201
|
Alicea C, Belkowski SM, Sliker JK, Zhu J, Liu-Chen LY, Eisenstein TK, Adler MW, Rogers TJ. Characterization of kappa-opioid receptor transcripts expressed by T cells and macrophages. J Neuroimmunol 1998; 91:55-62. [PMID: 9846819 DOI: 10.1016/s0165-5728(98)00151-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have found that the immature T cell lines R1.1 and DPK and the macrophage lines P388D1 and WEHI-3 also express kappa-opioid receptor (KOR) mRNA. Characterization of the KOR transcripts in both brain tissue and these T cells has revealed both the normal full-length as well as a truncated form of the mRNA. Our results show that the truncated transcript lacks the second exon. Primary macrophages express this truncated form of the transcript in the absence of detectable levels of the full-length form. These results suggest a degree of heterogeneity in the expression of the opioid receptors which has not previously been reported.
Collapse
Affiliation(s)
- C Alicea
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | |
Collapse
|
202
|
Affiliation(s)
- H Rosen
- Department of Molecular Virology, The Hebrew University Faculty of Medicine, Jerusalem, Israel
| | | | | |
Collapse
|
203
|
Franck J, Lindholm S, Raaschou P. Modulation of Volitional Ethanol Intake in the Rat by Central delta-Opioid Receptors. Alcohol Clin Exp Res 1998. [DOI: 10.1111/j.1530-0277.1998.tb03897.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
204
|
Georges F, Normand E, Bloch B, Le Moine C. Opioid receptor gene expression in the rat brain during ontogeny, with special reference to the mesostriatal system: an in situ hybridization study. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1998; 109:187-99. [PMID: 9729374 DOI: 10.1016/s0165-3806(98)00082-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The three main types of opioid receptors micro, delta and kappa are found in the central nervous system and periphery. In situ hybridization study was undertaken to determine the expression of mu, delta, kappa-opioid receptors mRNAs in the brain during pre- and postnatal development, especially in the mesostriatal system. By G13, mu and kappa-opioid receptor mRNA were detectable in the telencephalon; mu-opioid receptor mRNA was found in the striatal neuroepithelium and cortical plate and kappa-opioid receptor mRNA in the corroidal fissure. By G15, kappa-opioid receptor mRNA was detectable in the nucleus accumbens and dorsal striatum, and in the substantia nigra and ventral tegmental area, suggesting an early expression of the corresponding receptor on dopaminergic terminal fibers. For the mu-opioid receptor mRNA in the striatum, patches appeared at G20. Delta-opioid receptor mRNA was first detected at G21, in many areas including the accumbens nucleus and the dorsal striatum. At P8, delta-opioid receptor mRNA was detected in large-sized cells of the striatum, possibly cholinergic, suggesting a possible modulation by opioids of the striatal cholinergic neurons. Our results demonstrate the early appearance of mu and kappa-opioid receptor mRNA (G13) and the relatively late development of delta-opioid receptor mRNA (G21) in the brain. We also show a distinct pattern of expression for mu, delta and kappa-opioid receptor mRNAs in the mesostriatal system during the development.
Collapse
MESH Headings
- Animals
- Brain/growth & development
- Brain Chemistry/genetics
- Female
- In Situ Hybridization
- Neostriatum/growth & development
- Pregnancy
- RNA, Messenger/analysis
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Rats
- Receptors, Opioid/biosynthesis
- Receptors, Opioid/genetics
- Receptors, Opioid, delta/biosynthesis
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, kappa/biosynthesis
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, mu/biosynthesis
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- F Georges
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5541, Laboratoire d'Histologie Embryologie, 33076 Bordeaux Cedex, France
| | | | | | | |
Collapse
|
205
|
Seki T, Minami M, Nakagawa T, Ienaga Y, Morisada A, Satoh M. DAMGO recognizes four residues in the third extracellular loop to discriminate between mu- and kappa-opioid receptors. Eur J Pharmacol 1998; 350:301-10. [PMID: 9696421 DOI: 10.1016/s0014-2999(98)00240-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Previously, we reported that replacement of the region from the fifth transmembrane domain to the C-terminus of kappa-opioid receptor with the corresponding region of mu-opioid receptor gives high affinity for [D-Ala2, N-MePhe4, Gly-ol5]enkephalin (DAMGO), a mu-opioid receptor-selective ligand, to the resultant chimeric receptor, suggesting that the difference in the amino acid sequence within this region is critical for the discrimination between mu- and kappa-opioid receptors by DAMGO. In the present study, we constructed further six mu/kappa-chimeric receptors and revealed that at least two separate regions around the third extracellular loop are critical for the discrimination between mu- and kappa-opioid receptors by DAMGO. Furthermore, we constructed several mutant receptors by a site-directed mutagenesis technique and found that the difference between Glu297 of kappa-opioid receptor and Lys303 of mu-opioid receptor in one region, and the difference between Ser310, Tyr312 and Tyr313 of kappa-opioid receptor and Val316, Trp318 and His319 of mu-opioid receptor in the other region, are critical for the discrimination between these receptors by DAMGO. The mutant receptor, kappa (E297K + Y313H + Y312W + S310V), in which the Glu297, Ser310, Tyr312 and Tyr313 of kappa-opioid receptor were changed to Lys, Val, Trp and His, respectively, bound to DAMGO with high affinity (Kd = 8.7 +/- 1.2 nM) and efficiently mediated the inhibitory effect of DAMGO on intracellular cAMP accumulation. The present results showed that these four amino acid residues act as determinants for the discrimination between mu- and kappa-opioid receptors by DAMGO.
Collapse
MESH Headings
- Adenylyl Cyclases/metabolism
- Amino Acid Sequence
- Animals
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalins/pharmacology
- Extracellular Space/drug effects
- Extracellular Space/genetics
- Extracellular Space/metabolism
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Mutation
- Protein Structure, Secondary
- Radioligand Assay
- Rats
- Receptors, Opioid, kappa/chemistry
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/chemistry
- Receptors, Opioid, mu/genetics
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- T Seki
- Department of Molecular Pharmacology, Faculty of Pharmaceutical Sciences, Kyoto University, Japan
| | | | | | | | | | | |
Collapse
|
206
|
Akil H, Owens C, Gutstein H, Taylor L, Curran E, Watson S. Endogenous opioids: overview and current issues. Drug Alcohol Depend 1998; 51:127-40. [PMID: 9716935 DOI: 10.1016/s0376-8716(98)00071-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- H Akil
- Mental Health Research Institute, University of Michigan, Ann Arbor 48109, USA.
| | | | | | | | | | | |
Collapse
|
207
|
Bouvier C, Avram D, Peterson VJ, Hettinger B, Soderstrom K, Murray TF, Leid M. Catecholaminergic CATH.a cells express predominantly delta-opioid receptors. Eur J Pharmacol 1998; 348:85-93. [PMID: 9650835 DOI: 10.1016/s0014-2999(98)00132-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CATH.a cells are a catecholaminergic cell line of neuronal origin. The opioid receptor complement expressed by CATH.a cells was defined pharmacologically and by reverse transcription-polymerase chain reaction (RT-PCR). CATH.a cells were found to express mRNA encoding all three of the major subtypes of opioid receptors. The relative abundance of CATH.a cell opioid receptor transcripts was delta > kappa> mu. Pharmacological and functional data were in agreement with the results of RT-PCR inasmuch as delta-opioid receptor was identified as the most abundant opioid receptor subtype expressed by CATH.a cells. In addition, at least one of the opioid signalling pathways, inhibition of adenylyl cyclase activity, was found to be operant in this cell line. CATH.a cells should be of general utility for the study of opioid receptor signalling mechanisms in the context of catecholaminergic neurons.
Collapse
Affiliation(s)
- C Bouvier
- Laboratory of Molecular Pharmacology, College of Pharmacy, Oregon State University, Corvallis 97331, USA
| | | | | | | | | | | | | |
Collapse
|
208
|
Kalyuzhny AE, Wessendorf MW. Relationship of ?- and ?-opioid receptors to GABAergic neurons in the central nervous system, including antinociceptive brainstem circuits. J Comp Neurol 1998. [DOI: 10.1002/(sici)1096-9861(19980323)392:4<528::aid-cne9>3.0.co;2-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
209
|
Yakimova KS, Sann H, Pierau FK. Effects of kappa and delta opioid agonists on activity and thermosensitivity of rat hypothalamic neurons. Brain Res 1998; 786:133-42. [PMID: 9554984 DOI: 10.1016/s0006-8993(97)01456-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Extracellular recordings were made from 161 warm-sensitive, six cold-sensitive and 153 temperature-insensitive neurons in slices of the preoptic area/anterior hypothalamus (PO/AH) of rats, to investigate the effects of the kappa-receptor opioid agonist dynorphin A1-17 and the delta-receptor opioid agonist DPDPE on neuronal response characteristics. While 61% of the neurons exhibited kappa-receptors, delta-receptors were only present in 37% of the neurons. No co-localization was observed between kappa- and delta-receptors, whereas mu-receptors could be co-localized with kappa- as well as delta-receptors. Antagonistic effects on tonic activity were induced by different concentrations of the kappa-agonist dynorphin A1-17. At 0.5 nM, the excitatory effect was predominant, while 50% of the neurons were already inhibited at 5 nM and inhibition was the major effect at 100 nM. A significant increase in temperature sensitivity was observed in warm-sensitive neurons during administration of 0.5 nM dynorphin A1-17; in contrast, the temperature sensitivity was significantly decreased at the high dose of 100 nM. In most of the neurons responding to the delta-receptor agonist DPDPE (0.5-100 nM) the firing rate was decreased. The temperature sensitivity was only affected in warm-sensitive neurons, and was increased in the majority of neurons at 0.5 and 5 nM, but predominantly decreased at higher concentrations. The effects of low concentrations of dynorphin A1-17 and DPDPE were prevented by pre- and co-perfusion of the appropriate antagonists. The present results suggest that changes of the temperature sensitivity of warm-sensitive PO/AH neurons are an important mechanism for the effect of low doses of opioids on body temperature.
Collapse
MESH Headings
- Animals
- Dynorphins/pharmacology
- Electrophysiology
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- Hypothalamus/cytology
- Hypothalamus/drug effects
- Hypothalamus/physiology
- Hypothalamus, Anterior/drug effects
- In Vitro Techniques
- Male
- Neurons/drug effects
- Neurons/physiology
- Preoptic Area/drug effects
- Rats
- Rats, Wistar
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, mu/agonists
- Thermosensing/drug effects
- Thermosensing/physiology
Collapse
Affiliation(s)
- K S Yakimova
- Department of Pharmacology, Faculty of Medicine,'Zdrave' 2 str., 1431 Sofia, Bulgaria
| | | | | |
Collapse
|
210
|
Narita M, Tseng LF. Evidence for the existence of the beta-endorphin-sensitive "epsilon-opioid receptor" in the brain: the mechanisms of epsilon-mediated antinociception. JAPANESE JOURNAL OF PHARMACOLOGY 1998; 76:233-53. [PMID: 9593217 DOI: 10.1254/jjp.76.233] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recently, mu-, delta- and kappa-opioid receptors have been cloned and relatively well-characterized. In addition to three major opioid receptor types, more extensive studies have suggested the possible existence of other opioid receptor types that can be classified as non-mu, non-delta and non-kappa. Based upon anatomical and binding studies in the brain, the sensitive site for an endogenous opioid peptide, beta-endorphin, has been postulated to account for the unique characteristics of the opioid receptor defined as a putative epsilon-opioid receptor. Many epsilon-opioid receptors are functionally coupled to G-proteins. The functional epsilon-opioid receptors in the brain are stimulated by bremazocine and etorphine as well as beta-endorphin, but not by selective mu-, delta- or kappa-opioid receptor agonists. Epsilon-opioid receptor agonists injected into the brain produce profound antinociception. The brain sites most sensitive to epsilon-agonist-induced antinociception are located in the caudal medial medulla such as the nucleus raphe obscures, nucleus raphe pallidus and the adjacent midline reticular formation. The stimulation of epsilon-opioid receptors in the brain facilitates the descending enkephalinergic pathway, which probably originates from the brainstem terminating at the spinal cord. The endogenous opioid Met-enkephalin, released in the spinal cord by activation of supraspinal epsilon-opioid receptors, stimulates spinal delta2-opioid receptors for the production of antinociception. It is noteworthy that the epsilon-opioid receptor-mediated pain control system is different from that of other opioid systems. Although there appears to be no epsilon-selective ligand currently available, these findings provide strong evidence for the existence of the putative epsilon-opioid receptor and its unique function in the brain.
Collapse
Affiliation(s)
- M Narita
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee 53226, USA
| | | |
Collapse
|
211
|
Zhang X, Bao L, Shi TJ, Ju G, Elde R, Hökfelt T. Down-regulation of mu-opioid receptors in rat and monkey dorsal root ganglion neurons and spinal cord after peripheral axotomy. Neuroscience 1998; 82:223-40. [PMID: 9483516 DOI: 10.1016/s0306-4522(97)00240-6] [Citation(s) in RCA: 144] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To understand the role of opioids and their receptors in chronic pain following peripheral nerve injury, we have studied the mu-opioid receptor in rat and monkey lumbar 4 and 5 dorsal root ganglion neurons and the superficial dorsal horn of the spinal cord under normal circumstances and after peripheral axotomy. Our results show that many small neurons in rat and monkey dorsal root ganglia, and some medium-sized and large neurons in rat dorsal root ganglia, express mu-opioid receptor-like immunoreactivity. Most of these neurons contain calcitonin gene-related peptide. The mu-opioid receptor was closely associated with the somatic plasmalemma of the dorsal root ganglion neurons. Both mu-opioid receptor-immunoreactive nerve fibers and cell bodies were observed in lamina II of the dorsal horn. The highest intensity of mu-opioid receptor-like immunoreactivity was observed in the deep part of lamina II. Most mu-opioid receptor-like immunoreactivity in the dorsal horn originated from spinal neurons. A few mu-opioid receptor-positive peripheral afferent terminals in the rat and monkey dorsal horn were calcitonin gene-related peptide-immunoreactive. In addition to pre- and post-junctional receptors in rat and monkey dorsal horn neurons, mu-opioid receptors were localized on the presynaptic membrane of some synapses of primary afferent terminals in the monkey dorsal horn. Peripheral axotomy caused a reduction in the number and intensity of mu-opioid receptor-positive neurons in the rat and monkey dorsal root ganglia, and of mu-opioid receptor-like immunoreactivity in the dorsal horn of the spinal cord. The decrease in mu-opioid receptor-like immunoreactivity was more pronounced in the monkey than in the rat dorsal root ganglia and spinal cord. It is probable that there was a parallel trans-synaptic down-regulation of mu-opioid-like immunoreactivity in local dorsal horn neurons of the monkey. These data suggest that one factor underlying the well known insensitivity of neuropathic pain to opioid analgesics could be due to a marked reduction in the number of mu-opioid receptors in the axotomized sensory neurons and in interneurons in the dorsal horn of the spinal cord.
Collapse
Affiliation(s)
- X Zhang
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
212
|
Quigley DI, McDougall J, Darland T, Zhang G, Ronnekliev O, Grandy DK, Allen RG. Orphanin FQ is the major OFQ1-17-containing peptide produced in the rodent and monkey hypothalamus. Peptides 1998; 19:133-9. [PMID: 9437745 DOI: 10.1016/s0196-9781(97)00268-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In order to investigate the processing of OFQ containing peptides in the hypothalamus we have developed a sensitive and quantitative radioimmunoassay for OFQ. We fractionated rodent and monkey hypothalamic extracts by reversed-phase high performance liquid chromatography and found that the extracts contained multiple peaks of OFQ immunoreactivity with the major peak co-eluting with synthetic OFQ1-17. Mouse hypothalamic extracts were also fractionated by SDS-PAGE to determine the apparent molecular weights of molecules containing the OFQ peptide. Multiple peaks of OFQ immunoreactivity, ranging in size from approximately 1 to 30 kilodaltons, were detected by this method. These results suggest that OFQ1-17 is processed to smaller peptides in mouse and monkey hypothalamic neurons.
Collapse
Affiliation(s)
- D I Quigley
- Vollum Institute for Advanced Biomedical Research, Oregon Health Sciences University, Portland 97201, USA
| | | | | | | | | | | | | |
Collapse
|
213
|
Abstract
Despite the existence of a large body of information on the subject, the mechanisms of opiate tolerance and dependence are not yet fully understood. Although the traditional mechanisms of receptor down-regulation and desensitization seem to play a role, they cannot entirely explain the phenomena of tolerance and dependence. Therefore, other mechanisms, such as the presence of antiopiate systems and the coupling of opiate receptors to alternative G-proteins, should be considered. A further complication of studies of opiate tolerance and dependence is the multiplicity of endogenous opiate receptors and peptides. This review will focus on the endogenous opioid system--peptides, receptors, and coupling of receptors to intracellular signaling via G-proteins--in the context of their roles in tolerance and dependence. Opioid peptides include the recently discovered endomorphins and those encoded by three known genes--pro-opiomelanocortin, pro-enkephalin, and pro-dynorphin. They bind to three types of receptors--mu, delta, and kappa. Each of the receptor types is further divided into multiple subtypes. These receptors are widely known to be coupled to G-proteins of the Gi and Go subtypes, but an increasing body of results suggests coupling to other G-proteins, such as Gs. The coupling of opiate receptors to Gs, in particular, has implications for tolerance and dependence. Alterations at the receptor and transduction level have been the focus of many studies of opiate tolerance and dependence. In these studies, both receptor down-regulation and desensitization have been demonstrated in vivo and in vitro. Receptor down-regulation has been more easily observed in vitro, especially in response to morphine, a phenomenon which suggests that some factor which is missing in vitro prevents receptors from down-regulating in vivo and may play a critical role in tolerance and dependence. We suggest that antiopiate peptides may operate in vivo in this capacity, and we outline the evidence for the antiopiate properties of three peptides: neuropeptide FF, orphanin FQ/nociceptin, and Tyr-W-MIF-1. In addition, we provide new results suggesting that Tyr-W-MIF-1 may act as an antiopiate at the cellular level by inhibiting basal G-protein activation, in contrast to the activation of G-proteins by opiate agonists.
Collapse
Affiliation(s)
- L M Harrison
- Tulane University School of Medicine and Veterans Affairs Medical Center, New Orleans, LA 70112-1262, USA
| | | | | |
Collapse
|
214
|
Emmerson PJ, Archer S, El-Hamouly W, Mansour A, Akil H, Medzihradsky F. Synthesis and characterization of 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY)-labeled fluorescent ligands for the mu opioid receptor. Biochem Pharmacol 1997; 54:1315-22. [PMID: 9393674 DOI: 10.1016/s0006-2952(97)00374-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A series of opioid ligands utilizing the 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY) fluorophores 4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene++ +-3-propionic acid or 4,4-difluoro-5-(4-phenyl-1,3-butadienyl)-4-bora-3a,4a-diaza- s-indacene-3-propionic acid were synthesized and characterized for their ability to act as a suitable fluorescent label for the mu opioid receptor. All compounds displaced the mu opioid receptor binding of [3H]Tyr-D-Ala-Gly-(Me)Phe-Gly-ol in monkey brain membranes with high affinity. The binding of fluorescent ligands to delta and kappa receptors was highly variable. 5,7-Dimethyl-BODIPY naltrexamine, "6-BNX," displayed subnanomolar affinities for the mu and kappa opioid receptors (Ki 0.07 and 0.43 nM, respectively) and nanomolar affinity at the delta (Ki 1.4 nM) receptor. Using fluorescence spectroscopy, the binding of 6-BNX in membranes from C6 glioma cells transfected with the cloned mu opioid receptor was investigated. In these membranes containing a high receptor density (10-80 pmol/mg protein), 6-BNX labeling was saturable, mu opioid specific, stereoselective (as determined with the isomers dextrorphan and levorphanol), and more than 90% specific. The results describe a series of newly developed fluorescent ligands for the mu opioid receptor and the use of one of these ligands as a label for the cloned mu receptor. These ligands provide a new approach for studying the structural and biophysical nature of opioid receptors.
Collapse
Affiliation(s)
- P J Emmerson
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor 48109, USA
| | | | | | | | | | | |
Collapse
|
215
|
Bagnol D, Mansour A, Akil H, Watson SJ. Cellular localization and distribution of the cloned mu and kappa opioid receptors in rat gastrointestinal tract. Neuroscience 1997; 81:579-91. [PMID: 9300443 DOI: 10.1016/s0306-4522(97)00227-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Several pharmacological and electrophysiological studies have shown that the opioid receptors are widely distributed in the gastrointestinal tract. Despite such consensus, there are conflicting findings regarding their effects in intestinal function, and their precise site of action remained unclear. The aim of the present study was therefore to delineate the cellular localization of mu and kappa opioid receptors in rat gastrointestinal tract using polyclonal antibodies generated to C-terminal end of the cloned mu (63 amino acids) and kappa (41 amino acids) receptors. The distribution of mu differs from that of kappa receptors within the gastrointestinal wall, with a greater abundance of mu receptor-like immunoreactive fibres in all intestinal layers. Numerous neurons expressing mu receptor-like proteins were found in the submucosal plexus with comparatively few in the myenteric plexus. In contrast, a higher number of neurons expressing kappa receptor-like immunoreactivity were visualized in the myenteric plexus with a small number in the submucosal plexus. A high number of immunopositive neurons were found in the myenteric plexus of the stomach and the proximal colon with both antibodies. In the submucosal and mucosal layers. mu receptor-immunoreactive fibres were more abundant and distributed around the crypts, blood vessels and lymphatic nodes. Interestingly, numerous mu and fewer kappa receptor-immunoreactive interstitial cells are localized in the region of myenteric plexus and at the internal border of the circular muscle. Finally, smooth muscle cells did not demonstrate any mu- nor kappa-receptor immunoreactivity. These findings suggest that in the rat gastrointestinal tract, mu and kappa opioid receptors may directly influence neuronal and interstitial cell activity. This appears not to be the case for the smooth muscle cells. In the muscular layers, the anatomical data point to mu receptor actions being mediated by nerve terminals, whereas kappa receptor effects may be mediated by both nerve terminals and somatodendritic synaptic mechanisms. In contrast, in the submucosal and mucosal layers, mu receptors predominate and are localized on both nerve terminals and somatodendritic synaptic elements.
Collapse
Affiliation(s)
- D Bagnol
- Mental Health Research Institute, University of Michigan, Ann Arbor 48109-0720, USA
| | | | | | | |
Collapse
|
216
|
Sim LJ, Childers SR. Anatomical distribution of mu, delta, and kappa opioid- and nociceptin/orphanin FQ-stimulated [35S]guanylyl-5'-O-(gamma-thio)-triphosphate binding in guinea pig brain. J Comp Neurol 1997; 386:562-72. [PMID: 9378852 DOI: 10.1002/(sici)1096-9861(19971006)386:4<562::aid-cne4>3.0.co;2-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
An in vitro autoradiographic technique has recently been developed to visualize receptor-activated G-proteins by using agonist-stimulated [35S]guanylyl-5'-O-(gamma-thio)-triphosphate ([35S]GTPgammaS) binding in the presence of excess guanosine 5'-diphosphate. This technique was used to localize opioid-activated G-proteins in guinea pig brain, a species that contains the three major types of opioid receptors. This study used selective mu, delta, and kappa opioid agonists as well as nociceptin or orphanin FQ (N/OFQ) peptide, an endogenous ligand for an orphan opioid receptor-like (ORL1) receptor, to stimulate [35S]GTPgammaS binding in guinea pig brain sections. Opioid receptor specificity was confirmed by blocking agonist-stimulated [35S] GTPgammaS binding with the appropriate antagonists. In general, the distribution of agonist-stimulated [35S]GTPgammaS binding correlated with previous reports of receptor binding autoradiography, although quantitative differences suggest regional variations in receptor coupling efficiency. Mu, delta, and kappa opioid-stimulated [35S]GTPgammaS binding was found in the caudate-putamen, nucleus accumbens, amygdala, and hypothalamus. Mu-stimulated [35S]GTPgammaS binding predominated in the hypothalamus, amygdala, and brainstem, whereas kappa-stimulated [35S]GTPgammaS binding was particularly high in the substantia nigra and cortex and was moderate in the cerebellum. N/OFQ-stimulated [35S] GTPgammaS binding was highest in the cortex, hippocampus, and hypothalamus and exhibited a unique anatomical distribution compared with opioid-stimulated [35S]GTPgammaS binding. The present study extends previous reports on opioid and ORL1 receptor localization by anatomically demonstrating functional activity produced by mu, delta, and kappa opioid and ORL1 receptor activation of G-proteins.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- Analgesics/pharmacology
- Analgesics, Non-Narcotic/pharmacology
- Animals
- Autoradiography
- Brain Chemistry/physiology
- Cerebellum/chemistry
- Diencephalon/chemistry
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalin, Leucine/analogs & derivatives
- Enkephalin, Leucine/pharmacology
- Enkephalins/pharmacology
- Guanosine 5'-O-(3-Thiotriphosphate)/metabolism
- Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology
- Guinea Pigs/physiology
- Male
- Medulla Oblongata/chemistry
- Mesencephalon/chemistry
- Naloxone/pharmacology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Opioid Peptides/pharmacology
- Pons/chemistry
- Receptors, Opioid/agonists
- Receptors, Opioid/analysis
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/analysis
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/analysis
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/analysis
- Receptors, Opioid, mu/antagonists & inhibitors
- Sulfur Radioisotopes
- Telencephalon/chemistry
- Nociceptin
Collapse
Affiliation(s)
- L J Sim
- Department of Physiology and Pharmacology, Center for the Neurobiological Investigation of Drug Abuse, Bowman Gray School of Medicine, Wake Forest University, Winston-Salem, North Carolina 27157, USA
| | | |
Collapse
|
217
|
Schoffelmeer AN, Hogenboom F, Mulder AH. Kappa1- and kappa2-opioid receptors mediating presynaptic inhibition of dopamine and acetylcholine release in rat neostriatum. Br J Pharmacol 1997; 122:520-4. [PMID: 9351509 PMCID: PMC1564953 DOI: 10.1038/sj.bjp.0701394] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
1. The effects of selective opioid receptor agonists and antagonists on N-methyl-D-aspartate (NMDA, 10 microM)-induced release of [3H]-dopamine and [14C]-acetylcholine (ACh) from superfused neostriatal slices were studied to investigate the possible occurrence of functional kappa-opioid receptor subtypes in rat brain. 2. The kappa receptor agonists (-)-ethylketocyclazocine ((-)-EKC), U69593 and the endogenous opioid peptide dynorphin A1-13 caused a naloxone-reversible inhibition of NMDA-induced [3H]-dopamine release, with pD2 values of about 9, 8.5 and 8.2, respectively, whereas both the mu agonist Tyr-D-Ala-Gly-(NMe)Phe-Gly-ol (DAMGO) and the delta agonist D-Pen2-D-Pen5-enkephalin (DPDPE) were ineffective in this respect. The inhibitory effect of submaximally effective concentrations of dynorphin A1-13, U69593 and (-)-EKC on NMDA-induced [3H]-dopamine release were not changed by the delta1/delta2-opioid receptor antagonist naltrindole (up to a concentration of 1 microM, but reversed by the kappa receptor antagonist nor-binaltorphimine (nor-BNI), with an IC50) as low as 0.02 nM, indicating the involvement of U69593-sensitive kappa1-opioid receptors. 3. NMDA-induced [14C]-ACh release was reduced in a naloxone-reversible manner by DPDPE (pD2 about 7.2), dynorphin A1-13 (pD2 6.7) and EKC (pD2 6.2), but not by U69593 and DAMGO. The inhibitory effect of a submaximally effective concentration of DPDPE, unlike those of dynorphin A1-13 and (-)-EKC, on NMDA-induced [14C]-ACh release was antagonized by naltrindole with an IC50 of 1 nM, indicating the involvement of delta-opioid receptors in the inhibitory effect of DPDPE. On the other hand, the inhibitory effects of dynorphin A1-13 and (-)-EKC on [14C]-ACh release were readily antagonized by nor-BNI with an IC50 of about 3 nM. A 100 fold higher concentration of nor-BNI also antagonized the inhibitory effect of DPDPE, indicating the involvement of U69593-insensitive kappa2-opioid receptors in the inhibitory effects of dynorphin A1-13 and (-)-EKC. 4. Although naloxone benzoylhydrazone (NalBzoH), displaying high affinity towards the putative kappa3-opioid receptor, antagonized the inhibitory effects of dynorphin A1-13 and (-)-EKC on [3H]-dopamine and [14C]-ACh release as well as that of U69593 on [3H]-dopamine release, it displayed a low apparent affinity (IC50 about 100 nM) in each case. 5. In conclusion, whereas activation of kappa1-opioid receptors causes presynaptic inhibition of NMDA-induced dopamine release, kappa2 receptor activation results in inhibition of ACh release in rat neostriatum. As such, this study is the first to provide unequivocal in vitro evidence for the existence of functionally distinct kappa-opioid receptor subtypes in the brain.
Collapse
MESH Headings
- Acetylcholine/metabolism
- Animals
- Benzeneacetamides
- Dopamine/metabolism
- Dynorphins/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- Ethylketocyclazocine/pharmacology
- In Vitro Techniques
- Male
- N-Methylaspartate/pharmacology
- Naloxone/pharmacology
- Neostriatum/drug effects
- Neostriatum/metabolism
- Neural Inhibition/drug effects
- Peptide Fragments/pharmacology
- Pyrrolidines/pharmacology
- Rats
- Rats, Wistar
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, kappa/physiology
- Receptors, Opioid, mu/agonists
Collapse
Affiliation(s)
- A N Schoffelmeer
- Research Institute Neurosciences Vrije Universiteit, Department of Pharmacology, Free University, Medical Faculty, Amsterdam, The Netherlands
| | | | | |
Collapse
|
218
|
Robinson ES, Nutt DJ, Jackson HC, Hudson AL. Antisense oligonucleotides in psychopharmacology and behaviour: promises and pitfalls. J Psychopharmacol 1997; 11:259-69. [PMID: 9305419 DOI: 10.1177/026988119701100310] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Antisense oligonucleotides are used to study the expression and function of a diverse range of proteins. Areas for which antisense has been used for pharmacological investigation include receptors, neuropeptides and immediate early genes, particularly when specific ligands or markers are not yet available. Antisense oligonucleotides target a specific mRNA and block the expression of the protein by sequence specific hybridization. This technique has not only been shown to be a valuable pharmacological tool but also to have potential therapeutic applications. In this review we discuss the technology behind the technique including developments in methodology employed in antisense experiments. Although antisense provides a novel and highly specific tool, the reliability of the technique and many of the problems associated with antisense experiments are discussed. The main focus of this article is the use of antisense in psychopharmacology to investigate behavioural changes following antisense-mediated inhibition of the expression of specific brain proteins and receptors.
Collapse
Affiliation(s)
- E S Robinson
- Psychopharmacology Unit, School of Medical Sciences, Bristol, UK
| | | | | | | |
Collapse
|
219
|
Darlison MG, Greten FR, Harvey RJ, Kreienkamp HJ, Stühmer T, Zwiers H, Lederis K, Richter D. Opioid receptors from a lower vertebrate (Catostomus commersoni): sequence, pharmacology, coupling to a G-protein-gated inward-rectifying potassium channel (GIRK1), and evolution. Proc Natl Acad Sci U S A 1997; 94:8214-9. [PMID: 9223341 PMCID: PMC21583 DOI: 10.1073/pnas.94.15.8214] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The molecular evolution of the opioid receptor family has been studied by isolating cDNAs that encode six distinct opioid receptor-like proteins from a lower vertebrate, the teleost fish Catostomus commersoni. One of these, which has been obtained in full-length form, encodes a 383-amino acid protein that exhibits greatest sequence similarity to mammalian mu-opioid receptors; the corresponding gene is expressed predominantly in brain and pituitary. Transfection of the teleost cDNA into HEK 293 cells resulted in the appearance of a receptor having high affinity for the mu-selective agonist [D-Ala2, MePhe4-Gly-ol5]enkephalin (DAMGO) (Kd = 0.63 +/- 0.15 nM) and for the nonselective antagonist naloxone (Kd = 3.1 +/- 1.3 nM). The receptor had negligible affinity for U50488 and [D-Pen2, D-Pen5]enkephalin (DPDPE), which are kappa- and delta-opioid receptor selective agonists, respectively. Stimulation of transfected cells with 1 microM DAMGO lowered forskolin-induced cAMP levels, an effect that could be reversed by naloxone. Experiments in Xenopus oocytes have demonstrated that the fish opioid receptor can, in an agonist-dependent fashion, activate a coexpressed mouse G-protein-gated inward-rectifying potassium channel (GIRK1). The identification of six distinct fish opioid receptor-like proteins suggests that additional mammalian opioid receptors remain to be identified at the molecular level. Furthermore, our data indicate that the mu-opioid receptor arose very early in evolution, perhaps before the appearance of vertebrates, and that the pharmacological and functional properties of this receptor have been conserved over a period of approximately 400 million years implying that it fulfills an important physiological role.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cell Line
- Cloning, Molecular
- DNA, Complementary
- Evolution, Molecular
- Fishes/genetics
- G Protein-Coupled Inwardly-Rectifying Potassium Channels
- Humans
- Molecular Sequence Data
- Potassium Channels/metabolism
- Potassium Channels, Inwardly Rectifying
- Protein Binding
- Radioligand Assay
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Sequence Homology, Amino Acid
- Xenopus laevis
Collapse
Affiliation(s)
- M G Darlison
- Institut für Zellbiochemie und klinische Neurobiologie, Universität Hamburg, Martinistrasse 52, D-20246 Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
220
|
Fickel J, Bagnol D, Watson SJ, Akil H. Opioid receptor expression in the rat gastrointestinal tract: a quantitative study with comparison to the brain. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1997; 46:1-8. [PMID: 9191072 DOI: 10.1016/s0169-328x(96)00266-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The present study was undertaken to analyze the expression of two opioid receptor genes (mu and kappa) in different gastrointestinal regions of the rat. A combination of mRNA quantification and immunohistochemical visualization was used to characterize their expression. Using naive animals, RNA was extracted from tissues and used in RNase protection assays: both receptor mRNAs were expressed in all investigated areas but displayed different expression profiles across the various regions of the digestive tract. Stomach and proximal colon appeared to have the highest expression levels of both receptors, whereas the lowest expression levels were found in the duodenum. Expression levels for both receptors were always lower in the gastrointestinal tract compared to the brain. However, the kappa-receptor expression in the proximal colon represented 40% of the amount found in the brain, which is almost 4 times as high as the respective mu-receptor expression. In contrast to smooth muscle cells, myenteric plexus perikarya of the rat stomach and colon were immunoreactive with antibodies raised against the C-termini of both kappa- and mu-opioid receptors. Numerous nerve fibers were also immunoreactive for both mu- and kappa-receptors and distributed in the longitudinal and circular muscle layers. Small perikarya immunoreactive for mu-receptor were localized around the myenteric plexus and at the submucosal border of the circular muscle, whereas only few perikarya were immunoreactive for the kappa-receptor. We conclude that at least in rat stomach and colon, mu- and kappa-opioid receptors may directly control neuronal communication but seem to have no direct influence on smooth muscle cells.
Collapse
Affiliation(s)
- J Fickel
- University of Michigan, Mental Health Research Institute, Ann Arbor 48109-0720, USA
| | | | | | | |
Collapse
|
221
|
Gaibelet G, Capeyrou R, Dietrich G, Emorine LJ. Identification in the mu-opioid receptor of cysteine residues responsible for inactivation of ligand binding by thiol alkylating and reducing agents. FEBS Lett 1997; 408:135-40. [PMID: 9187354 DOI: 10.1016/s0014-5793(97)00407-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Inactivation by thiol reducing and alkylating agents of ligand binding to the human mu-opioid receptor was examined. Dithiothreitol reduced the number of [3H]diprenorphine binding sites. Replacement by seryl residues of either C142 or C219 in extracellular loops 1 and 2 of the mu receptor resulted in a complete loss of opioid binding. A disulfide bound linking C142 to C219 may thus be essential to maintain a functional conformation of the receptor. We also demonstrated that inactivation of ligand binding upon alkylation by N-ethylmaleimide occurred at two sites. Alteration of the more sensitive (IC50 = 20 microM) did not modify antagonists binding but decreased agonist affinity almost 10-fold. Modification of the less reactive site (IC50 = 2 mM) decreased the number of both agonist and antagonist binding sites. The alkylation site of higher sensitivity to N-ethylmaleimide was shown by mutagenesis experiments to be constituted of both C81 and C332 in transmembrane domains 1 and 7 of the mu-opioid receptor.
Collapse
MESH Headings
- Alkylating Agents/metabolism
- Alkylating Agents/pharmacology
- Alkylation
- Animals
- Binding Sites
- COS Cells
- Cloning, Molecular
- Cysteine/metabolism
- Diprenorphine/metabolism
- Disulfides/chemistry
- Disulfides/metabolism
- Dithiothreitol/metabolism
- Dithiothreitol/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalins/metabolism
- Ethylmaleimide/metabolism
- Ethylmaleimide/pharmacology
- Humans
- Ligands
- Mutagenesis, Site-Directed
- Narcotic Antagonists/metabolism
- Protein Binding
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/chemistry
- Receptors, Opioid, mu/metabolism
- Reducing Agents/metabolism
- Reducing Agents/pharmacology
- Sulfhydryl Reagents/metabolism
- Sulfhydryl Reagents/pharmacology
- Transfection
Collapse
Affiliation(s)
- G Gaibelet
- Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique, Unité Propre de Recherches No. 9062, Toulouse, France
| | | | | | | |
Collapse
|
222
|
Abstract
Most opioid receptor-mediated functions appear to be mediated through G protein interactions, therefore an understanding of opioid signalling requires knowledge of those interactions. This review chronicles the studies examining these interactions for all the opioid receptor subtypes, both in vivo and in vitro.
Collapse
Affiliation(s)
- K M Standifer
- Dept. of Pharmacological and Pharmaceutical Sciences, University of Houston, TX 77204-5515, USA
| | | |
Collapse
|
223
|
Rossi GC, Su W, Leventhal L, Su H, Pasternak GW. Antisense mapping DOR-1 in mice: further support for delta receptor subtypes. Brain Res 1997; 753:176-9. [PMID: 9125445 DOI: 10.1016/s0006-8993(97)00081-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In contrast to the pharmacological studies implicating delta-opioid receptor subtypes, cloning studies have identified only a single cDNA encoding a delta receptor, DOR-1. Antisense studies have established the importance of DOR-1 in delta analgesia in mice. Antisense mapping extends this approach to include oligodeoxynucleotides which are targeted against each of the exons of the gene. Five different antisense oligodeoxynucleotides based upon the three DOR-1 exons all block both spinal and supraspinal analgesic actions of the delta2 ligand [D-Ala2,Glu4]deltorphin, consistent with the suggestion that DOR-1 encodes the delta2 receptor. At the spinal level, [D-Pen2,D-Pen5]enkephalin (DPDPE) acts also acts through delta2 receptors and all the antisense probes block spinal DPDPE analgesia. However, supraspinally only the two antisense probes targeting exon 3 block DPDPE analgesia. The remaining three antisense probes based upon exons 1 and 2 are inactive. Thus, the delta receptors responsible for spinal and supraspinal DPDPE analgesia can be discriminated at the molecular level by antisense mapping.
Collapse
MESH Headings
- Analgesia
- Animals
- Cloning, Molecular
- DNA, Antisense/genetics
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- Exons
- Male
- Mice
- Mice, Inbred Strains
- Oligonucleotides, Antisense/genetics
- Oligopeptides/pharmacology
- Peptide Mapping/methods
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
Collapse
Affiliation(s)
- G C Rossi
- Cotzias Laboratory of Neuro-Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
224
|
Pepin MC, Yue SY, Roberts E, Wahlestedt C, Walker P. Novel "restoration of function" mutagenesis strategy to identify amino acids of the delta-opioid receptor involved in ligand binding. J Biol Chem 1997; 272:9260-7. [PMID: 9083060 DOI: 10.1074/jbc.272.14.9260] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A novel "restoration of function" mutagenesis strategy was developed to identify amino acid sequence combinations necessary to restore the ability to bind delta-selective ligands to an inactive delta/mu receptor chimera in which 10 amino acids of the third extracellular loop of the delta receptor were replaced by the corresponding amino acids from the mu receptor (delta/mu291-300). This chimera binds a nonselective opioid ligand but is devoid of affinity for delta-selective ligands. A library of mutants was generated in which some of the 10 amino acids of the mu sequence of delta/mu291-300 were randomly reverted to the corresponding delta amino acid. Using a ligand binding assay, we screened this library to select mutants with high affinity for delta-selective ligands. Sequence analysis of these revertants revealed that a leucine at position 300, a hydrophobic region (amino acids 295-300), and an arginine at position 291 of the human delta-opioid receptor were present in all revertants. Single and double point mutations were then introduced in delta/mu291-300 to evaluate the contribution of the leucine 300 and arginine 291 residues for the binding of delta-selective ligands. An increased affinity for delta-selective ligands was observed when the tryptophan 300 (mu residue) of delta/mu291-300 was reverted to a leucine (delta residue). Further site-directed mutagenesis experiments suggested that the presence of a tryptophan at position 300 may block the access of delta-selective ligands to their docking site.
Collapse
MESH Headings
- Amino Acid Sequence
- Analgesics/metabolism
- Benzomorphans/metabolism
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins
- Gene Library
- Genetic Techniques
- Humans
- Models, Molecular
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Receptors, Opioid, delta/chemistry
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/genetics
- Structure-Activity Relationship
Collapse
Affiliation(s)
- M C Pepin
- Astra Research Centre Montreal, Montreal, Québec H4S 1Z9, Canada
| | | | | | | | | |
Collapse
|
225
|
Molecular and Neuroanatomical Properties of the Endogenous Opioid System: Implications for Treatment of Opiate Addiction. ACTA ACUST UNITED AC 1997. [DOI: 10.1006/smns.1997.0108] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
226
|
|
227
|
Abstract
Opiates by an action at specific receptors can induce a highly selective alteration in the response of humans and animals to strong and otherwise aversive chemical, mechanical or thermal stimuli. Specific investigations in a variety of species from rodent to primate using microinjection techniques to examine the pharmacology of local drug action have shown potent antinociceptive actions to be mediated by a receptor specific action at a number of sites within the brain, including the periaqueductal gray (PAG: mu receptor), the rostral ventral medulla (mu/delta receptor) and the substantia nigra (mu receptor) and within the spinal dorsal horn (mu/delta/kappa receptor). Mechanistic studies have shown these actions in the different sites to be mediated by several discrete mechanisms. For example, in the PAG, the local opiate effect is likely mediated by the indirect activation of bulbospinal pathways, rostral projections to forebrain sites and by a local alteration in afferent input into the brainstem core. In the spinal cord, this effect is mediated by an action presynaptic to the primary afferent and by a post-synaptic effect to hyperpolarize projection neurons. In addition, it is now appreciated that mu and kappa receptors in the periphery can modulate the sensitized state of the small afferent terminal innervating inflamed tissue and exert an anti-hyperalgesic action. After systemic delivery of an opiate, it is thus clear that a wide array of central and peripheral systems serve to explain the powerful analgesic effect exerted by this class of agents.
Collapse
Affiliation(s)
- T L Yaksh
- Anesthesiology Research Laboratory, University of California, San Diego, USA
| |
Collapse
|
228
|
Law PY, McGinn TM, Campbell KM, Erickson LE, Loh HH. Agonist activation of delta-opioid receptor but not mu-opioid receptor potentiates fetal calf serum or tyrosine kinase receptor-mediated cell proliferation in a cell-line-specific manner. Mol Pharmacol 1997; 51:152-60. [PMID: 9016358 DOI: 10.1124/mol.51.1.152] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Activation by opioid receptors of cell proliferation was examined with fibroblast cell lines stably expressing either delta-opioid or mu-opioid receptors. Addition of [D-Ala2, D-Leu5]-enkephalin or [D-Pen2,D-Pen5]-enkephalin to Chinese hamster ovary (CHO) cells transfected with delta-opioid receptor cDNA resulted in an agonist concentration-dependent potentiation of fetal calf serum (FCS)-stimulated cell proliferation. This potentiation by delta-opioid agonists was antagonized by naloxone and was not observed with the kappa-opioid receptor selective agonist U50,488 or the mu-opioid receptor selective agonist [D-Ala2,N-MePhe4, Gly-ol5]-enkephalin. This delta-opioid agonist effect was not observed at FCS concentrations > 0.1% and could be blocked by pretreating cells with pertussis toxin, indicating that Gi/Go were involved in this action. In addition, delta-opioid agonists could potentiate CHO cell proliferation stimulated by those growth factors that are mediated by tyrosine kinase receptors (i.e., insulin, insulin-like growth factor 1, and fibroblast-derived growth factor b). This delta-opioid agonist potentiation of growth apparently was dependent on the level of delta-opioid receptors that were expressed and had cell-line selectivity. Activation of delta-opioid receptors expressed in Rat-1 or NIH3T3 fibroblast did not result in a modulation of the cell growth induced by FCS or by growth factors. Interestingly, in CHO cells transfected with mu-opioid receptor cDNA, activation with agonists did not produce a potentiation of FCS-stimulated proliferation. This lack of mu-opioid receptor effect was not due to the differences among CHO clones. In a CHO cell line transfected with both delta-opioid receptor cDNA and mu-opioid receptor cDNA, activation of delta-but not mu-opioid receptors resulted in a potentiation of growth. These data suggest that delta- and mu-opioid receptors in CHO cells activate similar but divergent second messenger pathways, resulting in the differential regulation of cell growth.
Collapse
Affiliation(s)
- P Y Law
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis 55455, USA
| | | | | | | | | |
Collapse
|
229
|
Okuda-Ashitaka E, Tachibana S, Houtani T, Minami T, Masu Y, Nishi M, Takeshima H, Sugimoto T, Ito S. Identification and characterization of an endogenous ligand for opioid receptor homologue ROR-C: its involvement in allodynic response to innocuous stimulus. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1996; 43:96-104. [PMID: 9037523 DOI: 10.1016/s0169-328x(96)00165-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We reported here purification and characterization of a novel heptadecapeptide in bovine brain as an endogenous ligand for ROR-C, an opioid receptor homologue cloned from rat cerebrum. The amino acid sequence of the peptide that we purified is identical to those recently identified as nociceptin in rat brain and orphanin FQ in porcine brain. The peptide inhibited the forskolin-induced cyclic AMP accumulation in ROR-C expressing Chinese hamster ovary cells. Studies on inhibitory activity of cyclic AMP accumulation and Northern blot analysis showed that the peptide and its precursor mRNA are present in a number of brain regions, less abundant in the spina cord, and negligible in the cerebellum. In situ hybridization analysis revealed that hybridization-positive neurons were distributed in the superficial layer (lamina I) of the dorsal horn and were also interspersed between the tract of Lissauer in the spinal cord. Intrathecal administration of the peptide into conscious mice induced allodynia, a pain response to innocuous tactile stimuli, in a beli-shaped manner. These results demonstrate that the peptide exists in the brain and spinal cord and plays an important role in pain transmission.
Collapse
Affiliation(s)
- E Okuda-Ashitaka
- Department of Medical Chemistry, Kansai Medical University, Moriguchi, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Obermeier H, Wehmeyer A, Schulz R. Expression of mu-, delta- and kappa-opioid receptors in baculovirus-infected insect cells. Eur J Pharmacol 1996; 318:161-6. [PMID: 9007528 DOI: 10.1016/s0014-2999(96)00743-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The mu-, delta- and kappa-opioid receptors have been expressed in Sf9 and 'High Five' insect cells using the baculovirus expression system. In both cell lines highest receptor levels (pmol/mg membrane protein) were observed 48 h after infection. Concomitant exposure to the narcotic antagonist naloxone (1 microM) enhanced the production of each receptor type. However, "High Five' cells differed from Sf9 cells in a 2-3-fold higher receptor density in the cell membrane and were therefore employed for receptor characterization. In membranes of 'High Five' cells opioid receptor levels ranged from 1.0 +/- 0.2 pmol/mg protein for the kappa-opioid receptor, 1.7 +/- 0.2 pmol/mg for the delta-opioid receptor to 2.1 +/- 0.5 pmol/mg for the mu-opioid receptor. The mu-, delta- and kappa-opioid receptor agonists [D-Ala2,N-methyl-Phe4-Gly-ol5]enkephalin ([3H]DAMGO), [D-Pen2,D-Pen5]enkephalin ([3H]DPDPE) and (5 alpha, 7 alpha, 8 beta)-(+)-N-methyl-N-(7-(1-pyrrolidinyl-1-oxaspiro(4,5)dec-8-yl) benzeneacetamide ([3H]U69,563) bound to the opioid receptors with Kd values of 3.4 +/- 0.3 nM, 4.5 +/- 0.1 nM and 1.2 +/- 0.3 nM, respectively, resembling those reported for opioid receptors expressed in mammalian cells. Testing the functionality of the receptors in 'High Five' cells, we found that high affinity agonist binding was strongly reduced in the presence of GTP gamma S/sodium, indicating their coupling to G proteins. Furthermore, activation of the three receptor types inhibited forskolin-stimulated cAMP formation. The results presented here suggest that the 'High Five' cell/baculovirus system provides a convenient method for high level expression of functionally intact opioid receptors as judged by receptor binding studies, their G-protein coupling and inhibition of adenylyl cyclase.
Collapse
MESH Headings
- Adenylyl Cyclases/metabolism
- Animals
- Baculoviridae/genetics
- Benzeneacetamides
- Cell Line
- Cyclic AMP/biosynthesis
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- GTP-Binding Proteins/metabolism
- Insecta
- Kinetics
- Protein Binding
- Pyrrolidines/pharmacology
- Receptors, Opioid/agonists
- Receptors, Opioid/biosynthesis
- Receptors, Opioid/metabolism
- Receptors, Opioid, delta/biosynthesis
- Receptors, Opioid, kappa/biosynthesis
- Receptors, Opioid, mu/biosynthesis
- Recombinant Proteins/agonists
- Recombinant Proteins/biosynthesis
- Recombinant Proteins/metabolism
Collapse
Affiliation(s)
- H Obermeier
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians University Munich, Germany
| | | | | |
Collapse
|
231
|
Steiner H, Gerfen CR. Dynorphin regulates D1 dopamine receptor-mediated responses in the striatum: relative contributions of pre- and postsynaptic mechanisms in dorsal and ventral striatum demonstrated by altered immediate-early gene induction. J Comp Neurol 1996; 376:530-41. [PMID: 8978468 DOI: 10.1002/(sici)1096-9861(19961223)376:4<530::aid-cne3>3.0.co;2-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Dynorphin, an endogenous kappa opioid receptor ligand, acts in the striatum to regulate the response of striatonigral neurons to D1 dopamine receptor stimulation. We investigated the relative contributions of both presynaptic kappa receptors on dopamine terminals and postsynaptic kappa receptors on striatal neurons by analyzing opioid regulation of D1 effects in the absence of presynaptic kappa receptors, after 6-hydroxydopamine depletion of striatal dopamine. D1-receptor-mediated immediate-early gene induction was measured by using in situ hybridization histochemistry. First, repeated treatment with the D1-receptor agonist SKF-38393 (2 mg/kg/day, 3-14 days) was used to increase dynorphin levels in rats with dopamine depletions. In the nucleus accumbens, increased dynorphin expression was accompanied by reduced induction of the immediate-early genes c-fos and zif 268 by SKF-38393. In contrast, in dorsal/lateral aspects of the dopamine-depleted striatum, this D1 response was sustained despite a large increase in dynorphin expression. These results are consistent with a requirement of dopamine terminals (presynaptic kappa receptors) for the inhibitory action of dynorphin in the dorsal/lateral striatum, but not in the ventral striatum. Second, the kappa receptor agonist spiradoline (1-10 mg/kg) reduced c-fos and zif 268 induction by SKF-39393 (2.5 mg/kg) preferentially in ventral parts of the dopamine-depleted striatum, which contain higher levels of kappa receptor mRNA and binding. These results also indicate that postsynaptic kappa receptors contribute to the inhibition of the D1 response at least in the ventral striatum. Together, these results indicate that dynorphin in the striatum functions to regulate dopamine input to striatonigral neurons, acting at both pre- and postsynaptic sites, and that the relative contributions of these mechanisms differ between dorsal and ventral striatal regions.
Collapse
Affiliation(s)
- H Steiner
- Laboratory of Neurophysiology, National Institute of Mental Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
232
|
Heteromultimerization of G-protein-gated inwardly rectifying K+ channel proteins GIRK1 and GIRK2 and their altered expression in weaver brain. J Neurosci 1996. [PMID: 8929423 DOI: 10.1523/jneurosci.16-22-07137.1996] [Citation(s) in RCA: 200] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The weaver (wv) gene (GIRK2) is a member of the G-protein-gated inwardly rectifying potassium (GIRK) channel family, known effectors in the signal transduction pathway of neurotransmitters such as acetylcholine, dopamine, opioid peptides, and substance P in modulation of neurotransmitter release and neuronal excitability. GIRK2 immunoreactivity is found in but not limited to brain regions known to be affected in wv mice, such as the cerebellar granule cells and dopaminergic neurons in the substantia nigra pars compacta. It is also observed in the ventral tegmental area, hippocampus, cerebral cortex, and thalamus. GIRK2 and GIRK1, a related family member, have overlapping yet distinct distributions in rat and mouse brains. In regions where both channel proteins are expressed, such as the cerebral cortex, hippocampus, and cerebellum, they can be co-immunoprecipitated, indicating that they interact to form heteromeric channels in vivo. In the brain of the wv mouse, GIRK2 expression is decreased dramatically. In regions where GIRK1 and GIRK2 distributions overlap, both GIRK1 and GIRK2 expressions are severely disrupted, probably because of their co-assembly. The expression patterns of these GIRK channel subunits provide a basis for consideration of the machinery for neuronal signaling as well as the differential effects of the wv mutation in various neurons.
Collapse
|
233
|
Abstract
Orphanin FQ (OFQ) has recently been reported to be an endogenous ligand for the opioid-like LC132 receptor. The effect of OFQ on high voltage-gated calcium channels (VGCCs) was examined in freshly dissociated rat pyramidal neurons using the whole-cell configuration of the patch-clamp technique. High-threshold Ba2+ currents were reversibly inhibited by OFQ. The depression of the currents was associated with a slowed rate of activation and a change in the activation I-V relationship at step potentials higher than +30 mV. In concentration-response experiments, a mean (+/-SEM) pEC50 value of 7.0 +/- 0.07 and a Hill coefficient of 1.5 +/- 0.08 (n = 5) were obtained. The near-maximum inhibition of the Ba2+ currents by OFQ (1 microM) amounted to 31 +/- 2.2% of control (n = 15). Opioid receptors could not account for the effects of OFQ on VGCCs, because naloxone, a broad spectrum mu-, delta-, and kappa-receptor antagonist, did not reduce the effectiveness of OFQ. When GTP-gamma-S was included in the pipette, the depression of the currents by OFQ was irreversible, whereas currents from neurons preincubated with pertussis toxin were not inhibited by OFQ, consistent with the involvement of a PTX-sensitive G-protein. When selective blockers of VGCCs were used, it was demonstrated that all subtypes of VGCCs were affected by OFQ. In conclusion, the effect of OFQ on VGCCs expressed in hippocampal CA3 and CA1 neurons may play an important role in the regulation of hippocampal cell excitability and neurotransmitter release.
Collapse
|
234
|
Liao YJ, Jan YN, Jan LY. Heteromultimerization of G-protein-gated inwardly rectifying K+ channel proteins GIRK1 and GIRK2 and their altered expression in weaver brain. J Neurosci 1996; 16:7137-50. [PMID: 8929423 PMCID: PMC6578936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/1996] [Accepted: 08/22/1996] [Indexed: 02/03/2023] Open
Abstract
The weaver (wv) gene (GIRK2) is a member of the G-protein-gated inwardly rectifying potassium (GIRK) channel family, known effectors in the signal transduction pathway of neurotransmitters such as acetylcholine, dopamine, opioid peptides, and substance P in modulation of neurotransmitter release and neuronal excitability. GIRK2 immunoreactivity is found in but not limited to brain regions known to be affected in wv mice, such as the cerebellar granule cells and dopaminergic neurons in the substantia nigra pars compacta. It is also observed in the ventral tegmental area, hippocampus, cerebral cortex, and thalamus. GIRK2 and GIRK1, a related family member, have overlapping yet distinct distributions in rat and mouse brains. In regions where both channel proteins are expressed, such as the cerebral cortex, hippocampus, and cerebellum, they can be co-immunoprecipitated, indicating that they interact to form heteromeric channels in vivo. In the brain of the wv mouse, GIRK2 expression is decreased dramatically. In regions where GIRK1 and GIRK2 distributions overlap, both GIRK1 and GIRK2 expressions are severely disrupted, probably because of their co-assembly. The expression patterns of these GIRK channel subunits provide a basis for consideration of the machinery for neuronal signaling as well as the differential effects of the wv mutation in various neurons.
Collapse
Affiliation(s)
- Y J Liao
- Howard Hughes Medical Institute, Department of Physiology, University of California, San Francisco 94143-0724, USA
| | | | | |
Collapse
|
235
|
Abstract
Polymerase chain reaction was used to determine whether opioid receptor-like sequences are present in species from the protostome and deuterostome branches of the metazoan kingdom. Multiple opioid receptor-like sequences were found in all vertebrates, but no specific fragments were obtained from any invertebrates. Delta, mu, kappa and ORL-1 receptors were identified from bovine DNA, and three different opioid receptor-like fragments were identified from the other vertebrates analyzed. The data suggest that the opioid receptor gene family has been highly conserved during vertebrate evolution and that, even in the primitive jawless fish, multiple members of the opioid receptor family appear to be present.
Collapse
Affiliation(s)
- X Li
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, 90024-1759, USA
| | | | | |
Collapse
|
236
|
Metzger TG, Paterlini MG, Portoghese PS, Ferguson DM. Application of the message-address concept to the docking of naltrexone and selective naltrexone-derived opioid antagonists into opioid receptor models. Neurochem Res 1996; 21:1287-94. [PMID: 8947918 DOI: 10.1007/bf02532369] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A binding site model for the opioid family of G-protein coupled receptors (GPCRs) is proposed based on the message-address concept of ligand recognition. Using ligand docking studies of the universal opioid antagonist, naltrexone, the structural basis for "message' recognition is explored across all three receptor types, mu, delta, and kappa. The binding mode proposed and basis for selectivity are also rationalized using the naltrexone-derived ligands, naltrindole (NTI) and norbinaltorphimine (nor BNI). These ligands are docked to the receptor according to the common naltrexone core or message. The resulting orientation places key "address' elements in close proximity to amino acid residues critical to selectivity among receptors types. Selectivity is explained by sequence differences in the mu, delta, and kappa receptors at these recognition points. Support for the model is derived from site directed mutagenesis studies and ligand binding data for the opioid receptors and other related GPCRs.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Binding Sites
- Conserved Sequence
- GTP-Binding Proteins/metabolism
- Models, Molecular
- Models, Structural
- Molecular Conformation
- Molecular Sequence Data
- Naltrexone/analogs & derivatives
- Naltrexone/chemistry
- Naltrexone/metabolism
- Narcotic Antagonists/chemistry
- Narcotic Antagonists/metabolism
- Protein Conformation
- Receptors, Opioid, delta/chemistry
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/chemistry
- Receptors, Opioid, kappa/metabolism
Collapse
Affiliation(s)
- T G Metzger
- Department of Medicinal Chemistry and Minnesota Supercomputer Institute, University of Minnesota, Minneapolis 55455, USA
| | | | | | | |
Collapse
|
237
|
Knoflach F, Reinscheid RK, Civelli O, Kemp JA. Modulation of voltage-gated calcium channels by orphanin FQ in freshly dissociated hippocampal neurons. J Neurosci 1996; 16:6657-64. [PMID: 8824306 PMCID: PMC6579277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/1996] [Revised: 08/06/1996] [Accepted: 08/09/1996] [Indexed: 02/02/2023] Open
Abstract
Orphanin FQ (OFQ) has recently been reported to be an endogenous ligand for the opioid-like LC132 receptor. The effect of OFQ on high voltage-gated calcium channels (VGCCs) was examined in freshly dissociated rat pyramidal neurons using the whole-cell configuration of the patch-clamp technique. High-threshold Ba2+ currents were reversibly inhibited by OFQ. The depression of the currents was associated with a slowed rate of activation and a change in the activation I-V relationship at step potentials higher than +30 mV. In concentration-response experiments, a mean (+/-SEM) pEC50 value of 7.0 +/- 0.07 and a Hill coefficient of 1.5 +/- 0.08 (n = 5) were obtained. The near-maximum inhibition of the Ba2+ currents by OFQ (1 microM) amounted to 31 +/- 2.2% of control (n = 15). Opioid receptors could not account for the effects of OFQ on VGCCs, because naloxone, a broad spectrum mu-, delta-, and kappa-receptor antagonist, did not reduce the effectiveness of OFQ. When GTP-gamma-S was included in the pipette, the depression of the currents by OFQ was irreversible, whereas currents from neurons preincubated with pertussis toxin were not inhibited by OFQ, consistent with the involvement of a PTX-sensitive G-protein. When selective blockers of VGCCs were used, it was demonstrated that all subtypes of VGCCs were affected by OFQ. In conclusion, the effect of OFQ on VGCCs expressed in hippocampal CA3 and CA1 neurons may play an important role in the regulation of hippocampal cell excitability and neurotransmitter release.
Collapse
Affiliation(s)
- F Knoflach
- F. Hoffmann-La Roche Ltd., Pharma Division, Preclinical Research, CH-4070 Basel, Switzerland
| | | | | | | |
Collapse
|
238
|
Devine DP, Taylor L, Reinscheid RK, Monsma FJ, Civelli O, Akil H. Rats rapidly develop tolerance to the locomotor-inhibiting effects of the novel neuropeptide orphanin FQ. Neurochem Res 1996; 21:1387-96. [PMID: 8947929 DOI: 10.1007/bf02532380] [Citation(s) in RCA: 102] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We examined the effects of intracerebroventricular (i.c.v.) administration of orphanin FQ (OFQ) on locomotor activity in rats. The rats were habituated to locomotor-testing boxes and then injected i.c.v. with OFQ (0 - 10 nmoles). Acute injections of OFQ produced dose-orderly reductions in horizontal locomotion and rearing activity. This suppression of motor activity was characterized by a disruption of balance and muscle control. Within minutes of i.c.v. injection of the higher doses of OFQ, the rats exhibited flaccid muscle tone. They each lay in an atypical posture, pressing the abdomen against the floor, and splaying the hindlimbs. When these rats locomoted, their gate was unsteady. They wobbled from side to side, and frequently fell over. Repeated daily injections of OFQ resulted in a rapid development of tolerance to the OFQ-induced suppression of locomotion and rearing activity. Tolerance to the observed impairments of motor control were also apparent. In the rats that were repeatedly treated with the highest dose (10 nmol) of OFQ, tolerance to the motoric effects was still apparent after 7 days without OFQ treatment.
Collapse
Affiliation(s)
- D P Devine
- Mental Health Research Institute, University of Michigan, Ann Arbor 48109-0720, USA
| | | | | | | | | | | |
Collapse
|
239
|
Drake CT, Patterson TA, Simmons ML, Chavkin C, Milner TA. Kappa opioid receptor-like immunoreactivity in guinea pig brain: ultrastructural localization in presynaptic terminals in hippocampal formation. J Comp Neurol 1996; 370:377-95. [PMID: 8799863 DOI: 10.1002/(sici)1096-9861(19960701)370:3<377::aid-cne8>3.0.co;2-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Physiological and pharmacological studies have suggested that kappa opioid receptors (KORs) may be located presynaptically in the guinea pig hippocampal formation. In the present study, KOR-like immunoreactivity (-LI) was examined by using a rabbit antibody raised against a synthetic peptide from the carboxyl terminus of a cloned rat kappa receptor (KT). The specificity of affinity-purified KT antibody was confirmed by Western blotting, enzyme-linked immunosorbent assay, immunolabeling of KORs expressed in Xenopus oocytes, and immunocytochemical preadsorption controls. Specificity also was demonstrated by the light microscopic distribution of KT-LI in sections through the forebrain and the pons, which was largely consistent with the distribution of KORs previously reported, and resembled that of immunoreactivity for dynorphin B, an endogenous ligand for KORs. Detailed analysis of the hippocampal formation revealed that KT-LI was located predominantly in thin processes in the granule cell and inner molecular layers of the dentate gyrus. A few KT-labeled processes were also present in stratum lacunosum-moleculare of the CA1 region and all layers of the CA3 region of the hippocampus. By electron microscopy, KT-LI was restricted to unmyelinated axons and axon terminals, and was associated with plasma membranes, large dense-core vesicles, and cytoplasmic surfaces of small vesicles. In the dentate gyrus, immunolabeled terminals formed asymmetric synapses with granule cell perikarya and large unlabeled dendrites. In the CA3 region of hippocampus, KT-LI was present in small unmyelinated axons. The results of this study 1) demonstrate the specificity of the KT antibody, 2) show that the distribution of KT labeling corresponds well with previous KOR and dynorphin localization in many regions, and 3) provide ultrastructural evidence that KORs are located presynaptically in the guinea pig hippocampal formation.
Collapse
Affiliation(s)
- C T Drake
- Department of Neurology and Neuroscience, Cornell University Medical College, New York, New York 10021, USA
| | | | | | | | | |
Collapse
|
240
|
Claude PA, Wotta DR, Zhang XH, Prather PL, McGinn TM, Erickson LJ, Loh HH, Law PY. Mutation of a conserved serine in TM4 of opioid receptors confers full agonistic properties to classical antagonists. Proc Natl Acad Sci U S A 1996; 93:5715-9. [PMID: 8650158 PMCID: PMC39126 DOI: 10.1073/pnas.93.12.5715] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The involvement of a conserved serine (Ser196 at the mu-, Ser177 at the delta-, and Ser187 at the kappa-opioid receptor) in receptor activation is demonstrated by site-directed mutagenesis. It was initially observed during our functional screening of a mu/delta-opioid chimeric receptor, mu delta2, that classical opioid antagonists such as naloxone, naltrexone, naltriben, and H-Tyr-Tic[psi,CH2NH]Phe-Phe-OH (TIPPpsi; Tic = 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid) could inhibit forskolin-stimulated adenylyl cyclase activity in CHO cells stably expressing the chimeric receptor. Antagonists also activated the G protein-coupled inward rectifying potassium channel (GIRK1) in Xenopus oocytes coexpressing the mu delta2 opioid receptor and the GIRK1 channel. By sequence analysis and back mutation, it was determined that the observed antagonist activity was due to the mutation of a conserved serine to leucine in the fourth transmembrane domain (S196L). The importance of this serine was further demonstrated by analogous mutations created in the mu-opioid receptor (MORS196L) and delta-opioid receptor (DORS177L), in which classical opioid antagonists could inhibit forskolin-stimulated adenylyl cyclase activity in CHO cells stably expressing either MORS196L or DORS177L. Again, antagonists could activate the GIRK1 channel coexpressed with either MORS196L or DORS177L in Xenopus oocytes. These data taken together suggest a crucial role for this serine residue in opioid receptor activation.
Collapse
Affiliation(s)
- P A Claude
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis, 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
241
|
Abstract
To help characterize kappa3 receptors and establish their relationship to traditional mu and delta receptors, we have generated a kappa3-selective monoclonal antibody. Monoclonal antibodies were raised against BE(2)-C cells, a human neuroblastoma cell line containing mu, kappa3, and delta opioid receptors. Of the 5,000 hybridoma cell lines screened, approximately 2,000 hybridomas tested positive against BE(2)-C membranes by ELISA, but only 98 of these were negative against a different neuroblastoma cell line lacking opioid receptors. Supernatants from one hybridoma, 8D8, inhibited up to 90% of 3H-NalBzoH (kappa3) binding without affecting 3H-DAMGO (mu) or 3H-naltrindole (delta) binding in BE(2)-C membranes. The selectivity of the antibody was further demonstrated by its blockade of the inhibition of cAMP accumulation in BE(2)-C cells by the kappa3 agonist NalBzoH but not the mu agonist morphine. Monoclonal antibody 8D8 (mAb8D8) also recognizes kappa3 receptors from mouse, rat, and calf brain. Administered intracerebroventricularly, mAb8D8 blocked kappa3 but not morphine (mu) analgesia in vivo. On Western blots, mAb8D8 recognized a protein with a molecular mass of approximately 70 kilodaltons in BE(2)-C. These studies demonstrate the selectivity of mAb8D8 for kappa3 receptors and provide additional support for the existence of this unique opioid receptor subtype.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Antibodies, Monoclonal
- Binding, Competitive/drug effects
- Blotting, Western
- Brain Neoplasms/immunology
- Cyclic AMP/metabolism
- Enzyme-Linked Immunosorbent Assay
- Male
- Mice
- Mice, Inbred Strains
- Neuroblastoma/immunology
- Opioid Peptides/metabolism
- Pain Measurement/drug effects
- Rats
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, kappa/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- A I Brooks
- The Cotzias Laboratory of Neuro-Oncology, Memorial Sloan-Kettering Cancer Center, New York 10021, USA
| | | | | | | | | |
Collapse
|
242
|
Opioids and the Control of Pain. Neurotherapeutics 1996. [DOI: 10.1007/978-1-59259-466-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
243
|
Leshner AI. Toward the year 2000. Subst Use Misuse 1996; 31:207-13. [PMID: 8834008 DOI: 10.3109/10826089609045808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
244
|
Randić M. Plasticity of excitatory synaptic transmission in the spinal cord dorsal horn. PROGRESS IN BRAIN RESEARCH 1996; 113:463-506. [PMID: 9009751 DOI: 10.1016/s0079-6123(08)61104-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- M Randić
- Department of Veterinary Physiology and Pharmacology, Iowa State University, Ames 50011, USA
| |
Collapse
|
245
|
Wang WW, Shahrestanifar M, Jin J, Howells RD. Studies on mu and delta opioid receptor selectivity utilizing chimeric and site-mutagenized receptors. Proc Natl Acad Sci U S A 1995; 92:12436-40. [PMID: 8618916 PMCID: PMC40372 DOI: 10.1073/pnas.92.26.12436] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Opioid receptors are members of the guanine nucleotide binding protein (G protein)-coupled receptor family. Three types of opioid receptors have been cloned and characterized and are referred to as the delta, kappa and mu types. Analysis of receptor chimeras and site-directed mutant receptors has provided a great deal of information about functionally important amino acid side chains that constitute the ligand-binding domains and G-protein-coupling domains of G-protein-coupled receptors. We have constructed delta/mu opioid receptor chimeras that were express in human embryonic kidney 293 cells in order to define receptor domains that are responsible for receptor type selectivity. All chimeric receptors and wild-type delta and mu opioid receptors displayed high-affinity binding of etorphine (an agonist), naloxone (an antagonist), and bremazocine (a mixed agonist/antagonist). In contrast, chimeras that lacked the putative first extracellular loop of the mu receptor did not bind the mu-selective peptide [D-Ala2,MePhe4,Gly5-ol]enkephalin (DAMGO). Chimeras that lacked the putative third extracellular loop of the delta receptor did not bind the delta-selective peptide, [D-Ser2,D-Leu5]enkephalin-Thr (DSLET). Point mutations in the putative third extracellular loop of the wild-type delta receptor that converted vicinal arginine residues to glutamine abolished DSLET binding while not affecting bremazocine, etorphine, and naltrindole binding. We conclude that amino acids in the putative first extracellular loop of the mu receptor are critical for high-affinity DAMGO binding and that arginine residues in the putative third extracellular loop of the delta receptor are important for high-affinity DSLET binding.
Collapse
MESH Headings
- Amino Acid Sequence
- Analgesics/metabolism
- Base Sequence
- Benzomorphans/metabolism
- Binding, Competitive
- Cell Line
- Cell Membrane/physiology
- Cell Membrane/ultrastructure
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalin, Leucine/analogs & derivatives
- Enkephalin, Leucine/metabolism
- Enkephalins/metabolism
- Etorphine/metabolism
- GTP-Binding Proteins/metabolism
- Humans
- Kidney
- Kinetics
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Naloxone/metabolism
- Oligodeoxyribonucleotides
- Polymerase Chain Reaction
- Protein Structure, Secondary
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/biosynthesis
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/biosynthesis
- Receptors, Opioid, mu/metabolism
- Recombinant Fusion Proteins/antagonists & inhibitors
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/metabolism
- Substrate Specificity
- Transfection
Collapse
Affiliation(s)
- W W Wang
- Department of Biochemistry and Molecular Biology, New Jersey Medical School, Newark 07103, USA
| | | | | | | |
Collapse
|
246
|
Kieffer BL. Recent advances in molecular recognition and signal transduction of active peptides: receptors for opioid peptides. Cell Mol Neurobiol 1995; 15:615-35. [PMID: 8719033 PMCID: PMC11563145 DOI: 10.1007/bf02071128] [Citation(s) in RCA: 297] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/1995] [Accepted: 04/15/1995] [Indexed: 02/01/2023]
Abstract
1. Opioid peptides are a family of structurally related neuromodulators which play a major role in the control of nociceptive pathways. These peptides act through membrane receptors of the nervous system, defined as mu, delta and kappa and endowed with overlapping but distinct pharmacological, anatomical and functional properties. 2. Recent cloning of an opioid receptor gene family has opened the way to the use of recombinant DNA technology at the receptor level. 3. This review focuses on the molecular cloning and functional characterization of opioid receptors and provides first insights into molecular aspects of opioid peptide recognition and signal transduction mechanisms, using the cloned receptors as investigation tools.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Brain/physiology
- Cloning, Molecular
- GTP-Binding Proteins/physiology
- Humans
- Mice
- Molecular Sequence Data
- Multigene Family
- Receptors, Opioid/chemistry
- Receptors, Opioid/genetics
- Receptors, Opioid/physiology
- Receptors, Opioid, delta/chemistry
- Receptors, Opioid, kappa/chemistry
- Receptors, Opioid, mu/chemistry
- Sequence Homology, Amino Acid
- Signal Transduction
Collapse
Affiliation(s)
- B L Kieffer
- Ecole Supérieure de Biotechnologie de Strabourg, Illkirch France
| |
Collapse
|
247
|
Gorodinsky A, Barg J, Belcheva MM, Levy R, McHale RJ, Vogel Z, Coscia CJ. Dynorphins modulate DNA synthesis in fetal brain cell aggregates. J Neurochem 1995; 65:1481-6. [PMID: 7561841 PMCID: PMC2581519 DOI: 10.1046/j.1471-4159.1995.65041481.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Previously, opioid peptide analogues, beta-endorphin, and synthetic opiates were found to inhibit DNA synthesis in 7-day fetal rat brain cell aggregates via kappa- and mu-opioid receptors. Here dynorphins and other endogenous opioid peptides were investigated for their effect on DNA synthesis in rat and guinea pig brain cell aggregates. At 1 microM, all dynorphins tested and beta-endorphin inhibited [3H]thymidine incorporation into DNA by 20-38% in 7-day rat brain cell aggregates. The putative epsilon-antagonist beta-endorphin (1-27) did not prevent the effect of beta-endorphin, suggesting that the epsilon-receptor is not involved in opioid inhibition of DNA synthesis. The kappa-selective antagonist norbinaltorphimine blocked dynorphin A or B inhibition of DNA synthesis, implicating a kappa-opioid receptor. In dose-dependency studies, dynorphin B was three orders of magnitude more potent than dynorphin A in the attenuation of thymidine incorporation, indicative of the mediation of its action by a discrete kappa-receptor subtype. The IC50 value of 0.1 nM estimated for dynorphin B is in the physiological range for dynorphins in developing brain. In guinea pig brain cell aggregates, the kappa-receptor agonists U50488, U69593, and dynorphin B reduced thymidine incorporation by 40%. When 21-day aggregates were treated with dynorphins, a 33-86% enhancement of thymidine incorporation was observed. Because both 7- and 21-day aggregates correspond to stages in development when glial cell proliferation is prevalent and glia preferentially express kappa-receptors in rat brain, these findings support the hypothesis that dynorphins modulate glial DNA synthesis during brain ontogeny.
Collapse
Affiliation(s)
- A Gorodinsky
- Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, MO 63104-1079, USA
| | | | | | | | | | | | | |
Collapse
|
248
|
Zhang S, Yu L. Identification of dynorphins as endogenous ligands for an opioid receptor-like orphan receptor. J Biol Chem 1995; 270:22772-6. [PMID: 7559404 DOI: 10.1074/jbc.270.39.22772] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
To identify the endogenous ligands for a cloned orphan receptor that shares high degrees of sequence homology with opioid receptors, this orphan receptor was expressed in Xenopus oocytes and in mammalian cell lines CHO-K1 and HEK-293. The coupling of the receptor to a G protein-activated K+ channel was used as a functional assay in oocytes. Endogenous opioid peptide dynorphins were found to activate the K+ channel by stimulating the orphan receptor. This activation was dose-dependent, with EC50 values at 45 and 37 nM for dynorphin A and dynorphin A-(1-13), respectively. The dynorphin effect was antagonized by the non-selective opioid antagonist naloxone but at rather high concentrations in the micromolar range. Naloxone also caused a rightward shift of the dose-response curve for dynorphin A, suggesting a competitive antagonism mechanism. In transiently transfected cells, 5 microM dynorphin A-(1-13) inhibited the forskolin-stimulated cyclic AMP increase by 51 and 35% in CHO-K1 and HEK-293 cells, respectively. Other classes of endogenous opioids, i.e. enkephalins and endorphins, caused very little activation of this receptor. These results suggest that this orphan receptor is a member of the opioid receptor family and that dynorphins are endogenous ligands for this receptor.
Collapse
Affiliation(s)
- S Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis 46202, USA
| | | |
Collapse
|
249
|
Simonin F, Gavériaux-Ruff C, Befort K, Matthes H, Lannes B, Micheletti G, Mattéi MG, Charron G, Bloch B, Kieffer B. kappa-Opioid receptor in humans: cDNA and genomic cloning, chromosomal assignment, functional expression, pharmacology, and expression pattern in the central nervous system. Proc Natl Acad Sci U S A 1995; 92:7006-10. [PMID: 7624359 PMCID: PMC41460 DOI: 10.1073/pnas.92.15.7006] [Citation(s) in RCA: 169] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Using the mouse delta-opioid receptor cDNA as a probe, we have isolated genomic clones encoding the human mu- and kappa-opioid receptor genes. Their organization appears similar to that of the human delta receptor gene, with exon-intron boundaries located after putative transmembrane domains 1 and 4. The kappa gene was mapped at position q11-12 in human chromosome 8. A full-length cDNA encoding the human kappa-opioid receptor has been isolated. The cloned receptor expressed in COS cells presents a typical kappa 1 pharmacological profile and is negatively coupled to adenylate cyclase. The expression of kappa-opioid receptor mRNA in human brain, as estimated by reverse transcription-polymerase chain reaction, is consistent with the involvement of kappa-opioid receptors in pain perception, neuroendocrine physiology, affective behavior, and cognition. In situ hybridization studies performed on human fetal spinal cord demonstrate the presence of the transcript specifically in lamina II of the dorsal horn. Some divergences in structural, pharmacological, and anatomical properties are noted between the cloned human and rodent receptors.
Collapse
MESH Headings
- Adenylyl Cyclases
- Aged
- Amino Acid Sequence
- Base Sequence
- Central Nervous System/chemistry
- Central Nervous System/physiology
- Chromosome Mapping
- Chromosomes, Human, Pair 8/genetics
- Cloning, Molecular
- DNA, Complementary/genetics
- Exons/genetics
- Genomic Library
- Humans
- In Situ Hybridization
- Ligands
- Middle Aged
- Molecular Sequence Data
- RNA, Messenger/isolation & purification
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/isolation & purification
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/isolation & purification
- Receptors, Opioid, mu/metabolism
- Recombinant Proteins/metabolism
- Sequence Homology, Amino Acid
- Signal Transduction
- Tissue Distribution
Collapse
Affiliation(s)
- F Simonin
- Ecole Supérieure de Biotechnologie, Parc d'innovation, Boulevard Sébastien Brandt, Illkirch-Graffenstaden, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Maderspach K, Takács J, Niewiadomska G, Csillag A. Postsynaptic and extrasynaptic localization of kappa-opioid receptor in selected brain areas of young rat and chick using an anti-receptor monoclonal antibody. JOURNAL OF NEUROCYTOLOGY 1995; 24:478-86. [PMID: 7595663 DOI: 10.1007/bf01181608] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
kappa-opioid receptors were visualized by light and electron microscopical immunohistochemistry in young rat and chick brains, using a monoclonal antibody KA8 (IgG1, kappa) raised against a kappa-opioid receptor preparation from frog brain, which recognizes selectively the kappa-type receptor with preference for the kappa-2 subtype. The most pronounced kappa-opioid receptor-like immunoreactivity was observed in the hypothalamic nuclei of the rat brain and in the chick optic tectum, in regions where the functional significance of kappa-opioid receptors is well documented. Both neurons and glia were stained, the former on both somata and dendrites. At the ultrastructural level, the receptor-like immunoreactivity was similar in both species. Immunoprecipitate decorated the inner surface of the plasma membrane of glial cells, neuronal somata and dendrites, in a discontinuous arrangement. In the cytoplasm, labelling was associated with ribosomes, polyribosomes and rough endoplasmic reticulum membranes but not with Golgi cisternae. In the neuropil, the immunoprecipitate was observed along the dendritic microtubules and was also associated with postsynaptic sites. Nuclei and axons were devoid of label and immunoreactivity was never visible presynaptically. Our findings indicate that the antibody used in the present study marks various forms of the kappa-opioid receptor protein including those synthesised in ribosomes, transported along dendritic microtubules and incorporated into postsynaptic and non-synaptic membranes. The antibody also recognizes glial opioid receptors. The observed subcellular distribution appears to be conserved in phylogenetically distant species.
Collapse
Affiliation(s)
- K Maderspach
- Biological Research Center, Hungarian Academy of Sciences, Szeged
| | | | | | | |
Collapse
|