1
|
Khoramjouy M, Ahmadi F, Faizi M, Shahhosseini S. Optimization binding studies of opioid receptors, saturation and competition, using [ 3H]-DAMGO. Pharmacol Rep 2021; 73:1390-1395. [PMID: 33871815 DOI: 10.1007/s43440-021-00265-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/24/2021] [Accepted: 04/07/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Opioid analgesics are prescribed for the moderate to severe pain in the clinic. New analogs of µ-opioid receptors are introduced because they may have less adverse effects and better efficacy. However, these new analogs have to be screened for their receptor affinity before entering clinical trial phases. A common method to do such screening is using radioligand-binding-assay, which is a fast and precise screening technique if the assays are done at an optimum condition. One of the main challenges in this type of screening is to separate free/unbound radioligands from bound radioligands. In this study, we applied a centrifugation method instead of a filtration method to separate free radioligands from bound radioligands, and also optimized the conditions for radioligand receptor binding studies of µ-opioid receptors, saturation, and the competition. METHODS We used the midbrain and brainstem of naltrexone-treated rats as a source of µ-opioid receptors, and [3H]-DAMGO as the radioligand. Naloxone was also used to determine non-specific binding. A given amount of membrane protein was incubated with an increasing amount of radioligand at 37 °C to saturate the receptors at equilibrium and the amount of radioligand saturated in the receptors were used in competition studies. RESULTS 160 µg membrane protein saturated with 20 nM [3H]-DAMGO at 37 °C for 35 min with Kd (15.06 nM, 95% CI 8.117-22.00) and Bmax (0.4750 pmol/mg, 95% CI 0.3839-0.5660). CONCLUSION Applying the centrifugation method instead of the filtration to separate free from bound radioligand produced repeatable and reliable results. The optimum conditions for radioligand binding were used in competition studies which resulted in the expected outcomes.
Collapse
Affiliation(s)
- Mona Khoramjouy
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ahmadi
- PET Radiopharmaceutical Sciences Section of the Molecular Imaging Branch of NIMH at NIH, Washington D.C, USA
| | - Mehrdad Faizi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soraya Shahhosseini
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy and Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Vali-e-Asr Ave., Niayesh Junction, P.O.Box 14155-6153, Tehran, Iran.
| |
Collapse
|
2
|
Ganglion-Specific Sensitivity of P2X3 Receptors to Leu-Enkephalin. NEUROPHYSIOLOGY+ 2020. [DOI: 10.1007/s11062-020-09869-y] [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]
|
3
|
Latif ZEH, Solli KK, Opheim A, Kunoe N, Benth JŠ, Krajci P, Sharma-Haase K, Tanum L. No increased pain among opioid-dependent individuals treated with extended-release naltrexone or buprenorphine-naloxone: A 3-month randomized study and 9-month open-treatment follow-up study. Am J Addict 2019; 28:77-85. [DOI: 10.1111/ajad.12859] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/15/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Zill-e-Huma Latif
- Department of R&D in Mental Health; Akershus University Hospital; Lørenskog Norway
| | - Kristin K. Solli
- Department of R&D in Mental Health; Akershus University Hospital; Lørenskog Norway
- Norwegian Center for Addiction Research; University of Oslo; Oslo Norway
| | - Arild Opheim
- Department of Addiction Medicine; Haukeland University Hospital; Bergen Norway
- The University of Bergen; Bergen Norway
| | - Nikolaj Kunoe
- Norwegian Center for Addiction Research; University of Oslo; Oslo Norway
| | - Jūratė Š. Benth
- Institute of Clinical Medicine; Campus Ahus; University of Oslo; Blindern Norway
- Health Services Research Unit; Akershus University Hospital; Lørenskog Norway
| | - Peter Krajci
- Department of Addiction Medicine; Oslo University Hospital; Oslo Norway
| | - Kamni Sharma-Haase
- Norwegian Center for Addiction Research; University of Oslo; Oslo Norway
| | - Lars Tanum
- Department of R&D in Mental Health; Akershus University Hospital; Lørenskog Norway
- Norwegian Center for Addiction Research; University of Oslo; Oslo Norway
| |
Collapse
|
4
|
Bowman SL, Shiwarski DJ, Puthenveedu MA. Distinct G protein-coupled receptor recycling pathways allow spatial control of downstream G protein signaling. J Cell Biol 2016; 214:797-806. [PMID: 27646272 PMCID: PMC5037407 DOI: 10.1083/jcb.201512068] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 08/12/2016] [Indexed: 12/22/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are recycled via a sequence-dependent pathway that is spatially and biochemically distinct from bulk recycling. Why there are two distinct recycling pathways from the endosome is a fundamental question in cell biology. In this study, we show that the separation of these two pathways is essential for normal spatial encoding of GPCR signaling. The prototypical β-2 adrenergic receptor (B2AR) activates Gα stimulatory protein (Gαs) on the endosome exclusively in sequence-dependent recycling tubules marked by actin/sorting nexin/retromer tubular (ASRT) microdomains. B2AR was detected in an active conformation in bulk recycling tubules, but was unable to activate Gαs. Protein kinase A phosphorylation of B2AR increases the fraction of receptors localized to ASRT domains and biases the downstream transcriptional effects of B2AR to genes controlled by endosomal signals. Our results identify the physiological relevance of separating GPCR recycling from bulk recycling and suggest a mechanism to tune downstream responses of GPCR signaling by manipulating the spatial origin of G protein signaling.
Collapse
Affiliation(s)
- Shanna Lynn Bowman
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | | | | |
Collapse
|
5
|
Alvarado-Bañuelos M, Barrios De Tomasi E, Juárez J. Changes in the incentive value of food after naltrexone treatment depend on a differential preference for a palatable food in male rats. Nutr Neurosci 2016; 20:416-423. [DOI: 10.1080/1028415x.2016.1162389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Mariana Alvarado-Bañuelos
- Laboratorio de Farmacología y Conducta, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Guadalajara, Jalisco CP 44130, México
| | - Eliana Barrios De Tomasi
- Laboratorio de Farmacología y Conducta, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Guadalajara, Jalisco CP 44130, México
| | - Jorge Juárez
- Laboratorio de Farmacología y Conducta, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Guadalajara, Jalisco CP 44130, México
| |
Collapse
|
6
|
Chakrabarti S, Madia PA, Gintzler AR. Selective up-regulation of functional mu-opioid receptor splice variants by chronic opioids. J Neurochem 2016; 136:1119-1130. [PMID: 26718622 DOI: 10.1111/jnc.13519] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/15/2015] [Accepted: 12/18/2015] [Indexed: 11/28/2022]
Abstract
We recently reported (Verzillo, et al. J. Neurochem: 130, 790-796, 2014) that chronic systemic morphine selectively up-regulates mRNA encoding two C-terminal μ-opioid receptor (MOR) splice variants, MOR-1C1 and MOR-1B2 (MOR-1B2/-1C1). Given the known disconnects between changes in levels of mRNA and corresponding protein, it is essential to directly demonstrate that chronic opioid treatment elevates functional MOR-1B2/-1C1 protein prior to inferring relevance of these MOR variants to spinal opioid tolerance mechanisms. Accordingly, we investigated the ability of chronic opioid exposure to up-regulate MOR protein in Chinese hamster ovary cells stably transfected with rat MOR variants MOR-1B2, MOR-1C1, or MOR-1 (considered to be the predominant MOR). Findings revealed that chronic treatment with the clinically relevant opioids morphine, oxycodone and hydrocodone substantially up-regulated membrane MOR-1B2/-1C1 protein. This up-regulation was abolished by the protein synthesis inhibitor cycloheximide, eliminating contributions from receptor redistribution. The increment in MOR-1B2/-1C1 protein was paralleled by a significant increment in opioid agonist-stimulated GTPγS-binding (reflective of increased aggregate MOR G protein coupling) indicating that up-regulated MOR-1B2/-1C1 represented functional receptors. Strikingly, these tolerance-associated adaptations of MOR-1B2/-1C1 differed considerably from those of MOR-1. Antithetical regulation of MOR-1B2/-1C1 and MOR-1 by chronic opioids has significant implications for the design of new therapeutic agents to counteract opioid analgesic tolerance and accompanying addiction. Since chronic opioids induce MOR-1B2/-1C1 up-regulation in spinal cord of males, but not females, elucidating cellular compartments and intracellular pathways mediating MOR-1B2/-1C1 up-regulation and defining their unique signaling attributes would enable a precision medicinal approach to pain management and addiction therapy. In the spinal cord of males, but not females, chronic morphine up-regulates mRNA encoding two mu-opioid receptor (MOR) variants, MOR-1B2 and MOR-1C1 (MOR-1B2/-1C1). We now demonstrate that chronic treatment with the clinically relevant opioids morphine, hydrocodone or oxycodone up-regulates MOR-1B2/-1C1 functional protein, which is dependent on de novo protein synthesis. Findings underscore the importance of unique signaling attributes of MOR variants to sexually dimorphic tolerance mechanisms.
Collapse
Affiliation(s)
- Sumita Chakrabarti
- Department of Obstetrics and Gynecology, State University of New York, Brooklyn, New York, USA
| | - Priyanka A Madia
- Department of Obstetrics and Gynecology, State University of New York, Brooklyn, New York, USA
| | - Alan R Gintzler
- Department of Obstetrics and Gynecology, State University of New York, Brooklyn, New York, USA
| |
Collapse
|
7
|
Bowman SL, Puthenveedu MA. Postendocytic Sorting of Adrenergic and Opioid Receptors: New Mechanisms and Functions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:189-206. [PMID: 26055059 DOI: 10.1016/bs.pmbts.2015.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The endocytic pathway tightly regulates the activity of G protein-coupled receptors (GPCRs). Much of our understanding of this relationship between GPCR endocytic trafficking and signaling comes from studies done on catecholamine and opioid receptors. After ligand-induced endocytosis, a key sorting step in the endosome determines whether receptors are recycled back to the cell surface, leading to recovery of signaling, or are degraded in the lysosome, leading to desensitization. Recycling of GPCRs, unlike that of many other proteins, is an active process driven by specific sequences on the receptor and proteins that interact with this sequence. Recent data suggest that sequence-dependent recycling plays complex roles in regulating both the timing and location of GPCR signaling. This chapter will describe our current understanding of the mechanisms regulating GPCR sorting in the endosome and discuss emerging ideas on their role in GPCR signaling, focusing on adrenergic and opioid receptors as prototypes.
Collapse
Affiliation(s)
- Shanna L Bowman
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
8
|
Reassessment of the Role of the Central Cholinergic System. J Mol Neurosci 2013; 53:352-8. [DOI: 10.1007/s12031-013-0164-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/24/2013] [Indexed: 12/18/2022]
|
9
|
Comparison of tolerance to morphine-induced respiratory and analgesic effects in mice. Toxicol Lett 2013; 217:251-9. [DOI: 10.1016/j.toxlet.2012.12.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 12/26/2012] [Accepted: 12/27/2012] [Indexed: 11/18/2022]
|
10
|
Opioid system and Alzheimer's disease. Neuromolecular Med 2012; 14:91-111. [PMID: 22527793 DOI: 10.1007/s12017-012-8180-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 04/04/2012] [Indexed: 12/15/2022]
Abstract
The opioid system may be involved in the pathogenesis of AD, including cognitive impairment, hyperphosphorylated tau, Aβ production, and neuroinflammation. Opioid receptors influence the regulation of neurotransmitters such as acetylcholine, norepinephrine, GABA, glutamate, and serotonin which have been implicated in the pathogenesis of AD. Opioid system has a close relation with Aβ generation since dysfunction of opioid receptors retards the endocytosis and degradation of BACE1 and γ-secretase and upregulates BACE1 and γ-secretase, and subsequently, the production of Aβ. Conversely, activation of opioid receptors increases the endocytosis of BACE1 and γ-secretase and downregulates BACE1 and γ-secretase, limiting the production of Aβ. The dysfunction of opioid system (opioid receptors and opioid peptides) may contribute to hyperphosphorylation of tau and neuroinflammation, and accounts for the degeneration of cholinergic neurons and cognitive impairment. Thus, the opioid system is potentially related to AD pathology and may be a very attractive drug target for novel pharmacotherapies of AD.
Collapse
|
11
|
Madia PA, Navani DM, Yoburn BC. [(35)S]GTPγS binding and opioid tolerance and efficacy in mouse spinal cord. Pharmacol Biochem Behav 2011; 101:155-65. [PMID: 22108651 DOI: 10.1016/j.pbb.2011.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 10/27/2011] [Accepted: 11/05/2011] [Indexed: 11/18/2022]
Abstract
The present study examined efficacy of a series of opioid agonists and then using chronic in vivo treatment protocols, determined tolerance to opioid agonist stimulated [(35)S]GTPγS (guanosine 5'-O-(3-[(35)S] thio)triphosphate) binding in mouse spinal cord membranes and compared it directly to spinal analgesic tolerance. The [(35)S]GTPγS binding assay was used to estimate efficacy (E(max) and τ; Operational Model of Agonism) of a series of opioid agonists for G-protein activation in mouse spinal cord. The rank order of opioid agonist efficacy determined in the [(35)S]GTPγS assay using the Operational Model and E(max) was similar. These efficacy estimates correlated with historical analgesic efficacy estimates. For tolerance studies, mice were continuously treated s.c. for 7days with morphine, oxycodone, hydromorphone, etorphine or fentanyl and [(35)S]GTPγS studies were conducted in spinal cord membranes. Other mice were tested in i.t. analgesia dose response studies (tailflick). Tolerance to DAMGO ([D-Ala(2),N-MePhe(4),Gly-ol(5)]enkephalin) or morphine stimulated [(35)S]GTPγS binding (decrease in E(max)) was observed following etorphine and fentanyl treatment only. These treatment protocols downregulate μ-opioid receptor density whereas morphine, oxycodone and hydromorphone do not. Spinal analgesic tolerance was observed following all treatment protocols examined (morphine, oxycodone and etorphine). Opioid antagonist treatment that specifically upregulates (chronic naltrexone) or downregulates (clocinnamox) μ-opioid receptor density produced a corresponding change in opioid agonist stimulated [(35)S]GTPγS binding. Although receptor downregulation and G-protein uncoupling are among potential mechanisms of opioid tolerance, the present results suggest that uncoupling in mouse spinal cord plays a minor role and that the [(35)S]GTPγS assay is particularly responsive to changes in μ-opioid receptor density.
Collapse
Affiliation(s)
- Priyanka A Madia
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, United States
| | | | | |
Collapse
|
12
|
Raehal KM, Schmid CL, Groer CE, Bohn LM. Functional selectivity at the μ-opioid receptor: implications for understanding opioid analgesia and tolerance. Pharmacol Rev 2011; 63:1001-19. [PMID: 21873412 DOI: 10.1124/pr.111.004598] [Citation(s) in RCA: 197] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Opioids are the most effective analgesic drugs for the management of moderate or severe pain, yet their clinical use is often limited because of the onset of adverse side effects. Drugs in this class produce most of their physiological effects through activation of the μ opioid receptor; however, an increasing number of studies demonstrate that different opioids, while presumably acting at this single receptor, can activate distinct downstream responses, a phenomenon termed functional selectivity. Functional selectivity of receptor-mediated events can manifest as a function of the drug used, the cellular or neuronal environment examined, or the signaling or behavioral measure recorded. This review summarizes both in vitro and in vivo work demonstrating functional selectivity at the μ opioid receptor in terms of G protein coupling, receptor phosphorylation, interactions with β-arrestins, receptor desensitization, internalization and signaling, and details on how these differences may relate to the progression of analgesic tolerance after their extended use.
Collapse
Affiliation(s)
- Kirsten M Raehal
- Molecular Therapeutics and Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | | | | | | |
Collapse
|
13
|
Patierno S, Anselmi L, Jaramillo I, Scott D, Garcia R, Sternini C. Morphine induces μ opioid receptor endocytosis in guinea pig enteric neurons following prolonged receptor activation. Gastroenterology 2011; 140:618-26. [PMID: 21070774 PMCID: PMC3033567 DOI: 10.1053/j.gastro.2010.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2010] [Revised: 10/18/2010] [Accepted: 11/02/2010] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS The μ opioid receptor (μOR) undergoes rapid endocytosis after acute stimulation with opioids and most opiates, but not with morphine. We investigated whether prolonged activation of μOR affects morphine's ability to induce receptor endocytosis in enteric neurons. METHODS We compared the effects of morphine, a poor μOR-internalizing opiate, and (D-Ala2,MePhe4,Gly-ol5) enkephalin (DAMGO), a potent μOR-internalizing agonist, on μOR trafficking in enteric neurons and on the expression of dynamin and β-arrestin immunoreactivity in the ileum of guinea pigs rendered tolerant by chronic administration of morphine. RESULTS Morphine (100 μmol/L) strongly induced endocytosis of μOR in tolerant but not naive neurons (55.7% ± 9.3% vs 24.2% ± 7.3%; P < .001) whereas DAMGO (10 μmol/L) strongly induced internalization of μOR in neurons from tolerant and naive animals (63.6% ± 8.4% and 66.5% ± 3.6%). Morphine- or DAMGO-induced μOR endocytosis resulted from direct interactions between the ligand and the μOR because endocytosis was not affected by tetrodotoxin, a blocker of endogenous neurotransmitter release. Ligand-induced μOR internalization was inhibited by pretreatment with the dynamin inhibitor, dynasore. Chronic morphine administration resulted in a significant increase and translocation of dynamin immunoreactivity from the intracellular pool to the plasma membrane, but did not affect β-arrestin immunoreactivity. CONCLUSIONS Chronic activation of μORs increases the ability of morphine to induce μOR endocytosis in enteric neurons, which depends on the level and cellular localization of dynamin, a regulatory protein that has an important role in receptor-mediated signal transduction in cells.
Collapse
Affiliation(s)
- Simona Patierno
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA,Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Laura Anselmi
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA,Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Ingrid Jaramillo
- Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA
| | - David Scott
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA,Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA,Department of Physiology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Rachel Garcia
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Catia Sternini
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA,Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA,Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
14
|
Tamagaki S, Suzuki T, Hagihira S, Hayashi Y, Mashimo T. Systemic daily morphine enhances the analgesic effect of intrathecal dexmedetomidine via up-regulation of alpha 2 adrenergic receptor subtypes A, B and C in dorsal root ganglion and dorsal horn. J Pharm Pharmacol 2010; 62:1760-7. [PMID: 21054403 DOI: 10.1111/j.2042-7158.2010.01192.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES It has been reported that the effect of intrathecally administered α2 adrenergic receptor (α2 AR) agonists is enhanced in mice that are chronically tolerant to systemic morphine. However, contributory factors have not been identified. Here we examined whether repeated systemic morphine affected the analgesic potency of intrathecal dexmedetomidine and the expression of subtype A, B and C α2 AR (α2A, α2B and α2C AR) in the dorsal root ganglion and dorsal horn in mice. METHODS After subcutaneous injection of morphine or saline for two weeks, dexmedetomidine was administered intrathecally to evaluate its antinociceptive effect. Also, the α2 AR subtypes and µ-opioid receptor mRNA expression in lumbar dorsal root ganglion was quantified using PCR, and α2A and α2C AR in lumbar dorsal root ganglion and dorsal horn were examined by immunohistochemistry. KEY FINDINGS Daily morphine enhanced the antinociceptive effect of intrathecal dexmedetomidine, increased all the α2 AR subtypes but decreased the µ-opioid receptor mRNA expression in dorsal root ganglion and increased immunoreactivity of α2A and α2C AR in dorsal root ganglion and dorsal horn. CONCLUSIONS These results suggest that systemic daily morphine enhances the analgesic effect of intrathecal dexmedetomidine via up-regulation of the α2A, α2B and α2C AR in lumbar dorsal root ganglion and dorsal horn.
Collapse
Affiliation(s)
- Shinji Tamagaki
- Department of Anesthesiology, Osaka University Medical School, Osaka, Japan
| | | | | | | | | |
Collapse
|
15
|
Zhang Y, Xiong W, Lin X, Ma X, Yu LC. Receptor trafficking induced by mu-opioid-receptor phosphorylation. Neurosci Biobehav Rev 2009; 33:1192-7. [PMID: 19747597 DOI: 10.1016/j.neubiorev.2009.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Revised: 03/10/2009] [Accepted: 03/11/2009] [Indexed: 10/21/2022]
Abstract
Opiates, including morphine, are widely used drugs for antinociception in clinics. Prolonged treatments of opioids induce both tolerance and dependence, which are the major side effects of opioid therapy. One of the mechanisms for the development of tolerance and dependence is implicated to be opioid-receptor trafficking. Here we review the current understandings of opioid-receptor phosphorylation, endocytosis and desensitization after repeated agonist treatments. Also, the role of G-protein coupled receptor kinases in opioid-receptor phosphorylation is discussed. How to associate these observations to physiological and behavioral changes in animal models and clinics is still under investigation.
Collapse
Affiliation(s)
- Yan Zhang
- Laboratory of Neurobiology, College of Life Sciences, Peking University, Beijing 100871, China.
| | | | | | | | | |
Collapse
|
16
|
Persistent inflammatory pain decreases the antinociceptive effects of the mu opioid receptor agonist DAMGO in the locus coeruleus of male rats. Neuropharmacology 2009; 56:1017-26. [PMID: 19265713 DOI: 10.1016/j.neuropharm.2009.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 02/14/2009] [Accepted: 02/19/2009] [Indexed: 12/18/2022]
Abstract
Persistent inflammatory nociception increases levels of endogenous opioids with affinity for delta opioid receptors in the ventromedial medulla and enhances the antinociceptive effects of the mu opioid receptor (MOPr) agonist [D-Ala2, N-Me-Phe4, Gly5-ol]enkephalin (DAMGO) [Hurley, R.W., Hammond, D.L., 2001. Contribution of endogenous enkephalins to the enhanced analgesic effects of supraspinal mu opioid receptor agonists after inflammatory injury. J. Neurosci. 21, 2536-2545]. It also increases levels of endogenous opioids that act at MOPr elsewhere in the CNS [Zangen, A., Herzberg, U., Vogel, Z., Yadid, G., 1998. Nociceptive stimulus induces release of endogenous beta-endorphin in the rat brain. Neuroscience 85, 659-662]. This study tested the hypothesis that a sustained release of endogenous opioids leads to a downregulation of MOPr in the locus coeruleus (LC) and induces a state of endogenous opioid tolerance. Four days after injection of complete Freund's adjuvant (CFA) in the left hindpaw of the rat, both the magnitude and duration of the antinociception produced by microinjection of DAMGO in the right LC were reduced. Saturation isotherms demonstrated a 50% decrease in MOPr B(max) in homogenates of the LC from CFA-treated rats; K(d) was unchanged. Receptor autoradiography revealed that this decrease was bilateral. The decreased efficacy of DAMGO in CFA-treated rats most likely results from a decreased number of MOPr in the LC. Microinjection of the MOPr antagonist D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP) in the LC did not exacerbate hyperalgesia in the ipsilateral hindpaw or produce hyperalgesia in the contralateral hindpaw of CFA-treated rats. The downregulation in MOPr is therefore unlikely to result from the induction of endogenous opioid tolerance in the LC. These results indicate that persistent inflammatory nociception alters the antinociceptive actions of MOPr agonists in the CNS by diverse mechanisms that are nucleus specific and likely to have different physiological implications.
Collapse
|
17
|
Dighe SV, Madia PA, Sirohi S, Yoburn BC. Continuous morphine produces more tolerance than intermittent or acute treatment. Pharmacol Biochem Behav 2009; 92:537-42. [PMID: 19248799 DOI: 10.1016/j.pbb.2009.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 01/30/2009] [Accepted: 02/13/2009] [Indexed: 10/21/2022]
Abstract
Dosing protocol and analgesic efficacy have been proposed to be important determinants of the magnitude of opioid tolerance. The present study examined the effect of acute, intermittent and continuous treatment with the low analgesic efficacy agonist morphine on analgesic tolerance. Mice were implanted s.c. with a 25 mg morphine pellet for 1-7 days. Other mice were implanted s.c. with two 25 mg, or one 75 mg morphine pellet for 7 days. The release of morphine from subcutaneous implanted pellets was quantitated using a spectrophotometric assay. In other studies, mice were injected with morphine once (18.5-185 mg/kg/day; approximately 10-100 times ED(50) for morphine analgesia) or once/day for 7 days. Controls were implanted with a placebo pellet or injected with saline. Analysis of drug release from a 25 mg pellet indicated that release was greatest during the first 24 h, declined and then remained relatively constant. The amount of morphine released over 7 days by a 75 mg pellet (23.9 mg) was more than that of a single 25 mg pellet (15.4 mg) but less than two 25 mg pellets (30.8 mg). Following treatment, morphine cumulative dose-response studies were conducted (tail flick). Continuous treatment with morphine using pellet implantation produced a dose-dependent shift in the morphine ED(50) by 3.3, 5.8 and 8.5 fold for one 25 mg pellet, one 75 mg pellet and two 25 mg pellets, respectively. Acute and intermittent morphine administration produced substantially less analgesic tolerance than continuous release of morphine by implant pellets. The maximum shift in the ED(50) was 1.6 for acute treatment and 2.7 for 7 day intermittent treatment; despite a larger total daily dose. The present results indicate that continuous treatment with morphine results in greater analgesic tolerance than acute or intermittent morphine treatment even at comparable daily doses. These results are consistent with the suggestion that intermittent dosing has reduced risk of producing opioid tolerance.
Collapse
Affiliation(s)
- Shveta V Dighe
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | | | | | | |
Collapse
|
18
|
Kumar P, Sunkaraneni S, Sirohi S, Dighe SV, Walker EA, Yoburn BC. Hydromorphone efficacy and treatment protocol impact on tolerance and mu-opioid receptor regulation. Eur J Pharmacol 2008; 597:39-45. [PMID: 18789923 DOI: 10.1016/j.ejphar.2008.08.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 08/18/2008] [Accepted: 08/21/2008] [Indexed: 11/26/2022]
Abstract
This study examined the antinociceptive (analgesic) efficacy of hydromorphone and hydromorphone-induced tolerance and regulation of mu-opioid receptor density. Initially s.c. hydromorphone's time of peak analgesic (tail-flick) effect (45 min) and ED50 using standard and cumulative dosing protocols (0.22 mg/kg, 0.37 mg/kg, respectively) were determined. The apparent analgesic efficacy (tau) of hydromorphone was then estimated using the operational model of agonism and the irreversible mu-opioid receptor antagonist clocinnamox. Mice were injected with clocinnamox (0.32-25.6 mg/kg, i.p.) and 24 h later, the analgesic potency of hydromorphone was determined. The tau value for hydromorphone was 35, which suggested that hydromorphone is a lower analgesic efficacy opioid agonist. To examine hydromorphone-induced tolerance, mice were continuously infused s.c. with hydromorphone (2.1-31.5 mg/kg/day) for 7 days and then morphine cumulative dose response studies were performed. Other groups of mice were injected with hydromorphone (2.2-22 mg/kg/day) once, or intermittently every 24 h for 7 days. Twenty-four hours after the last injection, mice were tested using morphine cumulative dosing studies. There was more tolerance with infusion treatments compared to intermittent treatment. When compared to higher analgesic efficacy opioids, hydromorphone infusions induced substantially more tolerance. Finally, the effect of chronic infusion (31.5 mg/kg/day) and 7 day intermittent (22 mg/kg/day) hydromorphone treatment on spinal cord mu-opioid receptor density was determined. Hydromorphone did not produce any change in mu-opioid receptor density following either treatment. These results support suggestions that analgesic efficacy is correlated with tolerance magnitude and regulation of mu-opioid receptors when opioid agonists are continuously administered. Taken together, these studies indicate that analgesic efficacy and treatment protocol are important in determining tolerance and regulation of mu-opioid receptors.
Collapse
Affiliation(s)
- Priyank Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | | | | | | | | | | |
Collapse
|
19
|
Hendriks-Balk MC, Peters SLM, Michel MC, Alewijnse AE. Regulation of G protein-coupled receptor signalling: focus on the cardiovascular system and regulator of G protein signalling proteins. Eur J Pharmacol 2008; 585:278-91. [PMID: 18410914 DOI: 10.1016/j.ejphar.2008.02.088] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 01/18/2008] [Accepted: 02/06/2008] [Indexed: 11/17/2022]
Abstract
G protein-coupled receptors (GPCRs) are involved in many biological processes. Therefore, GPCR function is tightly controlled both at receptor level and at the level of signalling components. Well-known mechanisms by which GPCR function can be regulated comprise desensitization/resensitization processes and GPCR up- and downregulation. GPCR function can also be regulated by several proteins that directly interact with the receptor and thereby modulate receptor activity. An additional mechanism by which receptor signalling is regulated involves an emerging class of proteins, the so-called regulators of G protein signalling (RGS). In this review we will describe some of these control mechanisms in more detail with some specific examples in the cardiovascular system. In addition, we will provide an overview on RGS proteins and the involvement of RGS proteins in cardiovascular function.
Collapse
Affiliation(s)
- Mariëlle C Hendriks-Balk
- Department Pharmacology and Pharmacotherapy, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
20
|
Sirohi S, Kumar P, Yoburn BC. Mu-opioid receptor up-regulation and functional supersensitivity are independent of antagonist efficacy. J Pharmacol Exp Ther 2007; 323:701-7. [PMID: 17698975 DOI: 10.1124/jpet.107.127019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic opioid antagonist treatment up-regulates opioid receptors and produces functional supersensitivity. Although opioid antagonists vary from neutral to inverse, the role of antagonist efficacy in mediating the chronic effects of opioid antagonists is not known. In this study, the effects of two putative inverse agonists (naltrexone, naloxone) and a putative neutral antagonist (6beta-naltrexol) were examined. Initially, peak effect (40 min, naltrexone and naloxone; 70 min, 6beta-naltrexol) and relative potency to antagonize morphine analgesia were determined (relative potencies = 1, 2, and 16, 6beta-naltrexol, naloxone, and naltrexone, respectively). Next, mice were infused for 7 days with naloxone (0.1-10 mg/kg/day), naltrexone (10 or 15 mg s.c. pellet), or 6beta-naltrexol (0.2-20 mg/kg/day), and spinal micro-opioid receptor density was examined, or morphine analgesia dose-response studies were conducted. All antagonists up-regulated mu-opioid receptors (60-122%) and induced supersensitivity (1.8-2.0-fold increase in morphine potency). There were no differences in antagonist potency to produce up-regulation or supersensitivity. These data suggest that opioid antagonist-induced mu-opioid receptor up-regulation and supersensitivity require occupancy of the receptor and that antagonist efficacy is not critical. Finally, the ED(50) to precipitate withdrawal jumping was examined in morphine-dependent mice. Naltrexone, naloxone, and 6beta-naltrexol produced withdrawal jumping, although potencies relative to 6beta-naltrexol were 211, 96, and 1, respectively. Thus, antagonist potency to precipitate opioid withdrawal was related to inverse agonist efficacy. Overall, the estimated relative potency of the opioid antagonists was a function of the outcome measured, and inverse agonist activity was not required for mu-opioid receptor up-regulation and supersensitivity.
Collapse
Affiliation(s)
- Sunil Sirohi
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, Queens, New York 11439, USA
| | | | | |
Collapse
|
21
|
Gago B, Fuxe K, Agnati L, Peñafiel A, De La Calle A, Rivera A. Dopamine D(4) receptor activation decreases the expression of mu-opioid receptors in the rat striatum. J Comp Neurol 2007; 502:358-66. [PMID: 17366605 DOI: 10.1002/cne.21327] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The dopaminergic and opioid peptide systems interact in many nuclei of the brain. In the striatum, dopamine/opioid peptide interactions modulate locomotor and motivated behaviors as well as reward, motivational, and tolerance processes in opiate dependence. Dopamine D(4) receptors (D(4) R) and mu-opioid receptors (MOR) are highly concentrated in the striosomes (islands) of the striatum, suggesting the existence of receptor-receptor interactions between them. In the present work we studied the role of D(4) R in modulating MOR expression in the islands by using immunohistochemistry and image analysis. The activation of D(4) R by the agonist PD168,077 (1 mg/kg) decreased MOR immunoreactivity (IR) in the striosomes 6 hours after drug treatment. MOR IR levels had recovered 12 hours later. Treatment with a D(4) R antagonist (L745,870, 1mg/kg) blocked downregulation of MOR IR, showing that the D(4) R agonist effects observed were specific. Furthermore, treatment with the D(2)/D(3) receptor agonist quinpirol (1 mg/kg) and D(2)/D(3) receptor antagonist raclopride (1 mg/kg) had no effect in MOR IR, suggesting that D(4) R is the only D2-like receptor producing an MOR downregulation in the islands. The decreases of MOR IR in the striosomes suggest that D(4) R activation may reduce MOR signaling. Increasing evidence has demonstrated that the islands in the striatum play a critical role in habit acquisition during drug addiction. D(4) R/MOR interactions could be crucial in such processes.
Collapse
Affiliation(s)
- Belén Gago
- Department of Cell Biology, School of Science, University of Málaga, 29071 Málaga, Spain
| | | | | | | | | | | |
Collapse
|
22
|
Pawar M, Kumar P, Sunkaraneni S, Sirohi S, Walker EA, Yoburn BC. Opioid agonist efficacy predicts the magnitude of tolerance and the regulation of mu-opioid receptors and dynamin-2. Eur J Pharmacol 2007; 563:92-101. [PMID: 17349996 PMCID: PMC1995431 DOI: 10.1016/j.ejphar.2007.01.059] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Revised: 01/16/2007] [Accepted: 01/17/2007] [Indexed: 11/29/2022]
Abstract
It has been proposed that opioid agonist efficacy may play a role in tolerance and the regulation of opioid receptor density. To address this issue, the present studies estimated the in vivo efficacy of three opioid agonists and then examined changes in spinal mu-opioid receptor density following chronic treatment in the mouse. In addition, tolerance and regulation of the trafficking protein dynamin-2 were determined. To evaluate efficacy, the method of irreversible receptor alkylation was employed and the efficacy parameter tau estimated. Mice were injected with the irreversible mu-opioid receptor antagonist clocinnamox (0.32-25.6 mg/kg, i.p), and 24 h later, the analgesic potency of s.c. morphine, oxycodone and etorphine were determined. Clocinnamox dose-dependently antagonized the analgesic effects of morphine, etorphine and oxycodone. The shift to the right of the dose-response curves was greater for morphine and oxycodone compared to etorphine and the highest dose of clocinnamox reduced the maximal effect of morphine and oxycodone, but not etorphine. The order of efficacy calculated from these results was etorphine>morphine>oxycodone. Other mice were infused for 7 days with oxycodone (10-150 mg/kg/day, s.c.) or etorphine (50-250 microg/kg/day, s.c.) and the analgesic potency of s.c. morphine determined. The low efficacy agonist (oxycodone) produced more tolerance than the high efficacy agonist (etorphine) at equi-effective infusion doses. In saturation binding experiments, the low efficacy opioid agonists (morphine, oxycodone) did not regulate the density of spinal mu-opioid receptors, while etorphine produced approximately 40% reduction in mu-opioid receptor density. Furthermore, etorphine increased spinal dynamin-2 abundance, while oxycodone did not produce any significant change in dynamin-2 abundance. Overall, these data indicate that high efficacy agonists produce less tolerance at equi-effective doses. Furthermore, increased efficacy was associated with mu-opioid receptor downregulation and dynamin-2 upregulation. Conversely, lower efficacy agonists produced more tolerance at equi-effective doses, but did not regulate mu-opioid receptor density or dynamin-2 abundance. Taken together, these studies indicate that agonist efficacy plays an important role in tolerance and regulation of receptors and trafficking proteins.
Collapse
MESH Headings
- Alkylation
- Analgesics, Opioid/metabolism
- Analgesics, Opioid/pharmacology
- Animals
- Binding, Competitive
- Cinnamates/pharmacology
- Dose-Response Relationship, Drug
- Down-Regulation/drug effects
- Drug Tolerance
- Dynamin II/metabolism
- Etorphine/metabolism
- Etorphine/pharmacology
- Male
- Mice
- Morphine/metabolism
- Morphine/pharmacology
- Morphine Derivatives/pharmacology
- Narcotic Antagonists/pharmacology
- Oxycodone/metabolism
- Oxycodone/pharmacology
- Pain Measurement
- Pain Threshold/drug effects
- Predictive Value of Tests
- Protein Processing, Post-Translational/drug effects
- Reaction Time/drug effects
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Spinal Cord/drug effects
- Spinal Cord/metabolism
Collapse
Affiliation(s)
- Mohit Pawar
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Priyank Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Soujanya Sunkaraneni
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Sunil Sirohi
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Ellen A. Walker
- Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Byron C. Yoburn
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
- * to whom reprint requests should be sent: Byron C. Yoburn, Ph.D., Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, , 718 990 1623 Phone, 718 990 6036 FAX
| |
Collapse
|
23
|
Johnson EE, Christie MJ, Connor M. The Role of Opioid Receptor Phosphorylation and Trafficking in Adaptations to Persistent Opioid Treatment. Neurosignals 2006; 14:290-302. [PMID: 16772732 DOI: 10.1159/000093044] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Indexed: 11/19/2022] Open
Abstract
Mu-opioid receptor activation underpins clinical analgesia and is the central event in the abuse of narcotics. Continued opioid use produces tolerance to the acute effects of the drug and adaptations that lead to physical and psychological dependence. Continued mu-receptor signaling provides the engine for these adaptations, with most evidence suggesting that chronic agonist treatment produces only limited alterations in primary mu-opioid receptor signaling. Here we examine agonist regulation of mu-opioid receptor function, and whether this is altered by chronic treatment. Receptor phosphorylation is thought to be the key initial event in agonist regulation of the mu-opioid receptor, providing a signal for acute receptor desensitization and also subsequent receptor resensitization. Morphine appears to produce qualitatively and quantitatively different mu-receptor phosphorylation than other agonists, but the consequences of this remain obscure, at least in neurons. There is no evidence that agonist-induced mu-opioid receptor phosphorylation changes in chronically morphine-treated animals, although receptor regulation appears to be altered. Thus, as receptor phosphorylation and resensitization appear to maintain continued signaling through the mu-opioid receptor, these two events are crucial in facilitating adaptations to chronic opioid treatment, and the possibility that agonist-specific phosphorylation can contribute to the development of different adaptations remains open.
Collapse
Affiliation(s)
- Emma E Johnson
- Pain Management Research Institute, Kolling Institute, University of Sydney at Royal North Shore Hospital, St. Leonards, Australia
| | | | | |
Collapse
|
24
|
Bernard V, Décossas M, Liste I, Bloch B. Intraneuronal trafficking of G-protein-coupled receptors in vivo. Trends Neurosci 2006; 29:140-7. [PMID: 16443287 DOI: 10.1016/j.tins.2006.01.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Revised: 11/09/2005] [Accepted: 01/12/2006] [Indexed: 02/01/2023]
Abstract
In vitro studies have widely demonstrated that the abundance and availability of G-protein-coupled receptors (GPCRs) at the cell surface is regulated by the neuronal environment and is the result of complex intraneuronal trafficking. However, this regulation is still poorly understood in vivo. Modulation of receptor availability at the neuronal membrane is a key event in the regulation of neuronal functions (e.g. neurotransmitter release or neuronal excitability in physiological, pathological or therapeutic conditions). We discuss the effects of duration of receptor stimulation (acute versus chronic) on the intraneuronal trafficking of GPCRs in vivo, and we show that this trafficking might differ according to subcellular compartment (soma, dendrites or axon terminals).
Collapse
Affiliation(s)
- Véronique Bernard
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5541, Laboratoire d'Histologie-Embryologie, Université Victor Ségalen-Bordeaux 2, 146 Rue Léo-Saignat, 33076 Bordeaux Cedex, France.
| | | | | | | |
Collapse
|
25
|
Narita M, Suzuki M, Narita M, Niikura K, Nakamura A, Miyatake M, Yajima Y, Suzuki T. mu-Opioid receptor internalization-dependent and -independent mechanisms of the development of tolerance to mu-opioid receptor agonists: Comparison between etorphine and morphine. Neuroscience 2006; 138:609-19. [PMID: 16417975 DOI: 10.1016/j.neuroscience.2005.11.046] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Revised: 11/10/2005] [Accepted: 11/19/2005] [Indexed: 12/30/2022]
Abstract
A growing body of evidences suggests that receptor desensitization is implicated in the development of tolerance to opioids, which is generally regulated by protein kinases and receptor trafficking proteins. In the present study, we demonstrated that repeated s.c. treatment with etorphine, but not morphine, produced a significant increase in protein levels of G protein-coupled receptor kinase 2, dynamin II, beta-arrestin 2 and phosphorylated-conventional protein kinase C in membranes of the mouse spinal cord, suggesting that the etorphine-induced mu-opioid receptor desensitization may result from G protein-coupled receptor kinase 2/dynaminII/beta-arrestin2-dependent phosphorylation of mu-opioid receptors. Unlike etorphine, morphine failed to change the levels of these trafficking proteins. Furthermore, we found that the level of glial fibrillary acidic protein in the mouse spinal cord was clearly increased by chronic in vivo and in vitro treatment with morphine, whereas no such effect was noted by etorphine. In the behavioral study, intraperitoneal pretreatment with the glial-modulating agent propentofylline suppressed the development of tolerance to morphine-induced antinociception. In addition, intrathecal injection of astrocytes and astrocyte-conditioned medium mixture, which were obtained from cultured astrocytes of the newborn mouse spinal cord, aggravated the development of tolerance to morphine. In contrast, these agents failed to affect the development of tolerance induced by etorphine. These findings provide direct evidence for the distinct mechanisms between etorphine and morphine on the development of tolerance to spinal antinociception. These findings raise the possibility that the increased astroglia response produced by chronic morphine could be associated with the lack of mu-opioid receptor internalization.
Collapse
Affiliation(s)
- M Narita
- Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
This paper is the 27th consecutive installment of the annual review of research concerning the endogenous opioid system, now spanning over 30 years of research. It summarizes papers published during 2004 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia; stress and social status; tolerance and dependence; learning and memory; eating and drinking; alcohol and drugs of abuse; sexual activity and hormones, pregnancy, development and endocrinology; mental illness and mood; seizures and neurologic disorders; electrical-related activity and neurophysiology; general activity and locomotion; gastrointestinal, renal and hepatic functions; cardiovascular responses; respiration and thermoregulation; and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, USA.
| | | |
Collapse
|
27
|
Zhang Q, Purohit V, Yoburn BC. Continuous opioid agonist treatment dose-dependently regulates mu-opioid receptors and dynamin-2 in mouse spinal cord. Synapse 2005; 56:123-8. [PMID: 15765525 DOI: 10.1002/syn.20137] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Continuous opioid agonist treatment produces tolerance and in some cases mu opioid receptor (muOR) down-regulation. Previous studies indicate that down-regulation of muOR is more likely with high-efficacy opioid agonists (e.g., etorphine), whereas lower efficacy agonists (e.g., morphine) do not regulate muOR density. It has been suggested that muOR down-regulation may depend upon increases in Dynamin-2 (DYN-2) proteins. Therefore, the present study examined the effect of various infusion doses of etorphine on muOR density, DYN-2 protein, and DYN-2 mRNA abundance in mouse spinal cord. Mice were implanted sc with an osmotic pump that infused etorphine (50-250 microg/kg/day). Controls were implanted with inert placebo pellets. At the end of 7 days, mice were sacrificed, spinal cord removed and processed for radioligand binding, quantitative Western blotting, or RT-PCR assay. Results indicate that etorphine induced dose-dependent regulation of muOR density, DYN-2 proteins, and mRNA abundance in mouse spinal cord. Higher infusion doses significantly down-regulated muOR density, increased DYN-2 protein abundance, and decreased DYN-2 mRNA. Analysis of these results indicated a significant correlation between muOR down-regulation and DYN-2 abundance in mouse spinal cord. Taken together, muOR regulation may depend on changes in DYN-2 abundance induced by high-efficacy opioid agonists in mouse spinal cord.
Collapse
Affiliation(s)
- Qiuyu Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, Queens, New York 11439, USA
| | | | | |
Collapse
|