You are on page 1of 5

Acta Anaesthesiologica Taiwanica 49 (2011) 21e25

Contents lists available at ScienceDirect

Acta Anaesthesiologica Taiwanica


journal homepage: www.e-aat.com

Review Article

Mu opioid receptors in pain management


Gavril Pasternak 1, 2 *, Ying-Xian Pan 1, 2
1
Department of Neurology, Memorial Sloan-Kettering Cancer Center, NY, USA.
2
Department of Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, NY, USA.

a r t i c l e i n f o a b s t r a c t

Article history: Most of the potent analgesics currently in use act through the mu opioid receptor. Although they are
Received 26 October 2010 classified as mu opioids, clinical experience suggests differences among them. The relative potencies of the
Received in revised form agents can vary from patient to patient, as well as the side-effect profiles. These observations, coupled with
30 November 2010
pharmacological approaches in preclinical models, led to the suggestion of multiple subtypes of mu
Accepted 3 December 2010
receptors. The explosion in molecular biology has led to the identification of a single gene encoding mu
opioid receptors. It now appears that this gene undergoes extensive splicing, in which a single gene can
Key words:
generate multiple proteins. Evidence now suggests that these splice variants may help explain the clinical
pain;
alternative splicing;
variability in responses among patients.
pharmacogenomics; Copyright Ó 2011, Taiwan Society of Anesthesiologists. Published by Elsevier Taiwan LLC. All rights
analgesics, opioid reserved.

1. Introduction need to be individualized. Opioid Rotation also illustrates differ-


ences among the mu opioids. In Opioid Rotation switching patients
Opiates have been used to manage pain for centuries. Opium, who are highly tolerant to one opioid to a different opioid often
which contains a number of alkaloids including morphine and restores the analgesic effectiveness and sensitivity.1,2 While all mu
codeine, was eaten, smoked, and used as a tincture. After their drugs display tolerance with prolonged dosing, cross-tolerance is
isolation, morphine and codeine were used individually. Thousands incomplete. That is, the tolerance to the second drug may not be
of analogs have been generated in an effort to avoid some of the as pronounced as the first, explaining why the switch is effective.
difficulties encountered with opiate use, such as respiratory All these features have raised questions about the mechanisms of
depression, sedation, and constipation. Most of these agents act these drugs, specifically how a single mu receptor could mediate
through mu opiate receptors, as defined by the selectivity of their these actions.
binding for the three classes of opioid receptors that have been
cloned. This is to be expected because the structure of morphine
was used as a template for the synthesis of most of them. Clinicians 2. Early studies of mu opiate receptor and multiplicity
have long observed that patients can have markedly varying
responses to mu opioids. Some patients may encounter severe side The first opioid receptor identified in binding assays in 197335
effects such as nausea and vomiting with one agent, whereas was the mu receptor. A receptor for the opiates had been predicted
having no difficulties at all with the other. For example, it is not years earlier from a variety of traditional pharmacological
uncommon to find patients with severe nausea/vomiting from approaches and the rigid structure activity studies of morphine and
morphine tolerating methadone without problems. Furthermore, related chemical scaffolds.6e8 Because the studies used morphine-
the analgesic activity of the opioids also can vary among patients. related probes, it is not surprising that the receptors were mu.
One patient may find one mu opioid to be quite effective, whereas However, several other families of opioid receptors have also been
a different drug may not work anywhere near as well. However, the identified (Table 1). In vivo studies led Martin to propose the exis-
same drug may not be the best for all patients. This variability tence of kappa receptors, in addition to the mu receptors.9,10 Delta
among patients has led to the accepted paradigm that analgesics receptors, which are selective for the enkephalins, were identified11
after the isolation and identification of endogenous opioids.12e14
Indeed, it turns out that the endogenous ligand for the kappa
* Corresponding author. Department of Neurology, Memorial Sloan-Kettering
receptors is another endogenous opioid, dynorphin A.15,16 Work is
Cancer Center, 1275 York Ave, New York, NY 10065, USA. Tel.: þ1 646 888 2165. in progress developing new selective delta and kappa opioids for
E-mail: pasterng@mskcc.org (G. Pasternak). clinical use, but for the most part the drugs available clinically are

1875-4597/$ e see front matter Copyright Ó 2011, Taiwan Society of Anesthesiologists. Published by Elsevier Taiwan LLC. All rights reserved.
doi:10.1016/j.aat.2010.12.008
22 G. Pasternak, Y.-X. Pan

Table 1
Classification of the opioid receptor families.

Receptor Drugs Analgesia Physical dependence Dysphoria Respiratory depression Gene


Mu Morphine Yes Yes No Yes MOR-1

Kappa Kappa1 Dynorphin A Yes No Yes No KOR-1


U50, 488H
Kappa2 Ethylketocyclazocine Yes No Yes No KOR-1/DOR-1
Kappa3 Naloxone benzoylhydrazone Yes No No No See below
Levorphanol

Delta Enkephalin Yes No ? No DOR-1


DPDPE

Three major families of opioid receptors have been proposed. Subtypes of all have been proposed using pharmacological and molecular biological approaches. The genes for all
have been established, except for kappa3. This subtype, which is important in the analgesic actions of a variety of opioid analgesics, including levorphanol and nalbuphine, is
most likely a heterodimer involving one of the truncated 6TM MOR-1 splice variants and an unknown second receptor.64 However, this remains to be confirmed. DPDPE:
[D-Pen2,D-Pen5]enkephalin.

mu. There are several mixed agonists/antagonists that interact with receptor binding assays. The new site was termed mu1, whereas
both mu and kappa receptors, but they have limited use. the original morphine-selective site was termed mu2.18 Under-
The first suggestion that there may be more than one class of standing the pharmacological significance of these two mu
mu receptors came from receptor binding studies in the mid 1970s receptors was facilitated by the development of selective antago-
that revealed a novel site with unusual characteristics.17 Although nists that could selectively block the mu1 site and not the mu2
mu drugs like morphine labeled it with high potency, confirming site.19e21 The naloxonazine-sensitive site corresponds to the mu1
its classification as a mu receptor, its ability to bind other drugs site, whereas the naloxonzine-insensitive mu site is mu2. When
clearly distinguished it from the original mu receptor seen in given in vivo, these antagonists blocked morphine analgesia, but

Fig. 1. Schematic of the human mu opioid receptor (OPRM1) gene structure and alternative splicing. A. Gene structure of the human OPRM1 gene. Exons and introns are showed by
boxes and horizontal lines, respectively. Intron size is indicated below the introns as kilobases (kb). Promoters are showed by arrows. B. Alternatively spliced variants of the human
OPRM1 gene. Exon composition for each alternatively spliced variant was showed by appropriate exon boxes. The lines between exons are introns that are spliced out during
splicing. Translation start and stop points are shown by bars below and above exon boxes, respectively. From the literature.68
Mu opioid receptors in pain management 23

not respiratory depression or the inhibition of gastrointestinal within the mRNA responsible for making a protein.46e48 Certain
transit or many of the signs of physical dependence.22e27 Together, exon targets were important in morphine analgesia, including
these studies implied that drugs such as morphine were acting exons 1 and 4. However, antisense probes targeting exons 2 and 3
through more than one mu receptor. had little effect on morphine although they significantly dimin-
ished the activity of heroin and morphine-6b-glucuronide. Thus, at
3. Molecular biology of mu opiate receptors the molecular level there does appear to be receptor differences for
these mu opiates.
The opiate receptors were first cloned in 1992 with the isolation The association of MOR-1 with morphine analgesia was further
of the delta receptor DOR-1 by two independent groups.28,29 This confirmed by the generation of several different knockout animals
initial report confirmed the existence of the receptors within the G- in which the MOR-1 gene was disrupted. In all of these different
protein coupled receptor family, one of the largest families of knockout animals, which targeted exons 1, 2, and/or 3, morphine
receptors known. Based on these structures, the other members of lost all activity.49e52 However, in one exon 1 knockout mouse,
the opiate receptor family were quickly cloned, including the heroin and M6G were still analgesic. This was quite confusing and it
mu3032 and the kappa1 receptors.33e35 Soon after, yet another was initially suggested that these drugs were activating alternative
receptor was identified with a strong homology to the opiate opioid receptors, such as delta or kappa1. This possibility was
receptors36e41 but which was subsequently found to have a previ- eliminated with the finding that the residual heroin and morphine-
ously unknown peptide as its endogenous ligand, orphanan FQ/ 6b-glucuronide analgesia remained in a triple knockout mouse,
nociceptin.42,43 which had the same disruption of exon 1 of MOR-1 and additional
The mu receptors are the most important class of receptors disruptions of the delta (DOR-1) and kappa1 (KOR-1) receptors.
clinically. Most of the drugs used clinically act through these What made this particular knockout mouse interesting was the
receptors and understanding their actions is important in defining continuous presence of a number of MOR-1 splice variants.52 Some
their pharmacology. Pharmacological studies suggested more than of these, under the control of the exon 11 promoter, do not contain
one class of mu opiate receptor. This has now been confirmed at the exon 1, perhaps explaining their continuous expression.
molecular level. Although only a single mu receptor gene has been The splice variants associated with the mu opiate receptor can
reported, the receptor undergoes extensive alternative splicing to be separated into two categories based on their promoter. The gene
generate a host of splice variants (Fig. 1). encoding MOR-1 is quite complex with two distinct promoters. The
After the cloning of MOR-1, much effort was placed on con- primary one is associated with exon 153,54 and generates a number
firming its pharmacological relevance. The first studies involved of variants that are spliced at the 30 end in mice, rats, and humans
using an antisense approach in which very short sequences of (Fig. 1).55e62 These are interesting from several perspectives. They
antisense targeted specific sequences of the mRNA, leading to its are full length seven transmembrane domain G-protein coupled
degradation.44,45 These initial reports quickly established that receptors. Structurally, they differ only at the very tip of the intra-
MOR-1 was critical in the production of morphine analgesia. cellular C-terminus. The initial opiate receptor cloned, MOR-1,
However, the antisense approach can be used more extensively to contains 12 amino acids at the very tip of the C-terminus encoded
perform antisense mapping to assess the presence of specific exons by exon 4 (Fig. 2). In the other C-terminus spliced variants, these 12

Fig. 2. Schematic of the human OPRM1 carboxyl terminal splice variants’ structures and amino acid sequences predicted from downstream exons of exon 3. The common receptor
structure of all the human carboxyl terminal splice variants is indicated by heavy lines across the lipid bilayer of the cell membrane. Splice junctions between exons are indicated by
arrows. The amino acid sequences predicted from different downstream exons are shown on the right along with the indicated splice variants. Potential phosphorylation sites are
indicated by the italic letters and the following symbols on the top of the letters. D: cAMP- and cGMP-dependent protein kinase phosphorylation site; p: tyrosine kinase phos-
phorylation site; *: casein kinase II phosphorylation site.
24 G. Pasternak, Y.-X. Pan

Table 2 Acknowledgments
Opioid analgesia in MOR-1 knockout mice.

Drug ED50 value (mg/kg, s.c.) This work was supported, in part, from grants from the National
Exon 1 KO Exon 11 KO
Institute on Drug Abuse to GWP (DA02615, DA07242, DA06241 and
DA00220) and YXP (DA013997 and DA029244).
WT KO Shift WT KO Shift
Morphine 5.0 >100 >20 1.6 2.6 1.6
Methadone 1.5 1.6 1.2 References
M6G 3.8 8.5 2.3 0.92 19.3 21
Heroin 1.2 4.0 2.7 0.58 3.2 6 1. Fine PG, Portenoy RK. Establishing “best practices” for opioid rotation:
Fentanyl 0.024 0.230 10 conclusions of an expert panel. J Pain Symptom Manage 2009;38:418e25.
2. Cherny N, Ripamonti C, Pereira J, Davis C, Fallon M, McQuay H, et al. Strategies
WT and KO mice with a disruption of exon 1 of MOR-1 were tested with the indi- to manage the adverse effects of oral morphine: an evidence-based report.
cated drug and their ED50 values determined.52 Other groups of mice were gener- J Clin Oncol 2001;19:2542e54.
ated with a disruption of exon 11. WT and KO mice were tested with the indicated 3. Pert CB, Snyder SH. Opiate receptor: demonstration in nervous tissue. Science
drug and their ED50 values determined.64 1973;179:1011e4.
ED50, median effective dose; WT, wildtype; KO, knockout. 4. Simon EJ, Hiller JM, Edelman I. Stereospecific binding of the potent narcotic
analgesic [3H]etorphine to rat-brain homogenate. Proc Natl Acad Sci U S A
1973;70:1947e9.
5. Terenius L. Characteristics of the “receptor” for narcotic analgesics in synaptic
amino acids are replaced with different sequences of varying plasma membrane from rat brain. Acta Pharmacol Toxicol 1973;33:377e84.
lengths. However, these differences are quite limited and the basic 6. Beckett AH, Casy AF. Synthetic analgesics: stereochemical considerations.
structure of the receptor with its seven transmembrane domains J Pharm Pharmacol 1954;6:986e1001.
7. Portoghese PS. A new concept on the mode of interaction of narcotic analgesics
and binding pocket are identical among them (Fig. 2). Thus, these with receptors. J Med Chem 1965;8:609e16.
splice variants share a common binding pocket and they all show 8. Goldstein A, Lowney LI, Pal BK. Stereospecific and nonspecific interactions of
high affinity and selectivity for mu opiates, as shown in receptor the morphine congener levorphanol in subcellular fractions of mouse brain.
Proc Natl Acad Sci U S A 1971;68:1742e7.
binding assays. Although the splicing has little effect on the ability 9. Martin WR. Opioid antagonists. Pharmacol Rev 1967;19:463e521.
of the mu opioids to bind to the receptor, the small structural 10. Gilbert PE, Martin WR. The effects of morphine and nalorphine-like drugs in
differences do impact the ability of mu opioids to activate the the nondependent, morphine-dependent and cyclazocine-dependent chronic
spinal dog. J Pharmacol Exp Ther 1976;198:66e82.
different splice variants. These differences in the activation profiles 11. Lord JAH, Waterfield AA, Hughes J, Kosterlitz HW. Endogenous opioid peptides:
of various mu opioids for the receptor variants may help explain the multiple agonists and receptors. Nature 1977;267:495e9.
subtle, but clear, differences among the various mu opioids seen 12. Pasternak GW. Opioids and opioid receptors: Neuroscience Research Program;
1974.
clinically. 13. Hughes J, Smith TW, Kosterlitz HW, Fothergill LA, Morgan BA, Morris HR.
The second set of variants is associated with exon 11, which is Identification of two related pentapeptides from the brain with potent opiate
located approximately 30 kilobases upstream of exon 1.57,63 These agonist activity. Nature 1975;258:577e9.
14. Terenius L. Effect of peptides and amino acids on dihydromorphine binding to
variants include a number with quite unique structures. Unlike the
the opiate receptor. J Pharm Pharmacol 1975;27:450e2.
exon 1-associated variants, which all encode full length, seven 15. Lowney LI, Gentleman SB, Goldstein A. A pituitary endorphin with novel
transmembrane domain receptors, many of the exon 11-associated properties. Life Sci 1979;24:2377e84.
variants generate truncated variants with only six transmembrane 16. Goldstein A, Tachibana S, Lowney LI, Hunkapiller M, Hood L. Dynorphin-(1-13),
an extraordinarily potent opioid peptide. Proc Natl Acad Sci U S A 1979;76:
domains. Despite their unique structure, there is much evidence 6666e70.
now to suggest that they are pharmacologically important. This 17. Pasternak GW, Snyder SH. Identification of a novel high affinity opiate receptor
derives from studies on mice in which all the exon 11-containing binding in rat brain. Nature 1975;253:563e5.
18. Wolozin BL, Pasternak GW. Classification of multiple morphine and enkephalin
variants are lost because of disruption of this region of the gene binding sites in the central nervous system. Proc Natl Acad Sci U S A
(Table 2).64 Interestingly, morphine analgesia was not affected. 1981;78:6181e5.
However, the analgesic actions of heroin and the morphine 19. Pasternak GW, Childers SR, Snyder SH. Multiple opiate receptors: evidence for
mediation of analgesia by a subpopulation of receptors. In: Way EL, editor.
metabolite morphine-6b-glucuronide were significantly reduced. Endogenous and exogenous opioid agonists and antagonists. Pergamon Press;
Looking at the drugs, it is apparent that the analgesic actions of mu 1979. p. 113e6.
opioids can be differentiated at the molecular level. 20. Pasternak GW, Childers SR, Snyder SH. Naloxazone, a long-acting opiate
antagonist: effects on analgesia in intact animals and on opiate receptor
binding in vitro. J Pharmacol Exp Ther 1980;214:455e62.
4. The future 21. Hahn EF, Carroll-Buatti M, Pasternak GW. Irreversible opiate agonists and
antagonists: the 14-hydroxydihydromorphinone azines. J Neurosci 1982;2:
572e6.
Pain management remains an art, dependent on the individu- 22. Hahn EF, Pasternak GW. Naloxonazine, a potent, long-acting inhibitor of opiate
alization of care. Not all patients can be managed with the same binding sites. Life Sci 1982;31:1385e8.
23. Ling GSF, Spiegel K, Nishimura S, Pasternak GW. Dissociation of morphine’s
drugs. Indeed, the complexity of the clinical responses has long been analgesic and respiratory depressant actions. Eur J Pharmacol 1983;86:
evident. Many factors may play a role, but evidence is accumulating 487e8.
that much of this variability may result from the complexity of mu 24. Wood PL, Pasternak GW. Specific mu2 opioid isoreceptor regulation of
nigrostraital neurons: in vivo evidence with naloxonazine. Neurosci Lett 1983;
receptors. Early work from our laboratory proposed two subtypes, 37:291e3.
mu1 and mu2, based on selective antagonists.18,20,65 Studies 25. Ling GSF, MacLeod JM, Lee S, Lockhart SH, Pasternak GW. Separation of
showing differences between morphine and morphine-6b-glucu- morphine analgesia from physical dependence. Science 1984;226:462e4.
26. Ling GSF, Spiegel K, Lockhart SH, Pasternak GW. Separation of opioid analgesia
ronide further implied a third subtype.48,66,67 However, the from respiratory depression: evidence for different receptor mechanisms.
molecular biology of the mu opioid receptor gene reveals the exis- J Pharmacol Exp Ther 1985;232:149e55.
tence of dozens of mu receptor splice variants. Correlating these 27. Ling GSF, Simantov R, Clark JA, Pasternak GW. Naloxonazine actions in vivo. Eur
J Pharmacol 1986;129:33e8.
splice variants to the pharmacologically defined receptors has
28. Kieffer BL, Befort K, Gaveriaux-Ruff C, Hirth CG. The d-opioid receptor: isolation
proven difficult. However, it seems likely that they play an impor- of a cDNA by expression cloning and pharmacological characterization. Proc
tant role in why patients respond so differently. As our under- Natl Acad Sci U S A 1992;89:12048e52.
standing progresses, the critical question is whether or not we can 29. Evans CJ, Keith Jr DE, Morrison H, Magendzo K, Edwards RH. Cloning of a delta
opioid receptor by functional expression. Science 1992;258:1952e5.
use this knowledge to develop novel and useful analgesic lacking 30. Chen Y, Mestek A, Liu J, Hurley JA, Yu L. Molecular cloning and functional
the detrimental actions of the currently available drugs. expression of a m-opioid receptor from rat brain. Mol Pharmacol 1993;44:8e12.
Mu opioid receptors in pain management 25

31. Thompson RC, Mansour A, Akil H, Watson SJ. Cloning and pharmacological 50. Sora I, Takahashi N, Funada M, Ujike H, Revay RS, Donovan DM, et al. Opiate
characterization of a rat m-opioid receptor. Neuron 1993;11:903e13. receptor knockout mice define m receptor roles in endogenous nociceptive
32. Wang JB, Imai Y, Eppler CM, Gregor P, Spivak CE, Uhl GR. m opiate receptor: responses and morphine-induced analgesia. Proc Natl Acad Sci U S A 1997;94:
cDNA cloning and expression. Proc Natl Acad Sci U S A 1993;90:10230e4. 1544e9.
33. Chen Y, Mestek A, Liu J, Yu L. Molecular cloning of a rat kappa opioid receptor 51. Loh HH, Liu HC, Cavalli A, Yang WL, Chen YF, Wei LN. m opioid receptor
reveals sequence similarities to the m and d opioid receptors. Biochem J knockout in mice: effects on ligand-induced analgesia and morphine lethality.
1993;295:625e8. Mol Brain Res 1998;54:321e6.
34. Li S, Zhu J, Chen C, Chen Y-W, Deriel JK, Ashby B, et al. Molecular cloning and 52. Schuller AG, King MA, Zhang J, Bolan E, Pan YX, Morgan DJ, et al. Retention of
expression of a rat kappa opioid receptor. Biochem J 1993;295:629e33. heroin and morphine-6 beta-glucuronide analgesia in a new line of mice
35. Meng F, Xie G-X, Thompson RC, Mansour A, Goldstein A, Watson SJ, et al. lacking exon 1 of MOR-1. Nat Neurosci 1999;2:151e6.
Cloning and pharmacological characterization of a rat kappa opioid receptor. 53. Liang Y, Mestek A, Yu L, Carr LG. Cloning and characterization of the promoter
Proc Natl Acad Sci U S A 1993;90:9954e8. region of the mouse m opioid receptor gene. Brain Res 1995;679:82e8.
36. Bunzow JR, Saez C, Mortrud M, Bouvier C, Williams JT, Low M, et al. Molecular 54. Mayer P, Schulzeck S, Kraus J, Zimprich A, Höllt V. Promoter region and
cloning and tissue distribution of a putative member of the rat opioid receptor alternatively spliced exons of the rat m-opioid receptor gene. J Neurochem
gene family that is not a m, d or kappa opioid receptor type. FEBS Lett 1996;66:2272e8.
1994;347:284e8. 55. Pan L, Xu J, Yu R, Xu MM, Pan YX, Pasternak GW. Identification and charac-
37. Chen Y, Fan Y, Liu J, Mestek A, Tian M, Kozak CA, et al. Molecular cloning, tissue terization of six new alternatively spliced variants of the human mu opioid
distribution and chromosomal localization of a novel member of the opioid receptor gene, Oprm. Neuroscience 2005;133:209e20.
receptor gene family. FEBS Lett 1994;347:279e83. 56. Pan Y-X, Xu J, Rossi GC, Leventhal L, Wan B-L, Zuckerman A, et al. Cloning and
38. Fukuda K, Kato S, Mori K, Nishi M, Takeshima H, Iwabe N, et al. cDNA cloning expression of a novel splice variant of the mouse mu-opioid receptor (MOR-1)
and regional distribution of a novel member of the opioid receptor family. FEBS gene. Soc Neurosci 1998;24:524.
Lett 1994;343:42e6. 57. Pan Y-X, Xu J, Mahurter L, Bolan EA, Xu MM, Pasternak GW. Generation of the
39. Mollereau C, Parmentier M, Mailleux P, Butour JL, Moisand C, Chalon P, et al. mu opioid receptor (MOR-1) protein by three new splice variants of the Oprm
ORL-1, a novel member of the opioid family: cloning, functional expression and gene. Proc Natl Acad Sci U S A 2001;98:14084e9.
localization. FEBS Lett 1994;341:33e8. 58. Pan YX, Xu J, Bolan EA, Abbadie C, Chang A, Zuckerman A, et al. Identification
40. Pan Y-X, Cheng J, Xu J, Pasternak GW. Cloning, expression and classification of and characterization of three new alternatively spliced mu opioid receptor
a kappa3-related opioid receptor using antisense oligodeoxynucleotides. Regul isoforms. Mol Pharmacol 1999;56:396e403.
Pept 1994;54:217e8. 59. Pan YX, Xu J, Mahurter L, Xu M, Gilbert AK, Pasternak GW. Identification and
41. Pan Y-X, Cheng J, Xu J, Rossi GC, Jacobson E, Ryan-Moro J, et al. Cloning and characterization of two new human mu opioid receptor splice variants, hMOR-
functional characterization through antisense mapping of a kappa3-related 1O and hMOR-1X. Biochem Biophys Res Commun 2003;301:1057e61.
opioid receptor. Mol Pharmacol 1995;47:1180e8. 60. Pan YX, Xu J, Bolan E, Moskowitz HS, Xu M, Pasternak GW. Identification of four
42. Meunier JC, Mollereau C, Toll L, Suaudeau C, Moisand C, Alvinerie P, et al. novel exon 5 splice variants of the mouse mu-opioid receptor gene: functional
Isolation and structure of the endogenous agonist of the opioid receptor like consequences of C-terminal splicing. Mol Pharmacol 2005;68:866e75.
ORL1 receptor. Nature 1995;377:532e5. 61. Pan YX, Xu J, Bolan E, Chang A, Mahurter L, Rossi G, et al. Isolation and
43. Reinscheid RK, Nothacker HP, Bourson A, Ardati A, Henningsen RA, Bunzow JR, expression of a novel alternatively spliced mu opioid receptor isoform, MOR-
et al. Orphanin FQ: a neuropeptide that activates an opioid like G protein- 1F. FEBS Lett 2000;466:337e40.
coupled receptor. Science 1995;270:792e4. 62. Pasternak DA, Pan L, Xu J, Yu R, Xu MM, Pasternak GW, et al. Identification of
44. Rossi GC, Pan Y-X, Cheng J, Pasternak GW. Blockade of morphine analgesia by three new alternatively spliced variants of the rat mu opioid receptor gene:
an antisense oligodeoxynucleotide against the mu receptor. Life Sci 1994;54: dissociation of affinity and efficacy. J Neurochem 2004;91:881e90.
L375e9. 63. Xu J, Xu M, Hurd YL, Pasternak GW, Pan YX. Isolation and characterization of
45. Uhl GR, Childers S, Pasternak GW. An opiate-receptor gene family reunion. new exon 11-associated N-terminal splice variants of the human mu opioid
Trends Neurosci 1994;17:89e93. receptor gene. J Neurochem 2009;108:962e72.
46. Standifer KM, Chien C-C, Wahlestedt C, Brown GP, Pasternak GW. Selective loss 64. Pan YX, Xu J, Xu M, Rossi GC, Matulonis JE, Pasternak GW. Involvement of exon
of d opioid analgesia and binding by antisense oligodeoxynucleotides to 11-associated variants of the mu opioid receptor MOR-1 in heroin, but not
a d opioid receptor. Neuron 1994;12:805e10. morphine, actions. Proc Natl Acad Sci U S A 2009;106:4917e22.
47. Rossi GC, Standifer KM, Pasternak GW. Differential blockade of morphine and 65. Pasternak GW, Childers SR, Snyder SH. Opiate analgesia: evidence for media-
morphine-6b-glucuronide analgesia by antisense oligodeoxynucleotides tion by a subpopulation of opiate receptors. Science 1980;208:514e6.
directed against MOR-1 and G-protein a subunits in rats. Neurosci Lett 66. Rossi GC, Leventhal L, Pan YX, Cole J, Su W, Bodnar RJ, et al. Antisense mapping
1995;198:99e102. of MOR-1 in rats: distinguishing between morphine and morphine-6beta-
48. Rossi GC, Pan Y-X, Brown GP, Pasternak GW. Antisense mapping the MOR-1 glucuronide antinociception. J Pharmacol Exp Ther 1997;281:109e14.
opioid receptor: evidence for alternative splicing and a novel morphine-6b- 67. Rossi GC, Brown GP, Leventhal L, Yang K, Pasternak GW. Novel receptor
glucuronide receptor. FEBS Lett 1995;369:192e6. mechanisms for heroin and morphine-6b-glucuronide analgesia. Neurosci Lett
49. Matthes HWD, Maldonado R, Simonin F, Valverde O, Slowe S, Kitchen I, et al. 1996;216:1e4.
Loss of morphine-induced analgesia, reward effect and withdrawal symptoms 68. Pan Y-X, Pasternak GW. Molecular biology of mu opioid receptors. In:
in mice lacking the m-opioid-receptor gene. Nature 1996;383:819e23. Pasternak GW, editor. The opiate receptors. 2nd ed. Humana Press; 2011.

You might also like