AJA Asian Journal of Anesthesiology

Advancing, Capability, Improving lives

Review Article
Volume 49, Issue 1, Pages 21-25
Gavril Pasternak 1.2 , Ying-Xian Pan 1.2
2077 Views


Abstract

Most of the potent analgesics currently in use act through the mu opioid receptor. Although they are classified as mu opioids, clinical experience suggests differences among them. The relative potencies of the agents can vary from patient to patient, as well as the side-effect profiles. These observations, coupled with pharmacological approaches in preclinical models, led to the suggestion of multiple subtypes of mu 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 generate multiple proteins. Evidence now suggests that these splice variants may help explain the clinical variability in responses among patients.

Keywords

pain; alternative splicing; pharmacogenomics; analgesics, opioid;


1. Introduction

Opiates have been used to manage pain for centuries. Opium, which contains a number of alkaloids including morphine and codeine, was eaten, smoked, and used as a tincture. After their isolation, morphine and codeine were used individually. Thousands of analogs have been generated in an effort to avoid some of the difficulties encountered with opiate use, such as respiratory depression, sedation, and constipation. Most of these agents act through mu opiate receptors, as defined by the selectivity of their 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 have long observed that patients can have markedly varying responses to mu opioids. Some patients may encounter severe side effects such as nausea and vomiting with one agent, whereas having no difficulties at all with the other. For example, it is not uncommon to find patients with severe nausea/vomiting from morphine tolerating methadone without problems. Furthermore, the analgesic activity of the opioids also can vary among patients. One patient may find one mu opioid to be quite effective, whereas a different drug may not work anywhere near as well. However, the same drug may not be the best for all patients. This variability among patients has led to the accepted paradigm that analgesics need to be individualized. Opioid Rotation also illustrates differences among the mu opioids. In Opioid Rotation switching patients who are highly tolerant to one opioid to a different opioid often restores the analgesic effectiveness and sensitivity.12 While all mu drugs display tolerance with prolonged dosing, cross-tolerance is incomplete. That is, the tolerance to the second drug may not be as pronounced as the first, explaining why the switch is effective. All these features have raised questions about the mechanisms of these drugs, specifically how a single mu receptor could mediate these actions.

2. Early studies of mu opiate receptor and multiplicity

The first opioid receptor identified in binding assays in 1973345 was the mu receptor. A receptor for the opiates had been predicted years earlier from a variety of traditional pharmacological approaches and the rigid structure activity studies of morphine and related chemical scaffolds.678 Because the studies used morphine-related probes, it is not surprising that the receptors were mu. However, several other families of opioid receptors have also been identified (Table 1). In vivo studies led Martin to propose the existence of kappa receptors, in addition to the mu receptors.910 Delta receptors, which are selective for the enkephalins, were identified11 after the isolation and identification of endogenous opioids.121314 Indeed, it turns out that the endogenous ligand for the kappa receptors is another endogenous opioid, dynorphin A.1516 Work is in progress developing new selective delta and kappa opioids for clinical use, but for the most part the drugs available clinically are mu. There are several mixed agonists/antagonists that interact with both mu and kappa receptors, but they have limited use.

The first suggestion that there may be more than one class of mu receptors came from receptor binding studies in the mid 1970s that revealed a novel site with unusual characteristics.17 Although mu drugs like morphine labeled it with high potency, confirming its classification as a mu receptor, its ability to bind other drugs clearly distinguished it from the original mu receptor seen in receptor binding assays. The new site was termed mu1, whereas the original morphine-selective site was termed mu2.18 Understanding the pharmacological significance of these two mu receptors was facilitated by the development of selective antagonists that could selectively block the mu1 site and not the mu2 site.192021 The naloxonazine-sensitive site corresponds to the mu1 site, whereas the naloxonzine-insensitive mu site is mu2. When given in vivo, these antagonists blocked morphine analgesia, but not respiratory depression or the inhibition of gastrointestinal transit or many of the signs of physical dependence.222324252627 Together, these studies implied that drugs such as morphine were acting through more than one mu receptor.

3. Molecular biology of mu opiate receptors

The opiate receptors were first cloned in 1992 with the isolation of the delta receptor DOR-1 by two independent groups.2829 This initial report confirmed the existence of the receptors within the G-protein coupled receptor family, one of the largest families of receptors known. Based on these structures, the other members of the opiate receptor family were quickly cloned, including the mu303132 and the kappa1 receptors.333435 Soon after, yet another receptor was identified with a strong homology to the opiate receptors363738394041 but which was subsequently found to have a previously unknown peptide as its endogenous ligand, orphanan FQ/nociceptin.4243

The mu receptors are the most important class of receptors clinically. Most of the drugs used clinically act through these receptors and understanding their actions is important in defining their pharmacology. Pharmacological studies suggested more than one class of mu opiate receptor. This has now been confirmed at the molecular level. Although only a single mu receptor gene has been reported, the receptor undergoes extensive alternative splicing to generate a host of splice variants (Fig. 1).

Fig. 1.
Download full-size image
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

After the cloning of MOR-1, much effort was placed on confirming its pharmacological relevance. The first studies involved using an antisense approach in which very short sequences of antisense targeted specific sequences of the mRNA, leading to its degradation.4445 These initial reports quickly established that MOR-1 was critical in the production of morphine analgesia. However, the antisense approach can be used more extensively to perform antisense mapping to assess the presence of specific exons within the mRNA responsible for making a protein.464748 Certain exon targets were important in morphine analgesia, including exons 1 and 4. However, antisense probes targeting exons 2 and 3 had little effect on morphine although they significantly diminished the activity of heroin and morphine-6β-glucuronide. Thus, at the molecular level there does appear to be receptor differences for these mu opiates.

The association of MOR-1 with morphine analgesia was further confirmed by the generation of several different knockout animals in which the MOR-1 gene was disrupted. In all of these different knockout animals, which targeted exons 1, 2, and/or 3, morphine lost all activity.49505152 However, in one exon 1 knockout mouse, heroin and M6G were still analgesic. This was quite confusing and it was initially suggested that these drugs were activating alternative opioid receptors, such as delta or kappa1. This possibility was eliminated with the finding that the residual heroin and morphine-6β-glucuronide analgesia remained in a triple knockout mouse, which had the same disruption of exon 1 of MOR-1 and additional disruptions of the delta (DOR-1) and kappa1 (KOR-1) receptors. What made this particular knockout mouse interesting was the continuous presence of a number of MOR-1 splice variants.52 Some of these, under the control of the exon 11 promoter, do not contain exon 1, perhaps explaining their continuous expression.

The splice variants associated with the mu opiate receptor can be separated into two categories based on their promoter. The gene encoding MOR-1 is quite complex with two distinct promoters. The primary one is associated with exon 15354 and generates a number of variants that are spliced at the 3′ end in mice, rats, and humans (Fig. 1).5556575859606162 These are interesting from several perspectives. They are full length seven transmembrane domain G-protein coupled receptors. Structurally, they differ only at the very tip of the intracellular C-terminus. The initial opiate receptor cloned, MOR-1, contains 12 amino acids at the very tip of the C-terminus encoded by exon 4 (Fig. 2). In the other C-terminus spliced variants, these 12 amino acids are replaced with different sequences of varying lengths. However, these differences are quite limited and the basic structure of the receptor with its seven transmembrane domains and binding pocket are identical among them (Fig. 2). Thus, these splice variants share a common binding pocket and they all show high affinity and selectivity for mu opiates, as shown in receptor binding assays. Although the splicing has little effect on the ability of the mu opioids to bind to the receptor, the small structural differences do impact the ability of mu opioids to activate the different splice variants. These differences in the activation profiles of various mu opioids for the receptor variants may help explain the subtle, but clear, differences among the various mu opioids seen clinically.

Fig. 2.
Download full-size image
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. Δ: cAMP- and cGMP-dependent protein kinase phosphorylation site; о: tyrosine kinase phosphorylation site; ∗: casein kinase II phosphorylation site.

The second set of variants is associated with exon 11, which is located approximately 30 kilobases upstream of exon 1.5763 These variants include a number with quite unique structures. Unlike the exon 1-associated variants, which all encode full length, seven transmembrane domain receptors, many of the exon 11-associated variants generate truncated variants with only six transmembrane domains. Despite their unique structure, there is much evidence now to suggest that they are pharmacologically important. This derives from studies on mice in which all the exon 11-containing variants are lost because of disruption of this region of the gene (Table 2).64 Interestingly, morphine analgesia was not affected. However, the analgesic actions of heroin and the morphine metabolite morphine-6β-glucuronide were significantly reduced. Looking at the drugs, it is apparent that the analgesic actions of mu opioids can be differentiated at the molecular level.

4. The future

Pain management remains an art, dependent on the individualization of care. Not all patients can be managed with the same drugs. Indeed, the complexity of the clinical responses has long been evident. Many factors may play a role, but evidence is accumulating that much of this variability may result from the complexity of mu receptors. Early work from our laboratory proposed two subtypes, mu1 and mu2, based on selective antagonists.182065 Studies showing differences between morphine and morphine-6β-glucuronide further implied a third subtype.486667 However, the molecular biology of the mu opioid receptor gene reveals the existence of dozens of mu receptor splice variants. Correlating these splice variants to the pharmacologically defined receptors has proven difficult. However, it seems likely that they play an important role in why patients respond so differently. As our understanding progresses, the critical question is whether or not we can use this knowledge to develop novel and useful analgesic lacking the detrimental actions of the currently available drugs.

Acknowledgments

This work was supported, in part, from grants from the National Institute on Drug Abuse to GWP (DA02615, DA07242, DA06241 and DA00220) and YXP (DA013997 and DA029244).


References

1
P.G. Fine, R.K. Portenoy
Establishing “best practices” for opioid rotation: conclusions of an expert panel
J Pain Symptom Manage, 38 (2009), pp. 418-425
2
N. Cherny, C. Ripamonti, J. Pereira, C. Davis, M. Fallon, H. McQuay, et al.
Strategies to manage the adverse effects of oral morphine: an evidence-based report
J Clin Oncol, 19 (2001), pp. 2542-2554
3
C.B. Pert, S.H. Snyder
Opiate receptor: demonstration in nervous tissue
Science, 179 (1973), pp. 1011-1014
4
E.J. Simon, J.M. Hiller, I. Edelman
Stereospecific binding of the potent narcotic analgesic [3H]etorphine to rat-brain homogenate
Proc Natl Acad Sci U S A, 70 (1973), pp. 1947-1949
5
L. Terenius
Characteristics of the “receptor” for narcotic analgesics in synaptic plasma membrane from rat brain
Acta Pharmacol Toxicol, 33 (1973), pp. 377-384
6
A.H. Beckett, A.F. Casy
Synthetic analgesics: stereochemical considerations
J Pharm Pharmacol, 6 (1954), pp. 986-1001
7
P.S. Portoghese
A new concept on the mode of interaction of narcotic analgesics with receptors
J Med Chem, 8 (1965), pp. 609-616
8
A. Goldstein, L.I. Lowney, B.K. Pal
Stereospecific and nonspecific interactions of the morphine congener levorphanol in subcellular fractions of mouse brain
Proc Natl Acad Sci U S A, 68 (1971), pp. 1742-1747
9
W.R. Martin
Opioid antagonists
Pharmacol Rev, 19 (1967), pp. 463-521
10
P.E. Gilbert, W.R. Martin
The effects of morphine and nalorphine-like drugs in the nondependent, morphine-dependent and cyclazocine-dependent chronic spinal dog
J Pharmacol Exp Ther, 198 (1976), pp. 66-82
11
J.A.H. Lord, A.A. Waterfield, J. Hughes, H.W. Kosterlitz
Endogenous opioid peptides: multiple agonists and receptors
Nature, 267 (1977), pp. 495-499
12
Pasternak GW. Opioids and opioid receptors: Neuroscience Research Program; 1974.
Article  
13
J. Hughes, T.W. Smith, H.W. Kosterlitz, L.A. Fothergill, B.A. Morgan, H.R. Morris
Identification of two related pentapeptides from the brain with potent opiate agonist activity
Nature, 258 (1975), pp. 577-579
14
L. Terenius
Effect of peptides and amino acids on dihydromorphine binding to the opiate receptor
J Pharm Pharmacol, 27 (1975), pp. 450-452
15
L.I. Lowney, S.B. Gentleman, A. Goldstein
A pituitary endorphin with novel properties
Life Sci, 24 (1979), pp. 2377-2384
16
A. Goldstein, S. Tachibana, L.I. Lowney, M. Hunkapiller, L. Hood
Dynorphin-(1-13), an extraordinarily potent opioid peptide
Proc Natl Acad Sci U S A, 76 (1979), pp. 6666-6670
17
G.W. Pasternak, S.H. Snyder
Identification of a novel high affinity opiate receptor binding in rat brain
Nature, 253 (1975), pp. 563-565
18
B.L. Wolozin, G.W. Pasternak
Classification of multiple morphine and enkephalin binding sites in the central nervous system
Proc Natl Acad Sci U S A, 78 (1981), pp. 6181-6185
19
G.W. Pasternak, S.R. Childers, S.H. Snyder
Multiple opiate receptors: evidence for mediation of analgesia by a subpopulation of receptors
E.L. Way (Ed.), Endogenous and exogenous opioid agonists and antagonists, Pergamon Press (1979), pp. 113-116
Article  
20
G.W. Pasternak, S.R. Childers, S.H. Snyder
Naloxazone, a long-acting opiate antagonist: effects on analgesia in intact animals and on opiate receptor binding in vitro
J Pharmacol Exp Ther, 214 (1980), pp. 455-462
21
E.F. Hahn, M. Carroll-Buatti, G.W. Pasternak
Irreversible opiate agonists and antagonists: the 14-hydroxydihydromorphinone azines
J Neurosci, 2 (1982), pp. 572-576
22
E.F. Hahn, G.W. Pasternak
Naloxonazine, a potent, long-acting inhibitor of opiate binding sites
Life Sci, 31 (1982), pp. 1385-1388
23
G.S.F. Ling, K. Spiegel, S. Nishimura, G.W. Pasternak
Dissociation of morphine’s analgesic and respiratory depressant actions
Eur J Pharmacol, 86 (1983), pp. 487-488
Article  
24
P.L. Wood, G.W. Pasternak
Specific mu2 opioid isoreceptor regulation of nigrostraital neurons: in vivo evidence with naloxonazine
Neurosci Lett, 37 (1983), pp. 291-293
25
G.S.F. Ling, J.M. MacLeod, S. Lee, S.H. Lockhart, G.W. Pasternak
Separation of morphine analgesia from physical dependence
Science, 226 (1984), pp. 462-464
26
G.S.F. Ling, K. Spiegel, S.H. Lockhart, G.W. Pasternak
Separation of opioid analgesia from respiratory depression: evidence for different receptor mechanisms
J Pharmacol Exp Ther, 232 (1985), pp. 149-155
27
G.S.F. Ling, R. Simantov, J.A. Clark, G.W. Pasternak
Naloxonazine actions in vivo
Eur J Pharmacol, 129 (1986), pp. 33-38
28
B.L. Kieffer, K. Befort, C. Gaveriaux-Ruff, C.G. Hirth
The d-opioid receptor: isolation of a cDNA by expression cloning and pharmacological characterization
Proc Natl Acad Sci U S A, 89 (1992), pp. 12048-12052
29
C.J. Evans, D.E. Keith Jr., H. Morrison, K. Magendzo, R.H. Edwards
Cloning of a delta opioid receptor by functional expression
Science, 258 (1992), pp. 1952-1955
30
Y. Chen, A. Mestek, J. Liu, J.A. Hurley, L. Yu
Molecular cloning and functional expression of a m-opioid receptor from rat brain
Mol Pharmacol, 44 (1993), pp. 8-12
31
R.C. Thompson, A. Mansour, H. Akil, S.J. Watson
Cloning and pharmacological characterization of a rat μ-opioid receptor
Neuron, 11 (1993), pp. 903-913
32
J.B. Wang, Y. Imai, C.M. Eppler, P. Gregor, C.E. Spivak, G.R. Uhl
μ opiate receptor: cDNA cloning and expression
Proc Natl Acad Sci U S A, 90 (1993), pp. 10230-10234
33
Y. Chen, A. Mestek, J. Liu, L. Yu
Molecular cloning of a rat kappa opioid receptor reveals sequence similarities to the m and d opioid receptors
Biochem J, 295 (1993), pp. 625-628
34
S. Li, J. Zhu, C. Chen, Y.-W. Chen, J.K. Deriel, B. Ashby, et al.
Molecular cloning and expression of a rat kappa opioid receptor
Biochem J, 295 (1993), pp. 629-633
35
F. Meng, G.-X. Xie, R.C. Thompson, A. Mansour, A. Goldstein, S.J. Watson, et al.
Cloning and pharmacological characterization of a rat kappa opioid receptor
Proc Natl Acad Sci U S A, 90 (1993), pp. 9954-9958
36
J.R. Bunzow, C. Saez, M. Mortrud, C. Bouvier, J.T. Williams, M. Low, et al.
Molecular cloning and tissue distribution of a putative member of the rat opioid receptor gene family that is not a μ, δ or kappa opioid receptor type
FEBS Lett, 347 (1994), pp. 284-288
37
Y. Chen, Y. Fan, J. Liu, A. Mestek, M. Tian, C.A. Kozak, et al.
Molecular cloning, tissue distribution and chromosomal localization of a novel member of the opioid receptor gene family
FEBS Lett, 347 (1994), pp. 279-283
38
K. Fukuda, S. Kato, K. Mori, M. Nishi, H. Takeshima, N. Iwabe, et al.
cDNA cloning and regional distribution of a novel member of the opioid receptor family
FEBS Lett, 343 (1994), pp. 42-46
39
C. Mollereau, M. Parmentier, P. Mailleux, J.L. Butour, C. Moisand, P. Chalon, et al.
ORL-1, a novel member of the opioid family: cloning, functional expression and localization
FEBS Lett, 341 (1994), pp. 33-38
40
Y.-X. Pan, J. Cheng, J. Xu, G.W. Pasternak
Cloning, expression and classification of a kappa3-related opioid receptor using antisense oligodeoxynucleotides
Regul Pept, 54 (1994), pp. 217-218
41
Y.-X. Pan, J. Cheng, J. Xu, G.C. Rossi, E. Jacobson, J. Ryan-Moro, et al.
Cloning and functional characterization through antisense mapping of a kappa3-related opioid receptor
Mol Pharmacol, 47 (1995), pp. 1180-1188
42
J.C. Meunier, C. Mollereau, L. Toll, C. Suaudeau, C. Moisand, P. Alvinerie, et al.
Isolation and structure of the endogenous agonist of the opioid receptor like ORL1 receptor
Nature, 377 (1995), pp. 532-535
43
R.K. Reinscheid, H.P. Nothacker, A. Bourson, A. Ardati, R.A. Henningsen, J.R. Bunzow, et al.
Orphanin FQ: a neuropeptide that activates an opioid like G protein-coupled receptor
Science, 270 (1995), pp. 792-794
Article  
44
G.C. Rossi, Y.-X. Pan, J. Cheng, G.W. Pasternak
Blockade of morphine analgesia by an antisense oligodeoxynucleotide against the mu receptor
Life Sci, 54 (1994), pp. L375-L379
Article  
45
G.R. Uhl, S. Childers, G.W. Pasternak
An opiate-receptor gene family reunion
Trends Neurosci, 17 (1994), pp. 89-93
46
K.M. Standifer, C.-C. Chien, C. Wahlestedt, G.P. Brown, G.W. Pasternak
Selective loss of d opioid analgesia and binding by antisense oligodeoxynucleotides to a d opioid receptor
Neuron, 12 (1994), pp. 805-810
47
G.C. Rossi, K.M. Standifer, G.W. Pasternak
Differential blockade of morphine and morphine-6b-glucuronide analgesia by antisense oligodeoxynucleotides directed against MOR-1 and G-protein a subunits in rats
Neurosci Lett, 198 (1995), pp. 99-102
48
G.C. Rossi, Y.-X. Pan, G.P. Brown, G.W. Pasternak
Antisense mapping the MOR-1 opioid receptor: evidence for alternative splicing and a novel morphine-6b-glucuronide receptor
FEBS Lett, 369 (1995), pp. 192-196
49
H.W.D. Matthes, R. Maldonado, F. Simonin, O. Valverde, S. Slowe, I. Kitchen, et al.
Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the m-opioid-receptor gene
Nature, 383 (1996), pp. 819-823
50
I. Sora, N. Takahashi, M. Funada, H. Ujike, R.S. Revay, D.M. Donovan, et al.
Opiate receptor knockout mice define μ receptor roles in endogenous nociceptive responses and morphine-induced analgesia
Proc Natl Acad Sci U S A, 94 (1997), pp. 1544-1549
51
H.H. Loh, H.C. Liu, A. Cavalli, W.L. Yang, Y.F. Chen, L.N. Wei
μ opioid receptor knockout in mice: effects on ligand-induced analgesia and morphine lethality
Mol Brain Res, 54 (1998), pp. 321-326
52
A.G. Schuller, M.A. King, J. Zhang, E. Bolan, Y.X. Pan, D.J. Morgan, et al.
Retention of heroin and morphine-6 beta-glucuronide analgesia in a new line of mice lacking exon 1 of MOR-1
Nat Neurosci, 2 (1999), pp. 151-156
53
Y. Liang, A. Mestek, L. Yu, L.G. Carr
Cloning and characterization of the promoter region of the mouse m opioid receptor gene
Brain Res, 679 (1995), pp. 82-88
54
P. Mayer, S. Schulzeck, J. Kraus, A. Zimprich, V. Höllt
Promoter region and alternatively spliced exons of the rat μ-opioid receptor gene
J Neurochem, 66 (1996), pp. 2272-2278
55
L. Pan, J. Xu, R. Yu, M.M. Xu, Y.X. Pan, G.W. Pasternak
Identification and characterization of six new alternatively spliced variants of the human mu opioid receptor gene, Oprm
Neuroscience, 133 (2005), pp. 209-220
56
Y.-X. Pan, J. Xu, G.C. Rossi, L. Leventhal, B.-L. Wan, A. Zuckerman, et al.
Cloning and expression of a novel splice variant of the mouse mu-opioid receptor (MOR-1) gene
Soc Neurosci, 24 (1998), p. 524
Article  
57
Y.-X. Pan, J. Xu, L. Mahurter, E.A. Bolan, M.M. Xu, G.W. Pasternak
Generation of the mu opioid receptor (MOR-1) protein by three new splice variants of the Oprm gene
Proc Natl Acad Sci U S A, 98 (2001), pp. 14084-14089
58
Y.X. Pan, J. Xu, E.A. Bolan, C. Abbadie, A. Chang, A. Zuckerman, et al.
Identification and characterization of three new alternatively spliced mu opioid receptor isoforms
Mol Pharmacol, 56 (1999), pp. 396-403
59
Y.X. Pan, J. Xu, L. Mahurter, M. Xu, A.K. Gilbert, G.W. Pasternak
Identification and characterization of two new human mu opioid receptor splice variants, hMOR-1O and hMOR-1X
Biochem Biophys Res Commun, 301 (2003), pp. 1057-1061
60
Y.X. Pan, J. Xu, E. Bolan, H.S. Moskowitz, M. Xu, G.W. Pasternak
Identification of four novel exon 5 splice variants of the mouse mu-opioid receptor gene: functional consequences of C-terminal splicing
Mol Pharmacol, 68 (2005), pp. 866-875
61
Y.X. Pan, J. Xu, E. Bolan, A. Chang, L. Mahurter, G. Rossi, et al.
Isolation and expression of a novel alternatively spliced mu opioid receptor isoform, MOR-1F
FEBS Lett, 466 (2000), pp. 337-340
62
D.A. Pasternak, L. Pan, J. Xu, R. Yu, M.M. Xu, G.W. Pasternak, et al.
Identification of three new alternatively spliced variants of the rat mu opioid receptor gene: dissociation of affinity and efficacy
J Neurochem, 91 (2004), pp. 881-890
63
J. Xu, M. Xu, Y.L. Hurd, G.W. Pasternak, Y.X. Pan
Isolation and characterization of new exon 11-associated N-terminal splice variants of the human mu opioid receptor gene
J Neurochem, 108 (2009), pp. 962-972
64
Y.X. Pan, J. Xu, M. Xu, G.C. Rossi, J.E. Matulonis, G.W. Pasternak
Involvement of exon 11-associated variants of the mu opioid receptor MOR-1 in heroin, but not morphine, actions
Proc Natl Acad Sci U S A, 106 (2009), pp. 4917-4922
65
G.W. Pasternak, S.R. Childers, S.H. Snyder
Opiate analgesia: evidence for mediation by a subpopulation of opiate receptors
Science, 208 (1980), pp. 514-516
Article  
66
G.C. Rossi, L. Leventhal, Y.X. Pan, J. Cole, W. Su, R.J. Bodnar, et al.
Antisense mapping of MOR-1 in rats: distinguishing between morphine and morphine-6beta-glucuronide antinociception
J Pharmacol Exp Ther, 281 (1997), pp. 109-114
67
G.C. Rossi, G.P. Brown, L. Leventhal, K. Yang, G.W. Pasternak
Novel receptor mechanisms for heroin and morphine-6b-glucuronide analgesia
Neurosci Lett, 216 (1996), pp. 1-4
68
Y.-X. Pan, G.W. Pasternak
Molecular biology of mu opioid receptors
G.W. Pasternak (Ed.), The opiate receptors (2nd ed.), Humana Press (2011)
Article  

References

Close