Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 430
Filtrar
1.
Toxicology ; 450: 152686, 2021 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-33486071

RESUMEN

6-Hydroxydopamine (6-OHDA) is a neurotoxin that destroy dopaminergic neurons and widely used to establish animal models of Parkinson's disease. Prostaglandins (PGs) are involved in various cellular processes, including the damage and repair of neuronal cells. However, the function of PGF2α in neuronal cells remains unclear. In this study, we investigated the effects of PGF2α against 6-OHDA-mediated toxicity in human neuroblastoma SH-SY5Y cells and elucidated its underlying molecular mechanism. When the cells were treated with 6-OHDA (50 µM) for 6 h, the expression levels of PGF2α synthetic enzymes; cyclooxygenase-2 and aldo-keto reductase 1C3 as PGF2α synthase were enhanced in an incubation-time-dependent manner. In addition, the production of PGF2α was increased in 6-OHDA-treated cells. Fluprostenol, a PGF2α receptor (FP) agonist (500 nM), suppressed 6-OHDA-induced cell death by decreasing the production of reactive oxygen species (ROS) and increasing the expression of the anti-oxidant genes. These fluprostenol-mediated effects were inhibited by co-treatment with AL8810, an FP receptor antagonist (1 µM) or transfection with FP siRNA (20 nM). Moreover, 6-OHDA-induced phosphorylation of extracellular signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase family, was inhibited by co-incubation with AL8810. Furthermore, fluprostenol itself enhanced ERK phosphorylation and further elevated the 6-OHDA-induced phosphorylation of ERK. In addition, 6-OHDA induced nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), activating anti-oxidant gene expression, was repressed by co-culturing with AL8810. These results indicate that PGF2α suppressed 6-OHDA-induced neuronal cell death by enhancing anti-oxidant gene expression via the FP receptor-ERK-Nrf2 signaling. Thus, FP receptor is a potential target for inhibition of ROS-mediated neuronal cell death.


Asunto(s)
Dinoprost/biosíntesis , Sistema de Señalización de MAP Quinasas/fisiología , Factor 2 Relacionado con NF-E2/metabolismo , Neuroprotección/fisiología , Oxidopamina/toxicidad , Receptores de Prostaglandina/metabolismo , Adrenérgicos/toxicidad , Línea Celular Tumoral , Dinoprost/agonistas , Relación Dosis-Respuesta a Droga , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Prostaglandinas F Sintéticas/farmacología
2.
Int J Mol Sci ; 21(12)2020 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-32585871

RESUMEN

The movement disorder Parkinson's disease (PD) is the second most frequently diagnosed neurodegenerative disease, and is associated with aging, the environment, and genetic factors. The intracellular aggregation of α-synuclein and the loss of dopaminergic neurons in the substantia nigra pars compacta are the pathological hallmark of PD. At present, there is no successful treatment for PD. Maackiain (MK) is a flavonoid extracted from dried roots of Sophora flavescens Aiton. MK has emerged as a novel agent for PD treatment that acts by inhibiting monoamine oxidase B. In this study, we assessed the neuroprotective potential of MK in Caenorhabditis elegans and investigated possible mechanism of this neuroprotection in the human SH-SY5Y cell line. We found that MK significantly reduced dopaminergic neuron damage in 6-hydroxydopamine (6-OHDA)-exposed worms of the BZ555 strain, with corresponding improvements in food-sensing behavior and life-span. In transgenic worms of strain NL5901 treated with 0.25 mM MK, the accumulation of α-synuclein was diminished by 27% (p < 0.01) compared with that in untreated worms. Moreover, in worms and the SH-SY5Y cell line, we confirmed that the mechanism of MK-mediated protection against PD pathology may include blocking apoptosis, enhancing the ubiquitin-proteasome system, and augmenting autophagy by increasing PINK1/parkin expression. The use of small interfering RNA to downregulate parkin expression in vivo and in vitro could reverse the benefits of MK in PD models. MK may have considerable therapeutic applications in PD.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Oxidopamina/toxicidad , Enfermedad de Parkinson/tratamiento farmacológico , Proteínas Quinasas/metabolismo , Pterocarpanos/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , alfa-Sinucleína/toxicidad , Adrenérgicos/toxicidad , Animales , Apoptosis , Autofagia , Caenorhabditis elegans/crecimiento & desarrollo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Neuroblastoma/etiología , Neuroblastoma/patología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Proteínas Quinasas/genética , Ubiquitina-Proteína Ligasas/genética
3.
J Physiol Sci ; 70(1): 16, 2020 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-32160868

RESUMEN

Respiratory disturbances present in Parkinson's disease (PD) are not well understood. Thus, studies in animal models aimed to link brain dopamine (DA) deficits with respiratory impairment are needed. Adult Wistar rats were lesioned with injection of 6-hydroxydopamine (6-OHDA) into the third cerebral ventricle. Two weeks after hypoxic test was performed in whole-body plethysmography chamber, phrenic (PHR) and hypoglossal (HG) nerve activities were recorded in normoxic and hypoxic conditions in anesthetized, vagotomized, paralyzed and mechanically ventilated rats. The effects of activation and blockade of dopaminergic carotid body receptors were investigated during normoxia in anesthetized spontaneously breathing rats. 6-OHDA injection affected resting respiratory pattern in awake animals: an increase in tidal volume and a decrease in respiratory rate had no effect on minute ventilation. Hypoxia magnified the amplitude and minute activity of the PHR and HG nerve of 6-OHDA rats. The ratio of pre-inspiratory to inspiratory HG burst amplitude was reduced in normoxic breathing. Yet, the ratio of pre-inspiratory time to total time of the respiratory cycle was increased during normoxia. 6-OHDA lesion had no impact on DA and domperidone effects on the respiratory pattern, which indicate that peripheral DA receptors are not affected in this model. Analysis of monoamines confirmed substantial striatal depletion of dopamine, serotonin and noradrenaline (NA) and reduction of NA content in the brainstem. In bilateral 6-OHDA model changes in activity of both nerves: HG (linked with increased apnea episodes) and PHR are present. Demonstrated respiratory effects could be related to specific depletion of DA and NA.


Asunto(s)
Encéfalo/fisiopatología , Nervio Hipogloso/fisiopatología , Hipoxia/fisiopatología , Enfermedad de Parkinson Secundaria/fisiopatología , Enfermedad de Parkinson/fisiopatología , Nervio Frénico/fisiopatología , Adrenérgicos/toxicidad , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Hipoxia/metabolismo , Masculino , Norepinefrina/metabolismo , Oxidopamina/toxicidad , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/metabolismo , Ratas , Ratas Wistar , Respiración
4.
Neuropharmacology ; 162: 107829, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31666199

RESUMEN

Parkinson's disease (PD) is characterized by progressive loss of midbrain dopaminergic neurons and treated with the dopamine precursor, 3,4-dihydroxy-l-phenylalanine (L-DOPA). Prolonged L-DOPA treatment is however associated with waning efficacy and the induction of L-DOPA induced dyskinesia (LID). GPR88 is an orphan G-protein Coupled Receptor (GPCR) expressed in dopaminoceptive striatal medium spiny neurons (MSNs) and their afferent corticostriatal glutamatergic neurons. Here, we studied the role of GPR88 in experimental parkinsonism and LID. Chronic L-DOPA administration to male GPR88 KO mice, subjected to unilateral 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle, resulted in more rotations than in their WT counterparts. Conversely, GPR88 KO mice had a lower abnormal involuntary movements (AIMs) score. These behavioral responses were accompanied by altered transcription of L-DOPA upregulated genes in lesioned GPR88 KO compared to WT striata. In accordance with a role for serotonin neurons in LID development, WT but not GPR88 KO striata exhibited 5-hydroxytryptamine displacement upon repeated L-DOPA treatment. Intact male GPR88 KO mice showed diminished tacrine-induced PD-like tremor and spontaneous hyperlocomotion. Dopamine and its metabolites were not increased in male GPR88 KO mice, but biosensor recordings revealed increased spontaneous/basal and evoked glutamate release in striata of male GPR88 KO mice. In conclusion, genetic deletion of GPR88 promotes l-DOPA-induced rotation and spontaneous locomotion yet suppresses the induction of LIDs and also reduces tremor. These data provide behavioral, neurochemical and molecular support that GPR88 antagonism may favour motor relief in PD patients without aggravating the induction of motor side effects.


Asunto(s)
Antiparkinsonianos/farmacología , Cuerpo Estriado/metabolismo , Discinesia Inducida por Medicamentos/genética , Levodopa/farmacología , Locomoción/efectos de los fármacos , Movimiento/efectos de los fármacos , Trastornos Parkinsonianos/genética , Receptores Acoplados a Proteínas G/genética , Adrenérgicos/toxicidad , Animales , Inhibidores de la Colinesterasa/toxicidad , Cuerpo Estriado/efectos de los fármacos , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Discinesia Inducida por Medicamentos/etiología , Discinesia Inducida por Medicamentos/metabolismo , Discinesia Inducida por Medicamentos/fisiopatología , Neuronas GABAérgicas , Ácido Glutámico/metabolismo , Locomoción/genética , Masculino , Haz Prosencefálico Medial , Ratones , Ratones Noqueados , Plasticidad Neuronal/genética , Oxidopamina/toxicidad , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/fisiopatología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Serotonina/metabolismo , Tacrina/toxicidad , Temblor
5.
Neurosci Lett ; 714: 134567, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31629033

RESUMEN

Emerging evidence continues to demonstrate that disrupted insulin signaling and altered energy metabolism may play a key role underpinning pathology in neurodegenerative conditions. Intranasally administered insulin has already shown promise as a memory-enhancing therapy in patients with Alzheimer's and animal models of the disease. Intranasal drug delivery allows for direct targeting of insulin to the brain, bypassing the blood brain barrier and minimizing systemic adverse effects. In this study, we sought to expand upon previous results that show intranasal insulin may also have promise as a Parkinson's therapy. We treated 6-OHDA parkinsonian rats with a low dose (3 IU/day) of insulin and assessed apomorphine induced rotational turns, motor deficits via a horizontal ladder test, and dopaminergic cell survival via stereological counting. We found that insulin therapy substantially reduced motor dysfunction and dopaminergic cell death induced by unilateral injection of 6-OHDA. These results confirm insulin's efficacy within this model, and do so over a longer period after model induction which more closely resembles Parkinson's disease. This study also employed a lower dose than previous studies and utilizes a delivery device, which could lead to an easier transition into human clinical trials as a therapeutic for Parkinson's disease.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Hipoglucemiantes/farmacología , Insulina/farmacología , Actividad Motora/efectos de los fármacos , Trastornos Parkinsonianos/fisiopatología , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Administración Intranasal , Adrenérgicos/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Movimiento/efectos de los fármacos , Oxidopamina/toxicidad , Enfermedad de Parkinson , Trastornos Parkinsonianos/patología , Porción Compacta de la Sustancia Negra/patología , Ratas , Tirosina 3-Monooxigenasa/metabolismo
6.
Oxid Med Cell Longev ; 2019: 8169125, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31827703

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc), and there is no cure for it at present. We have previously reported that the tetramethylpyrazine (TMP) derivative T-006 exhibited beneficial effects in Alzheimer's disease (AD) models. However, its effect on PD remains unclear. In the present study, we investigated the neuroprotective effects and underlying mechanisms of T-006 against 6-hydroxydopamine- (6-OHDA-) induced lesions in in vivo and in vitro PD models. Our results demonstrated that T-006 alleviated mitochondrial membrane potential loss and restored the energy metabolism and mitochondrial biogenesis that were induced by 6-OHDA in PC12 cells. In addition, animal experiments showed that administration of T-006 significantly attenuated the 6-OHDA-induced loss of tyrosine hydroxylase- (TH-) positive neurons in the SNpc, as well as dopaminergic nerve fibers in the striatum, and also increased the concentration of dopamine and its metabolites (DOPAC, HVA) in the striatum. Functional deficits were restored following T-006 treatment in 6-OHDA-lesioned mice, as demonstrated by improved motor coordination and rotational behavior. In addition, we found that the neuroprotective effects of T-006 were mediated, at least in part, by the activation of both the PKA/Akt/GSK-3ß and CREB/PGC-1α/NRF-1/TFAM pathways. In summary, our findings demonstrate that T-006 could be developed as a novel neuroprotective agent for PD, and the two pathways might be promising therapeutic targets for PD.


Asunto(s)
Adrenérgicos/toxicidad , Modelos Animales de Enfermedad , Hidrazonas/farmacología , Fármacos Neuroprotectores/farmacología , Oxidopamina/toxicidad , Enfermedad de Parkinson/prevención & control , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Biogénesis de Organelos , Consumo de Oxígeno/efectos de los fármacos , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
7.
Exp Neurol ; 322: 113036, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31425688

RESUMEN

The pathophysiology of Parkinson's disease (PD) and L-DOPA-induced dyskinesia (LID) is associated with aberrant neuronal activity and abnormal high levels of oscillatory activity and synchronization in several basal ganglia nuclei and the cortex. Previously, we have shown that the firing activity of neurons in the substantia nigra pars reticulata (SNr) is relevant in dyskinesia and may be driven by subthalamic nucleus (STN) hyperactivity. Conversely, low frequency oscillatory activity and synchronization in these structures seem to be more important in PD because they are not influenced by prolonged L-DOPA administration. The aim of the present study was to assess (through single-unit extracellular recording techniques under urethane anaesthesia) the neuronal activity of the entopeduncular nucleus (EPN) and its relationship with LID and STN hyperactivity, together with the oscillatory activity and synchronization between these nuclei and the cerebral cortex in 6-OHDA-lesioned rats that received long term L-DOPA treatment (or not). Twenty-four hours after the last L-DOPA injection the firing activity of EPN neurons in long term L-DOPA treated 6-OHDA-lesioned rats was more irregular and bursting compared to sham rats, being those alterations partially reversed by the acute challenge of L-DOPA. No correlation between EPN neurons firing activity and abnormal involuntary movements score was found. However, there was a significant correlation between the firing activity parameters of EPN and STN neurons recorded from long term L-DOPA treated 6-OHDA-lesioned rats. Low frequency oscillatory activity and synchronization both within the EPN and with the cerebral cortex were enhanced in 6-OHDA-lesioned animals. These changes were reversed by the acute L-DOPA challenge only in long term L-DOPA treated 6-OHDA-lesioned rats. Altogether, these results obtained from long term L-DOPA treated 6-OHDA-lesioned rats suggest (1) a likely relationship between STN and EPN firing patterns and spiking phases induced by changes after prolonged L-DOPA administration and (2) that the effect of L-DOPA on the firing pattern, low frequency oscillatory activity and synchronization in the EPN may have a relevant role in LID.


Asunto(s)
Discinesia Inducida por Medicamentos/fisiopatología , Núcleo Entopeduncular/efectos de los fármacos , Núcleo Entopeduncular/fisiopatología , Levodopa/farmacología , Trastornos Parkinsonianos/fisiopatología , Adrenérgicos/toxicidad , Animales , Antiparkinsonianos/farmacología , Masculino , Neuronas/efectos de los fármacos , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Ratas , Ratas Sprague-Dawley , Núcleo Subtalámico/efectos de los fármacos , Núcleo Subtalámico/fisiopatología
8.
Neuropharmacology ; 158: 107725, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31351976

RESUMEN

Selective blockade of serotonin 2A (5-HT2A) receptors is a promising strategy to reduce L-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesia and has shown efficacy in a Phase III clinical trial for dopaminergic psychosis in Parkinson's disease (PD). However, pre-clinical and clinical evidence suggest that, while this approach may be effective and well tolerated, there might be a ceiling beyond which no further therapeutic benefit might be achieved. There is mounting evidence that 5-HT2A receptors form a functional hetero-complex with metabotropic glutamate 2 (mGlu2) receptors, with antagonism of 5-HT2A receptors and activation of mGlu2 receptors producing similar effects on the Gi/Gq signalling ratio at the intra-cellular level. Based on this interaction between 5-HT2A and mGlu2 receptors, we hypothesised that activation of mGlu2 receptors would alleviate dyskinesia and psychosis in PD. LY-354,740 is a selective mGlu2/3 orthosteric agonist that was previously tested in the clinic. In experiments conducted in the 6-hydroxydopamine (6-OHDA)-lesioned rat and the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned marmoset, we found that mGlu2/3 activation with LY-354,740 significantly reduced the expression of dyskinesia and psychosis-like behaviours, while simultaneously enhancing l-DOPA therapeutic benefit. Moreover, mGlu2/3 activation with LY-354,740 attenuated the development of dyskinesia. These data indicate that activation of mGlu2/3 receptors is a therapeutic strategy that may provide relief for both motor and-non-motor treatment-related complications in PD.


Asunto(s)
Antiparkinsonianos/efectos adversos , Conducta Animal/efectos de los fármacos , Compuestos Bicíclicos con Puentes/farmacología , Discinesia Inducida por Medicamentos/etiología , Agonistas de Aminoácidos Excitadores/farmacología , Levodopa/efectos adversos , Trastornos Parkinsonianos/tratamiento farmacológico , Psicosis Inducidas por Sustancias/etiología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Adrenérgicos/toxicidad , Animales , Compuestos Bicíclicos con Puentes/uso terapéutico , Callithrix , Dopaminérgicos/toxicidad , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Agonistas de Aminoácidos Excitadores/uso terapéutico , Intoxicación por MPTP/tratamiento farmacológico , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Psicosis Inducidas por Sustancias/tratamiento farmacológico , Ratas , Receptores de Glutamato Metabotrópico/agonistas
9.
Toxicol In Vitro ; 60: 400-411, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31247335

RESUMEN

Neuronal exposure to 6-hydroxydopamine (6-OHDA), a hydroxylated analog of dopamine, constitutes a very useful strategy for studying the molecular events associated with neuronal death in Parkinson's disease. 6-OHDA increases oxidant levels and impairs mitochondrial respiratory chain, thus promoting neuronal injury and death. Despite the extensive use of 6-OHDA in animal models, the exact molecular events triggered by this neurotoxicant at the neuronal level have not been yet fully understood. Human IMR-32 neuroblastoma cells exposed to increasing concentrations of 6-OHDA displayed high levels of reactive oxygen species and increased plasma membrane permeability with concomitant cell viability diminution. As part of the neuronal response to 6-OHDA exposure, the nuclear translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) p65 subunit was observed. NFκB nuclear localization was also accompanied by an increase of IκB phosphorylation as well as a rise in cyclooxygenase-2 (COX-2) and the prostaglandin receptor, EP4, mRNA levels. Even though the canonical pathways participating in the modulation of NFκB have been extensively described, here we tested the hypothesis that 6-OHDA-induced injury can activate lipid signaling and, in turn, modulate the transcriptional response. 6-OHDA challenge triggered the activation of lipid signaling pathways and increased phosphatidic acid (PA), diacylglycerol and free fatty acid levels in human neuroblastoma cells. The inhibition of PA production was able to prevent the decrease in cell viability triggered by 6-OHDA, the nuclear translocation of NFκB p65 subunit and the rise in COX-2 mRNA expression. Our results indicate that the onset of the inflammatory process triggered by 6-OHDA involves the activation of PA signaling that, in turn, governs NFκB subcellular localization and COX-2 expression.


Asunto(s)
Adrenérgicos/toxicidad , FN-kappa B/metabolismo , Oxidopamina/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , Dopamina/metabolismo , Humanos , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad , Subtipo EP4 de Receptores de Prostaglandina E/genética
10.
Neuropharmacology ; 151: 98-111, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30959019

RESUMEN

Noradrenergic degeneration in the locus coeruleus (LC) seems a convergent neuropathological marker of different neurodegenerative diseases. Herein, we investigated the temporal development of apoptotic signaling activation in the LC, noradrenergic dysfunction and behavioral impairments in rats following the noradrenergic lesion of the LC. For this purpose, the dopamine reuptake inhibitor nomifensine was administered 1 h before the stereotaxic bilateral injections of 6-hydroxydopamine (6-OHDA; 5, 10 or 20 µg/hem) into the LC. The behavioral and neurochemical analyses were performed at 7, 21 and 42 days after 6-OHDA injections. All doses of 6-OHDA induced neuronal death in LC, but only the highest dose (20 µg/hem) disrupted the motor function. 6-OHDA (5 µg/hem) injection induced short-term memory deficits in all periods, olfactory discrimination and long-term memory impairments at 7 days, and depressive-like behaviors at 21 and 42 days after injection. Moreover, 6-OHDA infusion increased Bax/Bcl2 ratio and caspase 3 levels, and decreased the dopamine ß-hydroxylase immunocontent in the LC. Noradrenergic neurotransmission dysfunction was observed in the LC, olfactory bulb, prefrontal cortex, hippocampus and striatum. The intranasal (i.n.) noradrenaline (NA) infusion restored the impairments in the olfactory discrimination, short-term memory and depressive-like behavior of 6-OHDA-lesioned rats. In addition, these effects were blocked by the prior i.n. infusion of the ß3-adrenergic receptor antagonist SR59230A. These findings indicate that the 6-OHDA injection into the LC induced the apoptosis signaling activation, noradrenergic neurotransmission dysfunction and behavioral impairments that were restored via ß3-adrenergic receptors activation mediated by the i.n. NA administration.


Asunto(s)
Adrenérgicos/toxicidad , Neuronas Adrenérgicas/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Locus Coeruleus/efectos de los fármacos , Oxidopamina/toxicidad , Receptores Adrenérgicos beta 3/metabolismo , Animales , Discriminación en Psicología/efectos de los fármacos , Modelos Animales de Enfermedad , Locus Coeruleus/metabolismo , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Wistar , Reconocimiento en Psicología/efectos de los fármacos , Factores de Tiempo
11.
Mov Disord ; 34(5): 697-707, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31002755

RESUMEN

BACKGROUND: Levodopa-induced dyskinesias are an often debilitating side effect of levodopa therapy in Parkinson's disease. Although up to 90% of individuals with PD develop this side effect, uniformly effective and well-tolerated antidyskinetic treatment remains a significant unmet need. The pathognomonic loss of striatal dopamine in PD results in dysregulation and disinhibition of striatal CaV1.3 calcium channels, leading to synaptopathology that appears to be involved in levodopa-induced dyskinesias. Although there are clinically available drugs that can inhibit CaV1.3 channels, they are not adequately potent and have only partial and transient impact on levodopa-induced dyskinesias. METHODS: To provide unequivocal target validation, free of pharmacological limitations, we developed a CaV1.3 shRNA to provide high-potency, target-selective, mRNA-level silencing of striatal CaV1.3 channels and examined its ability to impact levodopa-induced dyskinesias in severely parkinsonian rats. RESULTS: We demonstrate that vector-mediated silencing of striatal CaV1.3 expression in severely parkinsonian rats prior to the introduction of levodopa can uniformly and completely prevent induction of levodopa-induced dyskinesias, and this antidyskinetic benefit persists long term and with high-dose levodopa. In addition, this approach is capable of ameliorating preexisting severe levodopa-induced dyskinesias. Importantly, motoric responses to low-dose levodopa remained intact in the presence of striatal CaV1.3 silencing, indicating preservation of levodopa benefit without dyskinesia liability. DISCUSSION: The current data provide some of the most profound antidyskinetic benefit reported to date and suggest that genetic silencing of striatal CaV1.3 channels has the potential to transform treatment of individuals with PD by allowing maintenance of motor benefit of levodopa in the absence of the debilitating levodopa-induced dyskinesia side effect. © 2019 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Antiparkinsonianos/efectos adversos , Canales de Calcio/genética , Discinesia Inducida por Medicamentos/prevención & control , Levodopa/efectos adversos , Neostriado/metabolismo , Trastornos Parkinsonianos/tratamiento farmacológico , Adrenérgicos/toxicidad , Animales , Modelos Animales de Enfermedad , Discinesia Inducida por Medicamentos/etiología , Discinesia Inducida por Medicamentos/terapia , Proteínas Fluorescentes Verdes , Sustancias Luminiscentes , Haz Prosencefálico Medial , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Sustancia Negra , Tirosina 3-Monooxigenasa/metabolismo
12.
Neurobiol Dis ; 125: 55-66, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30677495

RESUMEN

Parkinson's disease (PD) presents with a constellation of non-motor symptoms, notably increased anxiety, which are currently poorly treated and underrepresented in animal models of the disease. Human post-mortem studies report loss of catecholaminergic neurons in the pre-symptomatic phases of PD when anxiety symptoms emerge, and a large literature from rodent and human studies indicate that catecholamines are important mediators of anxiety via their modulatory effects on limbic regions such as the amygdala. On the basis of these observations, we hypothesized that anxiety in PD could result from an early loss of catecholaminergic inputs to the amygdala and/or other limbic structures. To interrogate this hypothesis, we bilaterally injected the neurotoxin 6-OHDA in the mouse basolateral amygdala (BL). This produced a restricted pattern of catecholaminergic (tyrosine-hydroxylase-labeled) denervation in the BL, intercalated cell masses and ventral hippocampus, but not the central amygdala or prefrontal cortex. We found that this circuit-specific lesion did not compromise performance on multiple measures of motor function (home cage, accelerating rotarod, beam balance, pole climbing), but did increase anxiety-like behavior in the elevated plus-maze and light-dark exploration tests. Fear behavior in the pavlovian cued conditioning and passive avoidance assays was, by contrast, unaffected; possibly due to preservation of catecholamine innervation of the central amygdala from the periaqueductal gray. These data provide some of the first evidence implicating loss of catecholaminergic neurotransmission in midbrain-amygdala circuits to increased anxiety-like behavior. Our findings offer an initial step towards identifying the neural substrates for pre-motor anxiety symptoms in PD.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Ansiedad/fisiopatología , Catecolaminas/antagonistas & inhibidores , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/psicología , Adrenérgicos/toxicidad , Amígdala del Cerebelo/efectos de los fármacos , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidopamina/toxicidad
13.
Behav Pharmacol ; 30(1): 89-94, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29847340

RESUMEN

The development of Parkinson's disease (PD) involves the degeneration of dopaminergic neurons caused by oxidative stress. Accumulating clinical evidence indicates that high blood levels of uric acid (UA), an intrinsic antioxidative substance, are associated with reduced risk of PD. However, this hypothesis has not been confirmed by in-vivo experiments. The present study investigated the effects of UA on behavioral abnormalities in the development of PD. We used unilateral 6-hydroxydopamine-lesioned mice, which were fed on a diet containing 1% UA and 2.5% potassium oxonate (an uricase inhibitor) to induce hyperuricemia. A significant elevation in UA levels was found in groups that were fed a UA diet. The 6-hydroxydopamine-lesioned mice showed impaired rotarod performance and increased apomorphine-induced contralateral rotations. These behavioral abnormalities were significantly reversed by feeding a UA diet for 1 week before and 5 weeks after surgery (subchronic hyperuricemia). These behavioral improvements occurred in parallel with recovery of tyrosine hydroxylase protein levels in the lesioned striatal side. The present study with a dietary hyperuricemia mice model confirms that UA exerts a neuroprotective effect on dopaminergic neuronal loss, improving motor dysfunction and ameliorating PD development.


Asunto(s)
Trastornos Mentales/sangre , Trastornos Mentales/etiología , Enfermedad de Parkinson Secundaria/complicaciones , Ácido Úrico/sangre , Adrenérgicos/toxicidad , Animales , Apomorfina/farmacología , Modelos Animales de Enfermedad , Hiperuricemia/sangre , Hiperuricemia/etiología , Masculino , Trastornos Mentales/dietoterapia , Ratones , Ratones Endogámicos ICR , Actividad Motora/efectos de los fármacos , Oxidopamina/toxicidad , Ácido Oxónico/administración & dosificación , Enfermedad de Parkinson Secundaria/inducido químicamente , Prueba de Desempeño de Rotación con Aceleración Constante , Tirosina 3-Monooxigenasa/metabolismo
14.
Exp Brain Res ; 237(1): 29-36, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30298296

RESUMEN

L-3,4-Dihydroxyphenylalanine (L-DOPA) is the most effective therapy for motor symptoms of Parkinson's disease (PD); however, with repeated administration, as many as 94% of PD patients develop complications such as L-DOPA-induced dyskinesia. We previously demonstrated that EMD-281,014, a highly selective serotonin 2A (5-HT2A) receptor antagonist, reduces the severity of dyskinesia in the parkinsonian marmoset, without interfering with L-DOPA anti-parkinsonian benefit. Here, we assessed the effects of EMD-281,014 on L-DOPA-induced abnormal involuntary movements (AIMs) in the 6-hydroxydopamine (6-OHDA)-lesioned rat. We first determined the pharmacokinetic profile of EMD-281,014, to administer doses leading to clinically relevant plasma levels in the behavioural experiments. Dyskinetic 6-OHDA-lesioned rats were then administered EMD-281,014 (0.01, 0.03 and 0.1 mg/kg) or vehicle in combination with L-DOPA and AIMs severity was evaluated. We also assessed the effect of EMD-281,014 on L-DOPA anti-parkinsonian action with the cylinder test. We found that the addition of EMD-281,014 (0.01, 0.03 and 0.1 mg/kg) to L-DOPA did not reduce AIMs severity (P > 0.05), when compared to vehicle. EMD-281,014 did not compromise L-DOPA anti-parkinsonian action. Our results suggest that the highly selective 5-HT2A receptor antagonist EMD-281,014 is well-tolerated by parkinsonian rats, but does not attenuate L-DOPA-induced AIMs. Our results highlight differences between rodent and primate models of PD when it comes to determining the anti-dyskinetic action of 5-HT2A receptor antagonists.


Asunto(s)
Discinesia Inducida por Medicamentos/tratamiento farmacológico , Indoles/uso terapéutico , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Piperazinas/uso terapéutico , Antagonistas del Receptor de Serotonina 5-HT2/uso terapéutico , Adrenérgicos/toxicidad , Animales , Antiparkinsonianos/efectos adversos , Área Bajo la Curva , Monoaminas Biogénicas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Discinesia Inducida por Medicamentos/etiología , Femenino , Lateralidad Funcional/efectos de los fármacos , Indoles/sangre , Levodopa/efectos adversos , Oxidopamina/toxicidad , Enfermedad de Parkinson Secundaria/inducido químicamente , Piperazinas/sangre , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Antagonistas del Receptor de Serotonina 5-HT2/sangre
15.
Neurotoxicology ; 70: 1-11, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30359634

RESUMEN

Mitochondrial dysfunction has been demonstrated to have a central role in Parkinson Disease (PD) pathophysiology. Some studies have indicated that PD causes an impairment in mitochondrial bioenergetics; however, the effects of PD on brain-region specific bioenergetics was never investigated before. This study aimed to evaluate mitochondrial bioenergetics in different rat brain structures in an in vitro model of PD using 6-OHDA. Rat brain slices of hippocampus, striatum, and cortex were exposed to 6-OHDA (100 µM) for 1 h and mitochondrial bioenergetic parameters, peroxide production, lactate dehydrogenase (LDH) and citrate synthase (CS) activities were analyzed. Hippocampus slices exposed to 6-OHDA presented increased peroxide production but, no mitochondrial adaptive response against 6-OHDA damage. Cortex slices exposed to 6-OHDA presented increased oxygen flux related to oxidative phosphorylation and energetic pathways exchange demonstrated by the increase in LDH activity, suggesting a mitochondrial compensatory response. Striatum slices exposed to 6-OHDA presented a decrease of oxidative phosphorylation and decrease of oxygen flux related to ATP-synthase indicating an impairment in the respiratory chain. The co-incubation of 6-OHDA with n-acetylcysteine (NAC) abolished the effects of 6-OHDA on mitochondrial function in all brain regions tested, indicating that the increased reactive oxygen species (ROS) production is responsible for the alterations observed in mitochondrial bioenergetics. The present results indicate a brain-region specific response against 6-OHDA, providing new insights into brain mitochondrial bioenergetic function in PD. These findings may contribute to the development of future therapies with a target on energy metabolism.


Asunto(s)
Encéfalo/metabolismo , Metabolismo Energético/fisiología , Mitocondrias/metabolismo , Oxidopamina/toxicidad , Consumo de Oxígeno/fisiología , Adrenérgicos/toxicidad , Animales , Encéfalo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Masculino , Mitocondrias/efectos de los fármacos , Técnicas de Cultivo de Órganos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Wistar
16.
Oxid Med Cell Longev ; 2018: 2131895, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30510616

RESUMEN

Parkinson's disease is a degenerative and progressive illness characterized by the degeneration of dopaminergic neurons. 6-hydroxydopamine (6-OHDA) is a widespread model for induction of molecular and behavioral alterations similar to Parkinson and has contributed for testing of compounds with neuroprotective potential. The Brazilian plant Anacardium microcarpum is used in folk medicine for treatment of several illnesses; however, the knowledge about toxicology and biological effects for this plant is very rare. The neuroprotective effect from hydroalcoholic extract and methanolic and acetate fraction of A. microcarpum on 6-OHDA-induced damage on chicken brain slices was investigated in this study. 6-OHDA decreased cellular viability measured by MTT reduction assay, induced lipid peroxidation by HPLC, stimulated Glutathione-S-Transferase and Thioredoxin Reductase activity, and decreased Glutathione Peroxidase activity and the total content of thiols containing compounds. The methanolic fraction of A. microcarpum presented the better neuroprotective effects in 6-OHDA-induced damage in relation with hydroalcoholic and acetate fraction. The presence of AKT and ERK1/2 pharmacological inhibitors blocked the protective effect of methanolic fraction suggesting the involvement of survival pathways in the neuroprotection by the plant. The plant did not prevent 6-OHDA autoxidation or 6-OHDA-induced mitochondrial dysfunction. Thus, the neuroprotective effect of the methanolic fraction of A. microcarpum appears to be attributed in part to chelating properties of extract toward reactive species and is dependent on ERK1/2 and AKT phosphorylation. This study contributes to the understanding of biochemical mechanisms implied in neuroprotective effects of the vegetal species A. microcarpum.


Asunto(s)
Anacardium/química , Regulación de la Expresión Génica/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oxidopamina/toxicidad , Enfermedad de Parkinson/tratamiento farmacológico , Extractos Vegetales/farmacología , Adrenérgicos/toxicidad , Animales , Pollos , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Femenino , Masculino , Mitocondrias/metabolismo , Mitocondrias/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo
17.
Exp Neurol ; 309: 79-90, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30076831

RESUMEN

Gene therapy is currently an irreversible approach, without possibilities to fine-tune or halt the expression of a therapeutic gene product. Especially when expressing neurotrophic factors to treat neurodegenerative disorders, options to regulate transgene expression levels might be beneficial. We thus developed an advanced single-genome inducible AAV vector for expression of GDNF, under control of the approved small molecule drug mifepristone. In the rat brain, GDNF expression can be induced over a wide range up to three hundred-fold over endogenous background, and completely returns to baseline within 3-4 weeks. When applied with appropriate serotype and titre, the vector is absolutely free of any non-induced background expression. In the BACHD model of Huntington's disease we demonstrate that the vector can be kept in a continuous ON-state for extended periods of time. In a model of Parkinson's disease we demonstrate that repeated short-term expression of GDNF restores motor capabilities in 6-OHDA-lesioned rats. We also report on sex-dependent pharmacodynamics of mifepristone in the rodent brain. Taken together, we show that wide-range and high-level induction, background-free, fully reversible and therapeutically active GDNF expression can be achieved under tight pharmacological control by this novel AAV - "Gene Switch" vector.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/terapia , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Ácido 3,4-Dihidroxifenilacético/metabolismo , Adrenérgicos/toxicidad , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ácido Homovanílico/metabolismo , Antagonistas de Hormonas/uso terapéutico , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Mifepristona/uso terapéutico , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Sinapsinas/genética , Sinapsinas/metabolismo , Sinucleínas/genética , Sinucleínas/metabolismo , Transducción Genética
18.
Neurosci Bull ; 34(3): 476-484, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29508251

RESUMEN

Previous studies have shown that electroacupuncture (EA) promotes recovery of motor function in Parkinson's disease (PD). However the mechanisms are not completely understood. Clinically, the subthalamic nucleus (STN) is a critical target for deep brain stimulation treatment of PD, and vesicular glutamate transporter 1 (VGluT1) plays an important role in the modulation of glutamate in the STN derived from the cortex. In this study, a 6-hydroxydopamine (6-OHDA)-lesioned rat model of PD was treated with 100 Hz EA for 4 weeks. Immunohistochemical analysis of tyrosine hydroxylase (TH) showed that EA treatment had no effect on TH expression in the ipsilateral striatum or substantia nigra pars compacta, though it alleviated several of the parkinsonian motor symptoms. Compared with the hemi-parkinsonian rats without EA treatment, the 100 Hz EA treatment significantly decreased apomorphine-induced rotation and increased the latency in the Rotarod test. Notably, the EA treatment reversed the 6-OHDA-induced down-regulation of VGluT1 in the STN. The results demonstrated that EA alleviated motor symptoms and up-regulated VGluT1 in the ipsilateral STN of hemi-parkinsonian rats, suggesting that up-regulation of VGluT1 in the STN may be related to the effects of EA on parkinsonian motor symptoms via restoration of function in the cortico-STN pathway.


Asunto(s)
Electroacupuntura/métodos , Enfermedad de Parkinson Secundaria/terapia , Núcleo Subtalámico/metabolismo , Regulación hacia Arriba/fisiología , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Adrenérgicos/toxicidad , Animales , Apomorfina/farmacología , Modelos Animales de Enfermedad , Agonistas de Dopamina/farmacología , Lateralidad Funcional/efectos de los fármacos , Masculino , Haz Prosencefálico Medial/lesiones , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidopamina/toxicidad , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/fisiopatología , Ratas , Ratas Sprague-Dawley , Núcleo Subtalámico/efectos de los fármacos , Núcleo Subtalámico/patología , Tirosina 3-Monooxigenasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
19.
Neuropharmacology ; 131: 116-127, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29197517

RESUMEN

L-DOPA-induced dyskinesia (LID) remains a significant problem in the management of Parkinson's disease (PD). In rodent and macaque models of PD, delta opioid receptor agonists have anti-parkinsonian actions while mu opioid antagonists can reduce the expression of LID. DPI-289 is a novel molecule with a unique combination of opioid receptor DAMA actions: delta agonist (Ki: 0.73 nM); mu antagonist (Ki: 12 nM). We demonstrated that DPI-289 has oral bioavailability and established its pharmacokinetic profile in both rat and primate. We hypothesised that these combined DAMA actions would provide an enhancement of L-DOPA effect without an associated increase in dyskinesia. In parkinsonian 6-OHDA lesioned rats and MPTP-lesioned macaques, DPI-289 provided anti-parkinsonian actions as monotherapy and an enhancement of L-DOPA benefit. Thus, acute administration of DPI-289 (3 mg/kg, p.o.) to 6-OHDA-lesioned rats produced a significant reduction in forelimb asymmetry (by 48%) that was maintained throughout the fifteen-day repeat-treatment period. Importantly, and in contrast to L-DOPA administration (6 mg/kg, i.p.), these benefits were not compromised by the development of abnormal involuntary movements. In the macaque, as monotherapy, DPI-289 (10 and 20 mg/kg) had significant, though incomplete, anti-parkinsonian actions lasting approximately 4 h. These benefits were not associated with dyskinesia. In fact, over the 6 h period of observation, DPI-289 (20 mg/kg) decreased parkinsonism by 19% and increased activity by 67% compared to vehicle treatment. By contrast, while high-dose L-DOPA (LDh) alone alleviated parkinsonism (for 3 h) this benefit was accompanied by significant dyskinesia that was disabling in nature. LDh provided a 50% reduction in parkinsonism over 6 h and 151% increase in activity. The combination of DPI-289 (20 mg/kg) and a low-dose of L-DOPA (LDl) provided anti-parkinsonian benefits greater than LDl alone without eliciting any significant dyskinesia. Treatment with LDl alone provided only transient statistically significant anti-parkinsonian benefit. However, the combination of LDl and DPI-289 reduced parkinsonism for 6 h (duration of monitoring), with parkinsonism being reduced by 35% and activity increased by 90% but with no increase in dyskinesia over that observed with LDl alone. Thus, DPI-289 has potential to improve the benefits of dopaminergic therapy in Parkinson's disease.


Asunto(s)
Benzamidas/uso terapéutico , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Levodopa/efectos adversos , Antagonistas de Narcóticos/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Piperazinas/uso terapéutico , Receptores Opioides delta/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Adrenérgicos/toxicidad , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/uso terapéutico , Animales , Benzamidas/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Discinesia Inducida por Medicamentos/etiología , Femenino , Cobayas , Macaca , Masculino , Ratones , Ratones Endogámicos C57BL , Movimiento/efectos de los fármacos , Antagonistas de Narcóticos/farmacocinética , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Trastornos Parkinsonianos/sangre , Trastornos Parkinsonianos/tratamiento farmacológico , Piperazinas/farmacología , Ratas Sprague-Dawley , Conducto Deferente/efectos de los fármacos , Conducto Deferente/metabolismo
20.
Brain Struct Funct ; 223(3): 1071-1089, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29081007

RESUMEN

Extinction of Pavlovian conditioning is a complex process that involves brain regions such as the medial prefrontal cortex (mPFC), the amygdala and the locus coeruleus. In particular, noradrenaline (NA) coming from the locus coeruleus has been recently shown to play a different role in two subregions of the mPFC, the prelimbic (PL) and the infralimbic (IL) regions. How these regions interact in conditioning and subsequent extinction is an open issue. We studied these processes using two approaches: computational modelling and NA manipulation in a conditioned place preference paradigm (CPP) in mice. In the computational model, NA in PL and IL causes inputs arriving to these regions to be amplified, thus allowing them to modulate learning processes in amygdala. The model reproduces results from studies involving depletion of NA from PL, IL, or both in CPP. In addition, we simulated new experiments of NA manipulations in mPFC, making predictions on the possible results. We searched the parameters of the model and tested the robustness of the predictions by performing a sensitivity analysis. We also present an empirical experiment where, in accord with the model, a double depletion of NA from both PL and IL in CPP with amphetamine impairs extinction. Overall the proposed model, supported by anatomical, physiological, and behavioural data, explains the differential role of NA in PL and IL and opens up the possibility to understand extinction mechanisms more in depth and hence to aid the development of treatments for disorders such as addiction.


Asunto(s)
Amígdala del Cerebelo/fisiología , Condicionamiento Clásico/fisiología , Comportamiento de Búsqueda de Drogas/fisiología , Extinción Psicológica/fisiología , Corteza Prefrontal/fisiología , Adrenérgicos/toxicidad , Animales , Aprendizaje por Asociación/fisiología , Simulación por Computador , Condicionamiento Clásico/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Norepinefrina/metabolismo , Oxidopamina/toxicidad , Piperazinas/toxicidad , Corteza Prefrontal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...