Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Neurosci ; 32(40): 13763-75, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23035088

RESUMEN

Recent evidence suggests that histone deacetylase (HDAC) inhibitors facilitate extinction of rewarding memory of drug taking. However, little is known about the role of chromatin modification in the extinction of aversive memory of drug withdrawal. In this study, we used conditioned place aversion (CPA), a highly sensitive model for measuring aversive memory of drug withdrawal, to investigate the role of epigenetic regulation of brain-derived neurotrophic factor (BDNF) gene expression in extinction of aversive memory. We found that CPA extinction training induced an increase in recruiting cAMP response element-binding protein (CREB) to and acetylation of histone H3 at the promoters of BDNF exon I transcript and increased BDNF mRNA and protein expression in the ventromedial prefrontal cortex (vmPFC) of acute morphine-dependent rats and that such epigenetic regulation of BDNF gene transcription could be facilitated or diminished by intra-vmPFC infusion of HDAC inhibitor trichostatin A or extracellular signal-regulated kinase (ERK) inhibitor U0126 (1,4-diamino-2,3-dicyano-1,4-bis(methylthio)butadiene) before extinction training. Correspondingly, disruption of the epigenetic regulation of BDNF gene transcription with U0126 or suppression of BDNF signaling with Trk receptor antagonist K252a or BDNF scavenger tyrosine kinase receptor B (TrkB)-Fc blocked extinction of CPA behavior. We also found that extinction training-induced activation of ERK and CREB and extinction of CPA behavior could be potentiated or suppressed by intra-vmPFC infusion of d-cycloserine, a NMDA receptor partial agonist or aminophosphonopentanoic acid, a NMDA receptor antagonist. We conclude that extinction of aversive memory of morphine withdrawal requires epigenetic regulation of BDNF gene transcription in the vmPFC through activation of the ERK-CREB signaling pathway perhaps in a NMDA receptor-dependent manner.


Asunto(s)
Aprendizaje por Asociación/fisiología , Reacción de Prevención/fisiología , Factor Neurotrófico Derivado del Encéfalo/genética , Represión Epigenética/fisiología , Extinción Psicológica/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Dependencia de Morfina/fisiopatología , Morfina/toxicidad , Corteza Prefrontal/fisiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Acetilación/efectos de los fármacos , Animales , Aprendizaje por Asociación/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Butadienos/farmacología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/fisiología , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Represión Epigenética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histonas/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor trkB/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología , Recompensa , Transcripción Genética
2.
J Neurosci ; 32(35): 12005-17, 2012 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-22933785

RESUMEN

Aversive memories associated with drug withdrawal may contribute to persistent drug seeking. Molecular mechanisms that are critical for aversive memory formation have yet to be elucidated. Recently, we showed in a rat conditioned place aversion (CPA) model that synaptic actin polymerization in the amygdala were required for aversive memory information. Here, we demonstrated that actin polymerization within the amygdala triggered transportation of activity-regulated cytoskeletal-associated protein (Arc/Arg3.1) into amygdalar synapses. Increased synaptic Arc/Arg3.1 expression contributed to aversive memory formation by regulating synaptic AMPA receptor (AMPAR) endocytosis, as in vivo knockdown of amygdalar Arc/Arg3.1 with Arc/Arg3.1-shRNA prevented both AMPAR endocytosis and CPA formation. We also demonstrated that conditioned morphine withdrawal led to induction of LTD in the amygdala through AMPAR endocytosis. We further demonstrated that Arc/Arg3.1-regulated AMPAR endocytosis was GluR2 dependent, as intra-amygdala injection of Tat-GluR2(3Y), a GluR2-derived peptide that has been shown to specifically block regulated, but not constitutive, AMPAR endocytosis, prevented AMPAR endocytosis, LTD induction, and aversive memory formation. Therefore, this study extends previous studies on the role of actin polymerization in synaptic plasticity and memory formation by revealing the critical molecular events involved in aversive memory formation as well as LTD induction, and by showing that Arc/Arg3.1 is a crucial mediator for actin polymerization functions, and, thus, underscores the unknown details of how actin polymerization mediates synaptic plasticity and memory.


Asunto(s)
Actinas/metabolismo , Amígdala del Cerebelo/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Memoria/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Síndrome de Abstinencia a Sustancias/metabolismo , Sinapsis/metabolismo , Regulación hacia Arriba , Actinas/genética , Animales , Reacción de Prevención , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/deficiencia , Endocitosis/genética , Células HEK293 , Humanos , Morfina/efectos adversos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/deficiencia , Plasticidad Neuronal/genética , Polimerizacion , Ratas , Ratas Sprague-Dawley , Sinapsis/genética , Regulación hacia Arriba/genética
3.
J Cell Sci ; 123(Pt 24): 4259-70, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21098639

RESUMEN

Distinct opioid receptor agonists have been proved to induce differential patterns of ERK activation, but the underlying mechanisms remain unclear. Here, we report that Ser363 in the δ-opioid receptor (δOR) determines the different abilities of the δOR agonists DPDPE and TIPP to activate ERK by G-protein- or ß-arrestin-dependent pathways. Although both DPDPE and TIPP activated ERK1/2, they showed different temporal, spatial and desensitization patterns of ERK activation. We show that that DPDPE employed G protein as the primary mediator to activate the ERK cascade in an Src-dependent manner, whereas TIPP mainly adopted a ß-arrestin1/2-mediated pathway. Moreover, we found that DPDPE gained the capacity to adopt the ß-arrestin1/2-mediated pathway upon Ser363 mutation, accompanied by the same pattern of ERK activation as that induced by TIPP. Additionally, we found that TIPP- but not DPDPE-activated ERK could phosphorylate G-protein-coupled receptor kinase-2 and ß-arrestin1. However, such functional differences of ERK disappeared with the mutation of Ser363. Therefore, the present study reveals a crucial role for Ser363 in agonist-specific regulation of ERK activation patterns and functions.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores Opioides delta/metabolismo , Serina/metabolismo , Animales , Arrestinas/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/enzimología , Encefalina D-Penicilamina (2,5)/farmacología , Activación Enzimática/efectos de los fármacos , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Ligandos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Proteínas Mutantes/metabolismo , Mutación/genética , Oligopéptidos/farmacología , Fosfolipasa C beta/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Relación Estructura-Actividad , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Tetrahidroisoquinolinas/farmacología , beta-Arrestinas
4.
J Neurosci ; 30(14): 5058-70, 2010 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-20371826

RESUMEN

Chronic exposure to opiates impairs hippocampal long-term potentiation (LTP) and spatial memory, but the underlying mechanisms remain to be elucidated. Given the well known effects of adenosine, an important neuromodulator, on hippocampal neuronal excitability and synaptic plasticity, we investigated the potential effect of changes in adenosine concentrations on chronic morphine treatment-induced impairment of hippocampal CA1 LTP and spatial memory. We found that chronic treatment in mice with either increasing doses (20-100 mg/kg) of morphine for 7 d or equal daily dose (20 mg/kg) of morphine for 12 d led to a significant increase of hippocampal extracellular adenosine concentrations. Importantly, we found that accumulated adenosine contributed to the inhibition of the hippocampal CA1 LTP and impairment of spatial memory retrieval measured in the Morris water maze. Adenosine A(1) receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine significantly reversed chronic morphine-induced impairment of hippocampal CA1 LTP and spatial memory. Likewise, adenosine deaminase, which converts adenosine into the inactive metabolite inosine, restored impaired hippocampal CA1 LTP. We further found that adenosine accumulation was attributable to the alteration of adenosine uptake but not adenosine metabolisms. Bidirectional nucleoside transporters (ENT2) appeared to play a key role in the reduction of adenosine uptake. Changes in PKC-alpha/beta activity were correlated with the attenuation of the ENT2 function in the short-term (2 h) but not in the long-term (7 d) period after the termination of morphine treatment. This study reveals a potential mechanism by which chronic exposure to morphine leads to impairment of both hippocampal LTP and spatial memory.


Asunto(s)
Adenosina/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Aprendizaje por Laberinto/fisiología , Morfina/administración & dosificación , Receptor de Adenosina A1/metabolismo , Adenosina/toxicidad , Agonistas del Receptor de Adenosina A1 , Animales , Esquema de Medicación , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Morfina/toxicidad , Conducta Espacial/efectos de los fármacos , Conducta Espacial/fisiología
5.
J Neurosci ; 29(39): 12244-54, 2009 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-19793983

RESUMEN

Aversive memories of drug withdrawal can generate a motivational state leading to compulsive drug taking. Changes in synaptic plasticity may be involved in the formation of aversive memories. Dynamic rearrangement of the cytoskeletal actin, a major structural component of the dendritic spine, regulates synaptic plasticity. Here, the potential involvement of actin rearrangements in the induction of aversive memories of morphine withdrawal was examined. We found that lesions of the amygdala or dorsal hippocampus (DH) but not nucleus accumbens (NAc) impaired conditioned place aversion (CPA) of acute morphine-dependent rats. Accordingly, conditioned morphine withdrawal induced actin rearrangements in the amygdala and the DH but not in the NAc. In addition, we found that conditioned morphine withdrawal also increased activity-regulated cytoskeletal-associated protein (Arc) expression in the amygdala but not in the DH, although actin rearrangements were observed in both areas. We further found that inhibition of actin rearrangements by intra-amygdala or intra-DH injections of latrunculin A, an inhibitor of actin polymerization, significantly attenuated CPA. Furthermore, we found that manipulation of amygdala beta-adrenoceptor activity by its antagonist propranolol and agonist clenbuterol differentially altered actin rearrangements in the DH. Therefore, our findings reveal that actin rearrangements in the amygdala and the DH are required for the acquisition and consolidation of the aversive memories of drug withdrawal and that the beta-noradrenergic system within the amygdala modulates aversive memory consolidation by regulating actin rearrangements but not Arc protein expression in the DH, which is distinct from its role in modulation of inhibitory avoidance memory.


Asunto(s)
Actinas/metabolismo , Amígdala del Cerebelo/patología , Reacción de Prevención/fisiología , Hipocampo/patología , Dependencia de Morfina/patología , Síndrome de Abstinencia a Sustancias/patología , Actinas/química , Amígdala del Cerebelo/química , Amígdala del Cerebelo/metabolismo , Animales , Hipocampo/química , Hipocampo/metabolismo , Masculino , Memoria/fisiología , Morfina/administración & dosificación , Ratas , Ratas Sprague-Dawley
6.
Acta Pharmacol Sin ; 31(9): 1065-70, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20729876

RESUMEN

The kappa-opioid receptor (KOR), a member of the opioid receptor family, is widely expressed in the central nervous system and peripheral tissues. Substantial evidence has shown that activation of KOR by agonists and endogenous opioid peptides in vivo may produce a strong analgesic effect that is free from the abuse potential and the adverse side effects of mu-opioid receptor (MOR) agonists, such as morphine. In addition, activation of the KOR has also been shown to exert an inverse effect on morphine-induced adverse actions, such as tolerance, reward, and impairment of learning and memory. Therefore, the KOR has received much attention in the effort to develop alternative analgesics to MOR agonists and agents for the treatment of drug addiction. However, KOR agonists also produce several severe undesirable side effects such as dysphoria, water diuresis, salivation, emesis, and sedation in nonhuman primates, which may limit the clinical utility of KOR agonists for pain and drug abuse treatment. This article will review the role of KOR activation in mediating antinociception and addiction. The possible therapeutic application of kappa-agonists in the treatment of pain and drug addiction is also discussed.


Asunto(s)
Analgésicos/uso terapéutico , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Analgésicos/farmacología , Animales , Humanos , Dolor/tratamiento farmacológico
7.
Acta Pharmacol Sin ; 31(4): 393-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20228826

RESUMEN

AIM: To examine the relationship between the RAVE (relative activity versus endocytosis) values of opiate agonists and their dependence liability by studying several potent analgesics with special profiles in the development of physical and psychological dependence. METHODS: The effects of (-)-cis-(3R,4S,2'R) ohmefentanyl (F9202), (+)-cis-(3R,4S,2'S) ohmefentanyl (F9204), dihydroetorphine (DHE) and morphine on [(35)S]GTP gamma S binding, forskolin-stimulated cAMP accumulation, and receptor internalization were studied in CHO cells stably expressing HA-tagged mu-opioid receptors (CHO-HA-MOR). cAMP overshoot in response to the withdrawal of these compound treatments was also tested. RESULTS: All four agonists exhibited the same rank order of activity in stimulation of [(35)S]GTP gamma S binding, inhibition of adenylyl cyclase (AC) and induction of receptor internalization: DHE>F9204>F9202>morphine. Based on these findings and the previous in vivo analgesic data obtained from our and other laboratories, the RAVE values of the four agonists were calculated. The rank order of RAVE values was morphine>F9202>F9204>DHE. For the induction of cAMP overshoot, the rank order was F9202>or=morphine>F9204>or=DHE. CONCLUSION: Taken in combination with previous findings of these compounds' liability to develop dependence, the present study suggests that the agonist with the highest RAVE value seems to have a relatively greater liability to develop psychological dependence relative to the agonist with the lowest RAVE value. However, the RAVE values of these agonists are not correlated with their probability of developing physical dependence or inducing cAMP overshoot, a cellular hallmark of dependence.


Asunto(s)
Analgésicos Opioides/farmacología , Etorfina/análogos & derivados , Fentanilo/análogos & derivados , Morfina/farmacología , Receptores Opioides mu/metabolismo , Animales , Células CHO , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colforsina/metabolismo , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Endocitosis/efectos de los fármacos , Etorfina/farmacología , Fentanilo/farmacología , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Trastornos Relacionados con Opioides/metabolismo , Receptores Opioides mu/agonistas
8.
Acta Pharmacol Sin ; 31(4): 387-92, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20228831

RESUMEN

AIM: To evaluate the influence of an initial heroin experience under a modified two-chained training schedule on drug-seeking behavior after a long abstinence period. METHODS: Rats were trained to respond for intravenous heroin (120 microg/kg) under a heterogeneous chained schedule of reinforcement using different responses in the first and second links of the chain. Animals received low-frequency drug administration training for four days and were then subjected to one month of abstinence in their home cages. Heroin-seeking behavior induced by re-exposure to the first chain associated context or discriminative stimuli was assessed after abstinence. RESULTS: Almost all animals could acquire operant skills quickly under the two-chained schedule training for four days, as measured in first active response latency, travel speed and goal-box enter latency. Both first chain associated context and discriminative stimulus could reinstate heroin-seeking behavior after one month abstinence. CONCLUSION: These observations suggest that an early experience of drug use is sufficient to maintain heroin-seeking behavior even after a one month abstinence.


Asunto(s)
Dependencia de Heroína/etiología , Heroína/administración & dosificación , Narcóticos/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Heroína/farmacología , Masculino , Narcóticos/farmacología , Ratas , Ratas Sprague-Dawley
9.
Acta Pharmacol Sin ; 31(12): 1547-52, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21102484

RESUMEN

AIM: to investigate the effects of ATPM-ET [(-)-3-N-Ethylaminothiazolo [5,4-b]-N-cyclopropylmethylmorphinan hydrochloride] on physical dependence and behavioral sensitization to morphine in mice. METHODS: the pharmacological profile of ATPM-ET was characterized using competitive binding and GTPγS binding assays. We then examined the antinociceptive effects of ATPM-ET in the hot plate test. Morphine dependence assay and behavioral sensitization assay were used to determine the effect of ATPM-ET on physical dependence and behavior sensitization to morphine in mice. RESULTS: the binding assay indicated that ATPM-ET ATPM-ET exhibited a high affinity to both κ- and µ-opioid receptors with K(i) values of 0.15 nmol/L and 4.7 nmol/L, respectively, indicating it was a full κ-opioid receptor agonist and a partial µ-opioid receptor agonist. In the hot plate test, ATPM-ET produced a dose-dependent antinociceptive effect, with an ED(50) value of 2.68 (2.34-3.07) mg/kg. Administration of ATPM-ET (1 and 2 mg/kg, sc) prior to naloxone (3.0 mg/kg, sc) injection significantly inhibited withdrawal jumping of mice. In addition, ATPM-ET (1 and 2 mg/kg, sc) also showed a trend toward decreasing morphine withdrawal-induced weight loss. ATPM-ET (1.5 and 3 mg/kg, sc) 15 min before the morphine challenge significantly inhibited the morphine-induced behavior sensitization (P<0.05). CONCLUSION: ATPM-ET may have potential as a therapeutic agent for the treatment of drug abuse.


Asunto(s)
Conducta Animal/efectos de los fármacos , Morfinanos/farmacología , Dependencia de Morfina/tratamiento farmacológico , Morfina/farmacología , Narcóticos/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Animales , Relación Dosis-Respuesta a Droga , Calor , Masculino , Ratones , Dependencia de Morfina/metabolismo , Dependencia de Morfina/psicología , Actividad Motora/efectos de los fármacos , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Dimensión del Dolor , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo
10.
Acta Pharmacol Sin ; 31(7): 784-90, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20562901

RESUMEN

AIM: To define the effect of adenosine A(1) receptor (A(1)R) on delta opioid receptor (DOR)-mediated signal transduction. METHODS: CHO cells stably expressing HA-tagged A(1)R and DOR-CFP fusion protein were used. The localization of receptors was observed using confocal microscope. DOR-mediated inhibition of adenylyl cyclase was measured using cyclic AMP assay. Western blots were employed to detect the phosphorylation of Akt and the DOR. The effect of A(1)R agonist N(6)-cyclohexyladenosine (CHA) on DOR down-regulation was assessed using radioligand binding assay. RESULTS: CHA 1 micromol/L time-dependently attenuated DOR agonist [D-Pen(2,5)]enkephalin (DPDPE)-induced inhibition of intracellular cAMP accumulation with a t(1/2)=2.56 (2.09-3.31) h. Pretreatment with 1 micromol/L CHA for 24 h caused a right shift of the dose-response curve of DPDPE-mediated inhibition of cAMP accumulation, with a significant increase in EC(50) but no change in E(max). Pretreatment with 1 micromol/L CHA for 1 h also induced a significant attenuation of DPDPE-stimulated phosphorylation of Akt. Moreover, CHA time-dependently phosphorylated DOR (Ser363), and this effect was inhibited by A(1)R antagonist 1,3-Dipropyl-8-cyclopentylxanthine (DPCPX) but not by DOR antagonist naloxone. However, CHA failed to produce the down-regulation of DOR, as neither receptor affinity (K(d)) nor receptor density (B(max)) of DOR showed significant change after chronic CHA exposure. CONCLUSION: Activation of A(1)R by its agonist caused heterologous desensitization of DOR-mediated inhibition of intracellular cAMP accumulation and phosphorylation of Akt. Activation of A(1)R by its agonist also induced heterologous phosphorylation but not down-regulation of DOR.


Asunto(s)
Agonistas del Receptor de Adenosina A1 , Adenosina/análogos & derivados , Receptores Opioides delta/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Adenosina/farmacología , Animales , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Semivida , Humanos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayo de Unión Radioligante , Receptores Opioides delta/metabolismo , Factores de Tiempo
11.
J Neurochem ; 108(1): 102-14, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19014372

RESUMEN

The opioid receptors are a member of G protein-coupled receptors that mediate physiological effects of endogenous opioid peptides and structurally distinct opioid alkaloids. Although it is well characterized that there is differential receptor desensitization and internalization properties following activation by distinct agonists, the underlying mechanisms remain elusive. We investigated the signaling events of delta-opioid receptor (deltaOR) initiated by two ligands, DPDPE and TIPP. We found that although both ligands inhibited adenylyl cyclase (AC) and activated ERK1/2, only DPDPE induced desensitization and internalization of the deltaOR. We further found that DPDPE, instead of TIPP, could activate GRK2 by phosphorylating the non-receptor tyrosine kinase Src and translocating it to membrane receptors. Activation of GRK2 led to the phosphorylation of serine residues in the C-terminal tail, which facilitates beta-arrestin1/2 membrane translocation. Meanwhile, we also found that DPDPE promoted beta-arrestin1 dephosphorylation in a Src-dependent manner. Thus, DPDPE appears to strengthen beta-arrestin function by dual regulations: promoting beta-arrestin recruitment and increasing beta-arrestin dephosphorylation at the plasma membrane in a Src-dependent manner. All effects initiated by DPDPE could be abolished or suppressed by PP2, an inhibitor of Src. Morphine, which has been previously shown to be unable to desensitize or internalize deltaOR, also behaved as TIPP in failure to utilize Src to regulate deltaOR signaling. These findings point to the existence of agonist-specific utilization of Src to regulate deltaOR signaling and reveal the molecular events by which Src modulates deltaOR responsiveness.


Asunto(s)
Receptores Opioides delta/metabolismo , Transducción de Señal/fisiología , Familia-src Quinasas/fisiología , Adenilil Ciclasas/metabolismo , Analgésicos Opioides/farmacología , Animales , Arrestinas/metabolismo , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Encefalina D-Penicilamina (2,5)/farmacología , Citometría de Flujo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfina/farmacología , Oligopéptidos/farmacología , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Tetrahidroisoquinolinas/farmacología , Transfección , beta-Arrestinas
12.
J Pharmacol Exp Ther ; 329(1): 306-13, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19136637

RESUMEN

ATPM [(-)-3-amino-thiazolo[5,4-b]-N-cyclopropylmethylmorphinan hydrochloride] was found to have mixed kappa- and mu-opioid activity and identified to act as a full kappa-agonist and a partial mu-agonist by in vitro binding assays. The present study was undertaken to characterize its in vivo effects on morphine antinociceptive tolerance in mice and heroin self-administration in rats. ATPM was demonstrated to yield more potent antinociceptive effects than (-)U50,488H (trans-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamide). It was further found that the antinociceptive effects of ATPM were mediated by kappa- and mu-, but not delta-opioid, receptors. In addition to its agonist profile on the mu-receptor, ATPM also acted as a mu-antagonist, as measured by its inhibition of morphine-induced antinociception. It is more important that ATPM had a greater ratio of the ED(50) value of sedation to that of antinociception than (-)U50,488 (11.8 versus 3.7), indicative of a less sedative effect than (-)U50,488H. In addition, ATPM showed less potential to develop antinociceptive tolerance relative to (-)U50,488H and morphine. Moreover, it dose-dependently inhibited morphine-induced antinociceptive tolerance. Furthermore, it was found that chronic treatment of rats for 8 consecutive days with ATPM (0.5 mg/kg s.c.) produced sustained decreases in heroin self-administration. (-)U50,488H (2 mg/kg s.c.) also produced similar inhibitory effect. Taken together, our findings demonstrated that ATPM, a novel mixed kappa-agonist and mu-agonist/-antagonist, could inhibit morphine-induced antinociceptive tolerance, with less potential to develop tolerance and reduce heroin self-administration with less sedative effect. kappa-Agonists with some mu-activity appear to offer some advantages over selective kappa-agonists for the treatment of heroin abuse.


Asunto(s)
Analgésicos Opioides/antagonistas & inhibidores , Analgésicos Opioides/farmacología , Dependencia de Heroína/tratamiento farmacológico , Dependencia de Heroína/psicología , Morfinanos/farmacología , Morfina/antagonistas & inhibidores , Morfina/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Músculos Abdominales/efectos de los fármacos , Analgésicos no Narcóticos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Calor , Masculino , Ratones , Dimensión del Dolor/efectos de los fármacos , Equilibrio Postural/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Receptores Opioides delta/efectos de los fármacos , Autoadministración
13.
Acta Pharmacol Sin ; 30(3): 282-90, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19262551

RESUMEN

AIM: The immediate early gene Arc (activity-regulated cytoskeletal-associated protein) mRNA and protein are induced by strong synaptic activation and rapidly transported into dendrites, where they localize at active synaptic sites. Thus, the Arc mRNA and protein are proposed as a marker of neuronal reactivity to map the neural substrates that are recruited by various stimuli. In the present study, we examined the expression of Arc protein induced by conditioned naloxone-precipitated drug withdrawal in different brain regions of acute morphine-dependent rats. The objective of the present study was to address the specific neural circuits involved in conditioned place aversion (CPA) that has not yet been well characterized. METHODS: Place aversion was elicited by conditioned naloxone-precipitated drug withdrawal following exposure to a single dose of morphine. An immunohistochemical method was employed to detect the expression of Arc, which was used as a plasticity marker to trace the brain areas that contribute to the formation of the place aversion. RESULTS: Marked increases in Arc protein levels were found in the medial and lateral prefrontal cortex, the sensory cortex, the lateral striatum and the amygdala. This effect was more pronounced in the basolateral amygdala (BLA), the central nucleus of the amygdala (CeA), and the bed nucleus of the striatal terminals (BNST) when compared with the control group. CONCLUSION: Our results suggest that these brain regions may play key roles in mediating the negative motivational component of opiate withdrawal.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/metabolismo , Condicionamiento Psicológico/fisiología , Proteínas del Citoesqueleto/metabolismo , Dependencia de Morfina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Proteínas del Citoesqueleto/genética , Humanos , Masculino , Morfina/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Proteínas del Tejido Nervioso/genética , Ratas , Ratas Sprague-Dawley
14.
Acta Pharmacol Sin ; 30(10): 1385-91, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19767765

RESUMEN

AIM: To evaluate the role of glutamate receptors in the dorsal hippocampus (DH) in the motivational component of morphine withdrawal. METHODS: NMDA receptor antagonist D-AP5 (5 microg/0.5 microL per side) or AMPA receptor antagonist NBQX (2 microg/0.5 microL per side) was microinjected into DH of rats. Conditioned place aversion (CPA) induced by naloxone-precipitated morphine withdrawal were assessed. RESULTS: Preconditioning microinjection of D-AP5 or NBQX into the DH impaired the acquisition of CPA in acute morphine-dependent rats. However, intra-DH microinjection of D-AP5 or NBQX after conditioning but before the testing session had no effect on the expression of CPA. CONCLUSION: Our results suggest that NMDA and AMPA receptors in the dorsal hippocampus are involved in the acquisition, but not in the expression, of the negative motivational components of acute morphine withdrawal in rats.


Asunto(s)
Condicionamiento Psicológico/efectos de los fármacos , Hipocampo/metabolismo , Morfina/farmacología , Receptores de Glutamato/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Masculino , Microinyecciones , Ratas , Ratas Sprague-Dawley
15.
Eur J Pharmacol ; 584(2-3): 306-11, 2008 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-18353307

RESUMEN

Analgesics such as morphine cause many side effects including addiction, but kappa-opioid receptor agonist can produce antinociception without morphine-like side effects. With the aim of developing new and potent analgesics with lower abuse potential, we studied the antinociceptive and physical dependent properties of a derivate of ICI-199441, an analogue of (-)U50,488H, named (2-(3,4-dichloro)-phenyl)-N-methyl-N-[(1S)-1-(2-isopropyl)-2-(1-(3-pyrrolinyl))ethyl] acetamides (LPK-26). LPK-26 showed a high affinity to kappa-opioid receptor with the Ki value of 0.64 nM and the low affinities to micro-opioid receptor and delta-opioid receptor with the Ki values of 1170 nM and >10,000 nM, respectively. It stimulated [(35)S]GTPgammaS binding to G-proteins with an EC50 value of 0.0094 nM. In vivo, LPK-26 was more potent than (-)U50,488H and morphine in analgesia, with the ED50 values of 0.049 mg/kg and 0.0084 mg/kg in hot plat and acetic acid writhing tests, respectively. Moreover, LPK-26 failed to induce physical dependence, but it could suppress naloxone-precipitated jumping in mice when given simultaneously with morphine. Taken together, our results show that LPK-26 is a novel selective kappa-opioid receptor agonist with highly potent antinociception effects and low physical dependence potential. It may be valuable for the development of analgesic and drug that can be used to reduce morphine-induced physical dependence.


Asunto(s)
Analgésicos Opioides/farmacología , Trastornos Relacionados con Opioides/prevención & control , Umbral del Dolor/efectos de los fármacos , Dolor/prevención & control , Pirroles/farmacología , Receptores Opioides kappa/agonistas , Ácido Acético , Animales , Conducta Animal/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Calor/efectos adversos , Humanos , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Dolor/etiología , Dolor/fisiopatología , Dimensión del Dolor , Unión Proteica , Ratas , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Factores de Tiempo , Transfección
16.
Biochem J ; 406(2): 215-21, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17542780

RESUMEN

Morphine is recommended as a first-line opioid analgesic in the pain management of cancer patients. Accumulating evidence shows that morphine has anti-apoptotic activity, but its impact on the therapeutic applications of antineoplastic drugs is not well known. The present study was undertaken to test the hypothesis that morphine might antagonize the pro-apoptotic activity of DOX (doxorubicin), a commonly used antitumour drug for the treatment of neuroblastoma, in cultured SH-SY5Y cells. In the present study we demonstrated that morphine suppressed DOX-induced inhibition of cell proliferation and programmed cell death in a concentration-dependent, and naloxone as well as pertussis toxin-irreversible, manner. Further studies showed that morphine inhibited ROS (reactive oxygen species) generation, and prevented DOX-mediated caspase-3 activation, cytochrome c release and changes of Bax and Bcl-2 protein expression. The antioxidant NAC (N-acetylcysteine) also showed the same effects as morphine on DOX-induced ROS generation, caspase-3 activation and cytochrome c release and changes in Bax (Bcl-2-associated X protein) and Bcl-2 protein expression. Additionally, morphine was found to suppress DOX-induced NF-kappaB (nuclear factor kappaB) transcriptional activation via a reduction of IkappaBalpha (inhibitor of nuclear factor kappaB) degradation. These present findings support the hypothesis that morphine can inhibit DOX-induced neuroblastoma cell apoptosis by the inhibition of ROS generation and mitochondrial cytochrome c release, as well as by blockade of NF-kappaB transcriptional activation, and suggests that morphine might have an impact on the antitumour efficiency of DOX.


Asunto(s)
Doxorrubicina/antagonistas & inhibidores , Morfina/farmacología , FN-kappa B/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Activación Transcripcional/efectos de los fármacos , Acetilcisteína/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Citoprotección/efectos de los fármacos , Doxorrubicina/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Quinasa I-kappa B/metabolismo , Neuroblastoma/patología , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Activación Transcripcional/genética
17.
Br J Pharmacol ; 174(17): 2842-2861, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28608532

RESUMEN

BACKGROUND AND PURPOSE: Exposure to opiates induces locomotor sensitization in rodents, which has been proposed to correspond to the compulsive drug-seeking behaviour. Numerous studies have demonstrated that locomotor sensitization can occur in a dopamine transmission-independent manner; however, the underlying mechanisms are unclear. EXPERIMENTAL APPROACH: Co-immunoprecipitation, BRET and cross-antagonism assays were used to demonstrate the existence of receptor heterodimers. Function of heterodimers was evaluated by behavioural studies of locomotor sensitization. KEY RESULTS: The dopamine D1 receptor antagonist SCH23390 antagonized the signalling initiated by stimulation of µ opioid receptors with agonists in transfected cells expressing two receptors and in striatal tissues from wild-type but not D1 receptor knockout (KO) mice, suggesting that SCH23390 modified µ receptor function via receptor heteromers, as the ability of an antagonist of one of the receptors to inhibit signals originated by stimulation of the partner receptor was a characteristic of receptor heteromers. The existence of µ receptor-D1 receptor heterodimers was further supported by biochemical and biophysical assays. In vivo, when dopamine release was absent (by destruction of the dopaminergic projection from the ventral tegmental area to the striatum), SCH23390 still significantly inhibited µ receptor agonist-induced behavioural responses in rats. Additionally, we demonstrated that D1 or µ receptor KO mice and thus unable to form µ receptor-D1 receptor heterodimers, failed to show locomotor sensitization to morphine. CONCLUSION AND IMPLICATIONS: Our results suggest that µ receptor-D1 receptor heterodimers may be involved in the dopamine-independent expression of locomotor sensitization to opiates.


Asunto(s)
Analgésicos Opioides/farmacología , Benzazepinas/farmacología , Antagonistas de Dopamina/farmacología , Receptores de Dopamina D1/metabolismo , Receptores Opioides mu/metabolismo , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Dopamina , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HEK293 , Humanos , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Receptores de Dopamina D1/genética
18.
Neuropharmacology ; 110(Pt A): 92-101, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27106167

RESUMEN

κ Opioid receptor system is widely implicated in the regulation of emotion. However, the findings about the role on anxiety in rodents are highly controversial, since both anxiogenic- and anxiolytic-like effects have been reported with κ opioid receptor activation. The mechanism and the underlying neuroanatomical substrates are unexplored. In the present study, we first investigated the effects of κ agonist U50,488H on anxiety-related behaviors over a wide range of doses, and we found that U50,488H produced dual effects in anxiety, with low dose being anxiogenic and high dose being anxiolytic. To assess the potential neuroanatomical substrates, we used phosphorylation of extracellular signal-related kinase1/2 (pERK1/2) to map the underlying neural circuits. We found that the anxiogenic effect of U50,488H was paralleled by an increase of pERK1/2 in the nucleus accumbens, whereas the anxiolytic effect was paralleled by an increase of pERK1/2 in the lateral septal nucleus. We then examined the behavioral consequences with locally microinjection of U50,488H, and we found that microinjection of U50,488H into the nucleus accumbens exerted anxiogenic-like effects, whereas microinjection of U50,488H into the lateral septal nucleus. Both effects can be abolished by κ antagonist nor-BNI pretreatment. To the best of our knowledge, the present work firstly provides the neuroanatomical sites that mediating the dual anxiogenic- and anxiolytic-like effects of U50,488H in mice. This study may help to explain current controversial role of κ receptor activation in anxiety-related behaviors in rodents, and may open new perspectives in the areas of anxiety disorders and κ receptor function.


Asunto(s)
Ansiedad/metabolismo , Encéfalo/metabolismo , Receptores Opioides kappa/metabolismo , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/administración & dosificación , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/toxicidad , Analgésicos no Narcóticos/administración & dosificación , Analgésicos no Narcóticos/toxicidad , Animales , Ansiedad/inducido químicamente , Ansiedad/prevención & control , Encéfalo/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Microinyecciones , Antagonistas de Narcóticos/administración & dosificación , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores
19.
Br J Pharmacol ; 172(20): 4847-63, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26211551

RESUMEN

BACKGROUND AND PURPOSE: ß-Arrestins function as signal transducers linking GPCRs to ERK1/2 signalling either by scaffolding members of ERK1/2s cascades or by transactivating receptor tyrosine kinases through Src-mediated release of transactivating factor. Recruitment of ß-arrestins to the activated GPCRs is required for ERK1/2 activation. Our previous studies showed that δ receptors activate ERK1/2 through a ß-arrestin-dependent mechanism without inducing ß-arrestin binding to the δ receptors. However, the precise mechanisms involved remain to be established. EXPERIMENTAL APPROACH: ERK1/2 activation by δ receptor ligands was assessed using HEK293 cells in vitro and male Sprague Dawley rats in vivo. Immunoprecipitation, immunoblotting, siRNA transfection, intracerebroventricular injection and immunohistochemistry were used to elucidate the underlying mechanism. KEY RESULTS: We identified a new signalling pathway in which recruitment of ß-arrestin2 to the EGFR rather than δ receptor was required for its role in δ receptor-mediated ERK1/2 activation in response to H-Tyr-Tic-Phe-Phe-OH (TIPP) or morphine stimulation. Stimulation of the δ receptor with ligands leads to the phosphorylation of PKCδ, which acts upstream of EGFR transactivation and is needed for the release of the EGFR-activating factor, whereas ß-arrestin2 was found to act downstream of the EGFR transactivation. Moreover, we demonstrated that coupling of the PKCδ/EGFR/ß-arrestin2 transactivation pathway to δ receptor-mediated ERK1/2 activation was ligand-specific and the Ser(363) of δ receptors was crucial for ligand-specific implementation of this ERK1/2 activation pathway. CONCLUSIONS AND IMPLICATIONS: The δ receptor-mediated activation of ERK1/2 is via ligand-specific transactivation of EGFR. This study adds new insights into the mechanism by which δ receptors activate ERK1/2.


Asunto(s)
Arrestinas/metabolismo , Receptores ErbB/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa C-delta/metabolismo , Receptores Opioides delta/metabolismo , Analgésicos Opioides/farmacología , Animales , Receptores ErbB/genética , Células HEK293 , Humanos , Masculino , Morfina/farmacología , Oligopéptidos/farmacología , Ratas Sprague-Dawley , Tetrahidroisoquinolinas/farmacología , Activación Transcripcional , beta-Arrestinas
20.
Br J Pharmacol ; 172(2): 482-91, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24597568

RESUMEN

BACKGROUND AND PURPOSE: Substantial evidence shows that negative reinforcement resulting from the aversive affective consequences of opiate withdrawal may play a crucial role in drug relapse. Understanding the mechanisms underlying the loss (extinction) of conditioned aversion of drug withdrawal could facilitate the treatment of drug addiction. EXPERIMENTAL APPROACH: Naloxone-induced conditioned place aversion (CPA) of Sprague-Dawley rats was used to measure conditioned aversion. An NMDA receptor antagonist and MAPK kinase inhibitor were applied through intracranial injections. The phosphorylation of ERK and cAMP response element-binding protein (CREB) was detected using Western blot. KEY RESULTS: The extinction of CPA behaviour increased the phosphorylation of ERK and CREB in the dorsal hippocampus (DH) and basolateral amygdala (BLA), but not in the central amygdala (CeA). Intra-DH injection of AP5 or intra-BLA injection of AP-5 or U0126 before extinction training significantly attenuated ERK and CREB phosphorylation in the BLA and impaired the extinction of CPA behaviour. Although intra-DH injections of AP-5 attenuated extinction training-induced activation of the ERK-CREB pathway in the BLA, intra-BLA injection of AP5 had no effect on extinction training-induced activation of the ERK-CREB pathway in the DH. CONCLUSIONS AND IMPLICATIONS: These results suggest that activation of ERK and CREB in the BLA and DH is involved in the extinction of CPA behaviour and that the DH, via a direct or indirect pathway, modulates the activity of ERK and CREB in the BLA through activation of NMDA receptors after extinction training. Understanding the mechanisms underlying the extinction of conditioned aversion could facilitate the treatment of drug addiction. LINKED ARTICLES: This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.


Asunto(s)
Reacción de Prevención/fisiología , Complejo Nuclear Basolateral/fisiología , Condicionamiento Psicológico/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Analgésicos Opioides/farmacología , Animales , Reacción de Prevención/efectos de los fármacos , Complejo Nuclear Basolateral/efectos de los fármacos , Complejo Nuclear Basolateral/metabolismo , Butadienos/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Masculino , Morfina/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA