; 15: 15 13-56

Olson RD, Kastin AJ, Olson GA. MIF-I suppresses depriva- tion-induced fluid consumption i n rats. Peptides 1980; 1:353-7

Ossipov MH, Kovelowski CJ, Vanderah T, Porreca F. Naltrin- dole, an opioid a antagonist, blocks the enhancement of morphine-antinociception induced by a CCKB antagonist in the rat. NeurosciLerf 1994;181:9-12

Panerai AE, Rovati LC, Cocco E, Sacerdote P, Mantegazza P. Dissociation of tolerance and dependence to morphine: a

Page 23: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 43 1

possible role for cholecystokinin. Bruin Res 1987;410: 52-60

Payza K, Akar CA. Yang HYT. Neuropeptide FF receptors: structure-activity relationship and effect of morphine. J Pharmucol Exp Ther 1993;267:88-94

Payza K, Yang HYT. Modulation of neuropeptide FF receptors by guanine nucleotides and cations in membranes of rat brain and spinal cord. J Neurochem 1993;60: 1894-9

Payza K. FMRFamide receptors in Helix aspersa. Peprides

Petrillo P, Gambino MC, Tavani A. Bremazocine induces anti- nociception, but prevents opioid-induced constipation and catatonia in rats and precipitates withdrawal in morphine- dependent rats. Life Sci 1984;35:915-27

Pittaway KM, Hill RG. Cholecystokinin and pain. In: Pain Headache, vol. 9. Basel: Karger, 1987;21346

Pittaway KM, Rodriguez RE, Hughes J, Hill RG. CCK-8 anal- gesia and hyperalgesia after intrathecal administration in the rat: comparison with CCK-related peptides. Neuropeptides 1987; 10:87-108

Poggioli R, Vergoni AV, Sandrini M, Barbafiera L, Marrama D, Bertolini A. Influence of the selective cholecystokinin antagonist L-364.7 18 on pain threshold and morphine anal- gesia. Phnrmcology 1991 ;42: 197-201

Pohl M, Collin E, Benoliel JJ, Bourgoin S, Cesselin F, Hamon M. Cholecystokinin (CCK)-like material and CCK mRNA levels in the rat brain and spinal cord after acute or repeated morphine treatment. Neuropeprides 1992;21: 193-200

Pohl M, Collin E, Bourgoin S er al. Expression of proenkeph- din A gene and presence of met-enkephalin in dorsal root ganglia of the adult rat. J Neurochem I994;63: 1226-34

Pournaghash S, Riley AL. Failure of cholecystokinin to precipi- tate withdrawal in morphine-treated rats. Pharmacol Bio- chem Behav 1991;38:479-84

Price DA, Greenberg MJ. Structure of a molluscan cardio- excitatory neuropeptide. Science 1977; 197:67&1

Price DD, von der Gruen A, Miller J, Rafii A, Price C. Potentiation of systemic morphine analgesia in humans by proglumide, a cholecystokinin antagonist. Anesth Anulg 1985;64:8014

Przewlocki R, Hassan AHS, Lason W, Epplen C, Herz A, Stein C. Gene expression and localization of opioid peptides in immune cells of inflamed tissue: functional role in antinoci- ception. Neuroscience 1992;48:491-500

Pu S, Zhuang H, Lu Z, Wu X, Han J. Cholecystokinin gene expression in rat amygdaloid neurons: normal distribution and effect of morphine tolerance. Mol Brain Res 1994a; 21: 183-9

Pu SF, Zhuang HX, Han JS. Cholecystokinin octapeptide (CCK-8) antagonizes morphine analgesia in nucleus accum- bens of the rat via the CCK-B receptor. Brain Res 3994b; 657: 159-64

Raffa RB. Supraspinal FMRFamide antagonizes morphine- induced horizontal, but not vertical locomotor activity. Pep- tides 1989;10:403-6

Raffa RB. The action of FMRFamide (Phe-Met-Arg-Phe-NH2) and related peptides on mammals. P eptides 1988;9:9 15-22

Raffa RB, Addo MF. Gill A. Supraspinal administration of [D- Met2]FMRFamide produces a naloxone-sensitive increase in heart rate in unrestrained spontaneously hypertensive rats. Neuropeptides 1992;21:201-5

Raffa RB, Connelly CD. Supraspinal antinociception produced by [D-Met2]-FMRFamide in mice. Neuropeprides 1992; 2 2 195-203

Raffa RB, Jacoby H. A-18-Famide and F-8-amide, endogenous mammalian equivalent of the molluscan neuropeptide FMRFamide (Phe-Met-Arg-Phe-NHJ, inhibit colonic bead expulsion in mice. Peptides 1989; 10:873-75

1987;8: 1065-74

Raffa RB, Kim A. Rice KC, De Costa BR. Codd EE, Rothman RB. Low affinity of FMRFamide and four FaRPs (FMRFa- mide-related peptides). including the mammalian-derived FaRPS F-8-Famide (NPFF) and A- I8-Famide. for opioid p, 3, K,, $?a. or qb receptors. Peprides 1994; 15:4014

Ramabadran K. Hyperalgesic effect of the selective kappa opi- oid agonist, U 50488 H, in mice. Jap J Pharmacol 1983; 33: 1289-92

Ramarao P, Jablonski HI, Rehder KR, Bhargava HN. Effect of rc-opioid receptor agonists on morphine analgesia in mor- phine-naive and morphine-tolerant rats. Eur J Pharmacol 1988; l56:23946

Rattray M, De Belleroche J. Morphine action on cholecystoki- nin octapeptide release from rat periaqueductal grey slices: sensitisation by naloxone. Neuropeprides 1987;lO: 189-200

Rattray M, Jordan CC. De Belleroche J. The novel CCK antag- onist L364.7 18 abolishes caerulein- but potentiates mor- phine-induced antinociception. Eur J Pharmacol 1988; 152:163-6

Reed GW, Olson GA, Olson RD. The Tyr-MIF-I family of peptides. Neurosci Behavior Rev 1994; 1 8 5 19-25

Rehfeld JL. Neuronal cholecystokinin. One or two multiple transmitters. J Neurochem 1985;44: 1-10

Reisine T, Bell GI. Molecular biology of opioid receptors. Trends Neurosci 1993;16:506-10

Rezayat M, Nikfar S, Zarrindast MR. CCK receptor activation may prevent tolerance to morphine in mice. Eur J Pharmu- col 1994;254:21-6

Rezvani A, Stokes KB, Rhoads DL, Way EL. Proglumide exhibits delta-opioid agonist properties. Alcohol Drug Res 1987;7: 1 3 5 4 6

Rodriguez RE, Sacristan MP. in vivo release of CCK-8 from the dorsal horn of the rat: inhibition by DAGOL. FEBS. Lert 1989;250:2 15-1 7

Rosen A, Brodin E. Effect of acute morphine treatment on pep- tide levels in the pen-aqueductal grey. Acra Physiol S c a d 1989;136:4934

Rossitch E, Ovelmen-Levitt J, Lyerly MA, Nashold BS. The deafferentation syndrome in the rat: effects of intraventricu- lar neurotensin and cholecystokinin. Exp Neurol 1987; 98:26+75

Rothman RB. A review of the role of anti-opioid peptides in morphine tolerance and dependence. Synapse 1992; 12: 129-38

Rothman RB, Brady LS, Xu H, Long JB. Chronic intracerebro- ventricular infusion of the antiopioid peptide, Phe-Leu-Phe- Gln-Pro-Gln-Arg-Phe-NH, (NPFF), dowregulates mu opi- oid binding sites in rat brain. Peprides 1993;14:1271-7

Rothman RB, Xu H, Yang HYT, Long JB. Upregulation of rat brain opioid receptors by the chronic administration of mor- phine: possible evidence for an anti-opiate model of toler- ance and dependence. NIDA. Res Motwgr 1991 ;105:264-70

Rovati LC, Sacerdote P, Panerai AE. Effects of proglumide on morphine analgesia and tolerance. Ann NY Acud Sci 1985; 448:630-32

Ruiz-Gay0 M, Durieux C, FourniC-Zaluski MC, Roques BP. Stimulation of a opioid receptors reduces the in vivo bind- ing of the CCK-B selective agonist ['HIpBC 264: evidence for a physiological regulation of CCKergic systems by endogenous enkephalins. J Neurochem 1992;59: 1805-1 1

Sandman CA, Kastin AJ. Intraventricular administration of MSH induces hyperalgesia in rats. Peptides 198 1 ;223 1-3

Schmauss C, Herz A. Intrathecally administered dynorphin (1- 17) modulates morphine-induced antinociception differ- ently in morphine-naive and morphine-tolerant rats. Eur J Phnrmucol1987;135:429-31

Schnur P, Raigoza VP, Sanchez MR, Kulkosky PJ. Cholecys-

Page 24: Opioid and anti-opioid peptides

432 F Cesselin

tokinin antagonizes morphine induced hypoactivity and hyperactivity in hamsters. Pharmacol Biochem Behav

Sheldon RJ, Nunan L, Porreca F. Differential modulation by (D- Penz, o-Pen5) enkephalin and dynorphin A (1-17) of the inhibitory bladder motility effects of selected mu agonists in vivo. J Pharmucol Exp Ther 1989;249:462-9

Sheldon RJ, Nunan L, Porreca F. Mu antagonist properties of kappa agonists in a model of rat urinary bladder motility in vivo. J Pharmucol Exp Ther 1987;243:234-40

Sheldon RJ, Nunan L, Porreca F. U 50488 H differentially antagonizes the bladder effects of p agonists at spinal sites. Eur J Pharmacol 1988;146:229-35

Shen KF, Crain SM. Antagonists at excitatory opioid receptors on sensory neurons in culture increase potency and specific- ity of opiate analgesics and attenuate development of toler- ancddependence. Bruin Res 1994;636:286-97

Shen KF, Crain SM. Chronic selective activation of excitatory opioid receptor functions in sensory neurons results in opi- oid dependence without tolerance. Bra in Res 1992; 597:74-83

Simonnet G, Devillers JP, Boisserie F. Pain facilitory systems activated by opiate receptor stimultion. Regul Peptides

Smock T, Fields HL. ACTH 1-24 blocks opiate-induced anal- gesia in the rat. Brain Res 198 1 ;212202-6

Stanfa LC, Dickenson AH. Cholecystokinin as a factor in the enhanced potency of spinal morphine following carrageenin inflammation. Br J Pharmacol 1993; 108:967-73

Stanfa L, Dickenson A, Xu XJ, Wiesenfeld-Hallin Z. Cholecys- tokinin and morphine analgesia: variations on a theme. Trends Pharmacol Sci 1994; I5:65-6

Stengaard-Pedersen K, Larsson LI. Localization and opiate receptor binding of enkephalin, CCK and ACTWPendor- phin in the rat central nervous system. Peprides 1981;2

Suberg SN, Culhane ES, Carstens E, Watkins LR. Behavioral and electrophysiological investigations of opiatdcholecys- tokinin interactions. Adv f a i n Res Ther 1985a;9:541-53

Suberg SN. Culhane ES, Carstens E, Watkins LR. The potentia- tion of morphine-induced inhibition of spinal transmission by proglumide, a putative cholecystokinin antagonist. Ann NYAcud Sci 1985b;448:660-62

Suh HH, Collins KA, Tseng F. Intrathecal cholecystokinin octa- peptide attenuates the antinociception and release of immu- noreactive Met-enkephalin induced by intraventricular Pendorphin in the rat. Neuropeptides 1992;21: 13 1-7

Suh HH, Tseng LF, Li CH. Beta-endorphin-( 1-27) antagonizes beta-endorphin- but not morphine-, D-Penz-D-PenS-enkeph- alin-, and U50.488H-induced analgesia in mice. Neuro- pharmacology 1988;27:957-63

Suh HH, Tseng LF. Differential effects of sulfated cholecystok- inin octapeptide and proglumide injected intrathecally on antinociception induced by /%endorphin and morphine administered intracerebroventricularly in mice. Eur J Phar-

Sullivan AF, Hewett K, Dickenson AH. Differential modula- tion of alpha(2)-adrenergic and opioid spinal antinocicep tion by cholecystokinin and cholecystokinin antagonists in the rat dorsal hom:an electrophysiological study. Bruin Res

Sweetnam PM. Wrathall JR, Neale JH. Localization of dynor- phin gene-product-immunoreactivity in neurons from spinal cord and dorsal root ganglia. Neuroscience 1986; 18:947-55

Tache Y, Lis M, Collu R. Effects of thyrotropin-releasing hor- mone on behavioral and hormonal changes induced by beta- endorphin. Life Sci 1977;21:84146

1986;25: 1067-70

1 994 ;54: 277-8

(SUPPI 1):3-19

mac01 1990; 179:329-38

1994;662: I4 1-7

Takemori AE, Portoghese PS. The mixed antinociceptive ago- nist-antagonist activity of @endorphin (1-27) in mice. Life Sci 1993;53: 1049-52

Takeshige C, Mera H, Hisarnitsu T, Tanaka M, Hishida F. Inhi- bition of the analgesia inhibitory system by D-phenylala- nine and proglumide. Brain Res Bull 1991 ;26:385-91

Tang J, Chou J, Iadarola M, Yang HYT, Costa E. Proglumide prevents and curtails acute tolerance to morphine in rats. Neuropharmacofogy 1984a;23:7 15- 18

Tang 1, Yang HYT, Costa E. Inhibition of spontaneous and opi- ate modifiate nociception by an endogenous neuropeptide with Phe-Met-Arg-Phe-NH2-like immunoreactivity. Proc

Terenius L. Somatostatin and ACTH are peptides with partial antagonist-like selectivity for opiate receptors. Eur J Phar- macol 1976;38:211-13

Terenius L, Gispen WH, De Wied D. ACTH-like peptides and opiate receptors in the rat brain:structure-activity studies. Eur J Pharmacol 1975;33:395-9

Tseng LF, Collins KA. Cholecystokinin administered intrathe- cally selectively antagonizes intracerebroventricular P-endorphin-induced tail-flick inhibition in the mouse. J Pharmucol Exp Ther 1992;260: 1086-92

Tulunay FC, Jen MF, Chang JK, Loh HH, Lee NM. Possible regulatory role of dynorphin on morphine and beta-endor- phin-induced analgesia. J Pharmacol Exp Ther 198 1 ;224: 2 9 6 8

Uhl GR, Childers S, Pastemak G. An opiate-receptor gene fam- ily reunion. Trends Neurosci 1994;17:89-93

Valverde 0, Maldonado R, Foumit-Zaluski MC, Roques BP. Cholecystokinin B antagonists strongly potentiate antinoci- ception mediated by endogenous enkephalins. J Pharmucol Exp Ther 1994;270:77-88

Vanderah TW, Lai J, Yamamura HI, Porreca F. Antisense oli- godesoxynucleotide to the CCKB receptor produces naltrin- dole- and tLeu(5)lenkephalin antiserum-sensitive enhance- ment of morphine antinociception. Neuroreport 1994;5:

Vanderhaeghen JJ, Signeau JC, Gepts W. New peptide in the vertebrate CNS reacting with gastrin antibodies. Nature (Lond) 1975;257:604-5

Walter R, Ritzmann RF, Bhargava HN, Flexner LB. Prolyl-leu- cyl-glycinamide, cyclo(leucylglycine), and derivatives block development of physical dependence on morphine in mice. Proc Nut1 Acad Sci USA 1979;76:5 18-20

Wang J, Ren M, Han J. Mobilization of calcium from intracel- Mar stores as one of the mechanisms underlying the antiop- ioid effect of cholecystokinin octapeptide. Peprides 1992; 13:947-51

Wang L, Gintzler AR. Bimodal opioid regulation of cyclic AMP formation:implications for positive and negative coupling of opiate receptors to adenylyl cyclase. J Neuro- chem 1994;63:1726-30

Wang XI, Fan SG, Ren MF, Han JS. Cholecystokinin octapep- tide suppressed [3H]etorphin binding in rat brain opiate receptor. Life Sci 1989;45: 1 17-23

Wang XI, Han JS. Modification by cholecystokinin octapeptide of the binding of p-, a-, and K-opioid receptors. J Neuro- chem 1990;55: 1379-82

Wang XJ, Wang XH, Han JS. Cholecystokinin octapeptide antagonized opioid analgesia mediated by mu- and kappa- but not delta-receptors in the spinal cord of the rat. Bruin Res 1990;523:5-10

Watkins LR, Kinscheck IB, Kaufman EFS, Miller J, Frenk H, Mayer DJ. Cholecystokinin antagonists selectively poten- tiate analgesia induced by endogenous opiates. Brain Res 1985a;327: 18 1-90

Natl A C U ~ Sci USA 1984b;81:5002-5

260 1-5

Page 25: Opioid and anti-opioid peptides

Opioid and anti-opioid peptides 433

Watkins LR, Kinscheck IB, Mayer DJ. Potentiation of mor- phine analgesia by the cholecystokinin antagonist proglu- mide. Brain Res 1985b;327, 169-80

Watkins LR, Kinscheck IB, Mayer DJ. Potentiation of opiate analgesia and apparent reversal of morphine tolerance by proglumide. Science 1984;224:395-6

Watkins LR, Kinscheck IB, Rosenquist G et al. The enhance- ment of opiate analgesia and the possible reversal of mor- phine tolerance by proglumide. Ann NY Acad Sci 1985c; 448:67&7

Weihe E, Hartschuh W, Weber E. Prodynorphin opioid pep- tides in small somatosensory primary afferents of guinea pig. Neurosci Lett 1985;58:347-52

Welin M, Harro J, Yukhananov R, Nyberg F, Oreland L. Chol- ecystokinin receptor binding in morphine analgesia: toler- ance, withdrawal and abstinence. Neuropeptides 1994;

Wesche DL, Frederickson RC. Diurnal differences in opioid peptide levels correlated with nociceptive sensitivity. Life Sci 1979;24:1861-68

Wiertelak EP, Maier SF. Watkins LR. Cholecystokinin anti- analgesia: safety cues abolish morphine analgesia. Science 1992;256:830-33

Wiertelak EP, Yang HYT, Mooney-Heiberger K, Maier SF, Watkins LR. The nature of conditioned anti-analgesia: spi- nal cord opiate and anti-opiate neurochemistry. Brain Res I994;634:2 14-26

Wiesenfeld-Hallin Z, Duranti R. Intrathecal cholecystokinin interacts with morphine but not substance P in modulating the nociceptive flexor reflex in the rat. Peptides 1987; 8: 153-8

Wiesenfeld-Hallin Z, Xu XI. Role of cholecystokinin type B receptor in nociception studied with peptide agonists and antagonists. In: Methods in Neurosciences, Volume II. New York Academic Press, 1993;148-69

Wiesenfeld-Hallin Z, Xu XJ, Hughes J, Horwell DC. Hokfelt T. PD 134308, a selective antagonist of cholecystokinin type B receptor, enhances the analgesic effect of morphine and synergically interacts with intrathecal galanin to depress spinal nociceptive reflexes. Proc Narl Acad Sci USA 1990;87:7105-109

Wiesenfeld-Hallin Z, Xu XJ, Hughes J, Horwell DC, Hokfelt T. Studies on the effect of systemic PD134308 (CAM 958) in spinal reflex and pain models with special reference to interaction with morphine and intrathecal galanin. Neuro- peprides 199 1 ; 19 (suppl):79-84

Wong F, Greenberg MJ, Tse SYH. Cardiovascular effects of intraventricular injection of FMRFamide, Met-enkephalin and their common analogues in the rat. Comp Biochem P hysiol 1985;8 1C: 175-9

Wood SK. Tulunay FC, Loh HH, Lee NM. Effect of dynor- phin-( I - 13) and related peptides on respiratory rate and morphine-induced respiratory rate depression. Eur J Phar-

Woodruff GN, Hughes J. Cholecystokinin antagonists. Annu Rev Pharmacol Toxic01 1991;31:469-501

Xu Q, Takahashi M, Kaneto H. Dependency on the brain func- tion of arginine vasopressin system of the development to and recovery from analgesic tolerance to morphine. Brain Res 1992577: 189-93

Xu XJ, Hao JX, Seiger A, Hughes J, Hokfelt T, Wiesenfeld- Hallin Z. Chronic pain-related behaviors in spinally injured rats; evidence for functional alterations of the endogenous cholecystokinin and opioid systems. Pain 1994a;56:271-7

Xu XJ, Hokfelt T. Hughes J, Wiesenfeld-Hallin Z. The CCK-B antagonist CI988 enhances the reflex-depressive effect of morphine in axotomized rats. Neuroreport 1994b;5:718-20

26:379-83

ma~011983;86:117-22

Xu XJ, Puke MJC, Verge VMK, Wiesenfeld-Hallin 2. Up- regulation of cholecystokinin in primary sensory neurons is associated with morphine insensitivity in experimental neuropathic pain in the rat. Neurosci Lett 1993;152: 129-32

Xu XJ, Wiesenfeld-Hallin Z, Hughes DC, Horwell DC, Hokfelt T. CI988, a selective antagonist of cholecystokinin B recep- tors prevents morphine tolerance in the rat. Br J Pharmacol 1992; l05:591-6

Yang HYT, Fratta W, Majane EA, Costa E. Isolation, sequenc- ing, synthesis and pharmacological characterization of two brain neuropeptides that modulate the action of morphine. P roc Natl Acud Sci USA 1985b;82:7757-761

Yehuda S, Kastin AJ, Coy DH. Antagonistic actions of MIF-I on the hypothermia and hypomobility induced by beta- endorphin or morphine. Int J Neurosci 1980; I 1 :3 17-20

Zabin MA, Innis RB, Warmsley JK, Snyder SH, Kuhar MJ. Autoradiographic localization of cholecystokinin receptors in rodent brain. J Neurosci 1983;3:877-906

Zadina JE, Kastin AJ. Interaction of Tyr-MIF-I at opiate recep- tor sites. Pharmacol Biochem Behav 1986;25: 1303-305

Zadina JE, Kastin AJ. Interactions between the antiopiate Tyr- MIF-I and the mu opiate morphiceptin at their respective binding sites in brain. Peptides I985;6:965-70

Zadina JE, Kastin AJ, Ge W, Brantl V. Hemorphins, cytochro- phins, and human beta-casomorphins bind to antiopiate (Tyr-MlF-I) as well as opiate binding sites in rat brain. Life Sci 1990;47:PL25-PL30

Zadina JE, Kastin AJ, Ge LJ, Gulden H, Bungart KJ. Chronic, but not acute, administration of morphine alters antiopiate (Tyr-MIF-I) binding sites in rat brain. Life Sci 1989;44: 5 5 5 4 1

Zadina JE, Kastin AJ, Kenigs V, Bruno C, Hackler L. Pro- longed analgesia after intracerebroventricular Tyr-W-MIF- 1 (Tyr-Pro-Trp-Gly-NH2). Neurosci Lett 1993; 155:220-22

Zadina JE, Kastin AJ. Kersh D, Wyatt A. Tyr-MIF-1 and hemorphin can act as opiate agonists as well as antagonists in the guinea pig ileum. Life Sci 19925 1369-85

Zadina JE, Kastin AJ, Krieg EF, Coy DH. Characterization of binding sites for N-Tyr-MIF- 1 (Tyr-Pro-Leu-Gly-NH2) in rat brain. Phannacol Biochem Behav 1982;17:1193-198

Zetler G. Analgesia and ptosis caused by caerulein and chole- cystokinin octapeptide (CCK-8). Neuropharmacology

Zhang L J , Han NL, Han JS. Regulation by lithium of the antag- onistic effect of cholecystokinin octapeptide on ohmefenta- nyl-induced antinociception. Neuropharmacology 1994; 33: 123-26

Zhang XY, Zadina JE, Ehrlich M, Kastin AJ. Tyr-MIF-1, but not MIF-I or morphine, decreases cyclic --dependent protein kinase activity extracted from HeLa cells. Mol Cell Neurosci 1992;3:348-53

Zhou Y , Sun YH, Shen JM, Han JS. Increased release of immu- noreactive CCK-8 by electroacupuncture and enhancement of electroacupuncture analgesia by CCK-B antagonist in rat spinal cord. Neuropeptides 1993a;24: 139-44

Zhou Y, Sun YH, Zhang ZW. Han JS. Accelerated expression of cholecystokinin in the brain of rats rendered tolerant to morphine. Neuroreport 1992;3: 1121-123

Zhou Y, Sun YH, Zhang ZW, Han JS. Increased release of immunoreactive cholecystokinin octapeptide by morphine and potentiation of p-opioid analgesia by CCKB receptor antagonist L-365.260 in rat spinal cord. Eur J Pharmacot 1993b;234: 147-54

Zhu J, Jhamandas K, Ymg HYT. Release of neuropeptide FF (FLFQPQRF-NH2) from rat spinal cord. Brain Res 1992; 592:326-32

1980;19:415-22