Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Eur J Neurosci ; 52(11): 4546-4562, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32725894

RESUMEN

Regional alterations in kinetics of catecholamine uptake are due in part to variations in clearance mechanisms. The rate of clearance is a critical determinant of the strength of catecholamine signaling. Catecholamine transmission in the nucleus accumbens core (NAcc) and basolateral amygdala (BLA) is of particular interest due to involvement of these regions in cognition and motivation. Previous work has shown that catecholamine clearance in the NAcc is largely mediated by the dopamine transporter (DAT), but clearance in the BLA is less DAT-dependent. A growing body of literature suggests that organic cation transporter 3 (OCT3) also contributes to catecholamine clearance in both regions. Consistent with different clearance mechanisms between regions, catecholamine clearance is more rapid in the NAcc than in the BLA, though mechanisms underlying this have not been resolved. We compared the expression of DAT and OCT3 and their contributions to catecholamine clearance in the NAcc and BLA. We found DAT protein levels were ~ 4-fold higher in the NAcc than in the BLA, while OCT3 protein expression was similar between the two regions. Immunofluorescent labeling of the two transporters in brain sections confirmed these findings. Ex vivo voltammetry demonstrated that the magnitude of catecholamine release was greater, and the clearance rate was faster in the NAcc than in the BLA. Additionally, catecholamine clearance in the BLA was more sensitive to the OCT3 inhibitor corticosterone, while clearance in the NAcc was more cocaine sensitive. These distinctions in catecholamine clearance may underlie differential effects of catecholamines on behavioral outputs mediated by these regions.


Asunto(s)
Complejo Nuclear Basolateral , Núcleo Accumbens , Complejo Nuclear Basolateral/metabolismo , Catecolaminas , Cationes , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Núcleo Accumbens/metabolismo
2.
Sci Signal ; 9(456): ra117, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27899527

RESUMEN

Agonists targeting the kappa opioid receptor (KOR) have been promising therapeutic candidates because of their efficacy for treating intractable itch and relieving pain. Unlike typical opioid narcotics, KOR agonists do not produce euphoria or lead to respiratory suppression or overdose. However, they do produce dysphoria and sedation, side effects that have precluded their clinical development as therapeutics. KOR signaling can be fine-tuned to preferentially activate certain pathways over others, such that agonists can bias signaling so that the receptor signals through G proteins rather than other effectors such as ßarrestin2. We evaluated a newly developed G protein signaling-biased KOR agonist in preclinical models of pain, pruritis, sedation, dopamine regulation, and dysphoria. We found that triazole 1.1 retained the antinociceptive and antipruritic efficacies of a conventional KOR agonist, yet it did not induce sedation or reductions in dopamine release in mice, nor did it produce dysphoria as determined by intracranial self-stimulation in rats. These data demonstrated that biased agonists may be used to segregate physiological responses downstream of the receptor. Moreover, the findings suggest that biased KOR agonists may present a means to treat pain and intractable itch without the side effects of dysphoria and sedation and with reduced abuse potential.


Asunto(s)
Dolor/tratamiento farmacológico , Prurito/tratamiento farmacológico , Receptores Opioides kappa/agonistas , Triazoles/farmacología , Animales , Masculino , Ratones , Dolor/metabolismo , Prurito/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores Opioides kappa/metabolismo , Triazoles/química , Arrestina beta 2/metabolismo
3.
Neuropharmacology ; 110(Pt A): 190-197, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27450094

RESUMEN

Acute ethanol exposure is known to stimulate the dopamine system; however, chronic exposure has been shown to downregulate the dopamine system. In rodents, chronic intermittent exposure (CIE) to ethanol also increases negative affect during withdrawal, such as, increases in anxiety- and depressive-like behavior. Moreover, CIE exposure results in increased ethanol drinking and preference during withdrawal. Previous literature documents reductions in CIE-induced anxiety-, depressive-like behaviors and ethanol intake in response to kappa opioid receptor (KOR) blockade. KORs are located on presynaptic dopamine terminals in the nucleus accumbens (NAc) and inhibit release, an effect which has been linked to negative affective behaviors. Previous reports show an upregulation in KOR function following extended CIE exposure; however it is not clear whether there is a direct link between KOR upregulation and dopamine downregulation during withdrawal from CIE. This study aimed to examine the effects of KOR modulation on dopamine responses to ethanol of behaving mice exposed to air or ethanol vapor in a repeated intermittent pattern. First, we showed that KORs have a greater response to an agonist after moderate CIE compared to air exposed mice using ex vivo fast scan cyclic voltammetry. Second, using in vivo microdialysis, we showed that, in contrast to the expected increase in extracellular levels of dopamine following an acute ethanol challenge in air exposed mice, CIE exposed mice exhibited a robust decrease in dopamine levels. Third, we showed that blockade of KORs reversed the aberrant inhibitory dopamine response to ethanol in CIE exposed mice while not affecting the air exposed mice demonstrating that inhibition of KORs "rescued" dopamine responses in CIE exposed mice. Taken together, these findings indicate that augmentation of dynorphin/KOR system activity drives the reduction in stimulated (electrical and ethanol) dopamine release in the NAc. Thus, blockade of KORs is a promising avenue for developing pharmacotherapies for alcoholism.


Asunto(s)
Dopamina/metabolismo , Regulación hacia Abajo/fisiología , Etanol/administración & dosificación , Receptores Opioides kappa/fisiología , Regulación hacia Arriba/fisiología , Animales , Bencenoacetamidas/farmacología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Pirrolidinas/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos
4.
Int J Mol Sci ; 17(8)2016 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-27472317

RESUMEN

The development of pharmacotherapeutics that reduce relapse to alcohol drinking in patients with alcohol dependence is of considerable research interest. Preclinical data support a role for nucleus accumbens (NAc) κ opioid receptors (KOR) in chronic intermittent ethanol (CIE) exposure-induced increases in ethanol intake. Nalmefene, a high-affinity KOR partial agonist, reduces drinking in at-risk patients and relapse drinking in rodents, potentially due to its effects on NAc KORs. However, the effects of nalmefene on accumbal dopamine transmission and KOR function are poorly understood. We investigated the effects of nalmefene on dopamine transmission and KORs using fast scan cyclic voltammetry in NAc brain slices from male C57BL/6J mice following five weeks of CIE or air exposure. Nalmefene concentration-dependently reduced dopamine release similarly in air and CIE groups, suggesting that dynorphin tone may not be present in brain slices. Further, nalmefene attenuated dopamine uptake rates to a greater extent in brain slices from CIE-exposed mice, suggesting that dopamine transporter-KOR interactions may be fundamentally altered following CIE. Additionally, nalmefene reversed the dopamine-decreasing effects of a maximal concentration of a KOR agonist selectively in brain slices of CIE-exposed mice. It is possible that nalmefene may attenuate withdrawal-induced increases in ethanol consumption by modulation of dopamine transmission through KORs.


Asunto(s)
Dopamina/metabolismo , Etanol/administración & dosificación , Naltrexona/análogos & derivados , Antagonistas de Narcóticos/farmacología , Núcleo Accumbens/metabolismo , Receptores Opioides kappa/metabolismo , Animales , Depresores del Sistema Nervioso Central/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Naltrexona/farmacología , Núcleo Accumbens/efectos de los fármacos
5.
Neuropsychopharmacology ; 41(9): 2263-74, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26860203

RESUMEN

Chronic early-life stress increases vulnerability to alcoholism and anxiety disorders during adulthood. Similarly, rats reared in social isolation (SI) during adolescence exhibit augmented ethanol intake and anxiety-like behaviors compared with group housed (GH) rats. Prior studies suggest that disruption of dopamine (DA) signaling contributes to SI-associated behaviors, although the mechanisms underlying these alterations are not fully understood. Kappa opioid receptors (KORs) have an important role in regulating mesolimbic DA signaling, and other kinds of stressors have been shown to augment KOR function. Therefore, we tested the hypothesis that SI-induced increases in KOR function contribute to the dysregulation of NAc DA and the escalation in ethanol intake associated with SI. Our ex vivo voltammetry experiments showed that the inhibitory effects of the kappa agonist U50,488 on DA release were significantly enhanced in the NAc core and shell of SI rats. Dynorphin levels in NAc tissue were observed to be lower in SI rats. Microdialysis in freely moving rats revealed that SI was also associated with reduced baseline DA levels, and pretreatment with the KOR antagonist nor-binaltorphimine (nor-BNI) increased DA levels selectively in SI subjects. Acute ethanol elevated DA in SI and GH rats and nor-BNI pretreatment augmented this effect in SI subjects, while having no effect on ethanol-stimulated DA release in GH rats. Together, these data suggest that KORs may have increased responsiveness following SI, which could lead to hypodopaminergia and contribute to an increased drive to consume ethanol. Indeed, SI rats exhibited greater ethanol intake and preference and KOR blockade selectively attenuated ethanol intake in SI rats. Collectively, the findings that nor-BNI reversed SI-mediated hypodopaminergic state and escalated ethanol intake suggest that KOR antagonists may represent a promising therapeutic strategy for the treatment of alcohol use disorders, particularly in cases linked to chronic early-life stress.


Asunto(s)
Dopamina/metabolismo , Etanol/administración & dosificación , Núcleo Accumbens/metabolismo , Receptores Opioides kappa/metabolismo , Aislamiento Social , Estrés Psicológico , Animales , Masculino , Naltrexona/análogos & derivados , Núcleo Accumbens/efectos de los fármacos , Ratas Long-Evans , Receptores Opioides kappa/antagonistas & inhibidores
6.
Artículo en Inglés | MEDLINE | ID: mdl-26625893

RESUMEN

BACKGROUND: Chronic ethanol exposure reduces dopamine transmission in the nucleus accumbens, which may contribute to the negative affective symptoms associated with ethanol withdrawal. Kappa opioid receptors have been implicated in withdrawal-induced excessive drinking and anxiety-like behaviors and are known to inhibit dopamine release in the nucleus accumbens. The effects of chronic ethanol exposure on kappa opioid receptor-mediated changes in dopamine transmission at the level of the dopamine terminal and withdrawal-related behaviors were examined. METHODS: Five weeks of chronic intermittent ethanol exposure in male C57BL/6 mice were used to examine the role of kappa opioid receptors in chronic ethanol-induced increases in ethanol intake and marble burying, a measure of anxiety/compulsive-like behavior. Drinking and marble burying were evaluated before and after chronic intermittent ethanol exposure, with and without kappa opioid receptor blockade by nor-binaltorphimine (10mg/kg i.p.). Functional alterations in kappa opioid receptors were assessed using fast scan cyclic voltammetry in brain slices containing the nucleus accumbens. RESULTS: Chronic intermittent ethanol-exposed mice showed increased ethanol drinking and marble burying compared with controls, which was attenuated with kappa opioid receptor blockade. Chronic intermittent ethanol-induced increases in behavior were replicated with kappa opioid receptor activation in naïve mice. Fast scan cyclic voltammetry revealed that chronic intermittent ethanol reduced accumbal dopamine release and increased uptake rates, promoting a hypodopaminergic state of this region. Kappa opioid receptor activation with U50,488H concentration-dependently decreased dopamine release in both groups; however, this effect was greater in chronic intermittent ethanol-treated mice, indicating kappa opioid receptor supersensitivity in this group. CONCLUSIONS: These data suggest that the chronic intermittent ethanol-induced increase in ethanol intake and anxiety/compulsive-like behaviors may be driven by greater kappa opioid receptor sensitivity and a hypodopaminergic state of the nucleus accumbens.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Trastornos del Sistema Nervioso Inducidos por Alcohol/metabolismo , Conducta Animal , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Etanol , Núcleo Accumbens/metabolismo , Receptores Opioides kappa/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Transmisión Sináptica , Consumo de Bebidas Alcohólicas/efectos adversos , Consumo de Bebidas Alcohólicas/fisiopatología , Consumo de Bebidas Alcohólicas/psicología , Trastornos del Sistema Nervioso Inducidos por Alcohol/fisiopatología , Trastornos del Sistema Nervioso Inducidos por Alcohol/psicología , Analgésicos Opioides/farmacología , Animales , Ansiedad/metabolismo , Ansiedad/fisiopatología , Ansiedad/psicología , Conducta Animal/efectos de los fármacos , Conducta Compulsiva , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Masculino , Ratones Endogámicos C57BL , Antagonistas de Narcóticos/farmacología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiopatología , Receptores Opioides kappa/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/fisiopatología , Síndrome de Abstinencia a Sustancias/psicología , Transmisión Sináptica/efectos de los fármacos
7.
Neuropharmacology ; 99: 735-49, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26188147

RESUMEN

Chronic alcohol consumption and withdrawal leads to anxiety, escalated alcohol drinking behavior, and alcohol dependence. Alterations in the function of key structures within the cortico-limbic neural circuit have been implicated in underlying the negative behavioral consequences of chronic alcohol exposure in both humans and rodents. Here, we used chronic intermittent ethanol vapor exposure (CIE) in male C57BL/6J mice to evaluate the effects of chronic alcohol exposure and withdrawal on anxiety-like behavior and basal synaptic function and neuronal excitability in prefrontal cortical and extended amygdala brain regions. Forty-eight hours after four cycles of CIE, mice were either assayed in the marble burying test (MBT) or their brains were harvested and whole-cell electrophysiological recordings were performed in the prelimbic and infralimbic medial prefrontal cortex (PLC and ILC), the lateral and medial central nucleus of the amygdala (lCeA and mCeA), and the dorsal and ventral bed nucleus of the stria terminalis (dBNST and vBNST). Ethanol-exposed mice displayed increased anxiety in the MBT compared to air-exposed controls, and alterations in neuronal function were observed in all brain structures examined, including several distinct differences between subregions within each structure. Chronic ethanol exposure induced hyperexcitability of the ILC, as well as a shift toward excitation in synaptic drive and hyperexcitability of vBNST neurons; in contrast, there was a net inhibition of the CeA. This study reveals extensive effects of chronic ethanol exposure on the basal function of cortico-limbic brain regions, suggests that there may be complex interactions between these regions in the regulation of ethanol-dependent alterations in anxiety state, and highlights the need for future examination of projection-specific effects of ethanol in cortico-limbic circuitry.


Asunto(s)
Trastornos Relacionados con Alcohol/fisiopatología , Amígdala del Cerebelo/efectos de los fármacos , Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Neuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Trastornos Relacionados con Alcohol/psicología , Amígdala del Cerebelo/fisiopatología , Animales , Trastornos de Ansiedad/inducido químicamente , Trastornos de Ansiedad/fisiopatología , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neuronas/fisiología , Técnicas de Placa-Clamp , Corteza Prefrontal/fisiopatología , Núcleos Septales/efectos de los fármacos , Núcleos Septales/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
8.
Drug Alcohol Depend ; 150: 24-30, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25765483

RESUMEN

BACKGROUND: Increasing evidence suggests that chronic ethanol exposure decreases dopamine (DA) neurotransmission in the nucleus accumbens (NAc), contributing to a hypodopaminergic state during withdrawal. However, few studies have investigated adaptations in presynaptic DA terminals after chronic intermittent ethanol (CIE) exposure. In monkeys and rats, chronic ethanol exposure paradigms have been shown to increase DA uptake and D2 autoreceptor sensitivity. METHODS: The current study examined the effects of ethanol on DA terminals in CIE exposed mice during two time-points after the cessation of CIE exposure. DA release and uptake were measured using fast scan cyclic voltammetry in NAc core slices from C57BL/6J mice, 0h and 72h following three weekly cycles (4 days of 16h ethanol vapor/8h room air/day+3 days withdrawal) of CIE vapor exposure. RESULTS: Current results showed that DA release was reduced, uptake rates were increased, and inhibitory D2-type autoreceptor activity was augmented following CIE exposure in mice. CONCLUSIONS: Overall, these CIE-induced adaptations in the accumbal DA system reduce DA signaling and therefore reveal several potential mechanisms contributing to a functional hypodopaminergic state during alcohol withdrawal.


Asunto(s)
Dopamina/metabolismo , Etanol/farmacología , Núcleo Accumbens/efectos de los fármacos , Terminales Presinápticos/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/efectos de los fármacos , Estimulación Eléctrica , Etanol/sangre , Masculino , Ratones , Núcleo Accumbens/metabolismo , Terminales Presinápticos/metabolismo , Receptores de Dopamina D2/efectos de los fármacos , Recompensa , Factores de Tiempo
9.
Neuropsychopharmacology ; 40(8): 1826-36, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25689882

RESUMEN

There are ∼ 1.6 million people who meet the criteria for cocaine addiction in the United States, and there are currently no FDA-approved pharmacotherapies. Amphetamine-based dopamine-releasing drugs have shown efficacy in reducing the motivation to self-administer cocaine and reducing intake in animals and humans. It is hypothesized that amphetamine acts as a replacement therapy for cocaine through elevation of extracellular dopamine levels. Using voltammetry in brain slices, we tested the ability of a single amphetamine infusion in vivo to modulate dopamine release, uptake kinetics, and cocaine potency in cocaine-naive animals and after a history of cocaine self-administration (1.5 mg/kg/infusion, fixed-ratio 1, 40 injections/day × 5 days). Dopamine kinetics were measured 1 and 24 h after amphetamine infusion (0.56 mg/kg, i.v.). Following cocaine self-administration, dopamine release, maximal rate of uptake (Vmax), and membrane-associated dopamine transporter (DAT) levels were reduced, and the DAT was less sensitive to cocaine. A single amphetamine infusion reduced Vmax and membrane DAT levels in cocaine-naive animals, but fully restored all aspects of dopamine terminal function in cocaine self-administering animals. Here, for the first time, we demonstrate pharmacologically induced, immediate rescue of deficits in dopamine nerve-terminal function in animals with a history of high-dose cocaine self-administration. This observation supports the notion that the DAT expression and function can be modulated on a rapid timescale and also suggests that the pharmacotherapeutic actions of amphetamine for cocaine addiction go beyond that of replacement therapy.


Asunto(s)
Anfetamina/administración & dosificación , Cocaína/administración & dosificación , Cuerpo Estriado/efectos de los fármacos , Inhibidores de Captación de Dopamina/administración & dosificación , Dopamina/metabolismo , Terminales Presinápticos/metabolismo , Animales , Condicionamiento Operante/efectos de los fármacos , Cuerpo Estriado/metabolismo , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Técnicas In Vitro , Infusiones Intravenosas , Masculino , Terminales Presinápticos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Autoadministración , Factores de Tiempo
10.
Proc Natl Acad Sci U S A ; 111(26): E2751-9, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24979798

RESUMEN

The majority of neurotransmitter systems shows variations in state-dependent cell firing rates that are mechanistically linked to variations in extracellular levels, or tone, of their respective neurotransmitter. Diurnal variation in dopamine tone has also been demonstrated within the striatum, but this neurotransmitter is unique, in that variation in dopamine tone is likely not related to dopamine cell firing; this is largely because of the observation that midbrain dopamine neurons do not display diurnal fluctuations in firing rates. Therefore, we conducted a systematic investigation of possible mechanisms for the variation in extracellular dopamine tone. Using microdialysis and fast-scan cyclic voltammetry in rats, as well as wild-type and dopamine transporter (DAT) knock-out mice, we demonstrate that dopamine uptake through the DAT and the magnitude of subsecond dopamine release is inversely related to the magnitude of extracellular dopamine tone. We investigated dopamine metabolism, uptake, release, D2 autoreceptor sensitivity, and tyrosine hydroxylase expression and activity as mechanisms for this variation. Using this approach, we have pinpointed the DAT as a critical governor of diurnal variation in extracellular dopamine tone and, as a consequence, influencing the magnitude of electrically stimulated dopamine release. Understanding diurnal variation in dopamine tone is critical for understanding and treating the multitude of psychiatric disorders that originate from perturbations of the dopamine system.


Asunto(s)
Ritmo Circadiano/fisiología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Dopamina/metabolismo , Espacio Extracelular/metabolismo , Animales , Western Blotting , Cromatografía Líquida de Alta Presión , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Masculino , Ratones , Ratones Noqueados , Microdiálisis , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa/metabolismo
11.
PLoS One ; 8(12): e83852, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24349553

RESUMEN

A large body of research has aimed to determine the neurochemical factors driving differential sensitivity to ethanol between individuals in an attempt to find predictors of ethanol abuse vulnerability. Here we find that the locomotor activating effects of ethanol are markedly greater in DBA/2J compared to C57BL/6J mice, although it is unclear as to what neurochemical differences between strains mediate this behavior. Dopamine elevations in the nucleus accumbens and caudate-putamen regulate locomotor behavior for most drugs, including ethanol; thus, we aimed to determine if differences in these regions predict strain differences in ethanol-induced locomotor activity. Previous studies suggest that ethanol interacts with the dopamine transporter, potentially mediating its locomotor activating effects; however, we found that ethanol had no effects on dopamine uptake in either strain. Ex vivo voltammetry allows for the determination of ethanol effects on presynaptic dopamine terminals, independent of drug-induced changes in firing rates of afferent inputs from either dopamine neurons or other neurotransmitter systems. However, differences in striatal dopamine dynamics did not predict the locomotor-activating effects of ethanol, since the inhibitory effects of ethanol on dopamine release were similar between strains. There were differences in presynaptic dopamine function between strains, with faster dopamine clearance in the caudate-putamen of DBA/2J mice; however, it is unclear how this difference relates to locomotor behavior. Because of the role of the dopamine system in reinforcement and reward learning, differences in dopamine signaling between the strains could have implications for addiction-related behaviors that extend beyond ethanol effects in the striatum.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Etanol/farmacología , Actividad Motora/efectos de los fármacos , Terminales Presinápticos/metabolismo , Animales , Cuerpo Estriado/citología , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Transmisión Sináptica/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA