Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
Acta Pharmacol Sin ; 40(5): 583-588, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30224637

RESUMEN

Increasing preclinical evidence demonstrates that dopamine D3 receptor (D3R) antagonists are a potential option for the treatment of drug addiction. The reinstatement of the addiction can be triggered by environmental stimuli that acquire motivational salience through repeated associations with the drug's effects. YQA14 is a novel D3R antagonist that has exhibited pharmacotherapeutic efficacy in reducing cocaine and amphetamine reward and relapse to drug seeking in mice. In this study we investigated the effects of YQA14 on morphine-induced context-specific locomotor sensitization in mice. We showed that repeated injection of YQA14 (6.25-25 mg/kg every day ip) prior to morphine (10 mg/kg every day sc) not only inhibited the acquisition, but also significantly attenuated the expression of morphine-induced locomotor sensitization. Furthermore, in the expression phase, one single injection of YQA14 (6.25-25 mg/kg, ip) dose-dependently inhibited the expression of morphine-induced behavioral sensitization. Moreover, YQA14 inhibited the expression of morphine-induced behavioral sensitization in wild mice (WT), but not in D3R knockout (D3R-/-) mice in the expression phase. In addition, D3R-/- mice also displayed the reduction in the expression phase compared with WT mice. In summary, this study demonstrates that blockade or knockout of the D3R inhibits morphine-induced behavior sensitization, suggesting that D3R plays an important role in the pathogenesis and etiology of morphine addiction, and it might be a potential target for clinical management of opioid addiction.


Asunto(s)
Benzoxazoles/uso terapéutico , Antagonistas de Dopamina/uso terapéutico , Dependencia de Morfina/prevención & control , Morfina/farmacología , Actividad Motora/efectos de los fármacos , Piperazinas/uso terapéutico , Receptores de Dopamina D3/antagonistas & inhibidores , Animales , Benzoxazoles/administración & dosificación , Benzoxazoles/farmacocinética , Antagonistas de Dopamina/administración & dosificación , Antagonistas de Dopamina/farmacocinética , Comportamiento de Búsqueda de Drogas , Inyecciones Intraperitoneales , Masculino , Ratones Noqueados , Morfina/administración & dosificación , Dependencia de Morfina/etiología , Dependencia de Morfina/psicología , Piperazinas/administración & dosificación , Piperazinas/farmacocinética , Receptores de Dopamina D3/genética
2.
Addict Biol ; 21(2): 374-86, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25522207

RESUMEN

Dopamine (DA) is thought to represent a teaching signal and has been implicated in the induction of addictive behaviours. Dysfunction of DA homeostasis leading to high or low DA levels is causally linked to addiction. Previously, it has been proposed that the transcription factors Nurr1 and Pitx3, which are critical for transcription of a set of genes involved in DA metabolism in the mesolimbic pathway, are associated with addiction pathology. Using quantitative real-time polymerase chain reaction, immunofluorescence and Western blotting, we studied the effects of single morphine administration, morphine dependence and withdrawal on the DA markers DA transporters (DAT), vesicular monoamine transporters (VMAT2) and DA 2 receptor subtype (DRD2), DA 1 receptor subtype as well as tyrosine hydroxylase (TH) in the ventral tegmental area (VTA) and/or nucleus accumbens (NAc). In addition, Nurr1 and Pitx3 expression was also measured. Present data showed a high degree of colocalization of Nurr1 and Pitx3 with TH(+) neurons in the VTA. We found that the increased Nurr1 and/or Pitx3 levels during morphine dependence and in morphine-withdrawn rats were associated to an increase of DAT, VMAT2 and DRD2. Altogether, present data indicate that morphine dependence and withdrawal induced consistent alterations of most of the DA markers, which was correlated with transcription factors involved in the maintenance of DA neurons in drug-reward pathways, suggesting that Nurr1 and Pitx3 regulation might be associated with controlling adaptation to chronic morphine and to morphine withdrawal-induced alterations of DA neurons activity in the mesolimbic pathway.


Asunto(s)
Dopamina/metabolismo , Dependencia de Morfina/etiología , Morfina/farmacología , Narcóticos/farmacología , Síndrome de Abstinencia a Sustancias/etiología , Enfermedad Aguda , Animales , Biomarcadores/metabolismo , Enfermedad Crónica , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Implantes de Medicamentos , Proteínas de Homeodominio , Masculino , Dependencia de Morfina/metabolismo , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Núcleo Accumbens/metabolismo , ARN Mensajero/metabolismo , Ratas Wistar , Receptores de Dopamina D2/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Factores de Transcripción , Área Tegmental Ventral/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
3.
J Neurosci ; 34(27): 9076-87, 2014 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-24990928

RESUMEN

Opioids are commonly used for pain relief, but their strong rewarding effects drive opioid misuse and abuse. How pain affects the liability of opioid abuse is unknown at present. In this study, we identified an epigenetic regulating cascade activated by both pain and the opioid morphine. Both persistent pain and repeated morphine upregulated the transcriptional regulator MeCP2 in mouse central nucleus of the amygdala (CeA). Chromatin immunoprecipitation analysis revealed that MeCP2 bound to and repressed the transcriptional repressor histone dimethyltransferase G9a, reducing G9a-catalyzed repressive mark H3K9me2 in CeA. Repression of G9a activity increased expression of brain-derived neurotrophic factor (BDNF). Behaviorally, persistent inflammatory pain increased the sensitivity to acquiring morphine-induced, reward-related behavior of conditioned place preference in mice. Local viral vector-mediated MeCP2 overexpression, Cre-induced G9a knockdown, and CeA application of BDNF mimicked, whereas MeCP2 knockdown inhibited, the pain effect. These results suggest that MeCP2 directly represses G9a as a shared mechanism in central amygdala for regulation of emotional responses to pain and opioid reward, and for their behavioral interaction.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Dolor Crónico/fisiopatología , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína 2 de Unión a Metil-CpG/fisiología , Morfina/farmacología , Narcóticos/farmacología , Recompensa , Animales , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/farmacología , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/etiología , Condicionamiento Clásico/fisiología , Cruzamientos Genéticos , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Emociones/fisiología , Epigénesis Genética , Conducta Exploratoria/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/uso terapéutico , Morfina/toxicidad , Dependencia de Morfina/etiología , Dependencia de Morfina/psicología , Narcóticos/uso terapéutico , Narcóticos/toxicidad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Refuerzo en Psicología , Transgenes
4.
J Biomed Sci ; 22: 19, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25890152

RESUMEN

BACKGROUND: Heroin use among young women of reproductive age has drawn much attention around the world. Although methadone is widely used in maintenance therapy for heroin/morphine addiction, the long-term effects of prenatal exposure to methadone and preventative therapy remain unclear. For revealing this question, female pregnant Sprague-Dawley rats were sub-grouped to receive (1) vehicle, (2) methadone 5 mg/kg at embryonic day 3 (E3) and then 7 mg/kg from E4 to E20, (3) dextromethorphan (DM) 3 mg/kg, and (4) methadone + DM (the rats received methadone followed by DM treatment), subcutaneously, twice a day from E3 to E20. The body weight, natural withdrawal, pain sensitivity, ED50, conditioned place preference and water maze were conducted at different postnatal stages (P1 to P79) of offspring. The quantitative real-time RT-PCR and electrophysiology were also used to measure the gene expression of opioid receptors in the spinal cord and changes of LTP/LTD in the hippocampus, separately. RESULTS: Prenatal exposure to methadone or DM did not affect survival rate, body weight, water maze and LTP or LTD of offspring. However, prenatal methadone significantly increased the withdrawal symptoms, pain sensitivity, addiction liability and decreased the mRNA expression of pain related opioid receptors. Co-administration of DM with methadone in the maternal rats effectively prevented these abnormalities of offspring induced by methadone. CONCLUSIONS: Our study clearly showed that co-administration of dextromethorphan with methadone in the maternal rats prevented the adverse effects induced by prenatal methadone exposure. It implies that dextromethorphan may have a potential to be used in combination with methadone for maintenance treatment in pregnant heroin-addicted women to prevent the adverse effects induced by methadone on offspring.


Asunto(s)
Antitusígenos/uso terapéutico , Dextrometorfano/uso terapéutico , Metadona/administración & dosificación , Dependencia de Morfina/tratamiento farmacológico , Narcóticos/administración & dosificación , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Animales , Femenino , Masculino , Metadona/toxicidad , Dependencia de Morfina/etiología , Dependencia de Morfina/fisiopatología , Narcóticos/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ratas , Ratas Sprague-Dawley
5.
Behav Pharmacol ; 23(3): 302-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22495184

RESUMEN

The present study was designed to determine the effect of N'-[6,7-dichloro-4-(4-methoxy-phenyl)-3-oxo-3,4-dihydroquinoxalin-2-yl] hydrazide (SJA 7019), a selective nonpeptide inhibitor of calpain, and sodium orthovanadate, a selective inhibitor of tyrosine phosphatase, on the development of physiological dependence, as assessed by precipitated morphine withdrawal behavior in mice. Subchronic morphine administration (5 mg/kg, intraperitoneally, twice daily for 5 days), followed by a single injection of naloxone was used to precipitate the opioid withdrawal syndrome in mice. Behavioral observations were made immediately after naloxone treatment. Withdrawal syndrome was assessed quantitatively in terms of the withdrawal severity score and the frequency of jumping, rearing, forepaw licking, and circling. Daily single administration of SJA 7019 (1.5, 3, and 6 mg/kg, intraperitoneally) or sodium orthovanadate (5, 10, and 20 mg/kg, intraperitoneally) was continued during the morphine treatment procedure. Administration of SJA 7019 as well as the sodium orthovanadate dose-dependently attenuated the naloxone-induced morphine withdrawal syndrome. Neither SJA 7019 nor sodium orthovanadate significantly affected locomotor activity or morphine-induced antinociception. Therefore, it may be concluded that treatment with SJA 7019 or sodium orthovanadate during the morphine exposure period attenuated the development of physiological dependence on morphine, possibly through mechanisms linked to activation of tyrosine phosphatase and calpain.


Asunto(s)
Calpaína/fisiología , Dependencia de Morfina/etiología , Proteínas Tirosina Fosfatasas/fisiología , Síndrome de Abstinencia a Sustancias/etiología , Analgésicos Opioides/farmacología , Animales , Calpaína/antagonistas & inhibidores , Femenino , Masculino , Ratones , Morfina/farmacología , Actividad Motora/efectos de los fármacos , Naloxona/farmacología , Conducta Estereotipada/efectos de los fármacos , Vanadatos/farmacología
6.
Bull Exp Biol Med ; 153(6): 835-8, 2012 Oct.
Artículo en Inglés, Ruso | MEDLINE | ID: mdl-23113297

RESUMEN

We studied the expression of mRNA for genes, whose protein products regulate the glutamate/NO/cGMP signal cascade in the frontal cortex, striatum, midbrain, and hippocampus of rats with various degrees of spontaneous morphine withdrawal syndrome. The concentration of Grin2a mRNA (subunit of the NMDA glutamate receptor) in the frontal cortex increased mainly in animals with severe abstinence. By contrast, the expression of mRNA for the Grin2b subunit in the striatum decreased in animals with mild abstinence. Variations in the content of mRNA for other products of the cascade (isoforms of NO-dependent guanylate cyclase and cGMP-dependent protein kinase) after morphine withdrawal were not related to the degree of abstinence. Our results suggest that the region-specific expression of mRNA for certain subunits of the NMDA glutamate receptor after morphine withdrawal can determine the degree of abstinence.


Asunto(s)
Cuerpo Estriado/metabolismo , Lóbulo Frontal/metabolismo , Dependencia de Morfina/genética , ARN Mensajero/genética , Receptores de N-Metil-D-Aspartato/genética , Síndrome de Abstinencia a Sustancias/genética , Animales , Cuerpo Estriado/fisiopatología , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Lóbulo Frontal/fisiopatología , Expresión Génica , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Morfina/efectos adversos , Dependencia de Morfina/etiología , Dependencia de Morfina/metabolismo , Dependencia de Morfina/fisiopatología , Óxido Nítrico/metabolismo , Especificidad de Órganos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/fisiopatología
7.
J Biomed Sci ; 17: 46, 2010 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-20529288

RESUMEN

BACKGROUND: Abuse of addictive substances is a serious problem that has a significant impact on areas such as health, the economy, and public safety. Heroin use among young women of reproductive age has drawn much attention around the world. However, there is a lack of information on effects of prenatal exposure to opioids on their offspring. In this study, an animal model was established to study effects of prenatal exposure to opioids on offspring. METHODS: Female pregnant Sprague-Dawley rats were sub-grouped to receive (1) vehicle, (2) 2-4 mg/kg morphine (1 mg/kg increment per week), (3) 7 mg/kg methadone, and (4) 3 mg/kg buprenorphine, subcutaneously, once or twice a day from E3 to E20. The experiments were conducted on animals 8-12 weeks old and with body weight between 250 and 350 g. RESULTS: Results showed that prenatal exposure to buprenorphine caused higher mortality than other tested substance groups. Although we observed a significantly lower increase in body weight in all of the opioid-administered dams, the birth weight of the offspring was not altered in all treated groups. Moreover, no obvious behavioral abnormality or body-weight difference was noted during the growing period (8-12 weeks) in all offspring. When the male offspring received morphine injection twice a day for 4 days, the prenatally opioid-exposed rats more quickly developed a tolerance to morphine (as shown by the tail-flick tests), most notably the prenatally buprenorphine-exposed offspring. However, the tolerance development to methadone or buprenorphine was not different in offspring exposed prenatally to methadone or buprenorphine, respectively, when compared with that of the vehicle controlled group. Similar results were also obtained in the female animals. CONCLUSIONS: Animals prenatally exposed to morphine, methadone, or buprenorphine developed tolerance to morphine faster than their controlled mates. In our animal model, prenatal exposure to buprenorphine also resulted in higher mortality and much less sensitivity to morphine-induced antinociception than prenatal exposure to morphine or methadone. This indicates that buprenorphine in higher doses may not be an ideal maintenance drug for treating pregnant women. This study provides a reference in selecting doses for clinical usage in treating pregnant heroin addicts.


Asunto(s)
Buprenorfina/administración & dosificación , Tolerancia a Medicamentos/fisiología , Metadona/administración & dosificación , Dependencia de Morfina/tratamiento farmacológico , Dependencia de Morfina/etiología , Morfina/administración & dosificación , Efectos Tardíos de la Exposición Prenatal/etiología , Analgésicos/administración & dosificación , Analgésicos/toxicidad , Animales , Buprenorfina/toxicidad , Modelos Animales de Enfermedad , Femenino , Dependencia de Heroína/tratamiento farmacológico , Humanos , Masculino , Metadona/toxicidad , Morfina/toxicidad , Dependencia de Morfina/fisiopatología , Dimensión del Dolor , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Sprague-Dawley
8.
J Neurosci ; 28(47): 12183-9, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19020012

RESUMEN

The Gbetagamma subunit has been implicated in many downstream signaling events associated with opioids. We previously demonstrated that a small molecule inhibitor of Gbetagamma-subunit-dependent phospholipase (PLC) activation potentiated morphine-induced analgesia (Bonacci et al., 2006). Here, we demonstrate that this inhibitor, M119 (cyclohexanecarboxylic acid [2-(4,5,6-trihydroxy-3-oxo-3H-xanthen-9-yl)-(9Cl)]), is selective for mu-opioid receptor-dependent analgesia and has additional efficacy in mouse models of acute tolerance and dependence. When administered by an intracerebroventricular injection in mice, M119 caused 10-fold and sevenfold increases in the potencies of morphine and the mu-selective peptide, DAMGO, respectively. M119 had little or no effect on analgesia induced by the kappa agonist U50,488 or delta agonists DPDPE or Deltorphin II. Similar results were obtained in vitro, as only activation of the mu-opioid receptor stimulated PLC activation, whereas no effect was seen with the kappa- and delta-opioid receptors. M119 inhibited mu-receptor-dependent PLC activation. In studies to further explore the in vivo efficacy of M119, systemic administration M119 also resulted in a fourfold shift increase in potency of systemically administered morphine. Of particular interest, M119 was also able to attenuate acute, antinociceptive tolerance and dependence in mice treated concomitantly with both M119 and morphine. These studies suggest that small organic molecules, such as M119, that specifically regulate Gbetagamma subunit signaling may have important therapeutic applications in enhancing opioid analgesia, while attenuating the development of tolerance and dependence.


Asunto(s)
Analgésicos/efectos adversos , Ciclohexanos/uso terapéutico , Tolerancia a Medicamentos/fisiología , Dependencia de Morfina/tratamiento farmacológico , Morfina/efectos adversos , Receptores Opioides mu/fisiología , Xantenos/uso terapéutico , Analgésicos/farmacología , Animales , Conducta Animal/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Ciclohexanos/química , Ciclohexanos/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Dependencia de Morfina/etiología , Dependencia de Morfina/fisiopatología , Naloxona/farmacología , Dimensión del Dolor/métodos , Fosfolipasa C beta/farmacología , Unión Proteica/efectos de los fármacos , Tiempo de Reacción/efectos de los fármacos , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección/métodos , Xantenos/química , Xantenos/farmacología
9.
Science ; 273(5275): 657-9, 1996 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-8662559

RESUMEN

Chronic morphine administration induces an up-regulation of several components of the cyclic adenosine 5'-monophosphate (cAMP) signal transduction cascade. The behavioral and biochemical consequences of opiate withdrawal were investigated in mice with a genetic disruption of the alpha and Delta isoforms of the cAMP-responsive element-binding protein (CREB). In CREBalphadelta mutant mice the main symptoms of morphine withdrawal were strongly attenuated. No change in opioid binding sites or in morphine-induced analgesia was observed in these mutant mice, and the increase of adenylyl cyclase activity and immediate early gene expression after morphine withdrawal was normal. Thus, CREB-dependent gene transcription is a factor in the onset of behavioral manifestations of opiate dependence.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Dependencia de Morfina/etiología , Morfina/administración & dosificación , Síndrome de Abstinencia a Sustancias/fisiopatología , Adenilil Ciclasas/metabolismo , Analgesia , Animales , Conducta Animal , Corteza Cerebral/enzimología , Tolerancia a Medicamentos , Regulación de la Expresión Génica , Marcación de Gen , Genes Inmediatos-Precoces , Locus Coeruleus/metabolismo , Ratones , Morfina/efectos adversos , Morfina/farmacología , Dependencia de Morfina/metabolismo , Mutación , Naloxona/farmacología , Receptores Opioides/metabolismo , Transducción de Señal
10.
Epilepsia ; 50(4): 789-800, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19183220

RESUMEN

PURPOSE: Herein we used electrophysiologic approaches in hippocampal area CA1 to estimate how morphine treatment alters the pentylenetetrazol (PTZ) effects. METHODS: Hippocampal slices taken from either control animals or animals made dependent via chronic morphine administration were examined. Changes in the population spike and epileptiform amplitudes were used as indices to quantify the effects of PTZ exposure in the control and morphine-dependent slices. Hippocampal slices taken either from control animals or from animals made dependent upon morphine were exposed to PTZ, either with or without morphine, naloxone, or morphine + naloxone. RESULTS: Morphine dependence increased a PTZ-induced long-term potentiation (LTP) of the population spike in CA1 in vitro. This LTP was not found in rats that had spontaneously withdrawn morphine or withdrawn with naloxone in vivo after chronic morphine intake. Morphine or naloxone in vitro blocked the PTZ-induced LTP changes in control and morphine-dependent slices. However, PTZ-induced multiple population spikes (epileptiform activity) in CA1 was not blocked by naloxone. DISCUSSION: It is concluded that dependence on morphine enhances PTZ-induced plastic and epileptic changes in CA1 excitability. We suggest that this model of neuronal activity in dependent slices could present an opportunity for studying the mechanisms underlying the increased likelihood of seizures in morphine users.


Asunto(s)
Convulsivantes/farmacología , Potenciales Evocados/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Dependencia de Morfina/patología , Pentilenotetrazol/farmacología , Animales , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Potenciales Evocados/efectos de los fármacos , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Morfina/administración & dosificación , Dependencia de Morfina/tratamiento farmacológico , Dependencia de Morfina/etiología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Narcóticos/administración & dosificación , Vías Nerviosas/fisiología , Ratas , Ratas Endogámicas , Estadísticas no Paramétricas , Síndrome de Abstinencia a Sustancias/patología , Síndrome de Abstinencia a Sustancias/fisiopatología
11.
Life Sci ; 234: 116784, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31445026

RESUMEN

Tobacco smoking is recognized as a life-threatening risk factor worldwide. Initiation of smoking primarily occurs during adolescence which is a critical developmental phase characterized by specific neurobehavioral alterations. The effect of adolescent nicotine exposure on vulnerability to opioid addiction has not been previously addressed. Furthermore, lateral paragigantocellularis (LPGi) is a key modulator of opiate effects. In this study we investigated the effect of adolescent nicotine treatment on development of morphine tolerance and dependence as well as LPGi neuronal responses to morphine during adulthood. Male Wistar rats received subcutaneous injections of either nicotine or saline during adolescence and then development of morphine tolerance and dependence was assessed during adulthood by tail-flick and withdrawal tests, respectively. In vivo single-unit recording was performed to examine the LPGi neuronal activities. Results indicated that adolescent nicotine exposure significantly facilitates the development of tolerance to analgesic effect of morphine and increases the expression of morphine withdrawal signs in adulthood. Also, it was observed that following adolescent nicotine treatment, the extent of morphine-induced excitation is attenuated in LPGi neurons of adult rats. Moreover, the onset of morphine-induced inhibition was increased in these animals. Neither the baseline, nor the regularity of firing was affected in our observations. It could be concluded that nicotine challenge during adolescence may enhance the future vulnerability to opioid addiction through induction of persistent neuroadaptations in LPGi neurons.


Asunto(s)
Tronco Encefálico/efectos de los fármacos , Dependencia de Morfina/etiología , Neuronas/efectos de los fármacos , Nicotina/efectos adversos , Envejecimiento , Animales , Tronco Encefálico/citología , Tronco Encefálico/fisiopatología , Masculino , Dependencia de Morfina/fisiopatología , Neuronas/patología , Trastornos Relacionados con Opioides/etiología , Trastornos Relacionados con Opioides/fisiopatología , Ratas Wistar , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/fisiopatología
12.
Neuropharmacology ; 54(3): 588-96, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18177675

RESUMEN

Using the C-fibre reflex as a nociceptive response elicited by a wide range of stimulus intensities in the rat, we recently reported that a single treatment with (+)-HA966, a glycine site-specific NMDA receptor antagonist: (1) potentiates morphine antinociception; and (2) reverses an established morphine tolerance. We presently aimed at determining whether our observation was likely to result from a direct effect on the spinal cord or an indirect effect of supraspinal origin. In a 2x2x2 experimental design, we compared the effects of 5 mg/kg morphine in: (1) sham-operated rats or animals whose brainstems had been transected at the level of the obex; (2) rats that were implanted with pellets, either 150 mg morphine or placebo; and (3) animals injected either with saline or 10 mg/kg (+)-HA966. The control C-fibre reflexes were similar in all groups of animals. As compared to "non-tolerant" rats, the depressive effect of morphine was weaker in "morphine-tolerant" animals where the threshold did not change following morphine but the gain of the stimulus-response curve decreased, albeit to a significantly lesser extent than in the "non-tolerant" group. Whether in "non-tolerant" or "tolerant" groups, the effects of morphine were stronger in "obex-transected" than in "sham-operated" animals. In all groups, the effects of morphine were potentiated by the preliminary administration of (+)-HA966. However, in the "morphine-tolerant" group, the preliminary administration of (+)-HA966 was more potent in the "sham-operated" than in the "obex-transected" groups. Since overall effects were very similar in "sham-operated" and "obex-transected" animals, we concluded for our model that the critical site for the expression of the neuronal plastic changes associated with morphine tolerance lies in the spinal cord.


Asunto(s)
Tolerancia a Medicamentos/fisiología , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Dependencia de Morfina/tratamiento farmacológico , Morfina/efectos adversos , Pirrolidinonas/uso terapéutico , Médula Espinal/efectos de los fármacos , Animales , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Electromiografía/métodos , Masculino , Dependencia de Morfina/etiología , Dependencia de Morfina/fisiopatología , Fibras Nerviosas Amielínicas/fisiología , Fibras Nerviosas Amielínicas/efectos de la radiación , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Reflejo/efectos de los fármacos , Médula Espinal/fisiología , Traumatismos de la Médula Espinal/fisiopatología
13.
Neuropharmacology ; 54(3): 475-86, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18082850

RESUMEN

Acute morphine administration produces analgesia and reward, but prolonged use may lead to analgesic tolerance in patients chronically treated for pain and to compulsive intake in opioid addicts. Moreover, long-term exposure may induce physical dependence, manifested as somatic withdrawal symptoms in the absence of the drug. We set up three behavioral paradigms to model these adaptations in mice, using distinct regimens of repeated morphine injections to induce either analgesic tolerance, locomotor sensitization or physical dependence. Interestingly, mice tolerant to analgesia were not sensitized to hyperlocomotion, whereas sensitized mice displayed some analgesic tolerance. We then examined candidate molecular modifications that could underlie the development of each behavioral adaptation. First, analgesic tolerance was not accompanied by mu opioid receptor desensitization in the periaqueductal gray. Second, cdk5 and p35 protein levels were unchanged in caudate-putamen, nucleus accumbens and prefrontal cortex of mice displaying locomotor sensitization. Finally, naloxone-precipitated morphine withdrawal did not enhance basal or forskolin-stimulated adenylate cyclase activity in nucleus accumbens, prefrontal cortex, amygdala, bed nucleus of stria terminalis or periaqueductal gray. Therefore, the expression of behavioral adaptations to chronic morphine treatment was not associated with the regulation of micro opioid receptor, cdk5 or adenylate cyclase activity in relevant brain areas. Although we cannot exclude that these modifications were not detected under our experimental conditions, another hypothesis is that alternative molecular mechanisms, yet to be discovered, underlie analgesic tolerance, locomotor sensitization and physical dependence induced by chronic morphine administration.


Asunto(s)
Adenilil Ciclasas/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Tolerancia a Medicamentos/fisiología , Locomoción/efectos de los fármacos , Dependencia de Morfina/etiología , Morfina/administración & dosificación , Narcóticos/administración & dosificación , Receptores Opioides mu/metabolismo , Analgésicos , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/anatomía & histología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Esquema de Medicación , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Unión Proteica/efectos de los fármacos , Factores de Tiempo
14.
Naunyn Schmiedebergs Arch Pharmacol ; 378(4): 407-20, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18548233

RESUMEN

Our previous studies have shown that morphine withdrawal increases the hypothalamic-pituitary-adrenocortical axis activity, which is dependent on a hyperactivity of noradrenergic pathways (nucleus tractus solitarius-A(2)) innervating the hypothalamic paraventricular nucleus. The extracellular signal-regulated kinase has been implicated in drug addiction, but its role in activation of paraventricular nucleus and nucleus tractus solitarius during morphine dependence remain poorly understood. We have determined the activation of extracellular signal-regulated kinase during morphine dependence and withdrawal as well as its involvement in morphine withdrawal-induced gene expression. We show that naloxone-induced morphine withdrawal activates extracellular signal-regulated kinases(1/2) and increases c-Fos expression in rat paraventricular nucleus and nucleus tractus solitarius-A(2) neurons. Activated extracellular signal-regulated kinases(1/2) was colocalized with c-Fos in both nuclei, and this response was blocked by SL327, a drug that prevents extracellular signal-regulated kinase activation. In the paraventricular nucleus from morphine-withdrawn rats, the number of neurons expressing CRF was increased. Immunohistochemical study showed a dramatic increase in c-Fos immunoreactivity within CRF-positive cells. These results suggest that extracellular signal-regulated kinases1/2 signaling pathway is necessary for morphine withdrawal-induced activation of brain areas associated with the stress system.


Asunto(s)
Encéfalo/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Morfina/toxicidad , Naloxona/farmacología , Síndrome de Abstinencia a Sustancias/fisiopatología , Aminoacetonitrilo/administración & dosificación , Aminoacetonitrilo/análogos & derivados , Aminoacetonitrilo/farmacología , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Hormona Liberadora de Corticotropina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Inmunoquímica , Inyecciones Subcutáneas , Masculino , Morfina/administración & dosificación , Dependencia de Morfina/etiología , Dependencia de Morfina/fisiopatología , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacología , Narcóticos/administración & dosificación , Narcóticos/toxicidad , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/patología , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/metabolismo , Núcleo Solitario/patología , Síndrome de Abstinencia a Sustancias/etiología , Factores de Tiempo
15.
Physiol Behav ; 93(3): 622-7, 2008 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-18067930

RESUMEN

Numerous studies have dealt with the role of opiate system in tasks aimed at measurement of cognitive behavior, but the role of morphine dependence on learning and memory is still controversial. In this study chronic exposure to morphine was employed to evaluate learning ability and spatial short-term memory (working memory) and long-term memory (reference memory) in the water maze task. Male albino rats were made dependent by chronic administration of morphine in drinking water that lasted at least 21 days. In Experiment 1, the performance of animals was evaluated in reference memory version of the water maze. Rats were submitted to a session of 6 trials for 6 consecutive days to find the submerged platform that was located in the center of a quadrant. Latency and traveled distance to find the platform were measured as indexes of learning. Memory retention was tested 24 h after the last training session in a probe trial (60 s) in which there was no platform and the time spent in each quadrant of the water maze was recorded. Results indicated that latency and traveled distance to find the platform were same in control and dependent rats during training days, but during the probe test morphine-dependent group spent significantly less time in the target quadrant. In Experiment 2, training on working memory version of the water maze task was started. Only two trials per day were given until the performance of animals was stabilized (at least 5 days). Final test was done at day 6. Acquisition-retention interval was 75 min. No significant differences were found on acquisition and retention trials between morphine and control groups. Our findings indicate that chronic exposure to morphine did not impair learning ability, but partially impaired retention of spatial long-term (reference) memory. Moreover, dependence on morphine did not affect either acquisition or retention of spatial short (working) memory.


Asunto(s)
Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Dependencia de Morfina/fisiopatología , Análisis de Varianza , Animales , Conducta Animal , Modelos Animales de Enfermedad , Masculino , Memoria/efectos de los fármacos , Morfina/efectos adversos , Dependencia de Morfina/etiología , Actividad Motora/fisiología , Narcóticos/efectos adversos , Ratas , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Factores de Tiempo
16.
Yakugaku Zasshi ; 128(3): 395-403, 2008 Mar.
Artículo en Japonés | MEDLINE | ID: mdl-18311059

RESUMEN

Signs characteristic of opiate withdrawal symptoms can be precipitated by an opiate antagonist after short-term infusion or even a single dose of an opiate both in humans and in animals. This phenomenon has been referred to as acute dependence. In contrast to extensive studies on chronic dependence, less is known about the neural mechanisms mediating acute dependence. It will benefit the development of appropriate therapies to facilitate opiate abstinence and reduced craving to better understand the mechanisms underlying acute opiate dependence and to determine whether there are dissociation and similarity between the early and fully developed stages of dependence. In the present study, we examined the influence of c-Fos expression in the amygdala in acquisition of conditioned place aversion (CPA) induced by naloxone-precipitated withdrawal from a single morphine exposure 24 h earlier. The effect of microinjection into the central amygdaloid nucleus (CeA) of various kinds of glutamatergic neurotransmission inhibitors was also investigated. Findings showed that CeA displayed significant increase in c-Fos expression in the acquisition of CPA. Furthermore, CPA was attenuated significantly and dose-dependently by microinjection into CeA of all glutamatergic neurotransmission inhibitors (NMDA receptor antagonist (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclo-hepten-5,10-imine maleate (MK-801), AMPA receptor antagonist 1-(4-aminophenyl)4-methyl-7,8-methylenedioxy-5H-2,3-benzodiazepine hydrochloride (GYKI52466), metabotropic glutamate receptor antagonist (+/-)-alpha-methyl-4-carboxyphenylglycine (MCPG), and glutamate release inhibitor riluzole). These findings suggest that CeA involves the acquisition of CPA induced by naloxone-precipitated withdrawal from a single morphine exposure, and the function of the glutamatergic system projected from the amygdala to nucleus accumbens plays a facilitative role in formation of morphine dependence.


Asunto(s)
Amígdala del Cerebelo/fisiología , Conducta Animal/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Dependencia de Morfina/etiología , Morfina/administración & dosificación , Morfina/efectos adversos , Naloxona/farmacología , Síndrome de Abstinencia a Sustancias/psicología , Amígdala del Cerebelo/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Glutamatos/fisiología , Humanos , Neurotransmisores/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas
17.
Drug Res (Stuttg) ; 68(1): 33-37, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28847022

RESUMEN

BACKGROUND: Dependence and tolerance to morphine are major problems which limit its chronic clinical application. PURPOSE: This study was aimed to investigate the attenuation effect of Cerebrolysin, a mixture of potent growth factors (BDNF, GDNF, NGF, CNTF etc,), on the development of Morphine-induced dependence and tolerance. METHODS: Male Wistar rats were selected randomly and divided into different groups (n=8) including: a control group, groups received additive doses of morphine (5-25 mg/kg, ip, at an interval of 12 h until tolerance completion), and groups pretreated with Cerebrolysin (40, 80 and 160 mg/kg, ip, before morphine administration). Development of tolerance was assessed by tail-flick test and the attenuation effect of Cerebrolysin on morphine-induced dependence was evaluated after injection of naloxone (4 mg/kg, ip, 12 h after the morning dose of morphine). Seven distinct withdrawal signs including: jumping, rearing, genital grooming, abdominal writhing, wet dog shake and teeth grinding were recorded for 45 min and total withdrawal score (TWS) was calculated. RESULTS: Results showed that administration of Cerebrolysin could prolonged development (10 and 14 days in administration of 80 mg/kg and 160 mg/kg Cerebrolysin) and completion (4, 10 and 14 days in administration of 40, 80 and 160 mg/kg Cerebrolysin, respectively) of tolerance. Results also indicated that administration of Cerebrolysin (40, 80 and 160 mg/kg) could significantly decreased the TWS value (62±2, 77±4 and 85±6%, respectively). CONCLUSION: In conclusion, it was found that pretreatment with Cerebrolysin could attenuated morphine-induced tolerance and dependence.


Asunto(s)
Aminoácidos/uso terapéutico , Analgésicos Opioides/efectos adversos , Dependencia de Morfina/prevención & control , Morfina/efectos adversos , Fármacos Neuroprotectores/uso terapéutico , Aminoácidos/farmacología , Animales , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Humanos , Masculino , Dependencia de Morfina/etiología , Fármacos Neuroprotectores/farmacología , Dolor/tratamiento farmacológico , Dimensión del Dolor , Ratas , Ratas Wistar , Factores de Tiempo
18.
Physiol Behav ; 191: 37-46, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29626506

RESUMEN

Drugs of abuse cause persistent alterations in synaptic plasticity that is thought to underlie addictive-like behaviors. Although, the perisynaptic glial cells are implicated in metabolic maintenance and support of the nervous systems, accumulating evidence suggests that glial cells exert a modulatory action on synaptic functions and participate in synaptic plasticity. However, it is well-documented that glial cells are associated with the acquisition of rewarding effects of abused drugs. The role of hippocampal glial cells in addictive-like behaviors remains poorly understood. In this study, we investigated the role of hippocampal glial cells in morphine-induced behavioral responses including morphine dependence, tolerance to the antinociceptive properties of morphine, and conditioned place preference (CPP). Male rats received subcutaneous (s.c.) morphine sulfate (10 mg/kg) at an interval of 12 h for 9 days. To suppress glial cells activity, the animals received microinjection of fluorocitrate (FC, a metabolic inhibitor of glial cells) into the CA1 region before each morphine administration. The animals were assessed for morphine dependence by monitoring naloxone hydrochloride-induced precipitation of somatic signs of morphine withdrawal. The tolerance to the antinociceptive effects of morphine and morphine-induced CPP were measured in a separate set of experimental groups. We found animals receiving FC before morphine injection demonstrated a significant reduction in several signs of morphine withdrawal such as freezing, defecation, chewing, explosive running, ptosis, activity, scratching, wet dog shake, and writhing. Inhibition of glial cells caused a significant reduction of tolerance to the antinociceptive effect of morphine. Finally, intra-CA1 administration of FC decreased morphine-induced CPP. Our findings suggest that hippocampal glial cells may be involved in morphine-induced behavioral responses.


Asunto(s)
Conducta Animal/efectos de los fármacos , Hipocampo/citología , Hipocampo/efectos de los fármacos , Morfina/farmacología , Narcóticos/farmacología , Neuroglía/efectos de los fármacos , Animales , Citratos/farmacología , Condicionamiento Operante/efectos de los fármacos , Inyecciones Subcutáneas , Masculino , Dependencia de Morfina/tratamiento farmacológico , Dependencia de Morfina/etiología , Naloxona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Ratas , Ratas Wistar , Factores de Tiempo
19.
J Neurosci ; 26(21): 5720-6, 2006 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-16723528

RESUMEN

Substantial evidence indicates that the ventral tegmental area (VTA) of the mesocorticolimbic dopaminergic (DA) system has a key role in mechanisms of opiate dependence. Although DA neurons have been studied extensively, little is known about their activity and their response to acute morphine during morphine dependence. We recorded the activity of VTA DA neurons in five groups of anesthetized rats: drug-naive (naive) rats, morphine-dependent [(MD) implanted with pellets] rats, and three groups of withdrawn rats. Withdrawals either were precipitated by naltrexone or occurred spontaneously 24 h or 15 d after pellet removal. We confirmed that acute morphine in naive rats produced a marked increase in the firing of VTA DA neurons. We also found that the basal firing rate of VTA DA neurons was markedly higher in MD than in naive rats; however, in MD rats, acute morphine failed to increase DA activity. We confirmed inhibition of VTA DA activity in MD rats in response to precipitated withdrawal; however, this inhibition resulted only in a normalization of the firing rate to that of naive animals. In rats that had spontaneous withdrawal after 24 h or 15 d, the activity of VTA DA neurons was similar to that of naive rats, and an acute injection of morphine failed to alter their activity. Our results indicate that VTA DA neurons show long-lasting tolerance to the acute effect of morphine after withdrawal. These findings show that VTA DA neural activity is unlikely to be a factor in the altered behavioral responses that occur with acute morphine or naltrexone administration after chronic opiate exposure.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Dopamina/efectos adversos , Dopamina/metabolismo , Dependencia de Morfina/fisiopatología , Neuronas/fisiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Área Tegmental Ventral/fisiopatología , Animales , Dependencia de Morfina/etiología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Dopaminérgicos/metabolismo , Área Tegmental Ventral/efectos de los fármacos
20.
Neuropsychopharmacology ; 32(3): 589-99, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16823388

RESUMEN

The negative physical and affective aspects of opioid abstinence contribute to the prolongation of substance abuse. Withdrawal treatment is successful only in a subset of subjects, yet little is known about the neurobiological causes of these individual differences. Here, we compare the somatic and motivational components of opioid withdrawal in animals with high reactivity (HR) vs low reactivity (LR) to novelty, a phenotype associated with differential vulnerability to drug abuse. During withdrawal, HR relative to LR showed increased teeth chattering and eye twitching episodes, somatic signs associated with adrenergic modulation. Given the role of noradrenergic circuitry of the extended amygdala in opioid withdrawal, we examined adrenergic receptor gene expression in the bed nucleus of stria terminalis (BST) and central nucleus of the amygdala. Relative to LR, HR rats exhibit a selective increase in beta(1) adrenergic receptor expression in lateral and medial BST. To uncover the functional relevance of this difference, we microinjected betaxolol, a selective beta(1) receptor antagonist, into dorsal BST and assessed somatic and affective responses during withdrawal. Betaxolol microinjection dose-dependently decreased teeth chattering episodes in HR to levels observed in LR animals. Moreover, the antagonist blocked conditioned place aversion, a measure of negative affect associated with withdrawal, in HR but not in LR animals. Our results reveal for the first time that reactivity to novelty predicts somatic and affective aspects of opiate dependence, and that beta(1) receptors in BST are implicated in opiate withdrawal but only in novelty-seeking individuals.


Asunto(s)
Dependencia de Morfina/etiología , Morfina/efectos adversos , Narcóticos/efectos adversos , Receptores Adrenérgicos beta 1/metabolismo , Núcleos Septales/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Animales , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Betaxolol/farmacología , Relación Dosis-Respuesta a Droga , Masculino , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 1/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda