Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
J Neuroimmune Pharmacol ; 7(2): 444-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22205542

ABSTRACT

Opioid abuse and dependency are international problems. Studies have shown that neuronal inflammation and degeneration might be related to the development of opioid addiction. Thus, using neuroprotective agents might be beneficial for treating opioid addiction. Memantine, an Alzheimer's disease medication, has neuroprotective effects in vitro and in vivo. In this study, we evaluated whether a low dose of memantine prevents opioid-induced drug-seeking behavior in rats and analyzed its mechanism. A conditioned-place-preference test was used to investigate the morphine-induced drug-seeking behaviors in rats. We found that a low-dose (0.2-1 mg/kg) of subcutaneous memantine significantly attenuated the chronic morphine-induced place-preference in rats. To clarify the effects of chronic morphine and low-dose memantine, serum and brain levels of cytokines and brain-derived neurotrophic factor (BDNF) were measured. After 6 days of morphine treatment, cytokine (IL-1ß, IL-6) levels had significantly increased in serum; IL-1ß and IL-6 mRNA levels had significantly increased in the nucleus accumbens and medial prefrontal cortex, both addiction-related brain areas; and BDNF levels had significantly decreased, both in serum and in addiction-related brain areas. Pretreatment with low-dose memantine significantly attenuated chronic morphine-induced increases in serum and brain cytokines. Low-dose memantine also significantly potentiated serum and brain BDNF levels. We hypothesize that neuronal inflammation and BDNF downregulation are related to the progression of opioid addiction. We hypothesize that the mechanism low-dose memantine uses to attenuate morphine-induced addiction behavior is its anti-inflammatory and neurotrophic effects.


Subject(s)
Drug-Seeking Behavior/drug effects , Memantine/pharmacology , Morphine Dependence , Neuroprotective Agents/pharmacology , Analgesics, Opioid/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/analysis , Enzyme-Linked Immunosorbent Assay , Interleukin-1beta/analysis , Interleukin-6/analysis , Male , Morphine/pharmacology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
2.
Life Sci ; 89(23-24): 875-7, 2011 Dec 05.
Article in English | MEDLINE | ID: mdl-21989208

ABSTRACT

AIMS: We have previously demonstrated that pretreatment with (+)-morphine given intrathecally attenuates the intrathecal (-)-morphine-produced tail-flick inhibition. The phenomenon has been defined as antianalgesia against (-)-morphine-produced analgesia. Present experiments were then undertaken to determine if the antianalgesic effect induced by (+)-morphine given spinally is mediated by the stimulation of the sigma-1 receptor in the mouse spinal cord. MAIN METHODS: Sigma-1 receptor ligands, N-[2-(3,4-Dichlorophenyl)ethyl]-N-methyl-2-(dimethylamino)ethylamine dihydrobromide (BD1047) and (+)-pentazocine were used to determine if (+)-morphine-induced antianalgesia is mediated by the stimulation of sigma-1 receptors in the mouse spinal cord. Tail-flick test was employed to measure the nociceptive response. All compounds were given intrathecally. KEY FINDINGS: Pretreatment with BD1047 (1-10 µg) or (+)-pentazocine (0.1-10 µg) dose-dependently reversed the attenuation of the (-)-morphine-produced tail-flick inhibition induced by (+)-morphine (10 pg). BD1047 and (+)-pentazocine injected alone did not affect (-)-morphine-produced tail-flick inhibition. SIGNIFICANCE: The finding indicates that (+)-morphine attenuates the (-)-morphine-produced tail-flick inhibition via the activation of the sigma-1 receptors in the mouse spinal cord. Sigma-1 receptors may play an important role in opioid analgesia in the mouse spinal cord.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Pain Measurement/drug effects , Receptors, sigma/drug effects , Spinal Cord/drug effects , Animals , Ethylenediamines/pharmacology , Male , Mice , Morphine/antagonists & inhibitors , Narcotic Antagonists/pharmacology , Nociception/drug effects , Nociception/physiology , Pentazocine/pharmacology , Reaction Time/drug effects , Receptors, sigma/physiology , Spinal Cord/physiology , Tail , Sigma-1 Receptor
3.
J Pharmacol Exp Ther ; 326(2): 614-22, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18492947

ABSTRACT

Cytochrome P450 genes catalyze formation of epoxyeicosatrienoic acids (EETs) from arachidonic acid. The effects of 5,6-EET, 8,9-EET, 11,12-EET, and 14,15-EET microinjected into the ventrolateral periaqueductal gray (vlPAG) on the thermally produced tail-flick response were studied in male Sprague-Dawley rats. 14,15-EET microinjected into vlPAG (3-156 pmol) dose-dependently inhibited the tail-flick response (ED50 = 32.5 pmol). In contrast, 5,6-EET, 8,9-EET, and 11,12-EET at a dose of 156 pmol were not active when injected into the vlPAG. 14,15-EET failed to displace the radiobinding of [3H][D-Ala2,NHPe4, Gly-ol5]enkephalin (mu-opioid receptor ligand) or [3H]naltrindole (delta-opioid receptor ligand) in crude membrane fractions of rat brain. Tail-flick inhibition produced by 14,15-EET from vlPAG was blocked by intra-vlPAG pretreatment with antiserum against beta-endorphin or Met-enkephalin or the mu-opioid receptor antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP) or the delta-opioid receptor antagonist naltrindole but not with dynorphin A[1-17] antiserum or the kappa-opioid receptor antagonist nor-binaltorphimine. In addition, tail-flick inhibition produced by 14,15-EET treatment was blocked by intrathecal pretreatment with Met-enkephalin antiserum, naltrindole, or CTOP but not with beta-endorphin antiserum. It is concluded that 1) 14,15-EET itself does not have any affinity for mu- or delta-opioid receptors and 2) 14,15-EET activates beta-endorphin and Met-enkephalin, which subsequently act on mu- and delta-opioid receptors to produce antinociception.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Analgesics/pharmacology , Enkephalin, Methionine/metabolism , Periaqueductal Gray/drug effects , beta-Endorphin/metabolism , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Dose-Response Relationship, Drug , Male , Microinjections , Pain Measurement/drug effects , Periaqueductal Gray/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects
4.
Eur J Pharmacol ; 587(1-3): 147-54, 2008 Jun 10.
Article in English | MEDLINE | ID: mdl-18448094

ABSTRACT

An unbiased conditioned place preference paradigm and the microdialysis technique was used to evaluate the effect of (+)-morphine pretreatment on the conditioned place preference produced by (-)-morphine and the increased release of the dopamine produced by mu-opioid ligand endomorphin-1, respectively, in the posterior nucleus accumbens shell of the male CD rat. (-)-Morphine (2.5-10 microg) microinjected into the posterior nucleus accumbens shell dose-dependently produced the conditioned place preference. Pretreatment with (+)-morphine (0.1-10 pg) given into the posterior accumbens shell for 45 min dose-dependently attenuated the conditioned place preference produced by (-)-morphine (5 microg) given into the same posterior accumbens shell. However, higher doses of (+)-morphine (0.1 and 1 ng) were less effective in attenuating the (-)-morphine-produced conditioned place preference. Thus, like given systemically, (+)-morphine given into the posterior nucleus accumbens shell also induces a U-shaped dose-response curve for attenuating the (-)-morphine-produced conditioned place preference. Microinjection of mu-opioid agonist endomorphin-1 (1-10 microg) given into the ventral tegmental area dose-dependently increased the release of the extracellular dopamine in the posterior nucleus accumbens shell in the urethane-anesthetized rats. The increased dopamine caused by endomorphin-1 (10 microg) was completed blocked by the (+)-morphine (10 pg) pretreatment given into ventral tegmental area. It is concluded that (+)-morphine attenuates the (-)-morphine-produced conditioned place preference and the mu-opioid receptor-mediated increase of extracellular dopamine in the posterior nucleus accumbens shell of the rat.


Subject(s)
Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Conditioning, Operant/drug effects , Dopamine/metabolism , Morphine/antagonists & inhibitors , Morphine/pharmacology , Nucleus Accumbens/metabolism , Receptors, Opioid, mu/drug effects , Analgesics, Opioid/chemistry , Anesthesia , Animals , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Electrochemistry , Male , Microinjections , Morphine/chemistry , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/anatomy & histology , Nucleus Accumbens/drug effects , Oligopeptides/administration & dosage , Oligopeptides/pharmacology , Rats , Stereoisomerism , Ventral Tegmental Area
5.
Eur J Pharmacol ; 571(2-3): 145-51, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17617400

ABSTRACT

We have previously demonstrated that (+)-morphine and (-)-morphine given spinally stereoselectively attenuate the spinally-administered (-)-morphine-produced tail-flick inhibition in the mouse. The phenomenon has been defined as antianalgesia. Present studies were then undertaken to determine if the systemic administration of (+)-morphine and (-)-morphine also stereoselectively attenuates the systemic (-)-morphine-produced tail-flick inhibition and the effects of (+)-morphine and (-)-morphine are mediated by the naloxone-sensitive sigma receptor activation in male CD-1 mice. Pretreatment with (+)-morphine at a dose of 0.01-10 ng/kg given subcutaneously dose-dependently attenuated the tail-flick inhibition produced by subcutaneously-administered (-)-morphine (5 mg/kg). Pretreatment with (-)-morphine (0.01-1.0 mg/kg) given subcutaneously also attenuates the (-)-morphine-produced tail-flick inhibition. The ED50 values for (+)-morphine and (-)-morphine for inhibiting the (-)-morphine-produced tail-flick inhibition were estimated to be 30.6 pg/kg and 97.5 microg/kg, respectively. The attenuation of the (-)-morphine-produced tail-flick inhibition induced by (+)-morphine or (-)-morphine pretreatment was reversed by the pretreatment with (+)-naloxone or by the sigma receptor antagonist BD1047 (N-[2-(3,4-dichlorophenyl)ethyl]-N-methyl-2-(dimethylamino)ethylamine dihydrobromide) given subcutaneously. Pretreatment with (+)-pentazocine, a selective sigma receptor agonist, (1-10 mg/kg) given subcutaneously also attenuates (-)-morphine-produced tail-flick inhibition, which was restored by (+)-naloxone (4 mg/kg) or BD1047 (10 mg/kg) pretreated subcutaneously. It is concluded that (+)-morphine exhibits extremely high stereoselective action over (-)-morphine given systemically in attenuating the systemic (-)-morphine-produced antinociception and the antianalgesic effect of (+)-morphine and (-)-morphine is mediated by activation of the naloxone-sensitive sigma receptor.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Pain Threshold/drug effects , Pain/prevention & control , Receptors, sigma/drug effects , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemistry , Analgesics, Opioid/therapeutic use , Animals , Dose-Response Relationship, Drug , Ethylenediamines/pharmacology , Injections, Subcutaneous , Male , Mice , Morphine/administration & dosage , Morphine/chemistry , Morphine/therapeutic use , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pain/metabolism , Pain/physiopathology , Pain Measurement , Pentazocine/pharmacology , Receptors, sigma/metabolism , Stereoisomerism
6.
Eur J Pharmacol ; 571(1): 1-7, 2007 Sep 24.
Article in English | MEDLINE | ID: mdl-17597599

ABSTRACT

We have previously demonstrated that (+)-morphine and (-)-morphine pretreated spinally for 45 min stereoselectively attenuates the tail-flick inhibition produced by (-)-morphine given spinally in the mouse. The present study is then undertaken to determine if the same phenomenon observed in the mouse spinal cord can also take place in the ventral periaqueductal gray of the rat. Pretreatment with (+)-morphine for 45 min at 0.3 to 3.3 fmol dose-dependently attenuated the tail-flick inhibition produced by (-)-morphine (9 nmol) given into the ventral periaqueductal gray. Likewise, pretreatment with (-)-morphine for 45 min at a higher dose (3-900 pmol), which given alone did not affect the baseline tail-flick latency, also dose-dependently attenuated the tail-flick inhibition produced by (-)-morphine. Thus, (+)-morphine is approximately 270,000-fold more potent than (-)-morphine in attenuating the (-)-morphine-produced tail-flick inhibition. The attenuation of the (-)-morphine-produced tail-flick inhibition induced by (+)-morphine or (-)-morphine was dose-dependently reversed by (+)-naloxone (27.5 to 110 pmol) pretreatment for 50 min given into the ventral periaqueductal gray. Pretreatment with the sigma receptor antagonist BD1047 (N-[2-(3,4-dichlorophenyl)ethyl]-N-methyl-2-(dimethylamino)ethylamine dihydrobromide) (11-45 nmol) for 45 min given into the ventral periaqueductal gray also reversed dose-dependently the attenuation of the (-)-morphine-produced tail-flick inhibition induced by (+)-morphine or (-)-morphine, indicating that the effects are mediated by the activation of the sigma receptors. Since (+)-morphine, (-)-morphine and (+)-naloxone do not have any affinity for the naloxone-inaccessible sigma receptors, we therefore propose that (+)-morphine and (-)-morphine attenuate the (-)-morphine-produced tail-flick inhibition via the activation of the naloxone-sensitive sigma receptor originally proposed by Tsao and Su [Tsao, L.T., Su, T.P., 1997. Naloxone-sensitive, haloperidol-sensitive, [(3)H](+)-SKF-1047-binding protein partially purified from rat liver and rat brain membranes: an opioid/sigma receptor. Synapse 25, 117-124].


Subject(s)
Morphine/pharmacology , Naloxone/pharmacology , Pain/prevention & control , Periaqueductal Gray/drug effects , Receptors, sigma/physiology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , Dose-Response Relationship, Drug , Ethylenediamines/administration & dosage , Ethylenediamines/pharmacology , Male , Microinjections , Models, Anatomic , Morphine/administration & dosage , Morphine/chemistry , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Pain/physiopathology , Pain Measurement/drug effects , Pain Measurement/methods , Periaqueductal Gray/anatomy & histology , Periaqueductal Gray/physiology , Rats , Receptors, sigma/antagonists & inhibitors , Stereoisomerism , Tail/physiopathology , Time Factors
7.
J Pharmacol Exp Ther ; 322(2): 813-21, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17494863

ABSTRACT

This study investigated three possible mechanisms by which the antinociceptive effects of the mu-opioid receptor (MOR) agonist [d-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO) and the delta-opioid receptor (DOR) agonist [d-Ala(2),Glu(4)]-deltorphin (deltorphin II) (DELT), microinjected into the rostral ventromedial medulla (RVM), are enhanced in rats with persistent inflammatory injury. Radioligand binding determined that neither the B(max) nor the K(d) values of [(3)H]DAMGO differed in RVM membranes from rats that received an intraplantar injection of saline or complete Freund's adjuvant (CFA) in one hindpaw 4 h, 4 days, or 2 weeks earlier. Likewise, neither the EC(50) nor the E(max) value for DAMGO-induced stimulation of guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS) binding differed in the RVM of saline- or CFA-treated rats at any time point. Microinjection of fixed dose combinations of DAMGO and DELT in the RVM of naive rats indicated that these agonists interact synergistically to produce antinociception when DAMGO is present in equal or greater amounts than DELT and, additively, when DELT is the predominant component. Thus, unlike the periphery or spinal cord, potentiation of MOR-mediated antinociception does not entail an increase in MOR number, affinity, or coupling. Rather, the data are concordant with our proposal that potentiation results from a synergistic interaction of exogenous MOR agonist with DOR-preferring enkephalins whose levels are increased in CFA-treated rats (J Neurosci 21:2536-2545, 2001). Virtually no specific [(3)H]DELT binding nor stimulation of [(35)S]GTPgammaS binding by DELT was obtained in RVM membranes from CFA- or saline-treated rats at any time point. The mechanisms responsible for the potentiation of DELT-mediated antinociception remain to be elucidated.


Subject(s)
Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Medulla Oblongata/drug effects , Pain/prevention & control , Receptors, Opioid, mu/agonists , Analgesics/metabolism , Analgesics/pharmacology , Animals , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Freund's Adjuvant/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hindlimb , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/prevention & control , Inflammation/chemically induced , Inflammation/complications , Inflammation/pathology , Male , Medulla Oblongata/chemistry , Medulla Oblongata/metabolism , Oligopeptides/pharmacology , Pain/etiology , Pain/metabolism , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/analysis , Receptors, Opioid, mu/metabolism
8.
Eur J Pharmacol ; 562(3): 221-6, 2007 May 21.
Article in English | MEDLINE | ID: mdl-17335800

ABSTRACT

An unbiased conditioned place preference paradigm was used to evaluate the effect of dextro-morphine on the morphine-produced reward in male CD rats. Morphine sulfate (1-10 mg/kg) given intraperitoneally dose-dependently produced the conditioned place preference. Pretreatment with dextro-morphine at a dose from 0.1 to 3 microg/kg given subcutaneously dose-dependently attenuated the morphine-produced conditioned place preference. However, dextro-morphine at a higher dose 100 microg/kg did not affect the morphine-produced conditioned place preference. Thus, dextro-morphine pretreatment induces a U-shaped dose-response curve for attenuating the morphine-produced conditioned place preference. The attenuation of the morphine-produced conditioned place preference was reversed by the pretreatment with the sigma(1) receptor antagonist BD1047 (N-[2-(3,4-Dichlorophenyl)ethyl]-N-methyl-2-(dimethylamino)ethylamine dihydrobromide. dextro-Morphine or BD1047 given alone did not affect the baseline place conditioning. It is concluded that dextro-morphine attenuated the morphine-produced conditioned place preference via the sigma(1) receptor activation.


Subject(s)
Analgesics, Opioid/pharmacology , Conditioning, Operant/drug effects , Morphine/pharmacology , Receptors, sigma/drug effects , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Ethylenediamines , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Rats , Receptors, sigma/metabolism , Reward , Stereoisomerism , Sigma-1 Receptor
9.
Eur J Pharmacol ; 554(2-3): 137-44, 2007 Jan 12.
Article in English | MEDLINE | ID: mdl-17112504

ABSTRACT

The effects of endomorphin-2 or endomorphin-1 microinjected into the centromedial amygdala on the thermally-induced tail-flick response were studied in male CD rats. Microinjection of endomorphin-2 (8.7-35.0 nmol) given into the centromedial amygdala time- and dose-dependently decreased the tail-flick latencies. On the other hand, endomorphin-1 (8-32.6 nmol) given into the same site did not cause any change of the tail-flick latency. However, endomorphin-1 (32.6 nmol) or endomorphin-2 (35.0 nmol) given into the basolateral site of amygdala did not affect the tail-flick latency. Pretreatment with the antiserum against dynorphin A(1-17) (200 microg) significantly reversed the decrease of the tail-flick latency induced by endomorphin-2. The decrease of the tail-flick latency induced by endomorphin-2 was also blocked by the endomorphin-2 selective micro-opioid receptor antagonist 3-methoxynaltrexone (6.4 pmol) and by the N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 (30 nmol), but not by the kappa-opioid receptor antagonist nor-binaltorphimine (6.6 nmol). It is concluded that endomorphin-2, but not endomorphin-1, given into the centromedial amygdala stimulates a 3-methoxynaltrexone-sensitive mu-opioid receptor subtype to induce the release of dynorphin A(1-17), which then acts on the NMDA receptor, but not kappa-opioid receptor for producing hyperalgesia. This conclusion is further supported by the additional findings that dynorphin A(1-17) (2.3 nmol) given into the centromedial amygdala also caused the decrease of the tail-flick latency, which was similarly blocked by the NMDA receptor antagonist MK-801 (30 nmol), but not kappa-opioid receptor antagonist nor-binaltorphimine (6.6 nmol).


Subject(s)
Amygdala/drug effects , Hyperalgesia/physiopathology , Oligopeptides/administration & dosage , Receptors, Opioid, mu/agonists , Amygdala/pathology , Amygdala/physiopathology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/toxicity , Animals , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Dynorphins/administration & dosage , Dynorphins/immunology , Dynorphins/pharmacology , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/prevention & control , Immune Sera/pharmacology , Male , Microinjections , Naltrexone/administration & dosage , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Oligopeptides/toxicity , Pain Measurement/methods , Rabbits , Rats , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/physiology , Time Factors
10.
Eur J Neurosci ; 24(9): 2575-80, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17100845

ABSTRACT

Glial stimulation by intrathecal injection of lipopolysaccharide (LPS) attenuated the tail-flick inhibition produced by morphine given intrathecally in the spinal cord of the male CD-1 mice. The phenomenon has been defined as antianalgesia. The effects of dextro-naloxone or levo-naloxone on the attenuation of morphine-produced tail-flick inhibition induced by LPS were then studied. Pretreatment with dextro-naloxone or levo-naloxone reversed the attenuation of the morphine-produced tail-flick inhibition induced by LPS. Pretreatment with dextro-naloxone or levo-naloxone alone did not affect the morphine-produced tail-flick inhibition. It is concluded that dextro-naloxone and levo-naloxone block the LPS-induced antianalgesia against morphine antinociception via a non-opioid mechanism.


Subject(s)
Analgesics, Opioid/administration & dosage , Lipopolysaccharides/administration & dosage , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Pain/physiopathology , Spinal Cord/drug effects , Animals , Injections, Spinal , Male , Mice , Morphine/administration & dosage , Neuroglia/drug effects , Neuroglia/metabolism , Pain/chemically induced , Spinal Cord/metabolism , Stereoisomerism
11.
Eur J Pharmacol ; 550(1-3): 91-4, 2006 Nov 21.
Article in English | MEDLINE | ID: mdl-17026985

ABSTRACT

We have previously demonstrated that intrathecal pretreatment with dextro-morphine or morphine attenuates the morphine-produced antinociception. The phenomenon has been defined as antianalgesia, which is mediated by a non-opioid receptor [Wu, H., Thompson, J., Sun, H., Terashvili, M., Tseng, L.F., 2005. Antianalgesia: stereo-selective action of dextro-morphine over levo-morphine on glia in the mouse spinal cord. J. Pharmacol. Exp. Ther. 314, 1101-1108]. To determine if p38 mitogen-activated protein kinase (MAPK) is involved in the antianalgesia, the effects of p38 MAPK inhibitor 4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)-1H-imidazole (SB203580) on the attenuation of the morphine-produced tail-flick inhibition induced by dextro-morphine or morphine were studied in male CD-1 mice. Intrathecal pretreatment with SB203580 (24.2 nmol) reversed the attenuation of the morphine-produced tail-flick inhibition induced by dextro-morphine (33 fmol) or morphine (0.3 nmol) pretreatment. The finding indicates that the antianalgesia induced by dextro-morphine or morphine is mediated by the activation of p38 MAPK in the mouse spinal cord.


Subject(s)
Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Morphine/antagonists & inhibitors , Morphine/pharmacology , Pyridines/pharmacology , Spinal Cord/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Analgesics, Opioid/chemistry , Animals , Male , Mice , Morphine/chemistry , Pain Measurement/drug effects , Reaction Time/drug effects , Stereoisomerism
12.
Eur J Pharmacol ; 531(1-3): 103-7, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16445907

ABSTRACT

We have demonstrated that the antianalgesia induced by dextro-morphine and levo-morphine is not mediated by the stimulation of mu-opioid receptors in male CD-1 mice. We now report that the dextro-morphine and levo-morphine attenuated antinociception produced by delta-opioid receptor agonist deltorphin II and kappa-opioid receptor agonist U50,488H given spinally in the male mu-opioid receptor knockout mice. The tail-flick response was used for the antinociceptive test. Intrathecal injection of levo-morphine (3 nmol) markedly inhibited the tail-flick response in wild type, partially in heterozygous, but not in homozygous mu-opioid receptor knockout mice. Intrathecal pretreatment with dextro-morphine (33 fmol) or levo-morphine (0.3 nmol) for 45 min also attenuated levo-morphine-produced antinociception in wide type mice. Intrathecal pretreatment with dextro-morphine (33 fmol) or levo-morphine (0.3 nmol) for 45 min attenuated the tail-flick inhibition produced by deltorphin II (12.8 nmol) and U50,488H (123.3 nmol) in wide type, heterozygous and homozygous mu-opioid receptor knockout mice. The findings provide additional evidence that mu-opioid receptors are not involved in the antianalgesia induced by dextro-morphine and levo-morphine.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics/pharmacology , Morphine/pharmacology , Oligopeptides/pharmacology , Receptors, Opioid, mu/genetics , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage , Analgesics/administration & dosage , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , Female , Genotype , Injections, Spinal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/administration & dosage , Morphine/chemistry , Nociceptors/drug effects , Nociceptors/physiopathology , Oligopeptides/administration & dosage , Pain/physiopathology , Pain/prevention & control , Pain Measurement/methods , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/physiology , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/physiology , Receptors, Opioid, mu/physiology , Stereoisomerism
13.
Neurosci Lett ; 386(1): 46-51, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-15982809

ABSTRACT

It is generally accepted that neuropathic pain is resistant to amelioration by morphine in clinical studies and insensitivity to intrathecal (i.t.) administered morphine in experimental models of neuropathic pain has been demonstrated. This study is to determine if endogenous dynorphin, substance P or cholecystokinin is involved in the lack of anti-allodynia of morphine in a partial sciatic nerve ligation (PSL) model of CD-1 mice. Mice exhibited tactile allodynia in the ipsilateral hind paw 1 day after PSL, and reached its maximal allodynic effect at 2 days and remained allodynic for 7 days. Morphine (3.0 nmol) given i.t. did not alter the tactile allodynic threshold in ipsilateral paw of mice pretreated i.t. with normal rabbit serum 2 days after PSL. However, the same dose of morphine (3.0 nmol) given i.t. reduced markedly allodynia in mice pretreated for 2h with antiserum against dynorphin A(1-17) (200 microg); the morphine-produced anti-allodynia developed slowly, reached its peak effect at 30 min and returned to an allodynic state in 60 min. Similarly, i.t. injection of morphine reduced the allodynia in PSL mice pretreated with antiserum against substance P (10 microg) or cholecystokinin (200 microg) for 2h. Intrathecal pretreatment with antiserum against dynorphin A(1-17), substance P or cholecystokinin for 2h injected alone did not affect the baseline mechanical tactile threshold in ipsilateral paw 2 days after PSL. The results indicate that endogenous dynorphin A(1-17), substance P and cholecystokinin are involved in PSL-induced neuropathic allodynia to attenuate the anti-allodynic effect of morphine.


Subject(s)
Cholecystokinin/antagonists & inhibitors , Dynorphins/antagonists & inhibitors , Immune Sera/pharmacology , Morphine/agonists , Neuralgia/drug therapy , Peripheral Nervous System Diseases/drug therapy , Substance P/antagonists & inhibitors , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/agonists , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , Cholecystokinin/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistance/drug effects , Drug Resistance/physiology , Drug Synergism , Dynorphins/metabolism , Injections, Spinal , Ligation , Male , Mice , Morphine/administration & dosage , Neuralgia/metabolism , Neuralgia/physiopathology , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/physiopathology , Sciatic Nerve/injuries , Sciatic Nerve/surgery , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Substance P/metabolism
14.
J Pharmacol Exp Ther ; 314(3): 1101-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15901793

ABSTRACT

We have previously shown that the naturally occurring levo-morphine at a subanalgesic picomolar dose pretreated i.t. induces antianalgesia against levo-morphine-produced antinociception. We now report that the synthetic stereo-enantiomer dextro-morphine, even at an extremely low femtomolar dose, induces antianalgesia against levo-morphine-produced antinociception using the tail-flick (TF) test in male CD-1 mice. Intrathecal pretreatment with dextro-morphine (33 fmol) time-dependently attenuated the i.t. levo-morphine-produced TF inhibition for 4 h and returned to the preinjection control level at 24 h. Intrathecal pretreatment with dextro-morphine (0.3-33 fmol), which injected alone did not affect the baseline TF latency, dose-dependently attenuated the TF inhibition produced by i.t.-administered levo-morphine (3.0 nmol). The ED(50) value for dextro-morphine to induce antianalgesia was estimated to be 1.07 fmol, which is 71,000-fold more potent than the ED(50) value of levo-morphine, indicating the high stereoselective action of dextro-morphine over levo-morphine for the induction of antianalgesia. Like levo-morphine, the dextro-morphine-induced antianalgesia against levo-morphine-produced TF inhibition was dose-dependently blocked by the nonopioid dextro-naloxone and its stereo-enantiomer levo-naloxone, a nonselective mu-opioid receptor antagonist. The antianalgesia induced by levo-morphine and dextro-morphine is reversed by the pretreatment with the glial inhibitor propentofylline (3.3-65 nmol), indicating that the antianalgesia is mediated by glial stimulation. The findings strongly indicate that the antianalgesia induced by levo-morphine and dextro-morphine is mediated by the stimulation of a novel nonopioid receptor on glial cells.


Subject(s)
Analgesia , Morphine/antagonists & inhibitors , Morphine/pharmacology , Neuroglia/drug effects , Spinal Cord/drug effects , Animals , Dose-Response Relationship, Drug , Injections, Spinal , Male , Mice , Naloxone/pharmacology , Stereoisomerism , Xanthines/pharmacology
15.
J Pharmacol Exp Ther ; 312(3): 1257-65, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15542622

ABSTRACT

The effects of pretreatment with endomorphin-1 (EM-1) and endomorphin-2 (EM-2) given into the ventral periaqueductal gray (vPAG) to induce antianalgesia against the tail-flick (TF) inhibition produced by morphine given into the vPAG were studied in rats. Pretreatment with EM-1 (3.5-28 nmol) given into vPAG for 45 min dose-dependently attenuated the TF inhibition produced by morphine (9 nmol) given into vPAG. Similarly, pretreatment with EM-2 (1.7-7.0 nmol) for 45 min also attenuated the TF inhibition induced by morphine; however, a high dose of EM-2 (14 nmol) did not attenuate the morphine-produced TF inhibition. The attenuation of morphine-produced TF inhibition induced by EM-2 or EM-1 pretreatment was blocked by pretreatment with mu-opioid antagonist (-)-naloxone (55 pmol) but not nonopioid (+)-naloxone (55 pmol). However, pretreatment with a morphine-6beta-glucuronide-sensitive mu-opioid receptor antagonist 3-methoxynaltrexone (6.4 pmol) selectively blocked EM-2- but not EM-1-induced antianalgesia. Pretreatment with dynorphin A(1-17) antiserum reversed only EM-2- but not EM-1-induced antianalgesia. Pretreatment with antiserum against beta-endorphin, [Met(5)]enkephalin, [Leu(5)]enkephalin, substance P or cholecystokinin, or with delta-opioid receptor antagonist naltrindole (2.2 nmol) or kappa-opioid receptor antagonist norbinaltorphimine (6.6 nmol) did not affect EM-2-induced antianalgesia. It is concluded that EM-2 selectively releases dynorphin A(1-17) by stimulation of a novel subtype of mu-opioid receptor, tentatively designated as mu(3) in the vPAG to induce antianalgesia, whereas the antianalgesia induced by EM-1 is mediated by the stimulation of another subtype of mu(1)- or mu(2)-opioid receptor.


Subject(s)
Analgesia , Oligopeptides/pharmacology , Periaqueductal Gray/drug effects , Receptors, Opioid, mu/drug effects , Animals , Cholecystokinin/metabolism , Dose-Response Relationship, Drug , Dynorphins/metabolism , Enkephalin, Leucine/metabolism , Enkephalin, Methionine/metabolism , Male , Morphine/antagonists & inhibitors , Naloxone/pharmacology , Rats , Substance P/metabolism , beta-Endorphin/metabolism
16.
Eur J Pharmacol ; 504(3): 177-83, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15541419

ABSTRACT

We previously demonstrated pretreatment with antiserum against dynorphin A1-17 attenuates endomorphin-2-induced analgesia and antianalgesia, suggesting that these endomorphin-2 effects are mediated by the release of dynorphin A1-17. Lumbar-cisternal spinal perfusion was used to measure the release of immunoreactive dynorphin A1-17 into spinal perfusates from urethane-anesthetized rats following endomorphin-2 or endomorphin-1 treatment within the perfusion solution. Treatment with endomorphin-2 (5-50 nmol) for 3 min caused a dose-dependent increase of immunoreactive dynorphin A1-17 in spinal perfusates, with a maximal increase detected between 24 and 48 min after endomorphin-2 treatment, while levels returned to baseline within 60 min. Endomorphin-2-induced release of immunoreactive dynorphin A1-17 was attenuated by pretreatment with mu-opioid receptor antagonist naloxone or 3-methoxynaltrexone. Endomorphin-1 induced a slight increase in immunoreactive dynorphin1-17 as well, but only at the highest dose used (50 nmol). Our results suggest that endomorphin-2 stimulated a specific subtype of mu-opioid receptor to induce the release of immunoreactive dynorphin A1-17 in spinal cords of rats.


Subject(s)
Dynorphins/metabolism , Oligopeptides/pharmacology , Spinal Cord/metabolism , Anesthesia , Animals , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Injections, Spinal , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oligopeptides/administration & dosage , Rats , Receptors, Opioid, mu/antagonists & inhibitors , Spinal Cord/drug effects
17.
Bioorg Med Chem ; 12(15): 4133-45, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15246090

ABSTRACT

Here we report the new drug design and synthesis of a series of 6,14-endoethenomorphinan-7-carboxamide derivatives as a putative epsilon opioid receptor agonist. One of these compounds, 17-(cyclopropylmethyl)-4,5alpha-epoxy-3,6beta-dihydroxy-6,14-endoethenomorphinan-7alpha-(N-methyl-N-phenethyl)carboxamide (TAN-821), showed agonistic activity for a putative epsilon opioid receptor (IC(50) = 71.71nM) in the rat vas deferens (RVD) preparations. TAN-821 stimulated the binding of the nonhydrolyzable guanosine 5'-triphosphate analog, guanosine 5'-(gamma-thio)-triphosphate (GTPgammaS), to the mouse pons/medulla membrane via the activation of putative epsilon opioid receptor. Moreover, TAN-821 given intracerebroventricularly (i.c.v.) produced a marked antinociception in the tail-flick test (ED(50) = 1.73 microg) and the hot-plate test (ED(50) = 2.05 microg) in a dose-dependent manner. The antinociception induced by TAN-821 administered i.c.v. was blocked by the i.c.v.-pretreatment with a putative epsilon opioid receptor partial agonist beta-endorphin [1-27], but not a mu opioid receptor antagonist beta-FNA, a delta opioid receptor antagonist NTI, or a kappa opioid receptor antagonist nor-BNI. The present results suggest that TAN-821 may be a useful tool for the investigation on the pharmacological properties of the putative epsilon opioid receptor.


Subject(s)
Acetamides/chemical synthesis , Acetamides/pharmacology , Morphinans/chemical synthesis , Receptors, Opioid/agonists , Animals , Binding Sites , Dose-Response Relationship, Drug , Drug Design , Guanosine 5'-O-(3-Thiotriphosphate)/agonists , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Mice , Mice, Inbred ICR , Molecular Conformation , Morphinans/pharmacology , Muscle, Smooth/drug effects , Pain Measurement/drug effects , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Time Factors
18.
Neurosci Lett ; 365(3): 157-61, 2004 Jul 29.
Article in English | MEDLINE | ID: mdl-15246539

ABSTRACT

An unbiased conditioned place preference paradigm was used to evaluate the reward effect of selective endogenous mu-opioid ligands, endomorphin-1 and endomorphin-2, in male CD-1 mice. Pre- and post-conditioning free-movement were measured on day 1 and day 5, respectively. Conditioning sessions were conducted twice daily from day 2 through day 4 consisting of the alternate injection of conditioning drug or vehicle. Intracerebroventricular (i.c.v.) injection of endomorphin-1 (0.3-10 microg) induced place preference in a dose-dependent manner; whereas, endomorphin-2 (1-10 microg) dose-dependently induced place aversion. Both endomorphin-1-induced place preference and endomorphin-2-induced place aversion were blocked by pretreatment i.c.v. with mu-opioid receptor antagonist, beta-funaltrexamine. Selective delta-opioid receptor antagonist, naltrindole, co-administered i.c.v. with endomorphin-1 or endomorphin-2 did not affect reward effect. However, endomorphin-2-induced place aversion, but not endomorphin-1-induced place preference, was blocked by the i.c.v.-administered selective kappa-opioid receptor antagonist, WIN 44,441-3. It is concluded that endomorphin-1 produces conditioned place preference, which is mediated by the stimulation of mu-, but not delta- or kappa-opioid receptors, while endomorphin-2 produces conditioned place aversion, which is mediated by the stimulation of mu- and kappa-, but not delta-opioid receptors.


Subject(s)
Conditioning, Operant/drug effects , Naltrexone/analogs & derivatives , Oligopeptides/pharmacology , Animals , Azocines/pharmacology , Injections, Intraventricular , Ligands , Male , Mice , Naltrexone/pharmacology , Oligopeptides/administration & dosage , Oligopeptides/metabolism , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism
19.
Bioorg Med Chem Lett ; 14(16): 4241-3, 2004 Aug 16.
Article in English | MEDLINE | ID: mdl-15261278

ABSTRACT

To newly synthesize a selective opioid receptor antagonist, 17-(cyclopropylmethyl)-4,5 alpha-epoxy-6 beta,21-epoxymethano-3-hydroxy-6,14-endoethenomorphinan-7 alpha-(N-phenethyl)carboxamide was first designed from an opioid receptor agonist TAN-821 on the basis of the accessory site concept. The designed compound antagonized the agonistic effects induced by an opioid receptor agonist beta-endorphin on the rat vas deference test. Moreover, the designed compound blocked the antinociception induced by beta-endorphin given intracerebroventricularly.


Subject(s)
Drug Design , Morphinans/chemical synthesis , Narcotic Antagonists , Animals , Male , Morphinans/chemistry , Morphinans/pharmacology , Rats , Receptors, Opioid , Vas Deferens/drug effects , beta-Endorphin/pharmacology
20.
J Pharmacol Exp Ther ; 310(1): 240-6, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14999057

ABSTRACT

Intrathecal (i.t.) pretreatment with a low dose (0.3 nmol) of morphine causes an attenuation of i.t. morphine-produced analgesia; the phenomenon has been defined as morphine-induced antianalgesia. The opioid-produced analgesia was measured with the tail-flick (TF) test in male CD-1 mice. Intrathecal pretreatment with low dose (0.3 nmol) of morphine time dependently attenuated i.t. morphine-produced (3.0 nmol) TF inhibition and reached a maximal effect at 45 min. Intrathecal pretreatment with morphine (0.009-0.3 nmol) for 45 min also dose dependently attenuated morphine-produced TF inhibition. The i.t. morphine-induced antianalgesia was dose dependently blocked by the nonselective mu-opioid receptor antagonist (-)-naloxone and by its nonopioid enantiomer (+)-naloxone, but not by endomorphin-2-sensitive mu-opioid receptor antagonist 3-methoxynaltrexone. Blockade of delta-opioid receptors, kappa-opioid receptors, and N-methyl-D-aspartate (NMDA) receptors by i.t. pretreatment with naltrindole, nor-binaltorphimine, and (-)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801), respectively, did not affect the i.t. morphine-induced antianalgesia. Intrathecal pretreatment with antiserum against dynorphin A(1-17), [Leu]-enkephalin, [Met]-enkephalin, beta-endorphin, cholecystokinin, or substance P also did not affect the i.t. morphine-induced antianalgesia. The i.t. morphine pretreatment also attenuated the TF inhibition produced by opioid muagonist [D-Ala2, N-Me-Phe4,Gly-ol5]-enkephalin, delta-agonist deltorphin II, and kappa-agonist U50,488H. It is concluded that low doses (0.009-0.3 nmol) of morphine given i.t. activate an antianalgesic system to attenuate opioid mu-, delta-, and kappa-agonist-produced analgesia. The morphine-induced antianalgesia is not mediated by the stimulation of opioid mu-, delta-, or kappa-receptors or NMDA receptors. Neuropeptides such as dynorphin A(1-17), [Leu]-enkephalin, [Met]-enkephalin, beta-endorphin, cholecystokinin, and substance P are not involved in this low-dose morphine-induced antianalgesia.


Subject(s)
Analgesia , Morphine/administration & dosage , Naltrexone/analogs & derivatives , Pain/drug therapy , Spinal Cord/drug effects , Animals , Dizocilpine Maleate/pharmacology , Drug Interactions , Drug Tolerance , Dynorphins/immunology , Dynorphins/metabolism , Enkephalins/immunology , Enkephalins/metabolism , Male , Mice , Morphine/therapeutic use , Naloxone/pharmacology , Naltrexone/pharmacology , Oligopeptides/metabolism , Pain Measurement/drug effects , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Substance P/immunology , Substance P/metabolism , beta-Endorphin/immunology , beta-Endorphin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...