Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pain Res Treat ; 2012: 867067, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666579

RESUMEN

Delta opioid agonists enhance antinociceptive effects of mu-opioid agonists in many preclinical assays of acute nociception, but delta/mu interactions in preclinical models of inflammation-associated pain have not been examined. This study examined interactions between the delta agonist SNC80 [(+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide] and the mu agonist analgesics methadone, morphine, and nalbuphine in an assay of capsaicin-induced thermal allodynia in rhesus monkeys. Thermal allodynia was produced by topical application of capsaicin to the tail. Antiallodynic effects of methadone, morphine, and nalbuphine were evaluated alone or in combination with fixed proportions of SNC80 identical to proportions previously shown to enhance acute thermal antinociceptive effects of these mu agonists in rhesus monkeys (0.9 : 1 SNC80/methadone; 0.29 : 1 SNC80/morphine; 3.6 : 1 SNC80/nalbuphine). Methadone, morphine, and nalbuphine each produced dose-dependent antiallodynia. SNC80 produced partial antiallodynia up to the highest dose tested (5.6 mg/kg). SNC80 produced a modest, enantioselective, and naltrindole-reversible enhancement of methadone-induced antiallodynia. However, SNC80 did not enhance morphine antiallodynia and only weakly enhanced nalbuphine antiallodynia. Overall, SNC80 produced modest or no enhancement of the antiallodynic effects of the three mu agonists evaluated. These results suggest that delta agonist-induced enhancement of mu agonist antiallodynia may be weaker and less reliable than previously demonstrated enhancement of mu agonist acute thermal nociception.

2.
J Pain ; 13(4): 317-27, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22424913

RESUMEN

UNLABELLED: The delta opioid receptor agonist SNC80 produces both antinociceptive and antidepressant effects in rodents. This profile suggests that SNC80 may also reverse prodepressant effects of pain. Accordingly, this study compared SNC80 effects in complementary assays of pain-stimulated and pain-depressed behavior in rats. Intraperitoneal injection of dilute acid served as an acute noxious visceral stimulus in rats to stimulate abdominal stretching (a pain-stimulated behavior) or depress intracranial self-stimulation of the medial forebrain bundle (ICSS; a pain-depressed behavior). When administered once per week to minimize acute tolerance, SNC80 (1-10 mg/kg IP) decreased acid-stimulated stretching but had little effect on acid-induced depression of ICSS. More frequent SNC80 administration produced tolerance to SNC80 effects on acid-stimulated stretching, but unmasked antinociception in the assay of acid-depressed ICSS. SNC80 did not facilitate ICSS in the absence of pain, and effects of SNC80 were not duplicated by ARM390, a reputed delta agonist congener of SNC80 that does not internalize delta receptors. These findings support continued consideration of delta agonists as candidate analgesics to treat prodepressant effects of pain and illustrate the potential for diametrically opposite effects of drug treatments on preclinical measures of pain-stimulated and pain-depressed behavior. PERSPECTIVE: The delta opioid agonist SNC80 blocked pain-related depression of intracranial self-stimulation in rats, suggesting that delta agonists may be useful to treat prodepressant effects of pain. Repeated SNC80 produced tolerance to SNC80 antinociception in a conventional assay of pain-stimulated behavior but unmasked SNC80 antinociception in an assay of pain-depressed behavior.


Asunto(s)
Analgésicos Opioides/farmacología , Benzamidas/farmacología , Depresión/tratamiento farmacológico , Dolor/tratamiento farmacológico , Dolor/psicología , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Animales , Depresión/etiología , Depresión/fisiopatología , Modelos Animales de Enfermedad , Electrodos Implantados , Masculino , Nociceptores/efectos de los fármacos , Nociceptores/fisiología , Dolor/complicaciones , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/fisiología , Autoestimulación/efectos de los fármacos , Autoestimulación/fisiología
3.
Bioorg Med Chem ; 19(14): 4330-7, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21684752

RESUMEN

N-Phenethyl-substituted ortho-a and para-a oxide-bridged phenylmorphans have been obtained through an improved synthesis and their binding affinity examined at the various opioid receptors. Although the N-phenethyl substituent showed much greater affinity for µ- and κ-opioid receptors than their N-methyl relatives (e.g., K(i)=167 nM and 171 nM at µ- and κ-receptors vs >2800 and 7500 nM for the N-methyl ortho-a oxide-bridged phenylmorphan), the a-isomers were not examined further because of their relatively low affinity. The N-phenethyl substituted ortho-b and para-b oxide-bridged phenylmorphans were also synthesized and their enantiomers were obtained using supercritical fluid chromatography. Of the four enantiomers, only the (+)-ortho-b isomer had moderate affinity for µ- and κ-receptors (K(i)=49 and 42 nM, respectively, and it was found to also have moderate µ- and κ-opioid antagonist activity in the [(35)S]GTP-γ-S assay (K(e)=31 and 26 nM).


Asunto(s)
Morfinanos/farmacología , Antagonistas de Narcóticos , Óxidos/química , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Conformación Molecular , Morfinanos/síntesis química , Morfinanos/química , Estereoisomerismo , Relación Estructura-Actividad
4.
Bioorg Med Chem ; 19(11): 3434-43, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21570305

RESUMEN

A new synthesis of N-methyl and N-phenethyl substituted ortho-c and para-c oxide-bridged phenylmorphans, using N-benzyl- rather than N-methyl-substituted intermediates, was used and the pharmacological properties of these compounds were determined. The N-phenethyl substituted ortho-c oxide-bridged phenylmorphan(rac-(3R,6aS,11aS)-2-phenethyl-2,3,4,5,6,11a-hexahydro-1H-3,6a-methanobenzofuro[2,3-c]azocin-10-ol (12)) was found to have the highest µ-opioid receptor affinity (K(i)=1.1 nM) of all of the a- through f-oxide-bridged phenylmorphans. Functional data ([³5S]GTP-γ-S) showed that the racemate 12 was more than three times more potent than naloxone as an µ-opioid antagonist.


Asunto(s)
Morfinanos/química , Antagonistas de Narcóticos , Óxidos/química , Cristalografía por Rayos X , Conformación Molecular , Morfinanos/síntesis química , Morfinanos/farmacología , Unión Proteica , Receptores Opioides/metabolismo , Estereoisomerismo
5.
Psychopharmacology (Berl) ; 216(3): 431-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21369752

RESUMEN

RATIONALE: Delta-opioid agonists enhance the antinociceptive efficacy of methadone and other mu-opioid agonists. However, relatively little is known about the degree to which delta agonists might enhance the abuse-related effects of mu agonists. OBJECTIVE: This study used a behavioral economic approach to examine effects of the delta agonist SNC80 [(+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide] on the reinforcing effects of methadone in a drug self-administration assay. Interactions between SNC80 and cocaine were also examined for comparison. METHODS: Rhesus monkeys (n = 4), surgically implanted with indwelling intravenous catheters, were tested in two phases. In phase 1, drug self-administration dose-effect curves for methadone (0.0032-0.1 mg/kg/injection (inj)) and cocaine (0.0032-0.32 mg/kg/inj) alone were determined under a fixed-ratio 10 (FR 10) schedule of reinforcement. In phase 2, FR values were increased every 3 days (FR 1-FR 1800) during availability of methadone alone (0.032 mg/kg/inj) and in combination with varying proportions of SNC80 (0.1:1, 0.3:1, and 0.9:1 SNC80/methadone) or of cocaine alone (0.032 mg/kg/inj) and in combination with varying proportions of SNC80 (0.33:1, 1:1, and 3:1 SNC80/cocaine). Demand curves related drug intake to FR price, and measures of reinforcement were derived. RESULTS: Methadone and cocaine alone each functioned as a reinforcer. SNC80 did not alter measures of reinforcement for either methadone or cocaine. CONCLUSIONS: SNC80 at proportions previously shown to enhance methadone-induced antinociception did not enhance the abuse-related effects of methadone. These results support the proposition that delta agonists may selectively enhance mu agonist analgesic effects without enhancing mu agonist abuse liability.


Asunto(s)
Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Metadona/farmacología , Trastornos Relacionados con Opioides/etiología , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Refuerzo en Psicología , Análisis de Varianza , Animales , Benzamidas/administración & dosificación , Cocaína/administración & dosificación , Cocaína/farmacología , Interpretación Estadística de Datos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Macaca mulatta , Masculino , Metadona/administración & dosificación , Trastornos Relacionados con Opioides/metabolismo , Trastornos Relacionados con Opioides/psicología , Dolor/psicología , Piperazinas/administración & dosificación , Receptores Opioides mu/agonistas , Autoadministración
6.
Biochem J ; 433(1): 205-13, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20942800

RESUMEN

eIF5A (eukaryotic translation initiation factor 5A) is the only cellular protein containing hypusine [Nϵ-(4-amino-2-hydroxybutyl)lysine]. eIF5A is activated by the post-translational synthesis of hypusine and the hypusine modification is essential for cell proliferation. In the present study, we report selective acetylation of the hypusine and/or deoxyhypusine residue of eIF5A by a key polyamine catabolic enzyme SSAT1 (spermidine/spermine-N1-acetyltransferase 1). This enzyme normally catalyses the N1-acetylation of spermine and spermidine to form acetyl-derivatives, which in turn are degraded to lower polyamines. Although SSAT1 has been reported to exert other effects in cells by its interaction with other cellular proteins, eIF5A is the first target protein specifically acetylated by SSAT1. Hypusine or deoxyhypusine, as the free amino acid, does not act as a substrate for SSAT1, suggesting a macromolecular interaction between eIF5A and SSAT1. Indeed, the binding of eIF5A and SSAT1 was confirmed by pull-down assays. The effect of the acetylation of hypusine on eIF5A activity was assessed by comparison of acetylated with non-acetylated bovine testis eIF5A in the methionyl-puromycin synthesis assay. The loss of eIF5A activity by this SSAT1-mediated acetylation confirms the strict structural requirement for the hypusine side chain and suggests a possible regulation of eIF5A by hypusine acetylation/deacetylation.


Asunto(s)
Acetiltransferasas/metabolismo , Lisina/análogos & derivados , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Acetilación , Animales , Bovinos , Línea Celular Tumoral , Humanos , Lisina/metabolismo , Unión Proteica , Factor 5A Eucariótico de Iniciación de Traducción
7.
Pharmacol Biochem Behav ; 97(2): 205-12, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20678514

RESUMEN

Mu-opioid receptor agonists such as fentanyl are effective analgesics, but their clinical use is limited by untoward effects. Adjunct medications may improve the effectiveness and/or safety of opioid analgesics. This study compared interactions between fentanyl and either the noncompetitive N-methyl-D-aspartate (NMDA) glutamate receptor antagonist ketamine or the delta-opioid receptor agonist SNC162 [(+)-4-[(alphaR)-alpha-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-phenyl)methyl]-N,N-diethylbenzamide] in two behavioral assays in rhesus monkeys. An assay of thermal nociception evaluated tail-withdrawal latencies from water heated to 50 and 54°C. An assay of schedule-controlled responding evaluated response rates maintained under a fixed-ratio 30 schedule of food presentation. Effects of each drug alone and of three mixtures of ketamine+fentanyl (22:1, 65:1, 195:1 ketamine/fentanyl) or SNC162+fentanyl (59:1, 176:1, 528:1 SNC162/fentanyl) were evaluated in each assay. All drugs and mixtures dose-dependently decreased rates of food-maintained responding, and drug proportions in the mixtures were based on relative potencies in this assay. Ketamine and SNC162 were inactive in the assay of thermal antinociception, but fentanyl and all mixtures produced dose-dependent antinociception. Drug interactions were evaluated using dose-addition and dose-ratio analysis. Dose-addition analysis revealed that interactions for all ketamine/fentanyl mixtures were additive in both assays. SNC162/fentanyl interactions were usually additive, but one mixture (176:1) produced synergistic antinociception at 50°C. Dose-ratio analysis indicated that ketamine failed to improve the relative potency of fentanyl to produce antinociception vs. rate suppression, whereas two SNC162/fentanyl mixtures (59:1 and 176:1) increased the relative potency of fentanyl to produce antinociception. These results suggest that delta agonists may produce more selective enhancement than ketamine of mu agonist-induced antinociception.


Asunto(s)
Analgésicos Opioides/farmacología , Anestésicos Disociativos/farmacología , Benzamidas/farmacología , Fentanilo/farmacología , Ketamina/farmacología , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu/efectos de los fármacos , Animales , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Calor , Macaca mulatta , Masculino , Esquema de Refuerzo
8.
Eur J Pharmacol ; 604(1-3): 58-65, 2009 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-19133255

RESUMEN

Delta opioid receptor agonists are under development for a variety of clinical applications, and some findings in rats raise the possibility that agents with this mechanism have abuse liability. The present study assessed the effects of the non-peptidic delta opioid agonist SNC80 in an assay of intracranial self-stimulation (ICSS) in rats. ICSS was examined at multiple stimulation frequencies to permit generation of frequency-response rate curves and evaluation of curve shifts produced by experimental manipulations. Drug-induced leftward shifts in ICSS frequency-rate curves are often interpreted as evidence of abuse liability. However, SNC80 (1.0-10 mg/kg s.c.; 10-56 mg/kg i.p.) failed to alter ICSS frequency-rate curves at doses up to those that produced convulsions in the present study or other effects (e.g. antidepressant effects) in previous studies. For comparison, the monoamine releaser d-amphetamine (0.1-1.0 mg/kg, i.p.) and the kappa agonist U69,593 (0.1-0.56 mg/kg, i.p.) produced dose-dependent leftward and rightward shifts, respectively, in ICSS frequency-rate curves, confirming the sensitivity of the procedure to drug effects. ICSS frequency-rate curves were also shifted by two non-pharmacological manipulations (reductions in stimulus intensity and increases in response requirement). Thus, SNC80 failed to facilitate or attenuate ICSS-maintained responding under conditions in which other pharmacological and non-pharmacological manipulations were effective. These results suggest that non-peptidic delta opioid receptor agonists have negligible abuse-related effects in rats.


Asunto(s)
Benzamidas/efectos adversos , Encéfalo/efectos de los fármacos , Trastornos Relacionados con Opioides/etiología , Piperazinas/efectos adversos , Receptores Opioides delta/agonistas , Autoestimulación/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Encéfalo/metabolismo , Estimulación Eléctrica , Piperazinas/farmacología , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente
9.
Eur J Pharmacol ; 602(1): 92-100, 2009 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-19027735

RESUMEN

Delta opioid agonists can selectively enhance the antinociceptive effects of mu opioid agonists without enhancing some other, potentially undesirable mu agonist effects. However, the degree of delta receptor efficacy required to produce this profile of interactions is unknown. To address this issue, the present study examined interactions produced by the mu agonist fentanyl and the intermediate-efficacy delta opioid MSF61 in rhesus monkeys. For comparison, interactions were also examined between fentanyl and the relatively high-efficacy delta agonist SNC243A and the delta antagonist naltrindole, which has negligible efficacy at delta receptors. Two different behavioral procedures were used: (a) a warm-water tail-withdrawal assay of thermal nociception, and (b) an assay of schedule-controlled responding for food reinforcement. Drug interactions within each procedure were evaluated using dose-addition analysis to compare experimental results with expected additivity. Drug interactions across procedures were evaluated using dose-ratio analysis to assess relative potencies to produce antinociception vs. response-rate suppression. As expected, dose-addition analysis found that fentanyl/SNC243A interactions were superadditive in the assay of antinociception but additive in the assay of schedule-controlled responding. Conversely, fentanyl/MSF61 interactions were generally additive in both procedures, and fentanyl/naltrindole interactions were additive or subadditive in both procedures. Dose-ratio analysis found that fentanyl alone produced antinociception and rate suppression with similar potencies. Some fentanyl/SNC243A mixtures produced antinociception with up to 4-fold greater potency than rate-suppression. However, fentanyl/MSF61 and fentanyl/naltrindole mixtures produced antinociception with lower potency than rate suppression. These results suggest that relatively high delta receptor efficacy is required for mu/delta antinociceptive synergy.


Asunto(s)
Analgésicos Opioides/farmacología , Macaca mulatta/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Fentanilo/farmacología , Calor , Ligandos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Dolor/tratamiento farmacológico , Dolor/metabolismo , Dimensión del Dolor , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas
10.
J Neurochem ; 106(1): 37-44, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18422943

RESUMEN

Transglutaminases (TGases) catalyze several reactions with protein substrates, including formation of gamma-glutamyl-epsilon-lysine cross-links and gamma-glutamylpolyamine residues. The resulting gamma-glutamylamines are excised intact during proteolysis. TGase activity is altered in several diseases, highlighting the importance of in situ enzymatic determinations. Previous work showed that TGase activity (as measured by an in vitro assay) and free gamma-glutamyl-epsilon-lysine levels are elevated in Huntington disease (HD) and that gamma-glutamyl-epsilon-lysine is increased in HD CSF. Although free gamma-glutamyl-epsilon-lysine was used in these studies as an index of in situ TGase activity, gamma-glutamylpolyamines may also be diagnostic. We have devised methods for the simultaneous determination of four gamma-glutamylamines in CSF: gamma-glutamyl-epsilon-lysine, gamma-glutamylspermidine, gamma-glutamylputrescine, and bis-gamma-glutamylputrescine and showed that all are present in normal human CSF at concentrations of approximately 150, 670, 40, and 240 nM, respectively. The high gamma-glutamylspermidine/gamma-glutamylputrescine and gamma-glutamylspermidine/bis-gamma-glutamylputrescine ratios presumably reflect in part the large spermidine to putrescine mole ratio in human brain. We also showed that all four gamma-glutamylamines are elevated in HD CSF. Our findings support the hypotheses that (i) gamma-glutamylpolyamines are reflective of TGase activity in human brain, (ii) polyamination is an important post-translational modification of brain proteins, and (iii) TGase-catalyzed modification of proteins is increased in HD brain.


Asunto(s)
Poliaminas Biogénicas/metabolismo , Encéfalo/enzimología , Glutamina/metabolismo , Enfermedad de Huntington/líquido cefalorraquídeo , Enfermedad de Huntington/enzimología , Transglutaminasas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Aminación , Poliaminas Biogénicas/sangre , Biomarcadores/análisis , Biomarcadores/líquido cefalorraquídeo , Encéfalo/fisiopatología , Femenino , Humanos , Lisina/metabolismo , Masculino , Persona de Mediana Edad , Neuroquímica/métodos , Procesamiento Proteico-Postraduccional , Putrescina/análogos & derivados , Putrescina/metabolismo , Espermidina/metabolismo , Regulación hacia Arriba/fisiología
11.
J Pharmacol Exp Ther ; 324(2): 714-24, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17986650

RESUMEN

The nonpeptidic delta-opioid agonist SNC80 [(+)-4-[alpha(R)-alpha-[(2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl]-(3-methoxybenzyl)-N,N-diethylbenzamide] produces many stimulant-like behavioral effects in rodents and monkeys, such as locomotor stimulation, generalization to cocaine in discrimination procedures, and antiparkinsonian effects. Tolerance to the locomotor-stimulating effects of SNC80 develops after a single administration of SNC80 in rats; it is not known whether cross-tolerance develops to the effects of other stimulant compounds. In the initial studies to determine whether SNC80 produced cross-tolerance to other stimulant compounds, it was discovered that amphetamine-stimulated locomotor activity was greatly enhanced in SNC80-pretreated rats. This study evaluated acute cross-tolerance between delta-opioid agonists and other locomotor-stimulating drugs. Locomotor activity was measured in male Sprague-Dawley rats implanted with radiotransmitters, and activity levels were recorded in the home cage environment. Three-hour SNC80 pretreatment produced tolerance to further delta-opioid receptor stimulation but also augmented greatly amphetamine-stimulated locomotor activity in a dose-dependent manner. Pretreatments with other delta-opioid agonists, (+)BW373U86 [(+)-4-[alpha(R)-alpha-[(2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl]-3-hydroxybenzyl]-N,N-diethylbenzamide] and oxymorphindole (17-methyl-6,7-dehydro-4,5-epoxy-3,14-dihydroxy-6,7,2',3'-indolomorphinan), also modified amphetamine-induced activity levels. SNC80 pretreatment enhanced the stimulatory effects of the dopamine/norepinephrine transporter ligands cocaine and nomifensine (1,2,3,4-tetrahydro-2-methyl-4-phenyl-8-isoquinolinanmine maleate salt), but not the direct dopamine receptor agonists SKF81297 [R-(+)-6-chloro-7,8-dihydroxy-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrobromide] and quinpirole [trans-(-)-(4alphaR)-4,4a, 5,6,7,8,8a,9-octahydro-5-propyl-1H-pyrazolo[3,4-g] quinoline monohydrochloride]. In conclusion, SNC80 enhanced the locomotor-stimulating effects of monoamine transporter ligands suggesting that delta-opioid receptor activation might alter the functional activity of monoamine transporters or presynaptic monoamine terminals.


Asunto(s)
Benzamidas/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Agonistas de Dopamina/farmacología , Actividad Motora/efectos de los fármacos , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Animales , Sinergismo Farmacológico , Masculino , Actividad Motora/fisiología , Agitación Psicomotora/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/fisiología
12.
Pharmacol Biochem Behav ; 85(2): 428-34, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17112570

RESUMEN

Non-peptidic delta opioid receptor agonists are being evaluated for a wide range of clinical applications; however, the clinical utility of piperazinyl benzamide delta agonists such as SNC80 may be limited by convulsant activity. The purpose of the present study was to evaluate the electroencephalographic and convulsant activity produced by a high dose of 10 mg/kg SNC80 IM in rhesus monkeys. EEG and behavioral activity were examined in four adult male rhesus monkeys after IM administration of SNC80. Monkeys were seated in a standard primate restraint chair, and EEG activity was recorded using an array of 16 needle electrodes implanted subcutaneously in the scalp in a bipolar (scalp-to-scalp) montage in a longitudinal direction, with bilateral frontal, central, temporal, and occipital leads. Behavior was recorded using video monitoring equipment. Initially, all monkeys were tested with 10 mg/kg SNC80, which is a relatively high dose 3-10-fold greater than doses necessary to produce a variety of other behavioral effects. Behavioral convulsions and EEG seizures were observed in one of the four monkeys. In this monkey, neither behavioral convulsions nor EEG seizures were observed when a lower dose of 3.2 mg/kg was administered nine weeks later or when the same dose of 10 mg/kg SNC80 was administered one year later. These results suggest that IM administration of SNC80 is less potent in producing convulsant effects than in producing other, potentially useful behavioral effects (e.g. antinociception) in rhesus monkeys.


Asunto(s)
Benzamidas/farmacología , Electroencefalografía/efectos de los fármacos , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Convulsiones/inducido químicamente , Animales , Macaca mulatta , Masculino
13.
Eur J Pharmacol ; 547(1-3): 92-100, 2006 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16934797

RESUMEN

Delta agonists such as SNC80 ((+)-4-[(aR)-a-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide) produce some cocaine-like behavioral effects and warrant evaluation as candidate "agonist" medications for cocaine abuse. The present study examined acute and chronic effects of the systemically active delta agonist SNC80 on cocaine- and food-maintained responding in rhesus monkeys. Acute SNC80 (0.32-3.2 mg/kg, i.m.) pretreatment dose-dependently decreased cocaine self-administration (0.0032 mg/kg/injection), but doses of SNC80 that decreased cocaine self-administration also decreased food-maintained responding. In chronic studies, SNC80 (0.32-3.2 mg/kg/h, i.v.) was delivered for 7 days, and food or cocaine (0.01 mg/kg/injection) was available during 4 daily components of food availability and 4 daily components of drug availability. Chronic SNC80 (1.8 mg/kg/h) tended to decrease cocaine self-administration but produced greater reductions in food-maintained responding. A higher dose of 3.2 mg/kg/h SNC80 eliminated both cocaine- and food-maintained responding and produced profound sedation in one monkey and was not tested in other monkeys. These findings indicate that SNC80 produced dose-dependent and non-selective reductions in cocaine self-administration. These results suggest that SNC80 is unlikely to be useful as a treatment for cocaine dependence.


Asunto(s)
Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Cocaína/administración & dosificación , Alimentos , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Análisis de Varianza , Animales , Trastornos Relacionados con Cocaína/fisiopatología , Trastornos Relacionados con Cocaína/prevención & control , Trastornos Relacionados con Cocaína/psicología , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Macaca mulatta , Masculino , Esquema de Refuerzo , Autoadministración , Factores de Tiempo
14.
J Pharmacol Exp Ther ; 317(3): 1337-48, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16537798

RESUMEN

delta-Opioid agonists produce convulsions and antidepressant-like effects in rats. It has been suggested that the antidepressant-like effects are produced through a convulsant mechanism of action either through overt convulsions or nonconvulsive seizures. This study evaluated the convulsive and seizurogenic effects of nonpeptidic delta-opioid agonists at doses that previously were reported to produce antidepressant-like effects. In addition, delta-opioid agonist-induced electroencephalographic (EEG) and behavioral changes were compared with those produced by the chemical convulsant pentylenetetrazol (PTZ). For these studies, EEG changes were recorded using a telemetry system before and after injections of the delta-opioid agonists [(+)-4-[(alphaR)-alpha-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-methoxyphenyl)methyl]-N,N-diethylbenz (SNC80) and [(+)-4-[alpha(R)-alpha-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-hydroxyphenyl)methyl]-N,N-diethylbenzamide [(+)-BW373U86]. Acute administration of nonpeptidic delta-opioid agonists produced bilateral ictal and paroxysmal spike and/or sharp wave discharges. delta-Opioid agonists produced brief changes in EEG recordings, and tolerance rapidly developed to these effects; however, PTZ produced longer-lasting EEG changes that were exacerbated after repeated administration. Studies with antiepileptic drugs demonstrated that compounds used to treat absence epilepsy blocked the convulsive effects of nonpeptidic delta-opioid agonists. Overall, these data suggest that delta-opioid agonist-induced EEG changes are not required for the antidepressant-like effects of these compounds and that neural circuitry involved in absence epilepsy may be related to delta-opioid agonist-induced convulsions. In terms of therapeutic development, these data suggest that it may be possible to develop delta-opioid agonists devoid of convulsive properties.


Asunto(s)
Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Pentilenotetrazol/farmacología , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Convulsiones/inducido químicamente , Animales , Relación Dosis-Respuesta a Droga , Electrodos , Electroencefalografía , Masculino , Ratas , Ratas Sprague-Dawley , Convulsiones/metabolismo
15.
Eur J Pharmacol ; 531(1-3): 151-9, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16442521

RESUMEN

Nonpeptidic delta-opioid receptor agonists produce antidepressant-like effects in rodents, and compounds that inhibit the breakdown of endogenous opioid peptides have antidepressant-like effects in animal models. In this study, the behavioral effects of the enkephalinase inhibitor, RB101 (N-[(R, S)-2-benzyl-3-[(S)(2-amino-4-methyl-thio)-butyldithio]-1-oxopropyl]-l-phenylalanine benzyl ester), were examined. Specifically, the effects of RB101 on convulsive activity, locomotor activity, and antidepressant-like effects in the forced swim test were studied in Sprague-Dawley rats, and the opioid receptor types mediating these effects were examined by antagonist studies. In addition, the effects of RB101 on brain-derived neurotrophic factor (BDNF) mRNA expression were evaluated in relation to its antidepressant effects. RB101 produced delta-opioid receptor-mediated antidepressant effects (32 mg/kg i.v. and 100 mg/kg i.p.) and increased locomotor activity (32 mg/kg i.v.) in rats. RB101 did not produce convulsions or seizures and did not alter BDNF mRNA expression. In conclusion, RB101 has the potential to produce antidepressant effects without convulsions.


Asunto(s)
Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Disulfuros/farmacología , Electroencefalografía/efectos de los fármacos , Neprilisina/antagonistas & inhibidores , Fenilalanina/análogos & derivados , Hormona Adrenocorticotrópica/sangre , Animales , Benzamidas/farmacología , Factor Neurotrófico Derivado del Encéfalo/sangre , Factor Neurotrófico Derivado del Encéfalo/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Lóbulo Frontal/metabolismo , Hipocampo/metabolismo , Inyecciones Intravenosas , Masculino , Actividad Motora/efectos de los fármacos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Fenilalanina/farmacología , Piperazinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Natación
16.
Psychopharmacology (Berl) ; 183(1): 31-40, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16220339

RESUMEN

RATIONALE: Selective delta opioid receptor agonists have been shown to produce antidepressant-like behavioral effects and increase brain-derived neurotrophic factor (BDNF) mRNA expression when given acutely, but the chronic effects of delta agonists have been less well characterized. OBJECTIVE: The present study examined the effects of chronic exposure to the delta agonist (+)BW373U86 (BW) on antidepressant-like behavior in the forced swim test and on BDNF mRNA expression in comparison to chronic treatment with the antidepressants fluoxetine, desipramine, bupropion, and tranylcypromine. METHODS: Sprague-Dawley rats were treated chronic ally with one of the above treatments and were tested for antidepressant effects in the forced swim test, and assayed for BDNF mRNA expression by in situ hybridization. RESULTS: Acute administration of 10 mg/kg BW produced a significant antidepressant-like effect in the forced swim test, while chronic (8- or 21-day) BW administration did not produce a significant antidepressant-like effect. When 10 mg/kg BW was administered for 8 days, it produced a significant increase in BDNF mRNA expression in the frontal cortex, while having no effect on BDNF expression when given for 21 days. Chronic bupropion and desipramine significantly decreased BDNF expression in the dentate gyrus of the hippocampus, while fluoxetine had no effect in any brain region. Chronic tranylcypromine produced a significant increase in BDNF expression in the CA1 region of the hippocampus. CONCLUSIONS: Chronic exposure to BW produces tolerance to most effects, although at differential rates. In addition, increased BDNF mRNA expression does not appear to be a common effect of chronic administration of various antidepressants.


Asunto(s)
Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Animales , Antidepresivos/administración & dosificación , Antidepresivos/farmacología , Benzamidas/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Bupropión/administración & dosificación , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Desipramina/administración & dosificación , Tolerancia a Medicamentos , Fluoxetina/administración & dosificación , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Piperazinas/administración & dosificación , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Natación , Factores de Tiempo
17.
J Pain ; 6(4): 261-74, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15820914

RESUMEN

UNLABELLED: Previous studies suggest that sex differences in morphine antinociception in rodents might be attributed to the activational effects of gonadal hormones. The present study determined whether hormonal modulation of opioid antinociception in adult rats extends to opioids other than the prototypic mu agonist morphine. Male and female rats were sham-gonadectomized (sham-GDX) or gonadectomized (GDX) and replaced with no hormone, estradiol (E2, females), progesterone (P4, females), E2+P4 (females), or testosterone (males). Approximately 28 days later, nociception was evaluated on the 50 degrees C hot plate and warm water tail withdrawal tests before and after subcutaneous administration of hydromorphone, buprenorphine, U50,488, or SNC 80. In sham-GDX (gonadally intact) rats, the mu agonists and U50,488 were less effective in females than in males in at least one nociceptive test, and the delta agonist SNC 80 was less effective in males than in females. In males, gonadectomy tended to decrease, and testosterone tended to increase antinociception produced by 3 of the 4 agonists. In females, gonadectomy and hormone treatment had more variable effects, although E2 tended to decrease mu opioid antinociception. The present results suggest that activational effects of gonadal hormones are relatively modest and somewhat inconsistent on antinociception produced by various opioid agonists in the adult rat. PERSPECTIVE: This study demonstrates that reproductive hormones such as testosterone in males and estradiol in females do not consistently modulate sensitivity to the analgesic effects of opioids in the adult organism.


Asunto(s)
Analgésicos Opioides/farmacología , Hormonas Esteroides Gonadales/metabolismo , Dolor/tratamiento farmacológico , Dolor/metabolismo , Receptores Opioides/efectos de los fármacos , Receptores Opioides/metabolismo , Caracteres Sexuales , Analgésicos Opioides/metabolismo , Animales , Estrógenos/metabolismo , Estrógenos/farmacología , Ciclo Estral/fisiología , Femenino , Hormonas Esteroides Gonadales/farmacología , Masculino , Orquiectomía , Ovariectomía , Dolor/fisiopatología , Dimensión del Dolor/efectos de los fármacos , Progesterona/metabolismo , Progesterona/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo , Testosterona/metabolismo , Testosterona/farmacología
18.
J Pharmacol Exp Ther ; 314(1): 221-31, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15792997

RESUMEN

Interactions between delta and mu opioid agonists in rhesus monkeys vary as a function of the behavioral endpoint. The present study compared interactions between the delta agonist SNC80 [(+)-4-[(alphaR)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide] and the mu agonist heroin in assays of schedule-controlled responding, thermal nociception, and drug self-administration. Both SNC80 (ED50 = 0.43 mg/kg) and heroin (ED50 = 0.088 mg/kg) produced a dose-dependent and complete suppression of response rates in the assay of schedule-controlled responding. Heroin also produced thermal antinociception (ED(5 degrees C) = 0.18 mg/kg) and maintained drug self-administration under both a fixed ratio schedule [dose-effect curve peak at 0.0032 mg/kg/injection (inj)] and under a food versus heroin concurrent-choice schedule (ED50 = 0.013 mg/kg/inj), whereas SNC80 did not produce thermal antinociception or maintain self-administration. Fixed ratio mixtures of SNC80 and heroin (1.6:1, 4.7:1, and 14:1 SNC80/heroin) produced additive effects in the assay of schedule-controlled responding and superadditive effects in the assay of thermal nociception. Also, SNC80 did not enhance the reinforcing effects of heroin, indicating that mixtures of SNC80 and heroin produced additive or infra-additive reinforcing effects. These results provide additional evidence to suggest that delta/mu interactions depend on the experimental endpoint and further suggest that delta agonists may selectively enhance the antinociceptive effects of mu agonists while either not affecting or decreasing the sedative and reinforcing effects of mu agonists.


Asunto(s)
Analgésicos Opioides/farmacología , Benzamidas/farmacología , Condicionamiento Operante/efectos de los fármacos , Alimentos , Heroína/farmacología , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Animales , Relación Dosis-Respuesta a Droga , Calor , Macaca mulatta , Masculino , Dimensión del Dolor/efectos de los fármacos , Esquema de Refuerzo , Refuerzo en Psicología , Autoadministración
19.
J Pharmacol Exp Ther ; 312(3): 1314-20, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15574687

RESUMEN

Previous studies have demonstrated that peptidic and nonpeptidic delta-opioid receptor agonists have different effects depending on the measure. For example, nonpeptidic delta-opioid agonists, but not peptidic agonists, produce convulsions in rats, and in vitro studies suggested that peptidic and nonpeptidic delta-opioid agonists might have differential mechanisms of receptor downregulation. The present study evaluated potential differences between peptidic and nonpeptidic delta-opioid agonists in their ability to activate G proteins using guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS) autoradiography experiments in rat brain slices. The peptidic agonist [d-Pen(2),d-Pen(5)]-enkephalin and the nonpeptidic agonist (+)BW373U86 [(+)-4-[alpha(R)-alpha-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-hydroxyphenyl)methyl]-N,N-diethylbenzamide] demonstrated concentration-dependent increases in [(35)S]GTPgammaS binding that were attenuated by the delta-opioid antagonist naltrindole. (+)BW373U86 was more potent and efficacious than the peptidic agonist, and this difference remained consistent across brain regions where significant stimulation was observed. In addition, multiple delta-opioid compounds were evaluated for their agonist activity in this assay. These data suggested that differences between peptidic and nonpeptidic delta-opioid agonists in behavioral studies were most likely caused by differences in agonist efficacy. Finally, these data also revealed that [(35)S]GTPgammaS autoradiography could be used to compare efficacy differences among agonists across various brain regions in rat brain slices.


Asunto(s)
Benzamidas/farmacología , Encéfalo/metabolismo , Encefalina D-Penicilamina (2,5)/farmacología , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Animales , Autorradiografía , Relación Dosis-Respuesta a Droga , Masculino , Ratas , Ratas Sprague-Dawley
20.
J Med Chem ; 47(10): 2624-34, 2004 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-15115403

RESUMEN

In our efforts toward developing a nonselective ligand that would block the effects of stimulants such as methamphetamine at dopamine (DA), serotonin (5-HT), and norepinephrine (NE) transporters, we synthesized a series of 3-(3,4-dichlorophenyl)-1-indanamine derivatives. Two of the examined higher affinity compounds had a phenolic hydroxyl group enabling preparation of a medium to long chain carboxylic acid ester that might eventually be useful for a long-acting depot formulation. The in vitro data indicated that (-)-(1R,3S)-trans-3-(3,4-dichlorophenyl)-6-hydroxy-N-methyl-1-indanamine ((-)-(1R,3S)-11) displays high-affinity binding and potent inhibition of uptake at all three biogenic amine transporters. In vivo microdialysis experiments demonstrated that intravenous administration of (-)-(1R,3S)-11 to rats elevated extracellular DA and 5-HT in the nucleus accumbens in a dose-dependent manner. Pretreating rats with 0.5 mg/kg (-)-(1R,3S)-11 elevated extracellular DA and 5-HT by approximately 150% and reduced methamphetamine-induced neurotransmitter release by about 50%. Ex vivo autoradiography, however, demonstrated that iv administration of (-)-(1R,3S)-11 produced a dose-dependent, persistent occupation of 5-HT transporter binding sites but not DA transporter sites.


Asunto(s)
Aminas Biogénicas/metabolismo , Indanos/síntesis química , Proteínas de Transporte de Membrana/metabolismo , Animales , Autorradiografía , Proteínas Portadoras/metabolismo , Cristalografía por Rayos X , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Líquido Extracelular/metabolismo , Técnicas In Vitro , Indanos/química , Indanos/farmacología , Ligandos , Glicoproteínas de Membrana/metabolismo , Estructura Molecular , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática , Núcleo Accumbens/metabolismo , Ratas , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Estereoisomerismo , Relación Estructura-Actividad , Simportadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA