Multiple opiate receptors: déjà vu all over again

https://doi.org/10.1016/j.neuropharm.2004.07.004Get rights and content

Abstract

The concept of multiple opioid receptors has changed dramatically since their initial proposal by Martin nearly 40 years ago. Three major opioid receptor families have now been proposed: mu, kappa and delta. Most of the opioid analgesics used clinically selectively bind to mu opioid receptors. Yet, clinicians have long appreciated subtle, but significant, differences in their pharmacology. These observations suggested more than one mu opioid receptor mechanism of action and led us to propose multiple mu opioid receptors over 20 years ago based upon a range of pharmacological and receptor binding approaches. A mu opioid receptor, MOR-1, was cloned about a decade ago. More recent studies have now identified a number of splice variants of this clone. These splice variants may help explain the pharmacology of the mu opioids and open interesting directions for future opioid research.

Introduction

The opiate field has a unique position in pharmacology and neuroscience. The opioid receptors were among the first neurotransmitter receptors to be identified and characterized in binding assays (Pert and Snyder, 1973, Simon et al., 1973, Terenius, 1973) and were one of the first to use a receptor to identify its endogenous ligand, the endogenous opioid peptides (Table 1) (Hughes et al., 1975, Hughes, 1975, Terenius and Wahlstrom, 1975, Pasternak et al., 1975a). Our understanding of these receptors and their endogenous ligands has mirrored the advancements of neuroscience, evolving from simple receptor binding assays, which were state of the art 30 years ago, to the cloning of these receptors and understanding them at the most basic molecular levels today.

From the beginning, our group has attempted to use the vast clinical experience with these agents to provide the direction for our laboratory work. Our focus over the years has been with the receptors, particularly the concept of multiple opioid receptors. The initial suggestion of multiple opioid receptors was based upon observations made with morphine and nalorphine combinations in patients (Lasagna and Beecher, 1954, Houde and Wallenstein, 1956). Nalorphine, in which the N-methyl group is substituted by an N-allyl group, was one of the earliest opioid antagonists. Its ability to reverse morphine actions was clearly demonstrated in clinical trials. However, as the relative dose of nalorphine to morphine was increased further, analgesia returned, illustrating a complexity in its pharmacology that had not been appreciated previously. Thus, nalorphine was a mixed agonist/antagonist capable of both reversing morphine analgesia and inducing analgesia itself at higher doses. These studies, which were performed almost half a century ago, led Bill Martin to suggest the existence of more than one opiate receptor, a concept he termed receptor dualism (Martin, 1967). In it, he proposed M and N receptors, corresponding to morphine and nalorphine receptors. Morphine was an agonist at the M receptor while nalorphine acted as an antagonist at the M receptor and an agonist at the N receptor.

Martin further refined his concept of multiple opioid receptors based upon the actions of series of drugs in the dog (Martin et al., 1976). In these studies, Martin again proposed a receptor for morphine, which he termed mu, as well as a receptor for the benzomorphan ketocyclazocine, which he termed kappa. He also suggested the existence of a third receptor, sigma. However, the concept of sigma receptors has evolved over the years (Pasternak et al., 1981, Wolozin et al., 1982, Walker et al., 1990). Although sigma1 receptors modulate opioid actions (Chien and Pasternak, 1994, Pasternak, 1994, Chien and Pasternak, 1995a, Chien and Pasternak, 1995b), they are no longer considered opioid receptors. Indeed, cloning studies of the sigma1 receptor from several species (Pan et al., 1998, Mei and Pasternak, 2001, Hanner et al., 1996, Kekuda et al., 1996, Seth et al., 1997) indicate a relatively small protein of approximately 25 kDa with two predicted transmembrane domains. Thus, the initial concept for multiple opioid receptors evolved from clinical trials with morphine/nalorphine combinations and classical pharmacological approaches. Their biochemical identification came afterwards.

Section snippets

Opioid binding sites

In 1973, opioid receptors were first demonstrated within the central nervous system using a rapid filtration binding assay (Pert and Snyder, 1973, Simon et al., 1973, Terenius, 1973). These early studies defined a saturable binding site which bound opioids with an unexpectedly high affinity in the nanomolar range. The sites were examined in extensive detail and were the first in which the binding of agonists and antagonists could be distinguished by sodium ions (Pert et al., 1973), divalent

Multiple mu receptors: clinical observations

We formally proposed the concept of multiple mu receptors over 20 years ago (Wolozin and Pasternak, 1981). Despite the fact that most of the opioids used clinically are mu-selective, as defined by their selectivity in receptor binding assays, clinicians have long observed differences among these drugs, requiring the individualization of therapy (Payne and Pasternak, 1992). Patients may respond far better to one mu opioid than another, both with respect to analgesic responsiveness and

Multiple mu receptors: molecular pharmacology

The first biochemical evidence suggesting multiple mu receptors came from binding studies almost 30 years ago (Pasternak and Snyder, 1975a). In these binding studies, we observed a novel, very high affinity site using radiolabeled agonists and antagonists. We subsequently termed this site mu1, as opposed to the mu-selective site we termed mu2 (Wolozin and Pasternak, 1981). Our major focus always has been the correlation of binding sites with function. The synthesis of several antagonists

Multiple mu receptors: molecular biology

The opioid receptors were first cloned in 1992, starting with the delta opioid receptor DOR-1 (Evans et al., 1992, Kieffer et al., 1992) and followed shortly afterwards by the mu MOR-1 and kappa KOR-1 receptors (Uhl et al., 1994, Chen et al., 1993a, Eppler et al., 1993, Thompson et al., 1993, Wang et al., 1993, Chen et al., 1993b, Li et al., 1993, Meng et al., 1993). They all have an extracellular N-terminus and intracellular C-terminus, with seven membrane-spanning domains that comprise the

Characterization of the MOR-1 splice variants

A common splicing pattern among the species involves the C-terminus and is well illustrated in the mouse (Fig. 6). All these variants contain exons 1, 2 and 3, which comprise all seven transmembrane domains that form the binding pocket. Thus, it is not surprising that all the variants retain high affinity and selectivity for mu opioids in receptor binding assays (Table 3). However, they differ structurally at the C-terminus, a region important in transduction of the signal following receptor

Conclusion

Early clinical studies provided the initial suggestion of discrete mu and kappa opioid receptors. The extensive clinical experience with a wide range of mu opioids raised the question of mu receptor heterogeneity. Preclinical behavioral studies supported these clinical observations and binding approaches also inferred multiple mu receptors. The recent cloning of a host of MOR-1 splice variants in mice, rats and humans underscores the complexity of the systems. Indeed, there are far more

Acknowledgements

This work has been supported by the National Institute on Drug Abuse through research grants (DA02615, DA06241 and DA07242) and a Senior Scientist Award (DA00220).

References (122)

  • T.E. Hebert et al.

    A peptide derived from a beta2-adrenergic receptor transmembrane domain inhibits both receptor dimerization and activation

    J. Biol. Chem

    (1996)
  • J. Hughes

    Isolation of an endogenous compound from the brain with pharmacological properties similar to morphine

    Brain Res

    (1975)
  • R. Kekuda et al.

    Cloning and functional expression of the human type 1 sigma receptor (hSigmaR1)

    Biochem. Biophys. Res. Commun

    (1996)
  • F.M. Leslie et al.

    Comparison of binding of [3H]-methionine-enkephalin, [3H]-naltrexone and [3H]-dihydromorphine in the mouse vas deferens and the myenteric plexus and brain of the guinea pig

    Eur. J. Pharmacol

    (1979)
  • G.S.F. Ling et al.

    Spinal and supraspinal opioid analgesia in the mouse: the role of subpopulations of opioid binding sites

    Brain Res

    (1983)
  • G.S.F. Ling et al.

    Naloxonazine actions in vivo

    Eur. J. Pharmacol

    (1986)
  • H.H. Loh et al.

    μ Opioid receptor knockout in mice: effects on ligand-induced analgesia and morphine lethality

    Mol. Brain Res

    (1998)
  • M.C. Luke et al.

    Irreversible opiate agonists and antagonists: V. Hydrazone and acylhydrazone derivatives of naltrexone

    Life Sci

    (1988)
  • J. Mei et al.

    Molecular cloning and pharmacological characterization of the rat sigma1 receptor

    Biochem. Pharmacol

    (2001)
  • Y.-X. Pan et al.

    Isolation and expression of a novel alternatively spliced mu opioid receptor isoform, MOR-1F

    FEBS Lett

    (2000)
  • Y.-X. Pan et al.

    Identification and characterization of two new human mu opioid receptor splice variants, hMOR-1O and hMOR-1X

    Biochem. Biophys. Res. Commun

    (2003)
  • G.W. Pasternak

    Anti-opioid activity of sigma1 systems

    Regulatory Peptides

    (1994)
  • G.W. Pasternak

    Incomplete cross tolerance and multiple mu opioid peptide receptors

    Trends Pharmacol. Sci

    (2001)
  • G.W. Pasternak

    Insights into mu opioid pharmacology—the role of mu opioid receptor subtypes

    Life Sci

    (2001)
  • G.W. Pasternak et al.

    Antisense mapping: assessing the functional significance of genes and splice variants

    Meth. Enzymol

    (2000)
  • G.W. Pasternak et al.

    Mapping of opioid receptors using antisense oligodeoxynucleotides: correlating their molecular biology and pharmacology

    Trends Pharmacol. Sci

    (1995)
  • G.W. Pasternak et al.

    An endogenous morphine like factor in mammalian brain

    Life Sci

    (1975)
  • G.W. Pasternak et al.

    Morphine-6-glucuronide, a potent mu agonist

    Life Sci

    (1987)
  • D. Paul et al.

    Differential blockade by naloxonazine of two μ opiate actions: analgesia and inhibition of gastrointestinal transit

    Eur. J. Pharmacol

    (1988)
  • D. Paul et al.

    Different μ receptor subtypes mediate spinal and supraspinal analgesia in mice

    Eur. J. Pharmacol

    (1989)
  • C.G. Pick et al.

    Genetic influences in opioid analgesic sensitivity in mice

    Brain Res

    (1991)
  • M.E.A. Reith et al.

    Strain differences in opiate receptors in mouse brain

    Eur. J. Pharmacol

    (1981)
  • G.C. Rossi et al.

    Antisense mapping the MOR-1 opioid receptor: evidence for alternative splicing and a novel morphine-6β-glucuronide receptor

    FEBS Lett

    (1995)
  • G.C. Rossi et al.

    Differential blockade of morphine and morphine-6β-glucuronide analgesia by antisense oligodeoxynucleotides directed against MOR-1 and G-protein α subunits in rats

    Neurosci. Lett

    (1995)
  • G.C. Rossi et al.

    Novel receptor mechanisms for heroin and morphine-6β-glucuronide analgesia

    Neurosci. Lett

    (1996)
  • P. Seth et al.

    Cloning and structural analysis of the cDNA and the gene encoding the murine type 1 sigma receptor

    Biochem. Biophys. Res. Commun

    (1997)
  • C. Abbadie et al.

    Immunohistochemical localization of the carboxy terminus of the novel mu opioid receptor splice variant MOR-1C within the human spinal cord

    Neuroreport

    (2000)
  • C. Abbadie et al.

    Differential distribution in rat brain of mu opioid receptor carboxy terminal splice variants MOR-1C and MOR-1-like immunoreactivity: evidence for region-specific processing

    J. Comp. Neurol

    (2000)
  • A. Baron et al.

    Opiate receptors in mice: genetic differences

    Life Sci

    (1975)
  • E.A. Bolan et al.

    Functional analysis of MOR-1 splice variants of the mu opioid receptor gene, Oprm

    Synapse

    (2004)
  • K.-J. Chang et al.

    Multiple opiate receptors: different regional distribution in the brain and differential binding of opiates and opioid peptides

    Mol. Pharmacol

    (1979)
  • Y. Chen et al.

    Molecular cloning and functional expression of a μ-opioid receptor from rat brain

    Mol. Pharmacol

    (1993)
  • Y. Chen et al.

    Molecular cloning of a rat kappa opioid receptor reveals sequence similarities to the μ and δ opioid receptors

    Biochem. J

    (1993)
  • N. Cherny et al.

    Strategies to manage the adverse effects of oral morphine: an evidence-based report

    J. Clin. Oncol

    (2001)
  • C.-C. Chien et al.

    Selective antagonism of opioid analgesia by a sigma system

    J. Pharmacol. Exp. Ther

    (1994)
  • J.A. Clark et al.

    Kappa opiate receptor multiplicity: evidence for two U50, 488-sensitive kappa1 subtypes and a novel kappa3 subtype

    J. Pharmacol. Exp. Ther

    (1989)
  • Y.-L. Du et al.

    A splice variant of the mu opioid receptor is present in human SHSY-5Y cells

    Soc. Neurosci

    (1997)
  • C.J. Evans et al.

    The opioid peptides

  • C.J. Evans et al.

    Cloning of a delta opioid receptor by functional expression

    Science

    (1992)
  • C.M. Flores et al.

    The pharmacogenetics of analgesia: toward a genetically-based approach to pain management

    Pharmacogenomics

    (2001)
  • Cited by (164)

    • Members of the same pharmacological family are not alike: Different opioids, different consequences, hope for the opioid crisis?

      2019, Progress in Neuro-Psychopharmacology and Biological Psychiatry
      Citation Excerpt :

      That is to say, a particular agonist with a higher κ:μ affinity ratio would show increased KOR-specific signaling patterns globally, as compared to an agonist with a lower κ:μ affinity ratio. When one also considers ligand-specific differences in affinity for opioid receptor subtype splice variants (Pasternak, 2004), region-specific differences in splice variant expression patterns (Abbadie et al., 2000a; Abbadie et al., 2000b; Abbadie et al., 2000c), and the possibility of differential non-canonical pathway activation across splice variants (q.v. Section 3.4.2), a truly enormous degree of nuance in biological response to various opioid agonists appears possible, influenced by individual genetics/epigenetics, tissue type, brain region, time, and prior drug exposure. It is important to note that receptor-mediated signaling and ligand biases are cell-specific.

    • Stress and Opioid Systems

      2017, Hormones, Brain and Behavior: Third Edition
    View all citing articles on Scopus
    View full text