451
|
|
452
|
Glass MJ, Kruzich PJ, Kreek MJ, Pickel VM. Decreased plasma membrane targeting of NMDA-NR1 receptor subunit in dendrites of medial nucleus tractus solitarius neurons in rats self-administering morphine. Synapse 2004; 53:191-201. [PMID: 15266550 DOI: 10.1002/syn.20049] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Opioid abuse is associated with repeated administration and escalation of dose that can result in profound adaptations in homeostatic processes. Potential cellular mechanisms and neural sites mediating opiate-dependent adaptations may involve NMDA-dependent synaptic plasticity within brain areas participating in behaviors related to consumption of natural reinforcers, as well as affective-autonomic integration, notably the medial nucleus tractus solitarius (mNTS). NMDA-dependent synaptic plasticity may be mediated by changes in the intracellular and surface targeting of NMDA receptors, particularly in postsynaptic sites including spines or small distal dendrites. High-resolution immunogold electron microscopic immunocytochemistry combined with morphometry were used to measure changes in targeting of the NMDA-NR1 (NR1) receptor subunit between intracellular and plasmalemmal sites in dendrites of neurons of the intermediate mNTS of rats self-administering escalating doses of morphine (EMSA). In control and EMSA rats, the density of plasmalemmal and cytosolic gold particles was inversely related to profile size. Collapsed across all NR1-labeled dendrites, rats self-administering morphine had a lower number of plasmalemmal gold particles per unit surface area (7.1 +/- 0.8 vs. 14.4 +/- 1 per 100 microm), but had a higher number of intracellular gold particles per unit cross-sectional area (169 +/- 6.1 vs. 148 +/- 5.1 per 100 microm2) compared to saline self-administering rats. Morphometric analysis showed that the decrease in plasma membrane labeling of NR1 was most robust in small dendritic profiles (<1 microm), where there was a reciprocal increase in the density of intracellular particles. These results indicate that the plasmalemmal distribution of the essential NR1 subunits in distal sites may prominently contribute to NMDA receptor-dependent modulation of neural circuitry regulating homeostatic processes, and targeting of these proteins can be prominently affected by morphine self-administration.
Collapse
Affiliation(s)
- Michael J Glass
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | | | | | |
Collapse
|
453
|
Billet F, Dourmap N, Costentin J. Involvement of corticostriatal glutamatergic terminals in striatal dopamine release elicited by stimulation of delta-opioid receptors. Eur J Neurosci 2004; 20:2629-38. [PMID: 15548206 DOI: 10.1111/j.1460-9568.2004.03723.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have previously shown that striatal dopamine release induced locally by a delta-opioid receptor agonist was totally inhibited by a glutamate N-methyl-D-aspartate receptor antagonist, indicating the involvement of glutamatergic receptors in this effect. The aim of the present study was to specify this mechanism. Firstly, we investigated the effect of [D-Pen2,D-Pen5]-enkephalin (DPDPE) on glutamate release in rats by intrastriatal microdialysis. The infusion of DPDPE (10 microm) enhanced the glutamate content in dialysate by approximately 34%, an effect which did not appear to result from inhibition of glutamate uptake. We then considered the consequences of a unilateral thermocoagulation of the frontal cortex on either glutamate or dopamine release induced by stimulation of delta-opioid receptors 2 days later. This lesion, which decreased the glutamate content in ipsilateral striatum by approximately 30%, totally prevented the increase in dialysate levels of glutamate induced by DPDPE. Moreover, whereas DPDPE (10 microm) was found to increase the striatal dopamine release in intact animals by approximately 59%, this effect was also completely suppressed by the cortical lesion. Finally, we studied the effect of the lesion on the [3H]-DPDPE binding to striatal membranes prepared from the whole striatum. In the ipsilateral striatum a significant decrease in this [3H]-DPDPE binding (by approximately 18%) was found 2 days after the lesion. Our results indicate that the increase in striatal dopamine release induced by DPDPE probably depends on glutamate release from corticostriatal glutamatergic afferents in response to the stimulation of delta-opioid receptors located on terminals of these neurons.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Cerebral Cortex/drug effects
- Cerebral Cortex/injuries
- Cerebral Cortex/physiology
- Chromatography, High Pressure Liquid/methods
- Dicarboxylic Acids/pharmacology
- Dopamine/metabolism
- Dose-Response Relationship, Drug
- Drug Interactions
- Electrocoagulation/methods
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Glutamates/metabolism
- Male
- Microdialysis/methods
- Neostriatum/drug effects
- Neostriatum/metabolism
- Neurotransmitter Uptake Inhibitors/pharmacology
- Pyrrolidines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/metabolism
- Staining and Labeling/methods
- Synaptosomes/metabolism
Collapse
Affiliation(s)
- Fabrice Billet
- Unité de Neuropsychopharmacologie Expérimentale, UMR 6036 CNRS, IFRMP no. 23, Faculté de Médecine et de Pharmacie de Rouen, 22 Boulevard Gambetta, 76183 Rouen Cédex 01, France
| | | | | |
Collapse
|
454
|
Khurdayan VK, Buch S, El-Hage N, Lutz SE, Goebel SM, Singh IN, Knapp PE, Turchan-Cholewo J, Nath A, Hauser KF. Preferential vulnerability of astroglia and glial precursors to combined opioid and HIV-1 Tat exposure in vitro. Eur J Neurosci 2004; 19:3171-82. [PMID: 15217373 PMCID: PMC4305445 DOI: 10.1111/j.0953-816x.2004.03461.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Human immunodeficiency virus (HIV)-1 infection can cause characteristic neural defects such as progressive motor dysfunction, striatal pathology and gliosis. Recent evidence suggests that HIV-induced pathogenesis is exacerbated by heroin abuse and that the synergistic neurotoxicity is a direct effect of heroin on the CNS, an alarming observation considering the high incidence of HIV infection with injection drug abuse. Although HIV infection results in neurodegeneration, neurons themselves are not directly infected. Instead, HIV affects microglia and astroglia, which subsequently contributes to the neurodegenerative changes. Opioid receptors are widely expressed by macroglia and macroglial precursors, and the activation of mu-opioid receptors can modulate programmed cell death, as well as the response of neural cells to cytotoxic insults. For this reason, we questioned whether opioid drugs might modify the vulnerability of macroglia and macroglial precursors to HIV-1 Tat protein. To address this problem, the effects of morphine and/or HIV Tat(1-72) on the viability of macroglia and macroglial precursors were assessed in mixed-glial cultures derived from mouse striatum. Our findings indicate that sustained exposure to morphine and Tat(1-72) viral protein induces the preferential death of glial precursors and some astrocytes. Moreover, the increased cell death is mediated by mu-opioid receptors and accompanied by the activation of caspase-3. Our results imply that opiates can enhance the cytotoxicity of HIV-1 Tat through direct actions on glial precursors and/or astroglia, suggesting novel cellular targets for HIV-opiate interactions.
Collapse
Affiliation(s)
- Valeriya K. Khurdayan
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Shreya Buch
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Nazira El-Hage
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Sarah E. Lutz
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Susan M. Goebel
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Indrapal N. Singh
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Pamela E. Knapp
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | | | - Avindra Nath
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Kurt F. Hauser
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
- Correspondence: Kurt F. Hauser, Ph.D., Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536-0298 USA, , Phone: (859) 323-6477, FAX: (859) 323-5946
| |
Collapse
|
455
|
Canales JJ. Catalase-independent early-gene expression in rat brain following acute ethanol exposure. Brain Res 2004; 1016:96-101. [PMID: 15234257 DOI: 10.1016/j.brainres.2004.04.078] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2004] [Indexed: 11/23/2022]
Abstract
Early-gene expression evoked by acute ethanol treatment was studied in rat brain by quantitative immunocytochemistry, with reference to ethanol metabolism by the enzyme catalase. Colocalization with mu-opioid receptor (MOR) sites was also examined. Ethanol challenges [1, 2.5, and 4 g/kg intraperitoneally (i.p.)] evoked dose-dependent increases in c-Fos expression in several brain regions, but overlap with MOR-rich sites was only partial. Strong inhibition of brain catalase activity (ca. 60%) with 3-amino-1,2,4-triazole (AT, 1 g/kg i.p.) did not alter ethanol-induced c-Fos nor Krox-24 expression in any of the brain regions analyzed. This evidence demonstrates that catalase-mediated metabolism is not a requisite for c-Fos nor Krox-24 induction in rat brain following acute ethanol treatment, and suggests that ethanol is by itself capable of eliciting strong neuronal and circuit-level adaptations in the nervous system.
Collapse
Affiliation(s)
- Juan J Canales
- Neurobiology Unit, Cavanilles Institute, University of Valencia, Poligono de la Coma s/n, Burjassot, 46980 Valencia, Spain.
| |
Collapse
|
456
|
DiPirro JM, Kristal MB. Placenta ingestion by rats enhances δ- and κ-opioid antinociception, but suppresses μ-opioid antinociception. Brain Res 2004; 1014:22-33. [PMID: 15212988 DOI: 10.1016/j.brainres.2004.04.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2004] [Indexed: 12/18/2022]
Abstract
Ingestion of placenta or amniotic fluid produces a dramatic enhancement of centrally mediated opioid antinociception in the rat. The present experiments investigated the role of each opioid receptor type (mu, delta, kappa) in the antinociception-modulating effects of Placental Opioid-Enhancing Factor (POEF-presumably the active substance). Antinociception was measured on a 52 degrees C hotplate in adult, female rats after they ingested placenta or control substance (1.0 g) and after they received an intracerebroventricular injection of a delta-specific ([D-Pen2,D-Pen5]enkephalin (DPDPE); 0, 30, 50, 62, or 70 nmol), mu-specific ([D-Ala2,N-MePhe4,Gly5-ol]enkephalin (DAMGO); 0, 0.21, 0.29, or 0.39 nmol), or kappa-specific (U-62066; spiradoline; 0, 100, 150, or 200 nmol) opioid receptor agonist. The results showed that ingestion of placenta potentiated delta- and kappa-opioid antinociception, but attenuated mu-opioid antinociception. This finding of POEF action as both opioid receptor-specific and complex provides an important basis for understanding the intrinsic pain-suppression mechanisms that are activated during parturition and modified by placentophagia, and important information for the possible use of POEF as an adjunct to opioids in pain management.
Collapse
MESH Headings
- Analgesics/administration & dosage
- Animals
- Behavior, Animal
- Differential Threshold
- Dose-Response Relationship, Drug
- Feeding Behavior
- Female
- Injections, Intraventricular
- Motor Activity
- Pain Threshold/drug effects
- Placenta
- Pregnancy
- Pyrrolidines/administration & dosage
- Rats
- Rats, Long-Evans
- Reaction Time/drug effects
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Sexual Behavior, Animal
Collapse
Affiliation(s)
- Jean M DiPirro
- Behavioral Neuroscience Program, Department of Psychology, University at Buffalo, Buffalo, NY 14260, USA.
| | | |
Collapse
|
457
|
Abstract
The reverse transcriptase-polymerase chain reaction (RT-PCR) was used to clone a cDNA fragment of a putative G-protein-coupled receptor from rat brain total RNA. Nucleotide sequencing of this cDNA fragment showed it to be homologous to that of the mu-opioid receptor splice variant MOR(1C) from mice. We used the cDNA to make an RNA probe for a ribonuclease protection assay (RPA). The results from the RPA showed a protected fragment of the size expected for MOR(1C) mRNA, as well as other RNase-protected fragments that may indicate the existence of other MOR1 transcripts. We then used the RNA probe for in situ hybridization (ISH) experiments. We detected strong autoradiographic labeling over much of the rat telencephalon, diencephalon, mesencephalon, cerebellum, spinal cord, and dorsal root ganglia. These findings suggest that MOR(1C), and possibly other MOR1 splice variants, are important components of the system by which the actions of opioids are transduced.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Brain/metabolism
- Cloning, Molecular/methods
- DNA, Complementary/biosynthesis
- DNA, Complementary/genetics
- Male
- Molecular Sequence Data
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Messenger/isolation & purification
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, mu/biosynthesis
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/isolation & purification
Collapse
Affiliation(s)
- Stephen A Schnell
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55405, USA.
| | | |
Collapse
|
458
|
Morinville A, Cahill CM, Aibak H, Rymar VV, Pradhan A, Hoffert C, Mennicken F, Stroh T, Sadikot AF, O'Donnell D, Clarke PBS, Collier B, Henry JL, Vincent JP, Beaudet A. Morphine-induced changes in delta opioid receptor trafficking are linked to somatosensory processing in the rat spinal cord. J Neurosci 2004; 24:5549-59. [PMID: 15201327 PMCID: PMC6729333 DOI: 10.1523/jneurosci.2719-03.2004] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2003] [Revised: 04/22/2004] [Accepted: 04/26/2004] [Indexed: 01/13/2023] Open
Abstract
An in vivo fluorescent deltorphin (Fluo-DLT) internalization assay was used to assess the distribution and regulation of pharmacologically available delta opioid receptors (deltaORs) in the rat lumbar (L4-5) spinal cord. Under basal conditions, intrathecal injection of Fluo-DLT resulted in the labeling of numerous deltaOR-internalizing neurons throughout dorsal and ventral horns. The distribution and number of Fluo-DLT-labeled perikaryal profiles were consistent with that of deltaOR-expressing neurons, as revealed by in situ hybridization and immunohistochemistry, suggesting that a large proportion of these cells was responsive to intrathecally administered deltaOR agonists. Pretreatment of rats with morphine for 48 hr resulted in a selective increase in Fluo-DLT-labeled perikaryal profiles within the dorsal horn. These changes were not accompanied by corresponding augmentations in either deltaOR mRNA or (125)I-deltorphin-II binding levels, suggesting that they were attributable to higher densities of cell surface deltaOR available for internalization rather than to enhanced production of the receptor. Unilateral dorsal rhizotomy also resulted in increased Fluo-DLT internalization in the ipsilateral dorsal horn when compared with the side contralateral to the deafferentation or to non-deafferented controls, suggesting that deltaOR trafficking in dorsal horn neurons may be regulated by afferent inputs. Furthermore, morphine treatment no longer increased Fluo-DLT internalization on either side of the spinal cord after unilateral dorsal rhizotomy, indicating that microOR-induced changes in the cell surface availability of deltaOR depend on the integrity of primary afferent inputs. Together, these results suggest that regulation of deltaOR responsiveness through microOR activation in this region is linked to somatosensory information processing.
Collapse
MESH Headings
- Animals
- Fluorescent Dyes/chemistry
- Fluorescent Dyes/pharmacology
- Guanosine 5'-O-(3-Thiotriphosphate)/metabolism
- In Situ Hybridization
- Lumbosacral Region
- Male
- Microscopy, Fluorescence
- Morphine/pharmacology
- Narcotics/pharmacology
- Oligopeptides/chemistry
- Oligopeptides/metabolism
- Oligopeptides/pharmacology
- Posterior Horn Cells/metabolism
- Protein Transport
- Radioligand Assay
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/biosynthesis
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Rhizotomy
- Spinal Cord/anatomy & histology
- Spinal Cord/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Anne Morinville
- Montreal Neurological Institute, McGill University, Montreal, Québec, Canada H3A 2B4
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
459
|
Borras MC, Becerra L, Ploghaus A, Gostic JM, DaSilva A, Gonzalez RG, Borsook D. FMRI Measurement of CNS Responses to Naloxone Infusion and Subsequent Mild Noxious Thermal Stimuli in Healthy Volunteers. J Neurophysiol 2004; 91:2723-33. [PMID: 15136603 DOI: 10.1152/jn.00249.2003] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aims of this study were to assess the effects of a μ-opioid antagonist, naloxone, on endogenous opioid systems and to evaluate the effect of naloxone on the CNS response to mild noxious heat. Doubled-blinded experiments were performed in a cross-over design in 10 healthy male volunteers. Functional magnetic resonance imaging (fMRI) data were collected before and during the infusion and also during thermal stimuli. Increased signal was observed in a number of cortical and subcortical brain regions for naloxone versus saline infusion. Cortical activation was induced in regions including cingulate, prefrontal cortex, and insula. Subcortical regions showing increased signal change included hippocampus and entorhinal cortex. A 46°C stimulus delivered to the back of the hand induced an overall increase in activation in a number of regions in the naloxone group that were not seen in the saline group (e.g., insula, orbitofrontal cortex, thalamus, and hippocampus). These results show that naloxone, even in the absence of psychophysical effects, produces activation in several brain regions that are known to have high levels of μ-opioid receptors and may be involved in endogenous analgesia. Our study is an example of how fMRI can measure subtle changes in brain activation induced by pharmacological agents without cognitive effects.
Collapse
Affiliation(s)
- M C Borras
- Center for Functional Pain Neuroimaging and Therapy Research, Athinoula A. Martinos Center for Biomedical Imaging, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | | | | | | | | | | | | |
Collapse
|
460
|
Abstract
Kappa (kappa)-opioid receptor agonists are particularly effective analgesics in experimental models of visceral pain. Their analgesic effects are mediated in the periphery. The molecular targets involved include peripherally located kappa-receptors and possibly, at least for some nonpeptidic kappa-agonists, additional nonopioid molecular targets such as sodium channels located on primary sensory afferents. Overall, these properties are expected to be of therapeutic interest in various visceral pain conditions, including abdominal surgery associated with postoperative pain and ileus, pancreatitis pain, dysmenorrhea, labor pain and functional disorders such as irritable bowel syndrome or dyspepsia. The first kappa-agonists to be developed were brain-penetrating organic small molecules. Their development was eventually discontinued due to central side effects such as sedation and dysphoria attributed to kappa-receptors located behind the blood-brain barrier. New drug discovery programs are now geared towards the design of peripherally-selective kappa-agonists. So far, most of the organic molecule-based peripheral kappa-agonists have achieved limited peripheral selectivity and a practically insufficient therapeutic window to justify full development. These compounds have been used in a small number of clinical pilot studies involving visceral pain. Although encouraging, the clinical data available so far with this class of compounds are too limited and fragmented to fully validate the therapeutic utility of kappa-agonists in visceral pain. Additional clinical studies with safer kappa-agonists (i.e. with higher peripheral selectivity) are still required. The most suitable tools to address this question in the future appear to be the newly discovered class of tetrapeptide-based kappa-agonists, which have shown unprecedented levels of peripheral selectivity.
Collapse
Affiliation(s)
- Pierre J-M Rivière
- Ferring Research Institute, Building 2, Room 442, 3550 General Atomics Court, San Diego, CA 92121, U.S.A.
| |
Collapse
|
461
|
Bai WZ, Ishida M, Arimatsu Y. Chemically defined feedback connections from infragranular layers of sensory association cortices in the rat. Neuroscience 2004; 123:257-67. [PMID: 14667460 DOI: 10.1016/j.neuroscience.2003.08.056] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The primary visual (V1), auditory (AI), and somatosensory (SI) cortices are reciprocally connected with their respective sensory association cortices. In the rat, we have previously demonstrated that some of the connections arising from the secondary somatosensory (SII) and parietal insular (PA) cortices and terminating in the SI, are characterized by the expression of latexin, a candidate protein of carboxypeptidase A inhibitor. Here, by using retrograde tracing and latexin-immunohistochemistry, we show that latexin-expressing neurons in other association cortices of different sensory modalities also contribute to the feedback projections to the corresponding primary sensory cortices. These are the lateral part of the secondary visual cortex (V2L), temporal association cortex, and the dorsal and ventral (AIIv) parts of the secondary auditory belt cortex. Within sublayer VIa of the V2L, AIIv and SII, the majority of the V1-, AI- and SI-projecting neurons respectively, are latexin-immunopositive. In contrast to feedback connections, far fewer latexin-expressing neurons participate in callosal or intrahemispheric feedforward connections. The latexin-expressing neurons constitute a virtually completely different population from corticothalamic neurons within the infragranular layers. Given that latexin might participate in the modulation of neuronal activity by controlling the protease activity, latexin-expressing feedback pathways would play a unique role in the modulation of sensory perception.
Collapse
Affiliation(s)
- W-Z Bai
- Mitsubishi Kagaku Institute of Life Sciences, 11 Minamiooya, Machida-shi, 194-8511, Tokyo, Japan
| | | | | |
Collapse
|
462
|
Zhang A, Xiong W, Bidlack JM, Hilbert JE, Knapp BI, Wentland MP, Neumeyer JL. 10-Ketomorphinan and 3-substituted-3-desoxymorphinan analogues as mixed kappa and micro opioid ligands: synthesis and biological evaluation of their binding affinity at opioid receptors. J Med Chem 2004; 47:165-74. [PMID: 14695830 DOI: 10.1021/jm0304156] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A series of 10-ketomorphinan analogues were synthesized, and their binding affinity at all three opioid receptors was investigated. In most cases, high affinity at micro and kappa receptors, and lower affinity at delta receptor was observed, resulting in good selectivity for micro and kappa receptors. A wide range of substituents can be accommodated on the nitrogen position. The N-(S)-tetrahydrofurfuryl analogue 11 displayed the highest affinity at all three receptors. The N-cyclobutylmethyl analogue 13 gave both high affinity and selectivity at kappa receptor, and N-2-phenylethyl analogue 18 exhibited good affinity and selectivity at micro receptor. Further modifications of the 3-substituent indicated that one H-bond donor was an essential requirement for good affinity at micro and kappa receptors. Similar modifications were investigated at the 3-OH group of morphinans: levorphanol (2a), cyclorphan (2b), and MCL-101 (2c) lacking the 10-keto group. The 3-amino bioisosteric analogues (40 and 41) displayed reasonably good affinity at micro and kappa receptors. The 3-carboxamido replacement (compounds 46-48) in the morphinan subseries resulted in similar affinities comparable to their corresponding 3-OH congeners. The high affinity of these carboxamido analogues, along with their greater lipophilicity and metabolic stability, make them promising candidates for further pharmacological investigation.
Collapse
Affiliation(s)
- Ao Zhang
- Alcohol and Drug Abuse Research Center, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, Massachusetts 02478, USA
| | | | | | | | | | | | | |
Collapse
|
463
|
Mrkusich EM, Kivell BM, Miller JH, Day DJ. Abundant expression of mu and delta opioid receptor mRNA and protein in the cerebellum of the fetal, neonatal, and adult rat. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2004; 148:213-22. [PMID: 14766199 DOI: 10.1016/j.devbrainres.2003.10.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/24/2003] [Indexed: 10/26/2022]
Abstract
Opioid receptor proteins and mRNAs have been localized to a variety of regions within the rat brain. It is generally accepted that within the lobes of the rat cerebellum, only delta opioid receptor (DOR) is expressed. This is in contrast to that observed in humans and rabbits which express both mu opioid receptor (MOR) and DOR. In this study, we report detection of MOR as well as DOR protein by immunohistochemical localization, and mRNA by fluorescent in situ hybridization (FISH) within Purkinje cells (PK) and the granular layer of neonatal (P6) and adult rat cerebellum. Expression of MOR mRNA was also detected within cells of the molecular layer, but at lower levels than those seen within the PK cells. Abundant expression of MOR and DOR mRNA was detected in the external germinal layer of the immature cerebellum of the fetal (E16) rat, supporting a role for MOR and DOR in regulating neurogenesis of the cerebellum. In addition, using exon-specific cRNA probes, exons 1 and 4, which are both found in the MOR-1 splice variant mRNA, were detected in PK cells in the cerebellum and also within deep cerebellar nuclei in the adult.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cerebellum/cytology
- Cerebellum/embryology
- Cerebellum/growth & development
- Cerebellum/metabolism
- Embryo, Mammalian
- Female
- Gene Expression Regulation, Developmental/physiology
- Immunohistochemistry/methods
- In Situ Hybridization, Fluorescence/methods
- Male
- Pregnancy
- Rats
- Rats, Wistar
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Eli M Mrkusich
- School of Biological Sciences, Victoria University of Wellington, Kelburn parade, P.O. Box 600, Wellington, New Zealand
| | | | | | | |
Collapse
|
464
|
Kivell BM, Day DJ, McDonald FJ, Miller JH. Developmental expression of mu and delta opioid receptors in the rat brainstem: evidence for a postnatal switch in mu isoform expression. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2004; 148:185-96. [PMID: 14766196 DOI: 10.1016/j.devbrainres.2003.12.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/16/2003] [Indexed: 11/22/2022]
Abstract
Opioid receptors are expressed in the brain during fetal and postnatal development, and the expression patterns vary with developmental age. To investigate the role of opioids in brain development, immunoblotting and immunohistochemical techniques were used to determine mu (MOR) and delta (DOR) opioid receptor expression levels and regional distributions in fetal, early postnatal and adult rat brainstem. Two immunoreactive bands were seen on Western blots of brainstem lysates for both MOR (50 and 70 kDa) and DOR (30 and 60 kDa). The expression levels of the isoforms changed dramatically between 6 and 15 days after birth. Total MOR protein was expressed at low levels in fetal and early postnatal animals with the 50-kDa band predominating. MOR expression then increased in the older animals and the 70-kDa isoform became dominant. Total DOR protein showed the opposite pattern, being high in the fetal and neonatal brainstem and low in the juvenile and adult. A postnatal switch in isoform expression for DOR was not evident in our study. In general, regional brainstem distributions in developing and adult animals were comparable to those reported in the literature, and both receptors were localized in the same areas where opioid receptor expression was high. It was concluded that MOR and DOR are developmentally regulated in the brainstem of the rat, that the isoform ratio switches postnatally from a fetal-neonatal pattern to a juvenile-adult pattern and that both receptors are generally expressed in the same brainstem regions from E16 to adult.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Blotting, Western/methods
- Brain Stem/embryology
- Brain Stem/growth & development
- Brain Stem/metabolism
- Cell Line, Tumor
- Embryo, Mammalian
- Female
- Gene Expression Regulation, Developmental
- Immunohistochemistry/methods
- Male
- Neuroblastoma
- Pregnancy
- Rats
- Rats, Wistar
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Bronwyn M Kivell
- School of Biological Sciences, Victoria University of Wellington, P.O. Box 600, Wellington, New Zealand
| | | | | | | |
Collapse
|
465
|
Zwick M, Molliver DC, Lindsay J, Fairbanks CA, Sengoku T, Albers KM, Davis BM. Transgenic mice possessing increased numbers of nociceptors do not exhibit increased behavioral sensitivity in models of inflammatory and neuropathic pain. Pain 2004; 106:491-500. [PMID: 14659533 DOI: 10.1016/j.pain.2003.09.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
At least two classes of neciceptors can be distinguished based on their growth factor requirements: glial cell-line derived neurotrophic factor (GDNF)- and nerve growth factor (NGF)-dependent primary afferent neurons. Based on numerous anatomical and biochemical differences, GDNF- and NGF-dependent neurons have been proposed to be involved in the development of different types of persistent pain. To examine this hypothesis we used two lines of transgenic mice that contained a supernormal number of either NGF- or GDNF-dependent neurons (referred to as NGF-OE and GDNF-OE mice, respectively). These mice were tested in a model of inflammatory pain (induced by injection of complete Freund's adjuvant) and neuropathic pain (using a spinal nerve ligation protocol). Contrary to expectations, neither line of transgenic mice became more hyperalgesic following induction of persistent pain. In fact, NGF-OE mice recovered more rapidly and became hypoalgesic despite extensive paw swelling in the inflammatory pain model. In the neuropathic pain model, only wildtype mice became hyperalgesic. Real-time PCR analysis showed that the NGF-OE and GDNF-OE mice exhibited changes in neuronal-specific mRNAs in the dorsal root ganglia but not the spinal cord dorsal horn. These results indicate that increasing the number of nociceptors results in potent compensatory mechanisms that may begin with changes in the sensory neurons themselves.
Collapse
Affiliation(s)
- Melissa Zwick
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536, USA Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Scaife Hall, Room S-843, 3550 Terrace Street, Pittsburgh, PA 15261, USA Department of Pharmaceutics, Pharmacology, and Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | |
Collapse
|
466
|
Yang TT, Hung CF, Lee YJ, Su MJ, Wang SJ. Morphine inhibits glutamate exocytosis from rat cerebral cortex nerve terminals (synaptosomes) by reducing Ca2+ influx. Synapse 2004; 51:83-90. [PMID: 14618675 DOI: 10.1002/syn.10290] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Morphine, a mu-opioid agonist, suppressed the Ca(2+)-dependent release of glutamate that was evoked by exposing cerebrocortical synaptosomes to the potassium channel blocker 4-aminopyridine. The presynaptic inhibition produced by morphine was concentration-dependent and blocked by the nonselective opioid receptor antagonist naloxone. As determined by examining the mechanism of mu-opioid receptor-mediated inhibition of glutamate release, morphine caused a significant reduction in 4-aminopyridine-evoked increase in the cytoplasmic free Ca(2+) concentration ([Ca(2+)](c)), but failed to alter both 4-aminopyridine-evoked depolarization of the synaptosomal plasma membrane potential and Ca(2+) ionophore (ionomycin)-induced glutamate release. In addition, morphine was not capable of producing further inhibition on 4AP-evoked glutamate release in synaptosomes pretreated with the cannabinoid CB(1) receptor agonist WIN 55212-2, which has been shown to depress glutamate release through a suppression of presynaptic voltage-dependent Ca(2+) channel activity. These data suggest that morphine exerts its inhibitory effect presynaptically, likely through the reduction of Ca(2+) influx into nerve terminals, and thereby inhibits the release of glutamate in the cerebral cortex. This may therefore indicate that mu-opioid receptor agonists have neuroprotective properties, especially in the excessive glutamate release that occurs under certain pathological conditions.
Collapse
Affiliation(s)
- Tsung-Tsair Yang
- Department of Mental Health, Cardinal Tien Hospital, Taipei, Taiwan 231
| | | | | | | | | |
Collapse
|
467
|
Sweet DC, Levine AS, Kotz CM. Functional opioid pathways are necessary for hypocretin-1 (orexin-A)-induced feeding. Peptides 2004; 25:307-14. [PMID: 15063013 DOI: 10.1016/j.peptides.2003.12.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2003] [Accepted: 12/22/2003] [Indexed: 11/18/2022]
Abstract
We investigated the interaction of the orexigenic neuropeptide, hypocretin-1 (Hcrt-1, also known as orexin-A), with endogenous opioids (also orexigenic neuropeptides). Rats were injected with naltrexone (NTX, nonspecific opioid antagonist) i.p., i.c.v., in the lateral hypothalamus (LH), and in the accumbens shell (AcbSh), and naloxone methiodide (nonspecific opioid antagonist unable to cross the blood brain barrier) was injected i.p. Rats were then injected with Hcrt-1 in the LH. Food intake was measured for up to 4h thereafter. Rats were also pretreated with NTX in the LH, with Hcrt-1 injected in the AcbSh. NTX suppressed Hcrt-1-induced feeding only when injected i.p., i.c.v., and in the AcbSh. These studies reveal the necessity for functional central opioidergic pathways involving the AcbSh, but not the LH in Hcrt-1-induced feeding.
Collapse
|
468
|
Coolen LM, Fitzgerald ME, Yu L, Lehman MN. Activation of μ opioid receptors in the medial preoptic area following copulation in male rats. Neuroscience 2004; 124:11-21. [PMID: 14960335 DOI: 10.1016/j.neuroscience.2003.10.045] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2003] [Indexed: 10/26/2022]
Abstract
The current study tested the hypothesis that sexual behavior is a biological stimulus for release of endogenous opioid peptides. In particular, activation of mu opioid receptors (MOR) in the medial preoptic area (MPOA), a key area for regulation of male sexual behavior, was studied in male rats. MOR endocytosis or internalization was used as a marker for ligand-induced receptor activation, utilizing confocal, electron, and bright microscopic analysis. Indeed, mating including one ejaculation induced receptor activation in the MPOA, demonstrated by increased immunoreactivity for MOR, increased numbers of endosome-like particles immunoreactive for MOR inside the cytoplasm of neurons, and increased percentage of neurons with three or more endosome-like particles inside the cytosol. Moreover, it was demonstrated that MOR activation occurred within 30 min following mating and was still evident after 6 h. Mating-induced internalization was prevented by treatment with the opioid receptor antagonist naloxone before mating, suggesting that mating-induced receptor activation is a result of action of endogenous MOR ligands. i.c.v. injections of MOR ligand [D-Ala(2), N-Me-Phe(4), Gly(5)-ol]-enkephalin resulted in internalization of the MOR in a similar manner observed following mating. Finally, mating induced Fos expression in MOR containing neurons in the MPOA. However, naloxone pretreatment did not prevent Fos activation of MOR neurons, suggesting that Fos induction was not the result of MOR activation. In summary, these results provide further evidence that endogenous opioid peptides are released in the MPOA during male sexual behavior.
Collapse
Affiliation(s)
- L M Coolen
- Department of Cell Biology, Neurobiology and Anatomy, University of Cincinnati College of Medicine, Vontz Center for Molecular Studies, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA.
| | | | | | | |
Collapse
|
469
|
Harlan RE, Kailas SR, Tagoe CEF, Garcia MM. Morphine actions in the rat forebrain: role of protein kinase C. Brain Res Bull 2004; 62:285-95. [PMID: 14709343 DOI: 10.1016/j.brainresbull.2003.09.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Acute administration of morphine induces expression of the immediate-early gene (IEG) c-Fos in dorsomedial striatum, portions of cerebral cortex, and in several midline-intralaminar thalamic nuclei, partly via a trans-synaptic mechanism that involves activation of glutamate receptors. Because activation of protein kinase C (PKC) may occur following the activation of glutamate receptors, we determined whether pharmacological inhibition of PKC would attenuate morphine-induced c-Fos expression, and whether acute administration of morphine would induce translocation of PKC. The selective PKC antagonist NPC 15437 given 30 min prior to morphine significantly decreased morphine-induced c-Fos expression in striatum and cingulate cortex, but not in centrolateral thalamus. In another experiment, rats were given an acute dose of morphine, and immunocytochemical analysis was performed for the betaI and betaII isoforms of PKC. Morphine induced a rapid and transient translocation of PKC betaII, but not betaI, from perinuclear spots to plasma membrane in numerous cortical and striatal neurons. Prior administration of naloxone blocked this response. Ultrastructural studies confirmed translocation from Golgi apparatus to plasma membrane 15 min after morphine injection. Double immunocytochemistry at the light microscopic level demonstrated co-localization of translocated PKC betaII and c-Fos in some cortical neurons 90 min after morphine injection. These results support a role for PKC, especially PKC betaII, in the rapid effects of morphine on the brain.
Collapse
Affiliation(s)
- Richard E Harlan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, LA 70112, USA.
| | | | | | | |
Collapse
|
470
|
Ossipov MH, Lai J, King T, Vanderah TW, Malan TP, Hruby VJ, Porreca F. Antinociceptive and nociceptive actions of opioids. ACTA ACUST UNITED AC 2004; 61:126-48. [PMID: 15362157 DOI: 10.1002/neu.20091] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although the opioids are the principal treatment options for moderate to severe pain, their use is also associated with the development of tolerance, defined as the progressive need for higher doses to achieve a constant analgesic effect. The mechanisms which underlie this phenomenon remain unclear. Recent studies revealed that cholecystokinin (CCK) is upregulated in the rostral ventromedial medulla (RVM) during persistent opioid exposure. CCK is both antiopioid and pronociceptive, and activates descending pain facilitation mechanisms from the RVM enhancing nociceptive transmission at the spinal cord and promoting hyperalgesia. The neuroplastic changes elicited by opioid exposure reflect adaptive changes to promote increased pain transmission and consequent diminished antinociception (i.e., tolerance).
Collapse
Affiliation(s)
- Michael H Ossipov
- Departments of Pharmacology, Anesthesiology and Chemistry, University of Arizona, Tucson, Arizona 85724, USA
| | | | | | | | | | | | | |
Collapse
|
471
|
Harris JA, Chang PC, Drake CT. Kappa opioid receptors in rat spinal cord: sex-linked distribution differences. Neuroscience 2004; 124:879-90. [PMID: 15026128 DOI: 10.1016/j.neuroscience.2003.12.042] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2003] [Indexed: 10/26/2022]
Abstract
Activation of kappa opioid receptors (KORs) in the spinal cord can diminish nociception. Humans and rodents show sex differences in the analgesia produced by KOR agonists, and female rats show fluctuations in KOR density and sensitivity across the estrous cycle. However, it is unclear whether there are sex differences in the amount and/or distribution of spinal KORs. In the present study, immunocytochemically labeled KORs were examined in laminae I and II of the lumbosacral spinal dorsal horn of male and normally cycling female Sprague-Dawley rats. The basic pattern of KOR labeling was determined in both sexes using qualitative electron microscopy (EM), and sex-linked differences in the density and subcellular distribution of KOR immunoreactivity were determined with quantitative EM and light microscopy. KOR labeling was visualized with immunoperoxidase for optimally sensitive detection, or with immunogold for precise subcellular localization. By EM, the general pattern of KOR immunoreactivity was similar in males and females. KOR immunoreactivity was common in dendrites, axons, and axon terminals, and was in a few glia and neuronal somata. Most KOR-immunoreactive (-ir) axons were fine-diameter and unmyelinated. Most KOR-ir terminals were small or medium-sized, and a minority formed asymmetric or symmetric synapses with unlabeled dendrites. KOR immunoreactivity was associated both with the plasma membrane and with cytoplasmic organelles, notably including dense core vesicles in terminals. Light microscopic densitometry revealed that KOR immunoreactivity was significantly denser in estrus and proestrus females than in males. By EM, the distribution of KOR-immunogold labeling within axon terminals differed, with a greater proportion of cytoplasmic KOR labeling in estrus females compared with males. In contrast, the abundance and types of KOR-immunoperoxidase-labeled profiles did not show sex-linked differences. We conclude that in both sexes, KORs are positioned to influence both pre- and postsynaptic neurotransmission and are present in morphologically heterogeneous neuron populations. These findings are consistent with complex consequences of KOR activation in the spinal cord. In addition, the presence of increased KOR density and proportionally elevated intracellular KORs in proestrus/estrus females suggests a basis for sex-linked differences in KOR-mediated antinociception.
Collapse
Affiliation(s)
- J A Harris
- Division of Neurobiology, Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 411 East 69th Street, New York, NY 10021, USA
| | | | | |
Collapse
|
472
|
Stumm RK, Zhou C, Schulz S, Höllt V. Neuronal types expressing μ- and δ-opioid receptor mRNA in the rat hippocampal formation. J Comp Neurol 2003; 469:107-18. [PMID: 14689476 DOI: 10.1002/cne.10997] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Opioids are thought to control the excitability of hippocampal principal neurons indirectly by inhibiting GABAergic interneurons. However, direct inhibition of hippocampal principal neurons by opioids has also been reported. To understand better the neuromodulatory role of opioids in rat hippocampal circuits, we analyzed types of micro- and delta-opioid receptor (MOR, DOR)-expressing hippocampal neurons. Most MOR-immunoreactive neurons in the granular and pyramidal cell layers exhibited multipolar morphologies characteristic of GABAergic neurons. Virtually all neurons in the hippocampal formation expressing high MOR mRNA levels cocontained the mRNA for glutamic acid decarboxylase (GAD). Most parvalbumin-, several calretinin-, and several pre-proenkephalin-containing neurons expressed the MOR gene in the hippocampal formation. Expression of high DOR mRNA levels was restricted to GAD-positive neurons in the principal cell layers, oriens layer and hilus. More than 90% of the parvalbumin-positive neurons in the hippocampal formation strongly expressed the DOR gene. Granule cells expressing vesicular glutamate transporter 1 (VGLUT1) mRNA contained very low MOR and DOR transcript levels. In VGLUT1-positive pyramidal cells, weak DOR but no MOR gene expression was detected. Whereas most somatostatinergic hilar neurons were negative for MOR and DOR mRNA, somatostatinergic oriens layer neurons frequently expressed these receptors. Taken together, weak expression of MOR and DOR genes in hippocampal principal cells is in concordance with direct opioid-mediated inhibition of principal cells. However, strong expression of the MOR and DOR genes in the hippocampus is restricted to gamma-aminobutyric acid (GABA)ergic neurons, with DORs being selectively expressed in the parvalbumin- and somatostatin-containing subpopulations. Activation of MOR and/or DOR in parvalbumin- and somatostatin-containing neurons, which provide GABAergic inhibition to the perisomatic and distal dendritic regions of principal cells, respectively, is likely to facilitate principal cell excitation.
Collapse
MESH Headings
- Animals
- Gene Expression Regulation/physiology
- Hippocampus/metabolism
- Male
- Neurons/classification
- Neurons/metabolism
- Neurons/physiology
- RNA, Messenger/biosynthesis
- Rats
- Rats, Wistar
- Receptors, Opioid, delta/biosynthesis
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, mu/biosynthesis
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- Ralf K Stumm
- Department of Pharmacology and Toxicology, Otto-von-Guericke University, Leipziger Strasse 44, 39120 Magdeburg, Germany.
| | | | | | | |
Collapse
|
473
|
Scott CJ, Mariani M, Clarke IJ, Tilbrook AJ. Effect of testosterone and season on proenkephalin messenger RNA expression in the preoptic area of the hypothalamus in the ram. Biol Reprod 2003; 69:2015-21. [PMID: 12930728 DOI: 10.1095/biolreprod.103.016956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Enkephalin appears to exert an inhibitory action on LH secretion, but whether testosterone regulates enkephalin gene expression is unknown. This study tested the hypothesis that testosterone and/or season modulate preproenkephalin mRNA expression in specific areas of the hypothalamus. Romney Marsh rams were castrated (wethers) either during the breeding season or nonbreeding season and received intramuscular injections of either oil or testosterone propionate (five/group). Blood samples were taken for the assay of plasma LH and testosterone. Preproenkephalin mRNA expression was quantified in hypothalamic sections by in situ hybridization. Mean plasma LH concentrations were reduced and the interpulse interval for LH pulses was greater in testosterone propionate-treated wethers compared with oil-treated wethers, with no change in LH pulse amplitude. Testosterone propionate treatment reduced proenkephalin expression in the diagonal band of Broca, the caudal preoptic area, and the bed nucleus of the stria terminalis. Seasonal differences in proenkephalin expression were observed in the bed nucleus of the stria terminalis, lateral septum, periventricular nucleus, and paraventricular nucleus. No differences were observed between treatments in seven other regions examined. We conclude that testosterone and season regulate proenkephalin mRNA levels in the preoptic area/hypothalamus in the ram in a region-specific manner.
Collapse
|
474
|
Mathon DS, Kamal A, Smidt MP, Ramakers GMJ. Modulation of cellular activity and synaptic transmission in the ventral tegmental area. Eur J Pharmacol 2003; 480:97-115. [PMID: 14623354 DOI: 10.1016/j.ejphar.2003.08.097] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The mesolimbic dopamine system, of which the cell bodies are located in the ventral tegmental area, has been implicated in the physiology of reward and the related pathophysiology of drug abuse. This area has been a site of significant interest to study the effects of drugs of abuse and neurotransmitter systems implicated in the rewarding effects of these compounds. One important aspect of synaptic transmission is the ability of synapses to strengthen or weaken their connection as a consequence of synaptic activity. Recently, it has become apparent that this phenomenon is also present in the ventral tegmental area and that this may bear important functional consequences for the ways in which drugs of abuse assert their effect. Here, we will review the effects of neurotransmitter systems and drugs of abuse on cellular activity and synaptic transmission in the ventral tegmental area.
Collapse
Affiliation(s)
- Daniel S Mathon
- Department of Pharmacology and Anatomy, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
475
|
Wilson BC, Ingram CD. Convergent effects of oxytocin and a δ-opioid agonist in the bed nuclei of the stria terminalis of the peripartum rat. Brain Res 2003; 991:267-70. [PMID: 14575903 DOI: 10.1016/j.brainres.2003.08.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In the bed nuclei of the stria terminalis (BST), opioids may suppress the facilitatory effect of oxytocin on its own release pre-partum. In vitro electrophysiological recording showed that in virgin, late-pregnant, and lactating rats, a delta-opioid agonist inhibited a high proportion of BST neurons, many of which were also oxytocin responsive. Response magnitude did not differ significantly between groups, suggesting that the postulated pre-partum increase in opioid tone does not involve postsynaptic changes in neuronal sensitivity.
Collapse
Affiliation(s)
- B C Wilson
- Department of Biology, Acadia University, B4P 2R6, Wolfville, Nova Scotia, Canada.
| | | |
Collapse
|
476
|
Lonergan T, Goodchild AK, Christie MJ, Pilowsky PM. Presynaptic Δ opioid receptors differentially modulate rhythm and pattern generation in the ventral respiratory group of the rat. Neuroscience 2003; 121:959-73. [PMID: 14580946 DOI: 10.1016/s0306-4522(03)00591-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The specific role of the Delta opioid receptor (DOR), in opioid-induced respiratory depression in the ventral respiratory group (VRG) is largely unknown. Here, we sought to determine (1) the relationship between DOR-immunoreactive (ir) boutons, bulbospinal and functionally identified respiratory neurons in the VRG and (2) the effects of microinjection of the selective DOR agonist, D-Pen 2,5-enkephalin (DPDPE), into different subdivisions of the VRG, on phrenic nerve discharge and mean arterial pressure. Following injections of retrograde tracer into the spinal cord or intracellular labelling of respiratory neurons, in Sprague-Dawley rats, brainstem sections were processed for retrograde or intracellular labelling and DOR-ir. Bulbospinal neurons were apposed by DOR-ir boutons regardless of whether they projected to single (cervical or thoracic ventral horn) or multiple (cervical and thoracic ventral horn) targets in the spinal cord. In the VRG, a total of 24 +/- 5% (67 +/- 13/223 +/- 49) of neurons projecting to the cervical ventral horn, and 37 +/- 3% (96 +/- 22/255 +/- 37) of neurons projecting to the thoracic ventral horn, received close appositions from DOR-ir boutons. Furthermore, DOR-ir boutons closely apposed six of seven intracellularly labelled neurons, whilst the remaining neuron itself possessed boutons that were DOR-ir. DPDPE was microinjected (10 mM, 60 nl, unilateral) into regions of respiratory field activity in the VRG of anaesthetised, vagotomised rats, and the effects on phrenic nerve discharge and mean arterial pressure were recorded. DPDPE depressed phrenic nerve amplitude, with little effect on phrenic nerve frequency in the Bötzinger complex, pre-Bötzinger complex and rVRG, the greatest effects occurring in the Bötzinger complex. The results indicate that the DOR is located on afferent inputs to respiratory neurons in the VRG. Activation of the DOR in the VRG is likely to inhibit the release of neurotransmitters from afferent inputs that modulate the pattern of activity of VRG neurons.
Collapse
Affiliation(s)
- T Lonergan
- Department of Pharmacology, University of Sydney, Camperdown, New South Wales, 2006, Australia
| | | | | | | |
Collapse
|
477
|
Kumazawa Y, Nishimura Y, Akamine T, Lin M, Asahara T, Shibuya H, Yamamoto T. Modulation of voltage-dependent potassium currents by opiates in facial motoneurons of neonatal rats. Neurosci Res 2003; 47:329-39. [PMID: 14568115 DOI: 10.1016/s0168-0102(03)00223-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We examined the modulation of rat facial motoneurons (FMNs) by opiates in a slice preparation (7-15 days old) using whole-cell patch clamp techniques. Although application of methionine enkephalin (ME) did not change the peak value of the transient outward current (A-current, IA), it reduced the persistent voltage-dependent K(+) currents (IKs) in a dose-dependent manner. The reduction was antagonized by naloxone (40 microM). IKs were reduced only by mu-selective agonist [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]enkephalin (DAMGO, 2-121.6 microM). This reduction was antagonized by naloxone (40 microM) or the mu-selective antagonist, D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Phe-Thr-NH(2) (CTOP, 1 microM). Agonists for other opiate receptors (delta- and kappa-opiate receptor) showed no effect on IKs. In accord with the effects on IKs, DAMGO (100 microM) prolonged the duration of the action potential evoked in Ca(2+)-free external solution containing 4-aminopiridine (1mM). These results suggest that the activation of mu-opiate receptors contributes to signal transduction in FMNs primarily by modulating action potential duration.
Collapse
Affiliation(s)
- Yuichi Kumazawa
- Department of Physiology, Faculty of Medicine, Mie University, Tsu, Mie 143-8540, Japan
| | | | | | | | | | | | | |
Collapse
|
478
|
Mennicken F, Zhang J, Hoffert C, Ahmad S, Beaudet A, O'Donnell D. Phylogenetic changes in the expression of delta opioid receptors in spinal cord and dorsal root ganglia. J Comp Neurol 2003; 465:349-60. [PMID: 12966560 DOI: 10.1002/cne.10839] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
To assess the validity of rodent models for investigating the role of delta opioid receptors (DOR) in analgesia, the distribution of DOR binding and mRNA were compared between rodent and primate spinal cord and dorsal root ganglia (DRG), using receptor autoradiography and in situ hybridization, respectively. In mouse and rat spinal cord, [(125)I]-deltorphin-labeled DOR binding sites were detected throughout the gray matter. In contrast, in primate and particularly in human spinal cord, DOR binding was mainly present in laminae I-II, with little to no binding in deeper layers. Accordingly, in rodent spinal cord, DOR mRNA was expressed by a large number of neurons distributed throughout the ventral and dorsal horns, whereas in the primate, DOR expression was significantly lower, as evidenced by a moderate number of labeled cells throughout the gray matter in monkey and by only few labeled cells in human, mainly in Clarke's column and lamina IX. Major species differences in DOR expression were also observed in primary afferent cells bodies. In rat DRG, intense DOR mRNA hybridization was primarily observed over large ganglion cells immunopositive for neurofilament 200. In contrast, in monkey and human DRG, DOR mRNA was primarily detected over small and medium-sized ganglion cells. These results demonstrate major differences in the expression and distribution of DOR in the spinal cord and DRG between mammalian species. Specifically, they point to a progressive specialization of DOR toward the regulation of primary somatosensory, namely nociceptive, inputs during phylogeny and suggest that the effects of DOR agonists in rodents may not be entirely predictive of their action in humans.
Collapse
Affiliation(s)
- Françoise Mennicken
- Molecular Sciences Department, AstraZeneca Research and Development Montreal, 7171 Frederick-Banting, St. Laurent, Quebec H4S 1Z9, Canada.
| | | | | | | | | | | |
Collapse
|
479
|
Hauser KF, Khurdayan VK, Goody RJ, Nath A, Saria A, Pauly JR. Selective vulnerability of cerebellar granule neuroblasts and their progeny to drugs with abuse liability. THE CEREBELLUM 2003; 2:184-95. [PMID: 14509568 PMCID: PMC4306667 DOI: 10.1080/14734220310016132] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Cerebellar development is shaped by the interplay of genetic and numerous environmental factors. Recent evidence suggests that cerebellar maturation is acutely sensitive to substances with abuse liability including alcohol, opioids, and nicotine. Assuming substance abuse disrupts cerebellar maturation, a central question is: what are the basic mechanisms underlying potential drug-induced developmental defects? Evidence reviewed herein suggests that the maturation of granule neurons and their progeny are intrinsically affected by several classes of substances with abuse liability. Although drug abuse is also likely to target directly other cerebellar neuron and glial types, such as Purkinje cells and Bergmann glia, findings in isolated granule neurons suggest that they are often the principle target for drug actions. Developmental events that are selectively disrupted by drug abuse in granule neurons and/or their neuroblast precursors include proliferation, migration, differentiation (including neurite elaboration and synapse formation), and programmed cell death. Moreover, different classes of drugs act through distinct molecular mechanisms thereby disrupting unique aspects of development. For example, drug-induced perturbations in: (i) neurotransmitter biogenesis; (ii) ligand and ion-gated receptor function and their coupling to intracellular effectors; (iii) neurotrophic factor biogenesis and signaling; and (iv) intercellular adhesion are all likely to have significant effects in shaping developmental outcome. In addition to identifying therapeutic strategies for drug abuse intervention, understanding the mechanisms by which drugs affect cellular maturation is likely to provide a better understanding of the neurochemical events that normally shape central nervous system development.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, Kentucky, 40536-0298, USA.
| | | | | | | | | | | |
Collapse
|
480
|
Phansuwan-Pujito P, Saleema L, Mukda S, Tongjaroenbuangam W, Jutapakdeegul N, Casalotti SO, Forge A, Dodson H, Govitrapong P. The opioid receptors in inner ear of different stages of postnatal rats. Hear Res 2003; 184:1-10. [PMID: 14553898 DOI: 10.1016/s0378-5955(03)00163-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There is increasing evidence that the opioid system has a role in hearing. To provide further evidence for such a role, the expression of opioid receptor mRNAs and proteins in the inner ear of rats was studied during development from birth (P0) to postnatal day 16 (P16). A semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) was employed to detect changes in the expression of delta- (DOR) kappa- (KOR) and mu- (MOR) opioid receptor mRNAs in rat cochleae at P0, P4, P8 and P16. Expression of DOR mRNA levels steadily increased from P0 to P8 with no further increases by P16. KOR mRNA was expressed at a relatively high level at P0 and P4 followed by a decrease while MOR mRNA was expressed at a low level at P0 and P4 followed by an increase by P8 and P16. Immunocytochemical labelling of inner ear sections revealed unique developmental and distribution patterns of opioid receptors. In the organ of Corti DOR immunoreactivity (DOR-IR) was detected in hair cells from P4. In contrast MOR-IR was present only in supporting cells at P0-P16. In the spiral ganglion all three receptor subtypes were expressed from P0 on nerve cell soma and qualitatively appeared to increase with age. Also DOR-IR and MOR-IR were detected at P8 and P16 in nerve fibers within the spiral ganglion. In the limbus DOR-IR was detected at P8 and P16 on cells proximal to the tectorial membrane while MOR-IR was detected more distally. In general these findings demonstrate that within the inner ear each receptor subtype follows specific temporal and spatial developmental patterns, some of which may be associated to the onset of hearing. The data provide further evidence that the opioid system may play a role in the development and functioning of the inner ear.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Animals, Newborn/genetics
- Animals, Newborn/metabolism
- Cochlea/growth & development
- Cochlea/metabolism
- Immunohistochemistry
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Pansiri Phansuwan-Pujito
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | | | | | | | | | | | | | | | | |
Collapse
|
481
|
Abstract
Opioids have been used and abused by humans for centuries. The mu opioid receptor represents the high affinity binding site for opioid narcotics with high abuse liability such as morphine, codeine and fentanyl. Heroin (diacetylmorphine), a semi-synthetic derivative of morphine, crosses the blood-brain barrier more readily than morphine due to its increased hydrophobicity. Once in the brain heroin is hydrolyzed to morphine, which acts at the mu opioid receptor and results in euphoria, thus conferring the reinforcing properties of heroin. Using molecular biology techniques, the mu opioid receptors from several species have been cloned. This article reviews recent progress in this area, with respect to the two major cellular functions of the mu opioid receptor: reduction of intracellular cAMP concentration by inhibiting adenylyl cyclase activity, and inhibition of neuronal firing by modulating membrane ion channels.
Collapse
Affiliation(s)
- L Yu
- Department of Medical and Molecualr Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
482
|
Pol O, Palacio JR, Puig MM. The expression of delta- and kappa-opioid receptor is enhanced during intestinal inflammation in mice. J Pharmacol Exp Ther 2003; 306:455-62. [PMID: 12724348 DOI: 10.1124/jpet.103.049346] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the gut, mu-, delta-, and kappa-opioid receptors are present in the submucous and myenteric plexi and in enterocytes. Using pharmacological methods, our group has shown that intestinal inflammation enhances the antitransit and antisecretory effects of systemic opioids. The aim of the present study was to evaluate whether the enhanced antisecretory effects of delta and kappa-agonists were associated with an increased transcription and/or expression of these receptors at central (brain and spinal cord) and/or peripheral sites (gut); we also evaluated the expression of delta- and kappa-opioid receptors in dissected sections of the gut containing the myenteric (MP/LM) or submucous (SP/M) plexi. The mRNA and protein levels of both opioid receptors were determined using a reverse-transcriptase polymerase chain reaction and immunoprecipitation/Western blot, respectively. Intestinal inflammation significantly augmented the transcription of delta-opioid receptors in the spinal cord (34%) and in the whole gut (102%). Also increased mRNA and protein levels of delta-opioid receptors in the MP/LM and SP/M preparations. The kappa-opioid receptors gene transcription was not altered by inflammation, whereas kappa-opioid receptors protein levels were significantly enhanced in the SP/M preparation. No changes in gene transcription or protein levels for delta- and kappa-opioid receptors could be demonstrated in the brain. These results suggest that local transcriptional and post-transcriptional changes of the delta- and kappa-opioid receptors genes could be responsible for the enhanced antisecretory effects of delta- and kappa-opioid agonists during intestinal inflammation.
Collapse
MESH Headings
- Animals
- Inflammation/metabolism
- Intestinal Diseases/metabolism
- Male
- Mice
- RNA, Messenger/metabolism
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Olga Pol
- Anesthesiology Research Unit, Institut Municipal Investigació Mèdica, Doctor Aiguader, 80, 08003 Barcelona, Spain.
| | | | | |
Collapse
|
483
|
Miranda-Paiva CM, Ribeiro-Barbosa ER, Canteras NS, Felicio LF. A role for the periaqueductal grey in opioidergic inhibition of maternal behaviour. Eur J Neurosci 2003; 18:667-74. [PMID: 12911762 DOI: 10.1046/j.1460-9568.2003.02794.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opiates are known to be involved in the regulation of various events surrounding parturition and lactation, such as maternal behaviour in rats. The onset of this behaviour has been closely linked to opiate action in the medial pre-optic area, where administration of morphine disrupts maternal behaviour during lactation. By combining the use of Fos protein immunohistochemical detection and pharmacological manipulations, in the present paper we show that the periaqueductal grey (PAG) is another region critically involved in the opioidergic blockade of maternal behaviour. According to our observations, a critical level of morphine-induced activation of the rostral lateral PAG appears to be required to inhibit maternal behaviour in lactating rats. This hypothesis was further confirmed in experiments showing that morphine's inhibitory effect on maternal responsiveness was blocked by unilateral naloxone injection into the rostral PAG, but not into nearby regions of the mesencephalic reticular nucleus. Therefore, only a partial inhibition of the opiate's effect on the rostral PAG was needed to block the inhibitory effect of morphine on maternal behaviour. Further studies are needed to ascertain whether the rostral lateral PAG plays a role in the natural onset of maternal behaviour, playing a complementary role to the medial pre-optic area, or merely inhibits maternal behaviour in response to this specific pharmacological challenge. Conversely, the present findings may well reflect a more general role of the PAG, seemingly providing an important piece of information for proposing a hitherto unexplored concept of the PAG as an important centre for the selection of adaptive behavioural responses.
Collapse
Affiliation(s)
- Cláudia M Miranda-Paiva
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, Avenue Orlando Marques Paiva 87, Cidade Universitária 05508-900 São Paulo, SP, Brazil
| | | | | | | |
Collapse
|
484
|
Winzer-Serhan UH, Chen Y, Leslie FM. Expression of opioid peptides and receptors in striatum and substantia nigra during rat brain development. J Chem Neuroanat 2003; 26:17-36. [PMID: 12954528 DOI: 10.1016/s0891-0618(03)00031-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We have used highly sensitive in situ hybridization to determine opioid receptor and peptide expression in embryonic and postnatal rat striatum, to follow the compartmentalization into patch and matrix structures, and have examined their developmental expression in the dopaminergic cell group of the substantia nigra (SN). Furthermore, opioid receptor binding sites were characterized in adjacent sections using highly selective ligands for the opioid receptor subtypes. The major findings of the study are: (1) striatal patches were first delineated by prodynorphin mRNA followed by mu opioid receptor mRNA expression at embryonic days 19 and 21, respectively; (2) in neonates, prodynorphin, mu and kappa opioid receptor mRNAs were transiently co-distributed within patches; (3) prodynorphin mRNA was co-expressed with mu but not kappa, receptor mRNA in neonatal patch neurons; (4) in the SN, kappa receptor and prodynorphin mRNAs were detected as early as embryonic days 15 and 19, respectively; (5) kappa receptor, but not prodynorphin, mRNA was expressed in dopaminergic neurons in the SN. The anatomical results are in agreement with the hypothesis that the endogenous opioid system has a trophic role during the development of striatal patch and matrix compartments and suggest the early regulation of dopamine release by kappa opioid receptors.
Collapse
Affiliation(s)
- Ursula H Winzer-Serhan
- Department of Medical Pharmacology and Toxicology, Texas A&M University System, Health Science Center, College Station, TX 77843-1114, USA.
| | | | | |
Collapse
|
485
|
Sun YG, Lundeberg T, Yu LC. Involvement of endogenous beta-endorphin in antinociception in the arcuate nucleus of hypothalamus in rats with inflammation. Pain 2003; 104:55-63. [PMID: 12855314 DOI: 10.1016/s0304-3959(02)00464-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Although exogenous administration of beta-endorphin to the arcuate nucleus of hypothalamus (ARC) had been shown to produce antinociception, the role of endogenous beta-endorphin of the ARC in nociceptive processing has not been studied directly. The aim of the present study was to investigate the effect of endogenous beta-endorphin in the ARC on nociception in rats with carrageenan-induced inflammation. The hindpaw withdrawal latency (HWL) to noxious thermal and mechanical stimulation was assessed by the hot-plate test and the Randall Selitto Test. Intra-ARC injection of naloxone had no significant influence on the HWL to thermal and mechanical stimulation in intact rats. The HWL decreased significantly after intra-ARC injection of 1 or 10 microg of naloxone in rats with inflammation, but not with 0.1 microg of naloxone. Furthermore, intra-ARC administration of the selective mu-opioid receptor antagonist beta-funaltrexamine (beta-FNA) decreased the nociceptive response latencies to both stimulation in a dose-dependent manner in rats with inflammation, while intra-ARC administration of the selective delta-opioid receptor antagonist naltrindole or the selective kappa-opioid receptor antagonist nor-binaltorphimine (nor-BNI) showed no influences on the nociceptive response latency. The antiserum against beta-endorphin, administered to the ARC, also dose-dependently reduced the HWL in rats with inflammation. The results indicate that endogenous beta-endorphin in the ARC plays an important role in the endogenous antinociceptive system in rats with inflammation, and that its effect is predominantly mediated by the mu-opioid receptor.
Collapse
Affiliation(s)
- Yan-Gang Sun
- Department of Physiology, College of Life Sciences, Peking University, Beijing 100871, People's Republic of China
| | | | | |
Collapse
|
486
|
Abstract
Although dynorphin has long been considered an endogenous opioid peptide with high affinity for the kappa-opioid receptor, its biological function remains uncertain. The high concentration of dynorphin peptides and kappa-opioid receptors in the hypothalamus suggest a possible role for dynorphin in neuroendocrine regulation. This review will summarize evidence that support a role for dynorphin in regulation of the developing hypothalamo-pituitary-adrenal (HPA) axis. Dynorphin can exert dual actions on adrenocorticotropin (ACTH) release: (i) via activation of hypothalamic kappa-opioid receptors leading to release of corticotropin-releasing hormone (CRH) and arginine vasopressin (AVP), and (ii) via a non-opioid mechanism that involves N-methyl-D-aspartate (NMDA) receptors and prostaglandins, and which is not dependent on CRH or AVP. The primary site of action of dynorphin and NMDA appears to be the fetal hypothalamus or a supra-hypothalamic site. The non-opioid mechanism does not mature until a few days prior to parturition and is active for only the brief perinatal period. In contrast, the opioid mechanism behaves as a constitutive system with sustained activity from prenatal to postnatal life. It is likely that the two mechanisms may respond to different stress stimuli and play a different role during development.
Collapse
Affiliation(s)
- Hazel H Szeto
- Department of Pharmacology, LC-405, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA.
| |
Collapse
|
487
|
Philippe D, Dubuquoy L, Groux H, Brun V, Chuoï-Mariot MTV, Gaveriaux-Ruff C, Colombel JF, Kieffer BL, Desreumaux P. Anti-inflammatory properties of the mu opioid receptor support its use in the treatment of colon inflammation. J Clin Invest 2003. [PMID: 12727924 DOI: 10.1172/jci200316750] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The physiologic role of the mu opioid receptor (MOR) in gut nociception, motility, and secretion is well established. To evaluate whether MOR may also be involved in controlling gut inflammation, we first showed that subcutaneous administration of selective peripheral MOR agonists, named DALDA and DAMGO, significantly reduces inflammation in two experimental models of colitis induced by administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) or peripheral expansion of CD4(+) T cells in mice. This therapeutic effect was almost completely abolished by concomitant administration of the opioid antagonist naloxone. Evidence of a genetic role for MOR in the control of gut inflammation was provided by showing that MOR-deficient mice were highly susceptible to colon inflammation, with a 50% mortality rate occurring 3 days after TNBS administration. The mechanistic basis of these observations suggests that the anti-inflammatory effects of MOR in the colon are mediated through the regulation of cytokine production and T cell proliferation, two important immunologic events required for the development of colon inflammation in mice and patients with inflammatory bowel disease (IBD). These data provide evidence that MOR plays a role in the control of gut inflammation and suggest that MOR agonists might be new therapeutic molecules in IBD.
Collapse
Affiliation(s)
- David Philippe
- Equipe Mixte INSERM 0114 sur la Physiopathologie des Maladies Inflammatoires Intestinales, Centre Hospitalier Universitaire, Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
488
|
Le Guen S, Gestreau C, Besson JM. Morphine withdrawal precipitated by specific mu, delta or kappa opioid receptor antagonists: a c-Fos protein study in the rat central nervous system. Eur J Neurosci 2003; 17:2425-37. [PMID: 12814374 DOI: 10.1046/j.1460-9568.2003.02678.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have recently shown concurrent changes in behavioural responses and c-Fos protein expression in the central nervous system in both naive and morphine-dependent rats after systemic administration of the opioid antagonist naloxone. However, because naloxone acts on the three major types of opioid receptors, the present study aimed at determining, in the same animals, both changes in behaviour and c-Fos-like immunoreactivity after intravenous injection of selective opioid antagonists, such as mu (beta-funaltrexamine, 10 mg/kg), delta (naltrindole, 4 mg/kg) or kappa (nor-binaltorphimine, 5 mg/kg) opioid receptor antagonists, in naive or morphine-dependent rats. In a first experimental series, only beta-funaltrexamine increased c-Fos expression in the eight central nervous system structures examined, whereas no effect was seen after naltrindole or nor-binaltorphimine administration in naive rats. These results suggest a tonic activity in the endogenous opioid peptides acting on mu opioid receptors in normal rats. A second experimental series in morphine-dependent rats showed that beta-funaltrexamine had the highest potency in the induction of classical signs of morphine withdrawal syndrome, as well as the increase in c-Fos expression in the 22 central nervous system structures studied, suggesting a major role of mu opioid receptors in opioid dependence. However, our results also demonstrated that naltrindole and, to a lesser extent, nor-binaltorphimine were able to induce moderate signs of morphine withdrawal and relatively weak c-Fos protein expression in restricted central nervous system structures. Therefore, delta and kappa opioid receptors may also contribute slightly to opioid dependence.
Collapse
Affiliation(s)
- Stéphanie Le Guen
- Laboratoire de Physiopharmacologie du Système Nerveux, Institut National de la Santé et de la Recherche Médicale (INSERM) and Ecole Pratique des Hautes Etudes (EPHE), Paris, France.
| | | | | |
Collapse
|
489
|
Philippe D, Dubuquoy L, Groux H, Brun V, Chuoï-Mariot MTV, Gaveriaux-Ruff C, Colombel JF, Kieffer BL, Desreumaux P. Anti-inflammatory properties of the mu opioid receptor support its use in the treatment of colon inflammation. J Clin Invest 2003; 111:1329-38. [PMID: 12727924 PMCID: PMC154442 DOI: 10.1172/jci16750] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The physiologic role of the mu opioid receptor (MOR) in gut nociception, motility, and secretion is well established. To evaluate whether MOR may also be involved in controlling gut inflammation, we first showed that subcutaneous administration of selective peripheral MOR agonists, named DALDA and DAMGO, significantly reduces inflammation in two experimental models of colitis induced by administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) or peripheral expansion of CD4(+) T cells in mice. This therapeutic effect was almost completely abolished by concomitant administration of the opioid antagonist naloxone. Evidence of a genetic role for MOR in the control of gut inflammation was provided by showing that MOR-deficient mice were highly susceptible to colon inflammation, with a 50% mortality rate occurring 3 days after TNBS administration. The mechanistic basis of these observations suggests that the anti-inflammatory effects of MOR in the colon are mediated through the regulation of cytokine production and T cell proliferation, two important immunologic events required for the development of colon inflammation in mice and patients with inflammatory bowel disease (IBD). These data provide evidence that MOR plays a role in the control of gut inflammation and suggest that MOR agonists might be new therapeutic molecules in IBD.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- CD4-Positive T-Lymphocytes/metabolism
- Colitis/chemically induced
- Colitis/drug therapy
- Colitis/mortality
- Colitis/pathology
- Colon/drug effects
- Colon/immunology
- Colon/pathology
- Cytokines/metabolism
- Disease Models, Animal
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/therapeutic use
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Oligopeptides/pharmacology
- Oligopeptides/therapeutic use
- Peroxidase/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/physiology
- Trinitrobenzenesulfonic Acid/toxicity
Collapse
Affiliation(s)
- David Philippe
- Equipe Mixte INSERM 0114 sur la Physiopathologie des Maladies Inflammatoires Intestinales, Centre Hospitalier Universitaire, Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
490
|
Aicher SA, Mitchell JL, Mendelowitz D. Distribution of mu-opioid receptors in rat visceral premotor neurons. Neuroscience 2003; 115:851-60. [PMID: 12435423 DOI: 10.1016/s0306-4522(02)00459-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Agonists of the mu-opioid receptor (MOR) can modulate the activity of visceral premotor neurons, including cardiac premotor neurons. Neurons in brainstem regions containing these premotor neurons also contain dense concentrations of the MOR1. This study examined the distribution of MOR1 within two populations of visceral premotor neurons: one located in the dorsal motor nucleus of the vagus and the other in the nucleus ambiguus. Visceral premotor neurons contained the retrograde tracer Fluoro-Gold following injections of the tracer into the pericardiac region of the thoracic cavity. MOR1 was localized using immunogold detection of an anti-peptide antibody. Visceral premotor neurons in both regions contained MOR1 at somatic and dendritic sites, although smaller dendrites were less likely to contain the receptor than larger dendrites, suggesting there may be selective trafficking of MOR1 within these neurons. MOR1 labeling in nucleus ambiguus neurons was more likely to be localized to plasma membrane sites, suggesting that ambiguus neurons may be more responsive to opioid ligands than neurons in the dorsal motor nucleus of the vagus. In addition, many of the dendrites of visceral premotor neurons were in direct apposition to other dendrites. MOR1 was often detected at these dendro-dendritic appositions that may be gap junctions. Together these findings indicate that the activity of individual visceral premotor neurons, as well as the coupling between neurons, may be regulated by ligands of the MOR.
Collapse
Affiliation(s)
- S A Aicher
- Neurological Sciences Institute, Oregon Health & Science University, 505 NW 185th Avenue, 97006, Beaverton, OR 97006, USA.
| | | | | |
Collapse
|
491
|
Jamot L, Matthes HWD, Simonin F, Kieffer BL, Roder JC. Differential involvement of the mu and kappa opioid receptors in spatial learning. GENES, BRAIN, AND BEHAVIOR 2003; 2:80-92. [PMID: 12884965 DOI: 10.1034/j.1601-183x.2003.00013.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In order to test the role of mu and kappa opioid receptors (Mu opioid receptor (MOR) and Kappa opioid receptor (KOR)) in hippocampal-dependent spatial learning, we analyzed genetically engineered null mutant mice missing the functional MOR or KOR gene. Compared to wild-type mice, the homozygous MOR null mutants exhibited an impairment in the ultimate level of spatial learning as shown in two distinct tasks, the 8-arm radial-maze and the Morris water-maze. Control behaviors were normal. The learning impairment could be associated with the impairment we found in the maintenance of long-term potentiation in mossy fibers in CA3. In comparison, there was no impairment in spatial learning in our KOR mutants or in mossy fibers (mf) in CA3 region long-term potentiation (LTP). Our work suggests that the MOR may play a positive role in learning and memory by increasing LTP in CA3 neurons.
Collapse
Affiliation(s)
- L Jamot
- The Samuel Lunenfeld Research Institute at Mt. Sinai Hospital, and the Department of Molecular and Medical Genetics, University of Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
492
|
Al-Khrasani M, Elor G, Yusuf Abbas M, Rónai AZ. The effect of endomorphins on the release of 3H-norepinephrine from rat nucleus tractus solitarii slices. REGULATORY PEPTIDES 2003; 111:97-101. [PMID: 12609755 DOI: 10.1016/s0167-0115(02)00257-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We used two, 3-min field stimulation cycles 30 min apart (S1, S2) in 3H-norepinephrine-loaded, superfused rat nucleus tractus solitarii-dorsal motor vagal nucleus (NTS-DVN) slices. The stimulation-induced release was expressed as the area above the baseline. Drugs were introduced 12 min before S2 and drug actions were characterized in terms of alterations of S2/S1 ratios. The S2/S1 ratio was 1.047 (0.946-1.159, n = 4, geometric mean and 95% confidence interval) in controls and 0.336 (0.230-0.490, n = 3), 0.726 (0.590-0.892, n = 4), 0.613 (0.594-0.683, n = 4) and 0.665 (0.500-0.886, n = 4) in the presence of 10(-6) M clonidine, D-Ala(2),MePhe(4),Gly(5)-ol-enkephalin (DAMGO), endomorphin-1 (Tyr-Pro-Trp-Phe-NH(2), EM-1) and -2 (Tyr-Pro-Phe-Phe-NH(2), EM-2) [the latter two in the presence of 10(-4) M diprotin A, an inhibitor of dipeptidyl-aminopeptidase IV (DAP-IV) enzyme]. The effect of DAMGO at 10(-5) M was significantly higher than at 10(-6) M, whereas the effect of endomorphins did not differ at the two concentration levels. Diprotin A potentiated only very modestly the action of endomorphins. These data (a) confirm the presence of functional mu-opioid receptors in the vagal complex, (b) render it likely that the enzymic degradation of endomorphins is not a highly effective process in brain slices and (c) may suggest that the apparent ceiling in the effect of endomorphins might be related to their partial agonist property.
Collapse
Affiliation(s)
- Mahmoud Al-Khrasani
- Faculty of Medicine, Department of Pharmacology and Pharmacotherapy, Semmelweis University, POB 370, H-1445, Budapest, Hungary.
| | | | | | | |
Collapse
|
493
|
Andrews ZB, Grattan DR. Opioid receptor subtypes involved in the regulation of prolactin secretion during pregnancy and lactation. J Neuroendocrinol 2003; 15:227-36. [PMID: 12588510 DOI: 10.1046/j.1365-2826.2003.00975.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Afferent endogenous opioid neuronal systems facilitate prolactin secretion in a number of physiological conditions including pregnancy and lactation, by decreasing tuberoinfundibular dopamine (TIDA) inhibitory tone. The aim of this study was to investigate the opioid receptor subtypes involved in regulating TIDA neuronal activity and therefore facilitating prolactin secretion during early pregnancy, late pregnancy and lactation in rats. Selective opioid receptor antagonists nor-binaltorphimine (kappa-receptor antagonist, 15 micro g/5 micro l), beta funaltrexamine (mu-receptor antagonist, 5 microg/5 microl) and naltrindole (delta-receptor antagonist, 5 microg/5 microl) or saline were administered intracerebroventricularly (i.c.v.) on day 8 of pregnancy during a nocturnal prolactin surge, on day 21 of pregnancy during the ante partum prolactin surge or on day 7 of lactation before the onset of a suckling stimulus. Serial blood samples were collected at regular time intervals, via chronic indwelling jugular cannulae, before and after drug administration and plasma prolactin was determined by radioimmunoassay. TIDA neuronal activity was measured using the 3,4-dihydroxyphenylacetic acid (DOPAC) : dopamine ratio in the median eminence 2 h 30 min after i.c.v. drug injection. In each experimental condition, plasma prolactin was significantly inhibited by both kappa- and mu-receptor antagonists, whereas the delta-receptor antagonist had no effect compared to saline-injected controls. Similarly, nor-binaltorphimine and beta funaltrexamine significantly increased the median eminence DOPAC : dopamine ratio during early and late pregnancy, and lactation whereas naltrindole had no effect compared to saline-injected controls. These data suggest that TIDA neuronal activity, and subsequent prolactin secretion, is regulated by endogenous opioid peptides acting at both kappa- and mu-opioid receptors during prolactin surges of early pregnancy, late pregnancy and lactation.
Collapse
MESH Headings
- Animals
- Female
- Injections, Intraventricular
- Lactation/metabolism
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Pituitary Gland/metabolism
- Pregnancy
- Pregnancy, Animal/metabolism
- Prolactin/blood
- Prolactin/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid/metabolism
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Z B Andrews
- Department of Anatomy and Structural Biology and Neuroscience Research Centre, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
494
|
Cahill CM, Morinville A, Hoffert C, O'Donnell D, Beaudet A. Up-regulation and trafficking of delta opioid receptor in a model of chronic inflammation: implications for pain control. Pain 2003; 101:199-208. [PMID: 12507715 DOI: 10.1016/s0304-3959(02)00333-0] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pharmacological and physiological evidence supports a role for delta (delta) opioid receptors in the nociceptive mechanisms of inflammation. However, few data exist regarding delta opioid receptor expression and localization in such conditions. In this study, we have assessed the distribution and function of delta opioid receptors in the rat spinal cord following induction of chronic inflammation by intraplantar injection of complete Freund's adjuvant (CFA). Intrathecal administration of the selective delta opioid receptor agonist, D-[Ala(2), Glu(4)] deltorphin, dose-dependently reversed thermal hyperalgesia induced by CFA. In situ hybridization and Western blotting experiments revealed an increase in delta opioid receptor mRNA and protein levels, respectively, in the dorsal lumbar spinal cord ipsilateral to the CFA injection site compared to the contralateral side and sham-injected controls. By electron microscopy, immunopositive delta opioid receptors were evident in neuronal perikarya, dendrites, unmyelinated axons and axon terminals. Quantification of immunopositive signal in dendrites revealed a twofold increase in the number of immunogold particles in the ipsilateral dorsal spinal cord of CFA-injected rats compared to the contralateral side and to sham-injected rats. Moreover, the relative frequency of immunogold particles associated with or in close proximity to the plasma membrane was increased in the ipsilateral dorsal spinal cord, indicating a more efficient targeting of delta opioid receptors to neuronal plasma membranes. These data demonstrate that CFA induces an up-regulation and increased membrane targeting of delta opioid receptors in the dorsal spinal cord which may account for the enhanced antinociceptive effects of delta opioid receptor agonists in chronic inflammatory pain models.
Collapse
MESH Headings
- Animals
- Arthritis/complications
- Behavior, Animal
- Chronic Disease
- Disease Models, Animal
- Freund's Adjuvant
- Gene Expression
- Hyperalgesia/chemically induced
- Hyperalgesia/metabolism
- Hyperalgesia/physiopathology
- Male
- Microscopy, Electron
- Neurogenic Inflammation/chemically induced
- Neurogenic Inflammation/metabolism
- Neurogenic Inflammation/physiopathology
- Neurons/metabolism
- Neurons/ultrastructure
- Nociceptors/drug effects
- Nociceptors/physiology
- Oligopeptides/pharmacology
- Pain, Postoperative/chemically induced
- Pain, Postoperative/metabolism
- Pain, Postoperative/physiopathology
- Protein Transport/physiology
- RNA, Messenger/analysis
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Spinal Cord/cytology
- Up-Regulation/physiology
Collapse
Affiliation(s)
- C M Cahill
- Canada Department of Neurology and Neurosurgery, Montreal Neurological Institute, 3801 University Street, Montreal, Quebec, Canada H3A 2B4
| | | | | | | | | |
Collapse
|
495
|
Johnson-Davis KL, Hanson GR, Keefe KA. Lack of effect of kappa-opioid receptor agonism on long-term methamphetamine-induced neurotoxicity in rats. Neurotox Res 2003; 5:273-81. [PMID: 12835119 DOI: 10.1007/bf03033385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
High-dose methamphetamine treatment induces long-term deficits in central monoamine systems. However, the mechanisms underlying these effects are unknown. Previous work has shown that the Kappa-opioid receptor agonist U-69593 [(+)-(5alpha,7alpha,8b)-(+)-N-methyl-N[7-(1-pyrrolidinyl)-1-oxaspiro[4.5]dec-8-yl] benzeneacetamide] attenuates the neurotoxic effects of methamphetamine on extracellular dopamine levels in mice, suggesting that endogenous Kappa-opioid receptor ligands, such as dynorphin, may protect against methamphetamine-induced toxicity and play a role in mediating the long-term consequences of methamphetamine. To further examine the role that dynorphin systems play in methamphetamine-induced neurotoxicity, we administered to male rats a total of four injections of methamphetamine (7.5 mg/kg, s.c.), with a 2-h interval between each dose. Rats were pretreated with either the Kappa-agonist U-69593 (0.32 mg/kg, s.c.) or vehicle, 15 min prior to the first and third methamphetamine injection. Furthermore, cages containing the U-69593 + methamphetamine-treated rats were placed on heating pads for 30 min after the first U-69593 injection to prevent the drug from blocking methamphetamine-induced hyperthermia. Rats were sacrificed 7 days after treatment. Striatal dopamine and serotonin contents were decreased approximately 75% and 55%, respectively, in the methamphetamine-treated rats and approximately 88% and 65%, respectively, in rats receiving the U-69593 + methamphetamine combination. There was a approximately 20% mortality rate in the rats treated with methamphetamine compared to approximately 75% mortality rate in rats treated with both U-69593 and methamphetamine. A similar rate of mortality was observed when combining a different Kappa-agonist, U-50488 [trans-(-)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamine], with methamphetamine. These data suggest that Kappa-agonists do not protect against methamphetamine-induced toxicity to monoamines in rats, and may potentiate mortality when co-administered with methamphetamine.
Collapse
Affiliation(s)
- Kamisha L Johnson-Davis
- Department of Pharmacology and Toxicology, University of Utah, 30 South 2000 East, Rm. 201, Salt Lake City, UT 84112, USA.
| | | | | |
Collapse
|
496
|
Abstract
Although a large superfamily of G-protein-coupled receptors serves multiple functions, little is known about their functional activation during ontogeny. To examine the functional activation of the mu-opioid receptor (MOR) and the delta-opioid receptor (DOR) during development, sections of mouse embryos and fetuses from e11.5 until birth were treated with DAMGO and DPDPE, respectively, and the ability of these drugs to induce G-protein coupling was assessed by using GTPgamma(35)S binding autoradiography. MOR activation was first detected in the caudate-putamen (CPU) at e12.5, and by e15.5, activity had not only increased in this region but also expanded to include the midbrain, medial habenula, hypothalamus, pons, and medulla. DOR activity first appeared at e17.5 in the hypothalamus, pons, medial habenula, and medulla and at p1 in the CPU at levels noticeably less than those of the MOR. In general, MOR and DOR activation lagged only slightly behind the appearance of MOR-1 and DOR-1 mRNA but delayed activation was particularly pronounced in the trigeminal ganglia, where MOR-1 gene expression was first detected at e13.5, but MOR activity was not observed even at birth. Thus, the data demonstrate temporal and often region-specific differences in the appearance and magnitude of functional activity in cell groups expressing either the MOR-1 or DOR-1 genes, suggesting that interaction between the opioid receptors, G-proteins, and other signaling cofactors is developmentally regulated.
Collapse
Affiliation(s)
- Joshua F Nitsche
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | |
Collapse
|
497
|
Schroeder JA, Niculescu M, Unterwald EM. Cocaine alters mu but not delta or kappa opioid receptor-stimulated in situ [35S]GTPgammaS binding in rat brain. Synapse 2003; 47:26-32. [PMID: 12422370 DOI: 10.1002/syn.10148] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic cocaine administration produces alterations in mu and kappa opioid receptor density as well as striatal and accumbens opioid-regulated adenylyl cyclase activity, suggesting a psychostimulant responsive interaction between opioidergic and dopaminergic systems. Stimulation of G-protein-coupled opioid receptors inhibits adenylyl cyclase production of cyclic AMP. The present study employed in situ [(35)S]GTPgammaS binding to measure opioid receptor-stimulated activation of G-proteins in response to acute and chronic cocaine exposure. Male Fischer rats received acute (1 or 3 days) or chronic (14 days) binge pattern cocaine administration. Three and 14 days of cocaine injections resulted in greater increases in the ability of the mu receptor agonist DAMGO to stimulate [(35)S]GTPgammaS binding in both the core and the shell of the nucleus accumbens, all regions of the caudate putamen and the cingulate cortex compared with saline-matched controls. The greatest increases in DAMGO-stimulated [(35)S]GTPgammaS binding were observed in the dorsal areas of the caudate putamen in animals that received 14 days of cocaine. No significant changes in delta (DPDPE), or kappa (dynorphin A(1-17)) receptor-stimulated [(35)S]GTPgammaS binding were found in any brain region in response to cocaine administration. These results demonstrate that binge pattern cocaine administration induce changes in mu but not delta or kappa opioid receptor-mediated G-protein activity. This study provides support for the hypothesis that the addictive properties of both psychostimulants and opiates may share common neurochemical signaling substrates.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Autoradiography
- Brain/drug effects
- Brain/metabolism
- Caudate Nucleus/drug effects
- Caudate Nucleus/metabolism
- Cocaine/administration & dosage
- Cocaine/pharmacology
- Dopamine Uptake Inhibitors/administration & dosage
- Dopamine Uptake Inhibitors/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Gyrus Cinguli/drug effects
- Gyrus Cinguli/metabolism
- Heterotrimeric GTP-Binding Proteins/metabolism
- Male
- Nucleus Accumbens/drug effects
- Nucleus Accumbens/metabolism
- Putamen/drug effects
- Putamen/metabolism
- Rats
- Rats, Inbred F344
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
- Sulfur Radioisotopes
- Time Factors
Collapse
Affiliation(s)
- Joseph A Schroeder
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA.
| | | | | |
Collapse
|
498
|
Ständer S, Gunzer M, Metze D, Luger T, Steinhoff M. Localization of mu-opioid receptor 1A on sensory nerve fibers in human skin. REGULATORY PEPTIDES 2002; 110:75-83. [PMID: 12468112 DOI: 10.1016/s0167-0115(02)00159-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Opioid peptides are endogenous neuromodulators that play a major role in the nociceptive pathway by interacting with opioid receptors. So far, four opioid receptors (micro-, delta-, kappa-, orphan-receptor) have been cloned with a wide distribution in the central and peripheral nervous system. In the present study, we give first evidence for the presence of the micro-opioid receptor (MOR) isoform 1A in nerve fibers of human skin. Immunohistochemical analysis revealed MOR immunoreactivity to be present in dermal and epidermal nerve fibers. Double-immunofluorescence staining revealed that MOR is present on calcitonin gene-related protein (CGRP)-positive sensory nerve fibers, while autonomic nerves of blood vessels, hair follicles, or skin glands were negative. In diseased skin such as psoriasis vulgaris, atopic dermatitis, and prurigo nodularis, distribution of MOR 1A immunoreactivity was similar to that of normal skin. These findings expand our knowledge about a direct regulatory role of cutaneous opioid receptors in the skin. Thus, peripheral cutaneous opioid receptors may be involved in the transmission of pain and pruritus, respectively. This is supported by previous observation that opioid receptor antagonists may significantly diminish experimentally evoked histamine-induced itch of the skin. Together, our findings contribute to the understanding of the high antipruritic potency of opioid receptor antagonists in various skin and systemic diseases.
Collapse
Affiliation(s)
- Sonja Ständer
- Department of Dermatology, University of Münster, Münster, Germany.
| | | | | | | | | |
Collapse
|
499
|
Hoopfer ED, Huang L, Denver RJ. Basic transcription element binding protein is a thyroid hormone-regulated transcription factor expressed during metamorphosis in Xenopus laevis. Dev Growth Differ 2002; 44:365-81. [PMID: 12392570 DOI: 10.1046/j.1440-169x.2002.00650.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Basic transcription element binding protein (BTEB) is a member of the Krüppel family of zinc finger transcription factors. It has been shown that BTEB plays a role in promoting neuronal process formation during postembryonic development. In the present study, the biochemical properties, transactivation function, and the developmental and hormone-regulated expression of BTEB in Xenopus laevis (xBTEB) are described. xBTEB binds the GC-rich basic transcription element (BTE) with high affinity and functions as a transcriptional activator on promoters containing multiple or single GC boxes. xBTEB mRNA levels increase in the tadpole brain, intestine and tail during metamorphosis, and are correlated with tissue-specific morphological and biochemical transformations. xBTEB mRNA expression can be induced precociously in premetamorphic tadpole tissues by treatment with thyroid hormone. In situ hybridization histochemistry showed that thyroid hormone upregulates xBTEB mRNA throughout the brain of premetamorphic tadpoles, with the highest expression found in the subventricular zones of the telencephalon, diencephalon, optic tectum, cerebellum and spinal cord. xBTEB protein parallels changes in its mRNA, and it was found that xBTEB is not expressed in mitotic cells in the developing brain, but is expressed just distal to the proliferative zone, supporting the hypothesis that this protein plays a role in neural cell differentiation.
Collapse
Affiliation(s)
- Eric D Hoopfer
- 3065C Natural Science Building, Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, MI 48109-1048, USA
| | | | | |
Collapse
|
500
|
Le Moine C, Fauchey V, Jaber M. Opioid receptor gene expression in dopamine transporter knock-out mice in adult and during development. Neuroscience 2002; 112:131-9. [PMID: 12044478 DOI: 10.1016/s0306-4522(02)00014-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dopamine transporter knock-out mice display locomotor hyperactivity due to increased extracellular striatal levels of dopamine. Hyperdopaminergic activity within this mesolimbic pathway is involved in the rewarding properties of morphine which are also increased in these mice. Due to the hyperdopaminergia, profound alterations in gene expression for dopamine receptors and neuropeptides are observed in the caudate putamen and nucleus accumbens. Here we investigated (1) the levels of mu-, delta- and kappa-opioid receptors mRNAs in normal mice from gestational day 13 (G13) to adult, and (2) the adaptive changes in the expression of these receptors in mice lacking the dopamine transporter. Our results show that, in wild-type mice, mu-opioid receptor mRNA expression appears early during development (G13) with a homogeneous distribution that evolves towards a patchy distribution in adult. Delta-opioid receptor mRNA appears only at G17 and kappa-opioid receptor mRNA is not observed before adulthood. The levels of delta-opioid receptor mRNA are not modified during development in knock-out mice compared to wild-type, but are increased in adult caudate putamen (+39%, P<0.05) and nucleus accumbens (+66%, P<0.05) at a time when these receptors are believed to be functional. The mu- and kappa-opioid receptors mRNA levels are not modified in the adult knock-out mice. In addition, we observed no differences in any opioid receptor mRNA level in dopamine transporter knock-out mice during prenatal ontogeny compared to wild-type. Our results constitute a detailed neuroanatomical description of opioid receptor mRNA expression from the time of their appearance during prenatal development until adulthood. Furthermore, we show here that chronic constitutive hyperdopaminergia only affects delta-opioid receptor mRNA levels in adult. Even if the propensity of knock-out mice to show increased rewarding properties to morphine seems to be mainly due to the substantial and further increase in hyperdopaminergic activity following drug treatment, the involvement of increased delta-opioid receptor mRNA levels in this behavior remains to be elucidated.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Animals, Newborn/growth & development
- Animals, Newborn/metabolism
- Dopamine Plasma Membrane Transport Proteins
- Embryo, Mammalian/metabolism
- Gene Expression/physiology
- Membrane Glycoproteins
- Membrane Transport Proteins/deficiency
- Membrane Transport Proteins/genetics
- Membrane Transport Proteins/physiology
- Mice
- Mice, Knockout/genetics
- Nerve Tissue Proteins
- RNA, Messenger/metabolism
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, mu/genetics
- Reference Values
Collapse
Affiliation(s)
- C Le Moine
- UMR CNRS 5541, Laboratoire d'Histologie Embryologie, Université Victor Segalen Bordeaux 2, Bordeaux, France.
| | | | | |
Collapse
|