Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 187
Filter
Add more filters










Publication year range
1.
Psychopharmacology (Berl) ; 239(7): 2187-2199, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35211768

ABSTRACT

RATIONALE: Tianeptine is a mu-opioid receptor (MOR) agonist with increasing reports of abuse in human populations. Preclinical data regarding the abuse potential and other opioid-like adverse effects of tianeptine at supratherapeutic doses are sparse. OBJECTIVES: The present study evaluated tianeptine in a rat model of abuse potential assessment and in mouse models of motor, gastrointestinal, and respiratory adverse effects. METHODS: Abuse potential was assessed in adult male Sprague-Dawley rats using an intracranial self-stimulation (ICSS) procedure to determine effects of acute and repeated tianeptine on responding for electrical brain stimulation. Male ICR mice were used to determine the effects of tianeptine in assays of locomotor behavior and gastrointestinal motility. Male Swiss-Webster mice were monitored for respiratory changes using whole-body plethysmography. RESULTS: In rats, acute tianeptine produced weak and delayed evidence for abuse-related ICSS facilitation at an intermediate dose (10 mg/kg, IP) and pronounced, naltrexone-preventable ICSS depression at a higher dose (32 mg/kg, IP). Repeated 7-day tianeptine (10 and 32 mg/kg/day, IP) produced no increase in abuse-related ICSS facilitation, only modest tolerance to ICSS depression, and no evidence of physical dependence. In mice, tianeptine produced dose-dependent, naltrexone-preventable locomotor activation. Tianeptine (100 mg/kg, SC) also significantly inhibited gastrointestinal motility and produced naloxone-reversible respiratory depression. CONCLUSIONS: Tianeptine presents as a MOR agonist with resistance to tolerance and dependence in our ICSS assay in rats, and it has lower abuse potential by this metric than many commonly abused opioids. Nonetheless, tianeptine produces MOR agonist-like acute adverse effects that include motor impairment, constipation, and respiratory depression.


Subject(s)
Opioid-Related Disorders , Respiratory Insufficiency , Analgesics, Opioid/pharmacology , Animals , Male , Mice , Mice, Inbred ICR , Naltrexone/pharmacology , Rats , Rats, Sprague-Dawley , Self Stimulation , Thiazepines
2.
Neurogastroenterol Motil ; 27(4): 468-80, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25703354

ABSTRACT

BACKGROUND: Gastrointestinal (GI) dysfunction is a major cause of morbidity in acquired immunodeficiency syndrome (AIDS). HIV-1-induced neuropathogenesis is significantly enhanced by opiate abuse, which increases proinflammatory chemokine/cytokine release, the production of reactive species, glial reactivity, and neuronal injury in the central nervous system. Despite marked interactions in the gut, little is known about the effects of HIV-1 in combination with opiate use on the enteric nervous system. METHODS: To explore HIV-opiate interactions in myenteric neurons, the effects of Tat ± morphine (0.03, 0.3, and 3 µM) were examined in isolated neurons from doxycycline- (DOX-) inducible HIV-1 Tat(1-86) transgenic mice or following in vitro Tat 100 nM exposure (>6 h). KEY RESULTS: Current clamp recordings demonstrated increased neuronal excitability in neurons of inducible Tat(+) mice (Tat+/DOX) compared to control Tat-/DOX mice. In neurons from Tat+/DOX, but not from Tat-/DOX mice, 0.03 µM morphine significantly reduced neuronal excitability, fast transient and late long-lasting sodium currents. There was a significant leftward shift in V(0.5) of inactivation following exposure to 0.03 µM morphine, with a 50% decrease in availability of sodium channels at -100 mV. Similar effects were noted with in vitro Tat exposure in the presence of 0.3 µM morphine. Additionally, GI motility was significantly more sensitive to morphine in Tat(+) mice than Tat(-) mice. CONCLUSIONS & INFERENCES: Overall, these data suggest that the sensitivity of enteric neurons to morphine is enhanced in the presence of Tat. Opiates and HIV-1 may uniquely interact to exacerbate the deleterious effects of HIV-1-infection and opiate exposure on GI function.


Subject(s)
Central Nervous System Sensitization/drug effects , Enteric Nervous System/drug effects , Morphine/toxicity , Neurons/drug effects , tat Gene Products, Human Immunodeficiency Virus/toxicity , Animals , Cells, Cultured , Enteric Nervous System/physiopathology , Gastrointestinal Motility/drug effects , Ileum/metabolism , Mice , Mice, Transgenic , Neurons/physiology , Receptors, Opioid, mu/metabolism , Sodium Channels/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics
3.
Neurogastroenterol Motil ; 26(10): 1361-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25257923

ABSTRACT

Opioid-induced constipation is a major clinical problem. The effects of morphine, and other narcotics, on the gastrointestinal tract persist over long-term use thus limiting the clinical benefit of these excellent pain relievers. The effects of opioids in the gut, including morphine, are largely mediated by the µ-opioid receptors at the soma and nerve terminals of enteric neurons. Recent studies demonstrate that regional differences exist in both acute and chronic morphine along the gastrointestinal tract. While tolerance develops to the analgesic effects and upper gastrointestinal motility upon repeated morphine administration, tolerance does not develop in the colon with chronic opioids resulting in persistent constipation. Here, we review the mechanisms by which tolerance develops in the small but not the large intestine. The regional differences lie in the signaling and regulation of the µ-opioid receptor in the various segments of the gastrointestinal tract. The differential role of ß-arrestin2 in tolerance development between central and enteric neurons defines the potential for therapeutic approaches in developing ligands with analgesic properties and minimal constipating effects.


Subject(s)
Analgesics, Opioid/adverse effects , Drug Tolerance , Enteric Nervous System/drug effects , Gastrointestinal Tract/drug effects , Morphine/adverse effects , Arrestins/metabolism , Constipation/chemically induced , Humans , Neurons/drug effects , Pain/drug therapy , Receptors, Opioid, mu/metabolism , beta-Arrestins
4.
J Pharmacol Exp Ther ; 345(3): 512-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23528610

ABSTRACT

The chronic use of opioids in humans, accompanied by the development of tolerance, is a dangerous phenomenon in its own right. However, chronic opioid use is often made more dangerous by the coconsumption of other substances. It has been observed that the blood level of opioids in postmortem analyses of addicts, who consumed ethanol along with the opioid, was much less than that observed in individuals who died from opioids alone. This relationship between ethanol and opioids led us to investigate the hypothesis that ethanol alters tolerance to opioids. In the present study, we report that ethanol significantly and dose-dependently reduced the antinociceptive tolerance produced by morphine and the cross-tolerance between [D-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin (DAMGO) and morphine in the mouse tail-flick test. The reversal of morphine tolerance was partially blocked by both the gamma receptor blocker bicuculline and by the γ-aminobutyric acid (GABA)(B) receptor blocker phaclofen and the administration of both inhibitors completely reversed the effects of ethanol on morphine tolerance. Diazepam, like ethanol, decreased morphine tolerance. However, this inhibition was reversed by the GABA(A) antagonist bicuculline but not by the GABA(B) antagonist phaclofen. These findings have important implications for individuals who abuse opioids and ethanol as well as suggest a mechanism to reduce the amount of opioid needed in chronic pain treatment.


Subject(s)
Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Morphine/antagonists & inhibitors , Morphine/pharmacology , Animals , Baclofen/analogs & derivatives , Baclofen/pharmacology , Bicuculline/pharmacology , Diazepam/pharmacology , Dose-Response Relationship, Drug , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , GABA Antagonists/pharmacology , Hypnotics and Sedatives/pharmacology , Immersion , Injections, Intraventricular , Male , Mice , Pain Measurement/drug effects , Receptors, GABA-A/drug effects , Receptors, GABA-B/drug effects
5.
J Pharmacol Exp Ther ; 332(3): 1127-35, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20008489

ABSTRACT

Differences in the mechanisms underlying tolerance and mu-opioid receptor desensitization resulting from exposure to opioid agonists of different efficacy have been suggested previously. The objective of this study was to determine the effects of protein kinase C (PKC) and G protein-coupled receptor kinase (GRK) inhibition on antinociceptive tolerance in vivo to opioid agonists of different efficacy. A rapid (8-h) tolerance-induction model was used where each opioid was repeatedly administered to naive mice. Animals were then challenged with the opioid after injection of a kinase inhibitor to determine its effects on the level of tolerance. Tolerance to meperidine, morphine, or fentanyl was fully reversed by the PKC inhibitor 12-(2-cyanoethyl)-6,7,12,13-tetrahydro-13-methyl-5-oxo-5H-indolo(2,3-a)pyrrolo(3,4-c)carbazole (Gö6976). However, in vivo tolerance to [d-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO) was not reversed by PKC inhibition. The novel small-molecule GRK inhibitors beta-adrenergic receptor kinase 1 inhibitor and 2-(8-[(dimethylamino) methyl]-6,7,8,9-tetrahydropyridol[1,2-a]indol-3-yl)-3-(1-methylindol-3-yl)maleimide (Ro 32-0432) did not reverse the tolerance to meperidine, fentanyl, or morphine but did reverse the tolerance to DAMGO. To correlate GRK-dependent DAMGO-induced tolerance with mu-opioid receptor desensitization, we used in vitro whole-cell patch-clamp recording from mouse locus coeruleus neurons and observed that the GRK inhibitors reduced DAMGO-induced desensitization of mu-opioid receptors, whereas the PKC inhibitor had no effect. These results suggest that tolerance induced by low- and moderate-efficacy mu-opioid receptor agonists is dependent on PKC, whereas tolerance induced by the high-efficacy agonist DAMGO is dependent on GRK.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/drug effects , G-Protein-Coupled Receptor Kinases/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Animals , Brain/physiology , Drug Interactions , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Fentanyl/pharmacology , In Vitro Techniques , Locus Coeruleus/drug effects , Locus Coeruleus/physiology , Male , Meperidine/pharmacology , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques
6.
Br J Pharmacol ; 158(1): 157-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19309357

ABSTRACT

BACKGROUND AND PURPOSE: The ability of an agonist to induce desensitization of the mu-opioid receptor (MOR) depends upon the agonist used. Furthermore, previous data suggest that the intracellular mechanisms underlying desensitization may be agonist-specific. We investigated the mechanisms underlying MOR desensitization, in adult mammalian neurons, caused by morphine (a partial agonist in this system) and DAMGO (a high-efficacy agonist). EXPERIMENTAL APPROACH: MOR function was measured in locus coeruleus neurons, by using whole-cell patch-clamp electrophysiology, in rat and mouse brain slices (both wild-type and protein kinase C (PKC)alpha knockout mice). Specific isoforms of PKC were inhibited by using inhibitors of the receptors for activated C-kinase (RACK), and in vivo viral-mediated gene-transfer was used to transfect neurons with dominant negative mutants (DNMs) of specific G-protein-coupled receptor kinases (GRKs). KEY RESULTS: Morphine-induced desensitization was attenuated by using RACK inhibitors that inhibit PKCalpha, but not by other isoform-specific inhibitors. Further, the PKC component of morphine-induced desensitization was absent in locus coeruleus neurons from PKCalpha knockout mice. The PKC-enhanced morphine-induced desensitization was not affected by over-expression of a GRK2 dominant negative mutant (GRK2 DNM). In contrast, DAMGO-induced MOR desensitization was independent of PKC activity but was reduced by over-expression of the GRK2 DNM but not by that of a GRK6 DNM. CONCLUSIONS AND IMPLICATIONS: In mature mammalian neurons, different MOR agonists can induce MOR desensitization by different mechanisms, morphine by a PKCalpha-mediated, heterologous mechanism and DAMGO by a GRK-mediated, homologous mechanism. These data represent functional selectivity at the level of receptor desensitization.


Subject(s)
Brain/enzymology , G-Protein-Coupled Receptor Kinase 2/physiology , Neurons/enzymology , Protein Kinase C-alpha/physiology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/physiology , Age Factors , Animals , Brain/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Female , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Male , Mice , Mice, Knockout , Neurons/drug effects , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar
7.
Eur J Neurosci ; 29(2): 307-18, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19200236

ABSTRACT

In morphine tolerance a key question that remains to be answered is whether mu-opioid receptor (MOPr) desensitization contributes to morphine tolerance, and if so by what cellular mechanisms. Here we demonstrate that MOPr desensitization can be observed in single rat brainstem locus coeruleus (LC) neurons following either prolonged (> 4 h) exposure to morphine in vitro or following treatment of animals with morphine in vivo for 3 days. Analysis of receptor function by an operational model indicated that with either treatment morphine could induce a profound degree (70-80%) of loss of receptor function. Ongoing PKC activity in the MOPr-expressing neurons themselves, primarily by PKCalpha, was required to maintain morphine-induced MOPr desensitization, because exposure to PKC inhibitors for only the last 30-50 min of exposure to morphine reduced the MOPr desensitization that was induced both in vitro and in vivo. The presence of morphine was also required for maintenance of desensitization, as washout of morphine for > 2 h reversed MOPr desensitization. MOPr desensitization was homologous, as there was no change in alpha(2)-adrenoceptor or ORL1 receptor function. These results demonstrate that prolonged morphine treatment induces extensive homologous desensitization of MOPrs in mature neurons, that this desensitization has a significant PKC-dependent component and that this desensitization underlies the maintenance of morphine tolerance.


Subject(s)
Drug Tolerance/physiology , Locus Coeruleus/drug effects , Morphine/pharmacology , Neurons/drug effects , Protein Kinase C/drug effects , Receptors, Opioid, mu/drug effects , Animals , Computer Simulation , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Male , Narcotics/pharmacology , Neurons/metabolism , Organ Culture Techniques , Protein Kinase C/metabolism , Rats , Rats, Wistar , Receptors, Opioid, mu/metabolism
8.
Br J Pharmacol ; 151(8): 1324-33, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17572699

ABSTRACT

BACKGROUND AND PURPOSE: Chronic morphine administration produces tolerance in vivo and attenuation of mu opioid receptor (MOR)-mediated G-protein activation measured in vitro, but the relationship between these adaptations is not clear. The present study examined MOR-mediated G-protein activation in the CNS of mice with different levels of morphine tolerance. EXPERIMENTAL APPROACH: Mice were implanted with morphine pellets, with or without supplemental morphine injections, to induce differing levels of tolerance as determined by a range of MOR-mediated behaviours. MOR function was measured using agonist-stimulated [(35)S]guanylyl-5'-O-(gamma-thio)-triphosphate ([(35)S]GTPgammaS) and receptor binding throughout the CNS. KEY RESULTS: Morphine pellet implantation produced 6-12-fold tolerance in antinociceptive assays, hypothermia and Straub tail, as measured by the ratio of morphine ED(50) values between morphine-treated and control groups. Pellet implantation plus supplemental injections produced 25-50-fold tolerance in these tests. In morphine pellet-implanted mice, MOR-stimulated [(35)S]GTPgammaS binding was significantly reduced only in the nucleus tractus solitarius (NTS) and spinal cord dorsal horn in tissue sections from morphine pellet-implanted mice. In contrast, MOR-stimulated [(35)S]GTPgammaS binding was significantly decreased in most regions examined in morphine pellet+morphine injected mice, including nucleus accumbens, caudate-putamen, periaqueductal gray, parabrachial nucleus, NTS and spinal cord. CONCLUSIONS AND IMPLICATIONS: Tolerance and the regional pattern of apparent MOR desensitization were influenced positively by the level of morphine exposure. These results indicate that desensitization of MOR-mediated G-protein activity is more regionally widespread upon induction of high levels of tolerance, suggesting that this response contributes more to high than low levels of tolerance to CNS-mediated effects of morphine.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance , GTP-Binding Proteins/metabolism , Morphine/pharmacology , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/administration & dosage , Animals , Binding Sites , Central Nervous System , Dose-Response Relationship, Drug , Guanosine 5'-O-(3-Thiotriphosphate) , Hypothermia/chemically induced , Male , Mice , Morphine/administration & dosage , Pain Measurement , Posterior Horn Cells , Solitary Nucleus , Tail/drug effects
9.
J Pharmacol Exp Ther ; 295(3): 1112-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11082447

ABSTRACT

Our laboratory demonstrated that morphine exhibits a modulatory control over the glyburide-binding site (sulfonylurea receptor) of the ATP-gated K(+) channel. This study evaluated the effect of chronic morphine administration on the sulfonylurea receptor during tolerance and physical dependence. ICR and Swiss-Webster mice were rendered tolerant to morphine by pellet implantation and were withdrawn by pellet removal. Alterations in the B(max) and K(D) were evaluated in mouse spinal cord using the radiolabeled ATP-gated K(+) channel blocker glyburide. The ED(50) for Swiss-Webster mice shifted from 13 to 451 mg/kg and thus they were more tolerant to morphine than ICR mice (ED(50) shift from 12 to 120 mg/kg). Swiss-Webster mice were also dependent to morphine only when the morphine pellet was in place, unlike ICR mice, which were dependent for 48 h after morphine pellet removal. Glyburide binding increased during chronic morphine treatment in Swiss-Webster mice by over 2-fold (from 294 to 635 fmol/mg of protein). This was not observed in ICR mice. In Swiss-Webster mice, chronic morphine treatment also significantly increased the K(D) by 3-fold (from 0.38 to 1.1 nM), whereas there was no change in affinity for ICR mice. Both strains of mice remained tolerant for 2 days after spontaneous withdrawal from morphine. However, the only increases in the B(max) and K(D) of glyburide were observed in Swiss-Webster mice that were highly tolerant to morphine. These results indicate that a high degree of tolerance is needed to alter ATP-gated potassium channels.


Subject(s)
ATP-Binding Cassette Transporters , Analgesics, Opioid/pharmacology , Glyburide/metabolism , Opioid-Related Disorders/etiology , Potassium Channels, Inwardly Rectifying , Animals , Drug Tolerance , Male , Methadone/pharmacology , Mice , Morphine/metabolism , Morphine/pharmacology , Motor Activity/drug effects , Potassium Channels/metabolism , Receptors, Drug/metabolism , Species Specificity , Sulfonylurea Receptors , Time Factors
10.
Br J Pharmacol ; 128(1): 220-6, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10498855

ABSTRACT

1. Opioid tolerance involves an alteration in the activity of intracellular kinases such as cyclic AMP-dependent protein kinase (PKA). Drugs that inhibit PKA reverse morphine antinociceptive tolerance. The hypothesis was tested that phospholipid pathways are also altered in morphine tolerance. Inhibitors of the phosphatidylinositol and phosphatidylcholine pathways were injected i.c.v. in an attempt to acutely reverse morphine antinociceptive tolerance. 2. Seventy-two hours after implantation of placebo or 75 mg morphine pellets, mice injected i.c.v. with inhibitor drug were challenged with morphine s.c. for generation of dose-response curves in the tail-flick test. Placebo pellet-implanted mice received doses of inhibitor drug having no effect on morphine's potency, in order to test for tolerance reversal in morphine pellet-implanted mice. Injection of the phosphatidylinositol-specific phospholipase C inhibitor ET-18-OCH3 significantly reversed tolerance, indicating a potential role for inositol 1,4,5-trisphosphate (IP3) and protein kinase C (PKC) in tolerance. Alternatively, phosphatidylcholine-specific phospholipase C increases the production of diacylglycerol and activation of PKC, without concomitant production of IP3. D609, an inhibitor of phosphatidylserine-specific phospholipase C, also reversed tolerance. Heparin is an IP3 receptor antagonist. Injection of low molecular weight heparin also reversed tolerance. PKC was also examined with three structurally dissimilar inhibitors. Bisindolylmaleimide I, Go-7874, and sangivamycin significantly reversed tolerance. 3. Chronic opioid exposure leads to changes in phospholipid metabolism that have a direct role in maintaining a state of tolerance. Evidence is accumulating that opioid tolerance disrupts the homeostatic balance of several important signal transduction pathways.


Subject(s)
Drug Tolerance , Morphine/pharmacology , Phosphatidylcholines/metabolism , Phosphatidylinositols/metabolism , Signal Transduction/drug effects , Analgesia , Animals , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/metabolism , Diglycerides/metabolism , Dose-Response Relationship, Drug , Heparin/chemistry , Heparin/pharmacology , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors , Male , Mice , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
11.
Pharmacol Biochem Behav ; 62(2): 381-8, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9972707

ABSTRACT

Opioid analgesic tolerance is associated with a disruption in Ca++ homeostasis. Drugs reducing Ca++ influx can prevent and reverse tolerance. The hypothesis was tested that both Ca++ influx and mobilization from intracellular pools maintains the expression of morphine tolerance. Ca++ modulating drugs were injected ICV at doses not affecting morphine's potency in placebo pellet-implanted mice, in order to determine whether tolerance would be reversed in morphine pellet-implanted mice. The Ca++ chelator EGTA significantly reversed tolerance. The Ca++ channel antagonists nifedipine and omega-conotoxin GVIA also reversed tolerance. The role of intracellular Ca++ was investigated using the membrane permeable intracellular Ca++ chelator EGTA-AM. EGTA-AM reversed tolerance at lower morphine doses, but not at higher morphine doses. Thus, mobilization of intracellular Ca++ contributes to the expression of tolerance. Finally, 1,4-dihydropyridine-sensitive Ca++ channels are known to stimulate Ca++-induced Ca++ release (CICR) from Ca++/caffeine-sensitive microsomal pools possessing ryanodine receptors. We examined whether blocking Ca++ mobilization from these pools with ryanodine would reverse morphine tolerance. Ryanodine's effects were similar to EGTA-AM. Tolerance was reversed at lower morphine doses, but not at higher doses. Thus, morphine tolerance appears to be associated with increases in Ca++ influx and mobilization from Ca++/caffeine-sensitive pools.


Subject(s)
Analgesics, Opioid/pharmacology , Calcium Channels/metabolism , Calcium/metabolism , Drug Tolerance/physiology , Morphine/pharmacology , Analgesia , Animals , Biological Transport , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Male , Mice , Microsomes/drug effects , Microsomes/metabolism
12.
Drug Alcohol Depend ; 45(1-2): 47-53, 1997 Apr 14.
Article in English | MEDLINE | ID: mdl-9179506

ABSTRACT

Modulation by opioids of ATP-gated potassium channels, which regulate in part intracellular calcium levels, was measured by the binding of [3H]glyburide. Scatchard analyses generated a KD for whole brain of vehicle-pretreated mice of 288 pM with a Bmax of 694 fmol/mg protein. In the spinal cord the KD was 0.94 nM and the Bmax was 184 fmol/mg protein. Acute morphine decreased the KD in brain and spinal cord with no change in Bmax. Morphine tolerance increased the KD in brain and spinal cord 2.6- and 2.9-fold, respectively, concurrent with 1.6- and 3.4-fold increases in Bmax. Modulation by morphine of glyburide-sensitive binding sites may contribute at least in part to tolerance to morphine via alterations in intracellular calcium levels in neurons.


Subject(s)
Brain/metabolism , Glyburide/metabolism , Hypoglycemic Agents/metabolism , Morphine/pharmacology , Narcotics/pharmacology , Spinal Cord/metabolism , Adenosine Triphosphate/physiology , Animals , Brain/drug effects , Calcium/metabolism , Drug Implants , Drug Tolerance , Injections, Subcutaneous , Ion Channel Gating/drug effects , Kinetics , Male , Mice , Morphine/administration & dosage , Narcotics/administration & dosage , Pain Measurement/drug effects , Potassium Channels/drug effects , Potassium Channels/metabolism , Spinal Cord/drug effects , Synaptosomes/drug effects , Synaptosomes/metabolism
14.
Pharmacol Biochem Behav ; 49(4): 1017-23, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7886070

ABSTRACT

Changes in the attitudes about neonatal pain and pain management have recently resulted in increases in the administration of opioids to neonates. Little is known, however, about the relative potencies of the various opioid agonists employed, especially in comparison to adult responses. The first objective in the present study was to compare the antinociceptive potency of four clinically relevant opioids in neonatal and adult rats. The second objective was to compare and contrast these agents in two different types of nociceptive tests: tonic (formalin-induced inflammation) and phasic (tail flick and hot plate). Our results indicate that the opioid agonists morphine, meperidine, and fentanyl, and the mixed agonist buprenorphine were all effective antinociceptive agents in both neonates and adults in each of the three tests employed, and that the relative potencies of these agents appeared to be similar in neonates and adults. In general, the pups were more sensitive to the antinociceptive agents when tested in the phasic nociceptive tests, and the drugs were more potent in the tonic test than either of the phasic tests.


Subject(s)
Aging/psychology , Analgesics/pharmacology , Narcotics/pharmacology , Pain Measurement/drug effects , Animals , Animals, Newborn , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Female , Formaldehyde , Hot Temperature , Male , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects
15.
Br J Sports Med ; 28(3): 164-6, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8000813

ABSTRACT

Previous research investigating the response of plasma beta-endorphins (beta-EP) to resistance exercise has resulted in equivocal findings. To examine further the effects of resistance exercise on beta-EP immunoreactivity, 10 male and 10 female college-age students participated in a series of controlled isotonic resistance exercises. The session consisted of three sets of eight repetitions at 80% of one repetition maximum (1-RM) for each of the following exercises: (1) bench press; (2) lateral pull-downs; (3) seated arm curls; and (4) military press. Blood plasma was sampled both before and after the lifting routine and beta-endorphin levels were determined by radioimmunoassay. A Students t test for paired samples indicated that mean(s.e.) plasma beta-endorphin levels after exercise (10.5(1.3) pg beta-EP ml-1) were significantly decreased as compared with pre-exercise (control) levels (16.5(1.2), P < 0.05). While the mechanism(s) contributing to the decrease in immunoreactivity is unclear, it may be the result of the synergistic effect of beta-EP clearance during rest intervals and changes in psychological states between sampling.


Subject(s)
Weight Lifting/physiology , beta-Endorphin/blood , Adult , Basketball/physiology , Basketball/psychology , Exercise/physiology , Exercise/psychology , Female , Humans , Isotonic Contraction/physiology , Male , Radioimmunoassay , Rest/physiology , Rest/psychology , Weight Lifting/psychology , beta-Endorphin/metabolism , beta-Lipotropin/blood , beta-Lipotropin/metabolism
16.
J Pharmacol Exp Ther ; 270(3): 1177-85, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7932169

ABSTRACT

In studying the interactions between handling mice and their subsequent analgesic response to an intrathecally (i.t.) administered mu-opioid agonist, DAMGO, it was found that suspending ICR mice by the tail for 1, 5, or 20 sec, 10 min before the tail-flick test, enhanced DAMGO by 5.3-, 7.4- and 23.6-fold, respectively, compared with mice maintained in a level posture. This enhancement was not accompanied by a change in the rostral flow of [3H]-DAMGO (25 ng, i.t.) to the brain (3.7% in 10 min), in its distribution along the neuraxis or in its systemic absorption. However, i.c.v. administration of beta-endorphin (1-27), an antagonist of epsilon opioid receptors, abolished the enhancement of i.t. DAMGO without affecting its basal analgesic potency. Pretreatment with the delta-opioid antagonist naltrindole (5.6 nmol, i.t.,-30 min) also blocked the enhancement of DAMGO without significantly affecting its basal analgesic potency. Alternatively, this same dose of naltrindole injected i.c.v. failed to block the enhancement of DAMGO in suspended mice. A 20-sec suspension failed to enhance i.t. kappa and delta-agonists, but it did enhance i.t. morphine. In mouse strain comparisons, i.t. DAMGO was more potent in C57BL/6J and DBA/2J mice than in C3H/HeJ and ICR mice, but DAMGO was enhanced by a 20-sec suspension in all strains tested. Thus suspending mice by the tail evoked a reflex enhancement of spinal mu agonist-induced analgesia that probably involved both the supraspinal release of beta-endorphin (an endogenous epsilon agonist) and the subsequent spinal release of an endogenous delta-receptor agonist in the reflex pathway.


Subject(s)
Analgesics/pharmacology , Enkephalins/pharmacology , Handling, Psychological , Opioid Peptides/physiology , Receptors, Opioid, mu/drug effects , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Male , Mice , Mice, Inbred Strains , Stimulation, Chemical , Tail
17.
Percept Mot Skills ; 77(3 Pt 1): 767-70, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8284151

ABSTRACT

Considerable research has shown significant increases in beta-endorphin levels after aerobic activity. These increases and their accompanying euphoric effect have been suggested as a possible psychophysiological mechanism underlying the exercise-dependence syndrome. The relationship between plasma beta-endorphin levels and a tendency towards exercise dependence, however, has not been established. To examine this relationship, 8 women trained in aerobic dance completed an exercise-dependence assessment prior to participation in a 45-min. session of continuous aerobic dance. Plasma beta-endorphin concentration was measured both prior to and following the aerobics routine. A Student t test for paired observations indicated that mean plasma beta-endorphin levels (+/- SE) were significantly higher after the aerobics routine (11.96 +/- 1.3 pg beta-EP.ml-1) than preexercise levels (8.62 +/- 1.4). However, beta-endorphin difference values (% change) were not significantly correlated with scores on the exercise-dependence survey. Those data suggest that scores on exercise dependence are not related to changes in plasma beta-endorphin levels after aerobic exercise.


Subject(s)
Behavior, Addictive/blood , Exercise/physiology , Physical Endurance/physiology , beta-Endorphin/blood , Adult , Behavior, Addictive/psychology , Exercise/psychology , Female , Humans , Physical Fitness/physiology , Physical Fitness/psychology
18.
J Sports Sci ; 11(6): 499-52, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8114174

ABSTRACT

Previous research investigating the response of plasma beta-endorphins (beta-EP) to resistance exercise has resulted in equivocal findings. To further examine the effects of resistance exercise on beta-endorphin immunoreactivity, six resistance-trained athletes participated in a three-set series of eight repetitions of isotonic exercise. All exercises were performed at 80% maximal effort. Blood was sampled from the group by venepuncture, both prior to and following the exercise bout, and beta-endorphin concentration was determined by radioimmunoassay. The results indicated that mean (+/- S.E.) plasma levels of beta-endorphins following exercise (18.04 +/- 3.4 pg beta-EP ml-1) were not significantly changed from pre-exercise (control) levels (19.59 +/- 2.4 pg beta-EP ml-1), although there was considerable inter-individual variability. Our results support previous research which has reported no significant changes in beta-endorphin immunoreactivity following resistance exercise, as well as reported findings of considerable variability in the beta-endorphin response to exercise.


Subject(s)
Exercise/physiology , Weight Lifting/physiology , beta-Endorphin/blood , Adult , Humans , Male , Radioimmunoassay
19.
Arukoru Kenkyuto Yakubutsu Ison ; 28(5): 379-93, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8267521

ABSTRACT

The cisterna magna of dogs anesthetized with sodium Surital was fitted with a cannula, and cerebrospinal fluid (CSF) was withdrawn before (control) and one hour after the s.c. injection of 10 mg/kg of morphine sulfate (morphine). The CSF from control and morphine-treated dogs was purified initially by gel filtration. Each fraction was submitted to opiate bioassay procedures, followed by high performance liquid chromatography (HPLC) purification on a mu-Bondapak C18 column. Two of the CSF fractions from HPLC purification showed greater opiate-like activity after morphine treatment than that in controls. One fraction contained morphine, the other an unknown peptide. This latter fraction produced a dose-dependent effect in the mouse tail-flick test. This fraction did not show radioimmunoreactivity to methionine (met)- or leucine (leu)-enkephalins, but showed a small amount of reactivity to beta-endorphin and dynorphin (1-13). Further purification of this fraction by HPLC yielded a fraction with five peaks, which upon amino acid analysis were found to contain small peptides. Met- and Leu-enkephalins, beta-endorphin and dynorphin (1-13)-like immunoreactivity in the fraction in which the respective standard was eluted by HPLC was significantly increased after a single administration of morphine. Based on these results, it is suggested that morphine at an antinociceptive dose causes the release of endogenous opioid peptides and may also stimulate the biosynthesis of their precursor molecules, pre-pro-opiomelanocortin, pre-pro-enkephalin A and pre-pro-enkephalin B.


Subject(s)
Endorphins/cerebrospinal fluid , Morphine/pharmacology , Amino Acids/analysis , Animals , Chromatography, High Pressure Liquid , Dogs , Dose-Response Relationship, Drug , Enkephalins/biosynthesis , Female , Guinea Pigs , Injections, Subcutaneous , Male , Mice , Morphine/administration & dosage , Protein Precursors/biosynthesis
20.
J Pharmacol Exp Ther ; 266(3): 1407-15, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8371146

ABSTRACT

To the authors' knowledge, the effect of i.t. administered calcium on thermoregulation in mice has not been investigated. Calcium administration (i.t.) induced hypothermia in mice. It was found that calcitonin gene-related peptide (CGRP) (i.t.) also produced hypothermia. Because opioids have well documented thermoregulatory effects, the authors evaluated whether the hypothermia induced by calcium and CGRP was the result of the release of opioids. Calcium induced hypothermia at different ambient temperatures (4 degrees C, 22 degrees C and 30 degrees C) in intact mice. Similarly treated spinalized mice maintained body temperature. Using laser Doppler flowmetry, there was a significant increase in blood flow in the tails of calcium-injected mice vs. those of vehicle-injected mice. Both naloxone and naltrindole failed to block the hypothermic effects of calcium (i.t.). Nor-binaltorphimine (i.t.) significantly blocked calcium (i.t.)-induced changes in body temperature. CGRP (i.t.) produced hypothermia for 15 hr postinjection, with the maximum decrease at 3 hr. CGRP induced hypothermia in intact and sham-lesioned mice but not in spinalized mice. CGRP (i.c.v.) also produced hypothermia (onset, 15-min postinjection) followed by the peak effect at 1 hr with recovery to baseline temperature by 2 hr. Subthreshold doses of calcium and CGRP given in combination produced greater than additive hypothermia. The hypothermic effects of CGRP were reversed by naloxone, naltrindole and nor-binaltorphimine. CGRP produced significant hypothermia in both morphine-tolerant and nontolerant mice. Chronic administration of CGRP in nontolerant and morphine-tolerant mice did not alter hypothermia after pretreatment with CGRP (i.t.).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Body Temperature Regulation/physiology , Calcitonin Gene-Related Peptide/pharmacology , Calcium/pharmacology , Endorphins/physiology , Hypothermia, Induced , Animals , Body Temperature Regulation/drug effects , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcium/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Injections, Spinal , Male , Mice , Mice, Inbred ICR , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...