Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Acta Pharmacol Sin ; 35(6): 738-51, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24858313

RESUMEN

AIM: 3-Methyl-6-chloro-7,8-hydroxy-1-(3-methylphenyl)-2,3,4,5-tetrahydro-1H-3-benzazepine (SKF83959) have been shown to affect several types of voltage-dependent channels in hippocampal pyramidal neurons. The aim of this study was to determine how modulation of a individual type of the channels by SKF83959 contributes to the overall excitability of CA1 pyramidal neurons during either direct current injections or synaptic activation. METHODS: Rat hippocampal slices were prepared. The kinetics of voltage-dependent Na(+) channels and neuronal excitability and depolarization block in CA1 pyramidal neurons were examined using whole-cell recording. A realistic mathematical model of hippocampal CA1 pyramidal neuron was used to simulate the effects of SKF83959 on neuronal excitability. RESULTS: SKF83959 (50 µmol/L) shifted the inactivation curve of Na(+) current by 10.3 mV but had no effect on the activation curve in CA1 pyramidal neurons. The effects of SKF83959 on passive membrane properties, including a decreased input resistance and depolarized resting potential, predicted by our simulations were in agreement with the experimental data. The simulations showed that decreased excitability of the soma by SKF83959 (examined with current injection at the soma) was only observed when the membrane potential was compensated to the control levels, whereas the decreased dendritic excitability (examined with current injection at the dendrite) was found even without membrane potential compensation, which led to a decreased number of action potentials initiated at the soma. Moreover, SKF83959 significantly facilitated depolarization block in CA1 pyramidal neurons. SKF83959 decreased EPSP temporal summation and, of physiologically greater relevance, the synaptic-driven firing frequency. CONCLUSION: SKF83959 decreased the excitability of CA1 pyramidal neurons even though the drug caused the membrane potential depolarization. The results may reveal a partial mechanism for the drug's anti-Parkinsonian effects and may also suggest that SKF83959 has a potential antiepileptic effect.


Asunto(s)
2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , Agonistas de Dopamina/farmacología , Hipocampo/citología , Células Piramidales/efectos de los fármacos , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Modelos Neurológicos , Células Piramidales/citología , Células Piramidales/metabolismo , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Transmisión Sináptica/efectos de los fármacos , Canales de Sodio Activados por Voltaje/metabolismo
2.
Acta Pharmacol Sin ; 34(9): 1149-55, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23892272

RESUMEN

AIM: SKF83959 (3-methyl-6-chloro-7,8-hydroxy-1-(3-methylphenyl)-2,3,4,5-tetrahydro-1H-3-benzazepine) is an atypical dopamine receptor-1 (D1 receptor) agonist, which exhibits many D1 receptor-independent effects. In the present work, we examined the effects of SKF83959 on monoaminergic transporters in vitro and its anti-depressant activity in vivo. METHODS: Human serotonin transporter (SERT), norepinephrine transporters (NET) or dopamine transporters (DAT) were stably expressed in CHO cells. The uptake kinetics of SERT, NET, and DAT were examined using [(3)H]-serotonin, [(3)H]-norepinephrine or [(3)H]-dopamine, respectively. A triple reuptake inhibitor DOV21947 was used as the positive control. Tail suspension test and forced swimming test were conducted in mice. SKF83959 or DOV21947 (2-8 mg/kg) were intraperitoneally injected 30 min before the tests. RESULTS: SKF83959 was a competitive inhibitor of SERT (K(i)=1.43±0.45 µmol/L), but a noncompetitive inhibitor of NET (K(i)=0.60±0.07 µmol/L) and DAT (K(i)=9.01±0.80 µmol/L). In contrast, DOV21947 was a competitive inhibitor of SERT (K(i)=0.89±0.24 µmol/L) and DAT (K(i)=1.47±0.31 µmol/L) and a noncompetitive inhibitor of NET (K(i)=0.18±0.04 µmol/L). In mice, both SKF83959 and DOV21947 elicited anti-depressant activity in a dose-dependent manner. CONCLUSION: SKF83959 functions as a novel triple reuptake inhibitor in vitro and exerts anti-depressant effects in vivo.


Asunto(s)
2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , Antidepresivos/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/antagonistas & inhibidores , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/antagonistas & inhibidores , Proteínas de Transporte de Serotonina en la Membrana Plasmática , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/uso terapéutico , Animales , Antidepresivos/uso terapéutico , Células CHO , Cricetinae , Cricetulus , Depresión/tratamiento farmacológico , Depresión/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
3.
J Neurosci Res ; 89(8): 1259-66, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21538463

RESUMEN

Dopamine (DA) profoundly modulates excitatory synaptic transmission and synaptic plasticity in the brain. In the present study the effects of SKF83959, the selective agonist of phosphatidylinositol (PI)-linked D(1) -like receptor, on the excitatory synaptic transmission were investigated in rat hippocampus. SKF83959 (10-100 µM) reversibly suppressed the field excitatory postsynaptic potential (fEPSP) elicited by stimulating the Schaffer's collateral-commissural fibers in CA1 area of hippocampal slices. However, the inhibition was not blocked by the D(1) receptor antagonist SCH23390, the D(2) receptor antagonist raclopride, the 5-HT(2A/2C) receptor antagonist mesulergine, or the α(1) -adrenoceptor antagonist prazosin. In addition, SKF83959 inhibited the afferent volley and significantly reduced the paired-pulse facilitation ratios. In dissociated hippocampal CA1 pyramidal neurons, SKF83959 had no detectable effect on glutamate-induced currents but potently inhibited voltage-activated Na(+) current (IC50 value = 26.9 ± 1.0 µM), which was not blocked by SCH23390 or by intracellular dialysis of GDP-ß-S. These results demonstrate that SKF83959 suppressed the excitatory synaptic transmission in hippocampal CA1 area, which was independent of D(1) -like receptor. The mechanism underlying the effect could be mainly inhibition of Na(+) channel in the afferent fibers. The suppression of excitatory synaptic transmission and the Na(+) channel by SKF83959 may contribute to its therapeutic benefits in Parkinson's disease.


Asunto(s)
2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores Dopaminérgicos/metabolismo , Transmisión Sináptica/efectos de los fármacos , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Animales , Benzazepinas/farmacología , Células Cultivadas , Potenciales Postsinápticos Excitadores/fisiología , Hipocampo/metabolismo , Masculino , Neuronas/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/fisiología
4.
Synapse ; 65(5): 379-87, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20803620

RESUMEN

RATIONALE: l-Stepholidine (l-SPD), a tetrahydroprotoberberine alkaloid, possesses a pharmacological profile of a D1/5-HT(1A) agonist and a D2 antagonist. This unique pharmacological profile makes it a promising novel antipsychotic candidate. Preliminary clinical trials and animal experiments suggest that l-SPD improves both positive and negative symptoms of schizophrenia without producing significant extrapyramidal side effects. To further explore the antipsychotic mechanisms of the drug, we studied the effects of l-SPD on the activity of dopamine (DA) neurons in the ventral tegmental area (VTA) using in vivo single-unit recording technique in rats. RESULT: We found that l-SPD increased VTA DA neurons firing rate and induced slow oscillation in firing pattern. Moreover, l-SPD, not clozapine, reversed d-amphetamine-induced inhibition which induced an excitation of VTA DA neurons. Furthermore, our data indicated that the excitatory effect of l-SPD is associated with its partial agonistic action for the 5-HT(1A) receptor since the 5-HT(1A) receptor antagonist WAY100635 could block the l-SPD-induced excitatory effect. However, activation of 5-HT(1A) receptor alone by specific agonist (±)-8-Hydroxy-2-(dipropylamino) tetralin (8-OH-DPAT) was insufficient to elicit excitation of VTA DA neurons, but the excitation of 8-OH-DPAT on VTA DA neurons was elicited in the presence of D2-like receptors antagonist raclopride. Collectively, these results indicate that l-SPD excited VTA DA neurons requiring its D2-like receptors antagonistic activity and 5-HT(1A) receptor agonistic activity. CONCLUSION: The present data demonstrate that D2 receptor antagonist/5-HT(1A) receptor agonistic dual properties modulate dopaminergic transmission in a unique pattern that may underlie the different therapeutic responses between l-SPD and other atypical antipsychotic drugs.


Asunto(s)
Berberina/análogos & derivados , Agonistas de Dopamina/farmacología , Dopamina/metabolismo , Neuronas/efectos de los fármacos , Receptor de Serotonina 5-HT1A/metabolismo , Área Tegmental Ventral/citología , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Berberina/farmacología , Dextroanfetamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Masculino , Inhibición Neural/efectos de los fármacos , Piperazinas/farmacología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología
5.
Acta Pharmacol Sin ; 30(6): 740-51, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19498417

RESUMEN

Neuronal nicotinic acetylcholine receptors (nAChRs) are the superfamily of ligand-gated ion channels and widely expressed throughout the central and peripheral nervous systems. nAChRs play crucial roles in modulating a wide range of higher cognitive functions by mediating presynaptic, postsynaptic, and extrasynaptic signaling. Thus far, nine alpha (alpha2-alpha10) and three beta (beta2, beta3, and beta4) subunits have been identified in the CNS, and these subunits assemble to form a diversity of functional nAChRs. Although alpha4beta2- and alpha7-nAChRs are the two major functional nAChR types in the CNS, alpha6*-nAChRs are abundantly expressed in the midbrain dopaminergic (DAergic) system, including mesocorticolimbic and nigrostriatal pathways, and particularly present in presynaptic nerve terminals. Recently, functional and pharmacological profiles of alpha6*-nAChRs have been assessed with the use of alpha6 subunit blockers such as alpha-conotoxin MII and PIA, and also by using alpha6 subunit knockout mice. By modulating DA release in the nucleus accumbens (NAc) and modulating GABA release onto DAergic neurons in the ventral tegmental area (VTA), alpha6*-nAChRs may play important roles in the mediation of nicotine reward and addiction. Furthermore, alpha6*-nAChRs in the nigrostriatal DAergic system may be promising targets for selective preventative treatment of Parkinson's disease (PD). Thus, alpha6*-nAChRs may hold promise for future clinical treatment of human disorders, such as nicotine addiction and PD. In this review, we mainly focus on the recent advances in the understanding of alpha6*-nAChR function, pharmacology and pathophysiology.


Asunto(s)
Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/metabolismo , Animales , Dopamina/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Nicotina/efectos adversos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/fisiopatología , Subunidades de Proteína , Receptores Nicotínicos/efectos de los fármacos , Recompensa , Tabaquismo/fisiopatología , Tabaquismo/rehabilitación , Área Tegmental Ventral/metabolismo
6.
Acta Pharmacol Sin ; 30(6): 851-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19498424

RESUMEN

AIM: Dopaminergic neurons in the substantia nigra pars compacta (SNc) play important roles in motor control and drug addiction. As the major afferent, GABAergic innervation controls the activity of SNc dopaminergic neurons. Although it is clear that nicotine modulates SNc dopaminergic neurons by activating subtypes of somatodendritic nicotinic acetylcholine receptors (nAChRs), the detailed mechanisms of this activation remain to be addressed. METHODS: In the current study, we recorded GABA(A) receptor-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) from dissociated SNc dopaminergic neurons that were obtained using an enzyme-free procedure. These neurons preserved some functional terminals after isolation, including those that release GABA. RESULTS: We found that both extra- and intra-cellular calcium modulates sIPSCs in these neurons. Furthermore, both nicotine and endogenous acetylcholine enhance the frequency of sIPSCs. Moreover, endogenous acetylcholine tonically facilitates sIPSC frequency, primarily by activating the alpha4beta2* nAChRs on the GABAergic terminals. CONCLUSION: Nicotine facilitates GABA release onto SNc dopaminergic neurons mainly via the activation of presynaptic alpha4beta2* nAChRs.


Asunto(s)
Dopamina/metabolismo , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Receptores de GABA-A/metabolismo , Acetilcolina/metabolismo , Animales , Calcio/metabolismo , Ácido Glutámico/metabolismo , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Neuronas/metabolismo , Ratas , Receptores Nicotínicos/efectos de los fármacos , Receptores Nicotínicos/metabolismo , Receptores Presinapticos/efectos de los fármacos , Receptores Presinapticos/metabolismo , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo
7.
J Neurosci ; 27(20): 5414-21, 2007 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-17507563

RESUMEN

Stimulation of the prefrontal cortex (PFC) has been shown to have an excitatory influence on dopamine (DA) neurons. We report here that, under nonstimulated conditions, the activity of DA neurons in the ventral tegmental area (VTA) also covaries, on a subsecond timescale, with the activity of PFC cells. Thus, in 67% of VTA DA neurons recorded in chloral hydrate-anesthetized rats, the firing of the cell displayed a slow oscillation (SO) that was highly coherent with the activity of PFC neurons. The SO was suppressed by transections immediately caudal to the PFC or by intra-PFC infusion of tetrodotoxin, suggesting that it depends on inputs derived from the PFC. Unexpectedly, the SO in most VTA DA neurons was reversed in phase relative to PFC cell activity, suggesting that at least part of PFC information is transferred to DA neurons indirectly through inhibitory relay neurons. These results, together with those reported previously, suggest that the PFC can act through multiple pathways to exert both excitatory and inhibitory influences on DA neurons. The observed functional coupling between DA and PFC neurons further suggests that these pathways not only allow a bidirectional control of DA neurons by the PFC, but also enable action potential-dependent DA release to be coordinated, on a subsecond timescale, with glutamate release from PFC terminals. Further understanding of this coordinated activity may provide important new insights into brain functions and disorders thought to involve both VTA DA and PFC neurons.


Asunto(s)
Potenciales de Acción/fisiología , Dopamina/fisiología , Neuronas/fisiología , Corteza Prefrontal/fisiología , Área Tegmental Ventral/fisiología , Animales , Masculino , Ratas , Ratas Sprague-Dawley
8.
J Neurochem ; 104(4): 946-56, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18005341

RESUMEN

3-methyl-6-chloro-7,8-hydroxy-1-(3-methylphenyl)-2,3,4,5-tetrahydro-1H-3-benzazepine (SKF83959), a selective agonist for the putative phosphatidylinositol (PI)-linked dopamine receptor (DAR), has been shown to possess potent anti-Parkinson disease effects but produces less dyskinesia and motor fluctuation that are frequently observed in Parkinson disease drug therapies. The present study was designed to detect the neuroprotection of SKF83959 and its potential mechanism for the effect in cultured rat cortical cells. The presence of SKF83959 with a dose range of 0.1-30 micromol/L improved H2O2-reduced cell viability in a dose-dependent manner. The anti-apoptotic action of SKF83959 was partially abolished by pre-application of the D1 antagonist SCH23390 (30 micromol/L) and the PI 3-kinase (PI 3-K) inhibitor LY294002 but not by the MEK1/2 inhibitor PD98059 (30 micromol/L). Moreover, SKF83959 treatment significantly inhibited H2O2-activated glycogen synthase kinase-3beta (GSK-3beta) which was associated with the drug's neuroprotective effect, but this inhibition was attenuated by SCH23390 and a selective PI 3-K inhibitor. Moreover, the application of either SKF83959 or a pharmacological inhibitor of GSK-3beta attenuated the inhibition by H2O2 on the expression of inducible NO synthase and production of NO. This indicates that D1-like receptor, presumably PI-linked D1 receptor, -mediated alteration of PI 3-K/Akt/GSK-3beta pathway is involved in the neuroprotection by SKF83959. In addition, SKF83959 also effectively decreased the level of the lipid peroxidation and increased the activity of GSH-peroxidase altered by H2O2. These results suggest that SKF83959 exerts its neuroprotective effect through both receptor-dependent and independent mechanisms: Inhibition of GSK-3beta and consequently increasing the expression of inducible NO synthase via putative PI-linked DAR; and its anti-oxidative activity which is independent of DAR.


Asunto(s)
2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , Antioxidantes/farmacología , Agonistas de Dopamina/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Fármacos Neuroprotectores/farmacología , Receptores de Dopamina D1/agonistas , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Relación Dosis-Respuesta a Droga , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley
9.
Mol Pain ; 4: 57, 2008 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19014677

RESUMEN

BACKGROUND: Clinical observation and experimental studies have indicated that a single exposure to morphine could induce tolerance and dependence. It has become a concern in clinical antinociceptive practice. However, the underling mechanism remains unknown. This study was designed to explore the changes of dopamine (DA) neuron activities in the ventral tegmental area (VTA) by employing a spectral analysis followed single morphine treatment. RESULTS: Acute morphine treatment significantly increased not only the firing rate and firing population but also the power of slow oscillation of DA neurons in naïve rats. These changes lasted at least for 3 days following the morphine treatment. During this period of time, responses of the DA neurons to subsequent morphine challenge were diminished. We further demonstrated a transient desensitization of opiate receptors as monitored by GTPgammaS binding to G-proteins. The present study provided first direct evidence for the temporal changes in the VTA DA neuron activities and opiate receptors desensitization. CONCLUSION: Prolonged VTA DA neuron activation and opiate receptors desensitization followed single morphine exposure may underlie the development of dependence and tolerance that may associate with the acute analgesic tolerance and acute addiction of morphine.


Asunto(s)
Dopamina , Morfina/farmacología , Neuronas/efectos de los fármacos , Animales , Tolerancia a Medicamentos , Morfina/administración & dosificación , Ratas , Receptores Opioides/efectos de los fármacos , Trastornos Relacionados con Sustancias/etiología , Factores de Tiempo
10.
Acta Pharmacol Sin ; 29(7): 781-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18565275

RESUMEN

AIM: Drug addiction is a chronic brain disease with constant relapse requiring long-term treatment. New pharmacological strategies focus on the development of an effective antirelapse drug. This study examines the effects of levotetrahydropalmatine (l-THP) on reducing heroin craving and increasing the abstinence rate among heroin-dependent patients. METHODS: In total, 120 heroin-dependent patients participated in the randomized, double-blinded, and placebocontrolled study using l-THP treatment. The participants remained in a ward during a 4-week period of l-THP treatment, followed by 4 weeks of observation after treatment. The patients were followed for 3 months after discharge. Outcome measures are the measured severity of the protracted abstinence withdrawal syndrome (PAWS) and the abstinence rate. RESULTS: Four weeks of l-THP treatment significantly ameliorated the severity of PAWS, specifically, somatic syndrome, mood states, insomnia, and drug craving, in comparison to the placebo group. Based on the 3 month follow-up observation, participants who survived the initial 2 weeks of l-THP medication and remained in the trial program had a significantly higher abstinence rate of 47.8% (95% confidence interval [CI]: 33%- 67%) than the 15.2% in the placebo group (95% CI: 7%-25%), according to a log- rank test (P<0.0005). CONCLUSION: l-THP significantly ameliorated PAWS, especially reducing drug craving. Furthermore, it increased the abstinence rate among heroin users. These results support the potential use of l-THP for the treatment of heroin addiction.


Asunto(s)
Analgésicos no Narcóticos/uso terapéutico , Alcaloides de Berberina/uso terapéutico , Dependencia de Heroína/tratamiento farmacológico , Dependencia de Heroína/psicología , Adulto , Método Doble Ciego , Determinación de Punto Final , Femenino , Dependencia de Heroína/rehabilitación , Humanos , Masculino , Pacientes Desistentes del Tratamiento , Proyectos Piloto , Prevención Secundaria , Resultado del Tratamiento
11.
Sci Rep ; 8(1): 3274, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29459754

RESUMEN

The initiation of psychostimulant sensitization depends on the mesocorticolimbic dopamine (DA) system. Although many cellular adaptations has been reported to be associated with this addictive behavior, the overall influence of these adaptations on the network regulation of DA neurons has not been established. Here, we profile a network-driven slow oscillation (SO) in the firing activity of ventral tegmental area (VTA) putative DA and non-DA neurons and their correlation with locomotor sensitization induced by repeated administration of cocaine. One day after the last cocaine injection, the power of SO (Pso) significantly increased both in DA and non-DA neurons. Interestingly, the Pso in DA neurons was positively correlated, while Pso in non-DA neurons was negatively correlated with the level of locomotor sensitization. On the other hand, the firing rates of DA and non-DA neurons were both elevated, but none exhibited any correlation with the level of sensitization. Fourteen days after the last injection, the Pso of DA neurons dissipated but still positively correlated with the level of sensitization. In contrast, the Pso in non-DA neurons lost correlation with locomotor sensitization. These results suggest that cocaine-induced locomotor sensitization is associated with long-term network adaptation in DA system and that DA and non-DA neurons may corporately facilitate/hamper the initiation of locomotor sensitization.


Asunto(s)
Potenciales de Acción , Cocaína/metabolismo , Inhibidores de Captación de Dopamina/metabolismo , Neuronas/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Animales , Conducta Animal , Masculino , Actividad Motora , Ratas Sprague-Dawley
12.
Curr Med Chem ; 14(28): 2996-3002, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18220736

RESUMEN

Tetrahydroprotoberberines (THPBs) represent a series of compounds extracted from the Chinese herb Corydalis ambigua and various species of Stephania. THPBs, dependent on the presence of hydroxyl groups in its structure, are divided into three types: nonhydroxyl-THPBs, monohydroxyl-THPBs and dihydroxyl-THPBs. THPBs are identified as a new category of dopamine receptor ligands. Among all THPBs, dihydroxyl-THPBs attracted particular attention because of their dual actions on dopamine (DA) receptors. They exhibit D(1) receptor agonistic activity while acting as D(2) receptor antagonists. This unique pharmacological profile made dihydroxyl-THPBs such as l-stepholidine (l-SPD) potential agents in the treatment of drug addiction, Parkinson's disease, and especially, schizophrenia. Clinical studies have shown that co-administration of l-SPD with a typical antipsychotic drug significantly enhances the therapeutic effects and remarkably reduces the tardive dyskinesia induced by the typical antipsychotic drug used with schizophrenic patients. Moreover, l-SPD alone was shown to have therapeutic value without inducing significant extrapyramidal side effects and also seemed to reduce the negative symptoms of schizophrenia. This is confirmed in experimental studies using animal models of schizophrenia, in which l-SPD improved social interaction and cognitive function, inhibited hyperactivity in schizophrenic animals. This review discusses the chemistry, pharmacology and clinical implications of l-THPBs in the drug development for psychosis and neurobiological diseases.


Asunto(s)
Alcaloides de Berberina/química , Alcaloides de Berberina/farmacología , Berberina/análogos & derivados , Trastornos Mentales/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , Berberina/química , Berberina/farmacología , Berberina/uso terapéutico , Alcaloides de Berberina/uso terapéutico , Dopamina/metabolismo , Humanos , Receptores Dopaminérgicos/metabolismo
13.
Sci Rep ; 7(1): 3809, 2017 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-28630404

RESUMEN

Major depressive disorder (MDD) is a common neuropsychiatric disorder characterized by diverse symptoms. Although several antidepressants can influence dopamine system in the medial prefrontal cortex (mPFC), but the role of D1R or D2R subtypes of dopamine receptor during anti-depression process is still vague in PFC region. To address this question, we investigate the antidepressant effect of levo-stepholidine (l-SPD), an antipsychotic medication with unique pharmacological profile of D1R agonism and D2R antagonism, and clarified its molecular mechanisms in the mPFC. Our results showed that l-SPD exerted antidepressant-like effects on the Sprague-Dawley rat CMS model of depression. Mechanism studies revealed that l-SPD worked as a specific D1R agonist, rather than D2 antagonist, to activate downstream signaling of PKA/mTOR pathway, which resulted in increasing synaptogenesis-related proteins, such as PSD 95 and synapsin I. In addition, l-SPD triggered long-term synaptic potentiation (LTP) in the mPFC, which was blocked by the inhibition of D1R, PKA, and mTOR, supporting that selective activation of D1R enhanced excitatory synaptic transduction in PFC. Our findings suggest a critical role of D1R/PKA/mTOR signaling cascade in the mPFC during the l-SPD mediated antidepressant process, which may also provide new insights into the role of mesocortical dopaminergic system in antidepressant effects.


Asunto(s)
Antidepresivos/farmacología , Berberina/análogos & derivados , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Trastorno Depresivo Mayor/tratamiento farmacológico , Antagonistas de Dopamina/farmacología , Corteza Prefrontal/metabolismo , Receptores Dopaminérgicos/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Animales , Berberina/farmacología , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/patología , Masculino , Corteza Prefrontal/patología , Ratas , Ratas Sprague-Dawley
14.
Neuropsychopharmacology ; 30(2): 261-7, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15578005

RESUMEN

l-Stepholidine (SPD) is a tetrahydroprotoberberine alkaloid and a mixed dopamine D1 agonist/D2 antagonist. Preliminary clinical trials suggest that SPD improves both positive and negative symptoms of schizophrenia without producing significant extrapyramidal side effects. Here, we report that SPD mimics the effect of the atypical antipsychotic drug clozapine, preferentially increasing Fos expression in corticolimbic areas. Thus, at 10 mg/kg (i.p.), SPD induced Fos expression in the medial prefrontal cortex (mPFC), nucleus accumbens (NAc), and lateral septal nucleus (LSN) without significantly affecting the dorsolateral striatum (DLSt). At higher doses (20-40 mg/kg), SPD also increased Fos expression in the DLSt. The increase, however, was less pronounced than the increase seen in the NAc. Within the NAc, SPD also induced more Fos expression in the shell than in the core. In all subcortical areas examined, the Fos expression induced by SPD was mimicked by the D2 antagonist sulpiride and reversed by the D2 agonist quinpirole, suggesting that the effect is due to blockade of D2-like receptors by SPD. In the mPFC, however, the effect was not mimicked by sulpride or reversed by quinpirole. It was also not mimicked by the D1 agonist SKF38393 or SKF38393 plus sulpride, and not reversed by the D1 antagonist SCH23390. These results suggest that, in the mPFC, SPD may induce Fos expression through a non-DA mechanism. Whether the mechanism involves an interaction of SPD with other neurotransmitters such as 5-HT and norepinephrine remains to be determined.


Asunto(s)
Antipsicóticos/farmacología , Berberina/análogos & derivados , Berberina/farmacología , Clozapina/farmacología , Agonistas de Dopamina/farmacología , Antagonistas de Dopamina/farmacología , Haloperidol/farmacología , Prosencéfalo/metabolismo , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Animales , Antagonistas de los Receptores de Dopamina D2 , Inmunohistoquímica , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Prosencéfalo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D2/agonistas , Núcleos Septales/efectos de los fármacos , Núcleos Septales/metabolismo
15.
Neuroreport ; 13(16): 2085-9, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12438931

RESUMEN

The present study investigated the influence of (-)-stepholidine, an effective dopamine D1 receptor agonist and D2 receptor antagonist, on the development of neural precursor cells. Incubation of striatal neural precursor cells with stepholidine resulted in significant increase in the number of proliferating precursor cell spheres when in the presence of fibroblast growth factor-2. This action can be blocked by application of haloperidol. Treatment with stepholidine also increased the number of microtubule-associated protein-2-immunoreactive cells in the cultures and promoted marked increases in tyrosine hydroxylase expression. These findings suggest that stepholidine is involved in the regulation of proliferation of precursor cells. The effect appears to be mediated by dopamine receptors. Stepholidine also promotes the differentiation of precursor cells, however, this action may be independent of its effect on dopaminergic receptors.


Asunto(s)
Berberina/análogos & derivados , Berberina/farmacología , Cuerpo Estriado/efectos de los fármacos , Agonistas de Dopamina/farmacología , Antagonistas de Dopamina/farmacología , Receptores Dopaminérgicos/efectos de los fármacos , Receptores Dopaminérgicos/metabolismo , Células Madre/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Cuerpo Estriado/metabolismo , Antagonistas de los Receptores de Dopamina D2 , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Haloperidol/farmacología , Inmunohistoquímica , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
16.
Pharmacol Biochem Behav ; 75(2): 289-94, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12873618

RESUMEN

The effect of l-12-chloroscoulerine (l-CSL), a novel ligand with dual dopamine D1 receptor agonistic and D2 receptor antagonistic actions, on the development of morphine-induced conditioned place preference (CPP) was investigated in mice. Morphine (10 mg/kg)-induced place preference was dose dependently suppressed by coadministration of l-CSL (5, 10 and 20 mg/kg), which induced neither place preference nor place aversion when administered alone at a dose of 20 mg/kg. The D1 receptor antagonist SCH23390 (0.1 mg/kg) suppressed, whereas the D2 receptor agonist (+/-)-2-(N-phenylethyl-N-propyl)-amino-5-hydroxytetralin (PPHT) (0.5 mg/kg) had no influence on the development of morphine-induced place preference. However, SCH23390 (0.1 mg/kg) did not affect, whereas PPHT (0.5 mg/kg) reversed the suppressive effect of l-CSL on the development of morphine-induced place preference. These results indicate that l-CSL suppresses the development of place preference of morphine by blocking D2 receptors.


Asunto(s)
Alcaloides de Berberina/farmacología , Condicionamiento Operante/efectos de los fármacos , Antagonistas de Dopamina/farmacología , Morfina/antagonistas & inhibidores , Morfina/farmacología , Antagonistas de Narcóticos , Narcóticos/farmacología , Receptores Dopaminérgicos/efectos de los fármacos , Animales , Benzazepinas/farmacología , Antagonistas de los Receptores de Dopamina D2 , Ligandos , Masculino , Ratones , Receptores de Dopamina D1/antagonistas & inhibidores
18.
PLoS One ; 7(12): e51507, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251560

RESUMEN

Dopamine (DA) neurons in the ventral tegmental area (VTA) not only participate in reward processing, but also respond to aversive stimuli. Although GABA neurons in this area are actively involved in regulating the firing of DA neurons, few data exist concerning the responses of these neurons to aversive sensory input. In this study, by employing extracellular single-unit recording and spectral analysis techniques in paralyzed and ventilated rats, we found that the firing pattern in 44% (47 of 106) of GABA cells in the VTA was sensitive to the sensory input produced by the ventilation, showing a significant ventilation-associated oscillation in the power spectra. Detailed studies revealed that most ventilation-sensitive GABA neurons (38 of 47) were excited by the stimuli, whereas most ventilation-sensitive DA neurons (11 of 14) were inhibited. When the animals were under anesthesia or the sensory pathways were transected, the ventilation-associated oscillation failed to appear. Systemic administration of non-competitive N-methyl-D-aspartase (NMDA) receptor antagonist MK-801 completely disrupted the association between the firing of GABA neurons and the ventilation. Interestingly, local MK-801 injection into the VTA dramatically enhanced the sensitivity of GABA neurons to the ventilation. Our data demonstrate that both GABA and DA neurons in the VTA can be significantly modulated by sensory input produced by the ventilation, which may indicate potential functional roles of VTA in processing sensation-related input.


Asunto(s)
Neuronas GABAérgicas/fisiología , Sensación/fisiología , Área Tegmental Ventral/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Estado de Conciencia/efectos de los fármacos , Estado de Conciencia/fisiología , Maleato de Dizocilpina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/fisiología , Neuronas GABAérgicas/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Corteza Somatosensorial/fisiología , Ventilación , Área Tegmental Ventral/efectos de los fármacos
20.
Neuropharmacology ; 59(7-8): 567-72, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20804776

RESUMEN

Tetrahydroberberine (THB) exhibits neuroprotective effects but its targets and underlying mechanisms are largely unknown. Emerging evidence indicates that ATP-sensitive potassium (K(ATP)) channels in the substantia nigra pars compacta (SNc) promote Parkinson disease (PD) pathogenesis, thus blocking K(ATP) channels may protect neurons against neuronal degeneration. In the present study, we tested a hypothesis that THB blocks K(ATP) channels in dopaminergic (DA) neurons acutely dissociated from rat SNc. Using perforated patch-clamp recording in current-clamp mode, the functional K(ATP) channels can be opened by persistent perfusion of rotenone, an inhibitor of complex I of the mitochondrial respiratory chain. Bath-application of THB reversibly blocks opened K(ATP) channels in a concentration-dependent manner, which is comparable to a classical K(ATP) channel blocker, Tol. Compared to THB analogs, l-stepholidine (l-SPD) or l-tetrahydropalmatine (l-THP), THB exhibits more profound blockade in K(ATP) channels. In addition, exposure of THB alone to the recorded neuron increases action potential firing, and THB also restores rotenone-induced membrane hyperpolarization in the presence of dopamine D2 receptor antagonist (sulpiride), suggesting that THB exhibits an excitatory effect on SNc DA neurons through the block of K(ATP) channels. Collectively, the blockade of neuronal K(ATP) channels by THB in SNc DA neurons is a novel pharmacological mechanism of THB, which may contribute to its neuroprotective effects in PD.


Asunto(s)
Berberina/análogos & derivados , Dopamina/metabolismo , Canales KATP/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Sustancia Negra/efectos de los fármacos , Animales , Berberina/farmacología , Técnicas In Vitro , Neuronas/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Receptores de Dopamina D2/fisiología , Sustancia Negra/citología , Sustancia Negra/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA