Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biol Psychiatry ; 90(6): 385-398, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-33965197

RESUMEN

BACKGROUND: Polymorphisms in GRM3, the gene encoding the mGlu3 metabotropic glutamate receptor, are associated with impaired cognition and neuropsychiatric disorders such as schizophrenia. Limited availability of selective genetic and molecular tools has hindered progress in developing a clear understanding of the mechanisms through which mGlu3 receptors regulate synaptic plasticity and cognition. METHODS: We examined associative learning in mice with trace fear conditioning, a hippocampal-dependent learning task disrupted in patients with schizophrenia. Underlying cellular mechanisms were assessed using ex vivo hippocampal slice preparations with selective pharmacological tools and selective genetic deletion of mGlu3 receptor expression in specific neuronal subpopulations. RESULTS: mGlu3 receptor activation enhanced trace fear conditioning and reversed deficits induced by subchronic phencyclidine. Mechanistic studies revealed that mGlu3 receptor activation induced metaplastic changes, biasing afferent stimulation to induce long-term potentiation through an mGlu5 receptor-dependent, endocannabinoid-mediated, disinhibitory mechanism. Selective genetic deletion of either mGlu3 or mGlu5 from hippocampal pyramidal cells eliminated effects of mGlu3 activation, revealing a novel mechanism by which mGlu3 and mGlu5 interact to enhance cognitive function. CONCLUSIONS: These data demonstrate that activation of mGlu3 receptors in hippocampal pyramidal cells enhances hippocampal-dependent cognition in control and impaired mice by inducing a novel form of metaplasticity to regulate circuit function, providing a clear mechanism through which genetic variation in GRM3 can contribute to cognitive deficits. Developing approaches to positively modulate mGlu3 receptor function represents an encouraging new avenue for treating cognitive disruption in schizophrenia and other psychiatric diseases.


Asunto(s)
Receptores de Glutamato Metabotrópico , Esquizofrenia , Animales , Cognición , Hipocampo/metabolismo , Potenciación a Largo Plazo , Ratones , Receptores de Glutamato Metabotrópico/metabolismo , Esquizofrenia/genética
2.
ACS Pharmacol Transl Sci ; 2(3): 198-209, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-31259318

RESUMEN

Metabotropic glutamate (mGlu) receptor type 5 (mGlu5) positive allosteric modulators (PAMs) enhance hippocampal long-term potentiation (LTP) and have cognition-enhancing effects in animal models. These effects were initially thought to be mediated by potentiation of mGlu5 modulation of N-methyl-d-aspartate receptor (NMDAR) currents. However, a biased mGlu5 PAM that potentiates Gαq-dependent mGlu5 signaling, but not mGlu5 modulation of NMDAR currents, retains cognition-enhancing effects in animal models, suggesting that potentiation of NMDAR currents is not required for these in vivo effects of mGlu5 PAMs. However, it is not clear whether the potentiation of NMDAR currents is critical for the ability of mGlu5 PAMs to enhance hippocampal LTP. We now report the characterization of effects of two structurally distinct mGlu5 PAMs, VU-29 and VU0092273, on NMDAR currents and hippocampal LTP. As with other mGlu5 PAMs that do not display observable bias for potentiation of NMDAR currents, VU0092273 enhanced both mGlu5 modulation of NMDAR currents and induction of LTP at the hippocampal Schaffer collateral (SC)-CA1 synapse. In contrast, VU-29 did not potentiate mGlu5 modulation of NMDAR currents but induced robust potentiation of hippocampal LTP. Interestingly, both VU-29 and VU0092273 suppressed evoked inhibitory postsynaptic currents (eIPSCs) in CA1 pyramidal cells, and this effect was blocked by the cannabinoid receptor type 1 (CB1) antagonist AM251. Furthermore, AM251 blocked the ability of both mGlu5 PAMs to enhance LTP. Finally, both PAMs failed to enhance LTP in mice with the restricted genetic deletion of mGlu5 in CA1 pyramidal cells. Taken together with previous findings, these results suggest that enhancement of LTP by mGlu5 PAMs does not depend on mGlu5 modulation of NMDAR currents but is mediated by a previously established mechanism in which mGlu5 in CA1 pyramidal cells induces endocannabinoid release and CB1-dependent disinhibition.

3.
Biol Psychiatry ; 85(12): 989-1000, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31003787

RESUMEN

BACKGROUND: The prefrontal cortex (PFC) integrates information from multiple inputs to exert top-down control allowing for appropriate responses in a given context. In psychiatric disorders such as posttraumatic stress disorder, PFC hyperactivity is associated with inappropriate fear in safe situations. We previously reported a form of muscarinic acetylcholine receptor (mAChR)-dependent long-term depression in the PFC that we hypothesize is involved in appropriate fear responding and could serve to reduce cortical hyperactivity following stress. However, it is unknown whether this long-term depression occurs at fear-related inputs. METHODS: Using optogenetics with extracellular and whole-cell electrophysiology, we assessed the effect of mAChR activation on the synaptic strength of specific PFC inputs. We used selective pharmacological tools to assess the involvement of M1 mAChRs in conditioned fear extinction in control mice and in the stress-enhanced fear-learning model. RESULTS: M1 mAChR activation induced long-term depression at inputs from the ventral hippocampus and basolateral amygdala but not from the mediodorsal nucleus of the thalamus. We found that systemic M1 mAChR antagonism impaired contextual fear extinction. Treatment with an M1 positive allosteric modulator enhanced contextual fear extinction consolidation in stress-enhanced fear learning-conditioned mice. CONCLUSIONS: M1 mAChRs dynamically modulate synaptic transmission at two PFC inputs whose activity is necessary for fear extinction, and M1 mAChR function is required for proper contextual fear extinction. Furthermore, an M1 positive allosteric modulator enhanced the consolidation of fear extinction in the stress-enhanced fear-learning model, suggesting that M1 positive allosteric modulators may provide a novel treatment strategy to facilitate exposure therapy in the clinic for the treatment of posttraumatic stress disorder.


Asunto(s)
Miedo/fisiología , Neuronas/fisiología , Corteza Prefrontal/fisiología , Receptor Muscarínico M1/fisiología , Trastornos por Estrés Postraumático/fisiopatología , Animales , Complejo Nuclear Basolateral/fisiología , Condicionamiento Clásico , Extinción Psicológica/fisiología , Hipocampo/fisiología , Depresión Sináptica a Largo Plazo , Núcleo Talámico Mediodorsal/fisiología , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Transmisión Sináptica/fisiología
4.
Trends Pharmacol Sci ; 40(4): 240-252, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30824180

RESUMEN

The metabotropic glutamate (mGlu) receptors are a family of G-protein-coupled receptors (GPCRs) that regulate cell physiology throughout the nervous system. The potential of mGlu receptors as therapeutic targets has been bolstered by current research that has provided insight into the diverse modes of mGlu activation and signaling. In particular, the allosteric modulation of mGlu receptors represents a major area of focus in studies of basic pharmacology as well as drug development, largely due to the high subtype specificity achievable by targeting allosteric sites on mGlu receptors. These provide sophisticated regulation of neuronal excitability and synaptic transmission to influence behavioral output. Here, we review how these allosteric mechanisms have been leveraged preclinically to demonstrate the therapeutic potential of allosteric modulators for neurological and neuropsychiatric disorders, such as autism, cognitive impairment, Parkinson's disease (PD), stress, and schizophrenia.


Asunto(s)
Desarrollo de Medicamentos , Trastornos Mentales/tratamiento farmacológico , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Regulación Alostérica , Sitio Alostérico , Animales , Humanos , Trastornos Mentales/fisiopatología , Receptores de Glutamato Metabotrópico/metabolismo , Transmisión Sináptica
5.
Neuropharmacology ; 144: 19-28, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30326237

RESUMEN

Stress can precipitate or worsen symptoms of many psychiatric illnesses. Dysregulation of the prefrontal cortex (PFC) glutamate system may underlie these disruptions and restoring PFC glutamate signaling has emerged as a promising avenue for the treatment of stress disorders. Recently, we demonstrated that activation of metabotropic glutamate receptor subtype 3 (mGlu3) induces a postsynaptic form of long-term depression (LTD) that is dependent on the activity of another subtype, mGlu5. Stress exposure disrupted this plasticity, but the underlying signaling mechanisms and involvement in higher-order cognition have not yet been investigated. Acute stress was applied by 20-min restraint and early reversal learning was evaluated in an operant-based food-seeking task. We employed whole-cell patch-clamp recordings of layer 5 prelimbic (PL)-PFC pyramidal cells to examine mGlu3-LTD and several mechanistically distinct mGlu5-dependent functions. Acute stress impaired both mGlu3-LTD and early reversal learning. Interestingly, potentiating mGlu5 signaling with the mGlu5 positive allosteric modulator (PAM) VU0409551 rescued stress-induced deficits in both mGlu3-LTD and reversal learning. Other aspects of PL-PFC mGlu5 function were not disrupted following stress; however, signaling downstream of mGlu5-Homer interactions, phosphoinositide-3-kinase (PI3K), Akt, and glycogen synthase kinase 3ß was implicated in these phenomena. These findings demonstrate that acute stress disrupts early reversal learning and PL-PFC-dependent synaptic plasticity and that potentiating mGlu5 function can restore these impairments. These findings provide a framework through which modulating coordinated mGlu3/mGlu5 signaling may confer benefits for the treatment of stress-related psychiatric disorders.


Asunto(s)
Corteza Cerebral/metabolismo , Plasticidad Neuronal/fisiología , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Aprendizaje Inverso/fisiología , Estrés Psicológico/metabolismo , Animales , Conducta Apetitiva/efectos de los fármacos , Conducta Apetitiva/fisiología , Fármacos del Sistema Nervioso Central/farmacología , Corteza Cerebral/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Masculino , Ratones Endogámicos C57BL , Plasticidad Neuronal/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Receptor del Glutamato Metabotropico 5/agonistas , Restricción Física , Aprendizaje Inverso/efectos de los fármacos , Estrés Psicológico/psicología , Técnicas de Cultivo de Tejidos
6.
J Pharmacol Exp Ther ; 365(2): 291-300, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29523700

RESUMEN

Mutations in the MeCP2 gene are responsible for the neurodevelopmental disorder Rett syndrome (RTT). MeCP2 is a DNA-binding protein whose abundance and ability to complex with histone deacetylase 3 is linked to the regulation of chromatin structure. Consequently, loss-of-function mutations in MeCP2 are predicted to have broad effects on gene expression. However, to date, studies in mouse models of RTT have identified a limited number of gene or pathway-level disruptions, and even fewer genes have been identified that could be considered amenable to classic drug discovery approaches. Here, we performed RNA sequencing (RNA-seq) on nine motor cortex and six cerebellar autopsy samples from RTT patients and controls. This approach identified 1887 significantly affected genes in the motor cortex and 2110 genes in the cerebellum, with a global trend toward increased expression. Pathway-level analysis identified enrichment in genes associated with mitogen-activated protein kinase signaling, long-term potentiation, and axon guidance. A survey of our RNA-seq results also identified a significant decrease in expression of the CHRM4 gene, which encodes a receptor [muscarinic acetylcholine receptor 4 (M4)] that is the subject of multiple large drug discovery efforts for schizophrenia and Alzheimer's disease. We confirmed that CHRM4 expression was decreased in RTT patients, and, excitingly, we demonstrated that M4 potentiation normalizes social and cognitive phenotypes in Mecp2+/- mice. This work provides an experimental paradigm in which translationally relevant targets can be identified using transcriptomics in RTT autopsy samples, back-modeled in Mecp2+/- mice, and assessed for preclinical efficacy using existing pharmacological tool compounds.


Asunto(s)
Terapia Molecular Dirigida , Receptor Muscarínico M4/metabolismo , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/genética , Análisis de Secuencia de ARN , Animales , Autopsia , Cerebelo/metabolismo , Humanos , Ratones , Corteza Motora/metabolismo , Receptor Muscarínico M4/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patología
7.
Curr Opin Pharmacol ; 38: 31-36, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29486374

RESUMEN

Accumulating evidence suggests that a dysregulation of the glutamatergic system exists in the brains of schizophrenia patients. The metabotropic glutamate (mGlu) receptors are being investigated as novel drug targets for this disease, and have shown promise in both preclinical and clinical studies. Activation of mGlu5 receptors may be efficacious for several symptom domains (positive, negative, and cognitive) and the potential for targeting mGlu5 receptors has been bolstered by recent research on mitigating toxicity profiles associated with mGlu5 activation. Additionally, genetic profiling of schizophrenia patients suggests that genes encoding for mGlu1 and mGlu3 receptors are altered, prompting preclinical studies that have demonstrated potential antipsychotic and cognitive enhancing effects of agents that activate mGlu1 and mGlu3 receptors, respectively. Development of subtype-specific drugs for the mGlu receptors, such as allosteric modulators, could provide a path forward for more efficacious and tolerable therapeutics for schizophrenia.


Asunto(s)
Receptores de Glutamato Metabotrópico/metabolismo , Esquizofrenia/metabolismo , Animales , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Humanos , Esquizofrenia/tratamiento farmacológico
8.
ACS Chem Neurosci ; 9(9): 2210-2217, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-29227625

RESUMEN

Rett syndrome and MECP2 Duplication syndrome are neurodevelopmental disorders attributed to loss-of-function mutations in, or duplication of, the gene encoding methyl-CpG-binding protein 2 (MeCP2), respectively. We recently reported decreased expression and function of the metabotropic glutamate receptor 7 (mGlu7) in a mouse model of Rett syndrome. Positive allosteric modulation of mGlu7 activity was sufficient to improve several disease phenotypes including cognition. Here, we tested the hypothesis that mGlu7 expression would be reciprocally regulated in a mouse model of MECP2 Duplication syndrome, such that negative modulation of mGlu7 activity would exert therapeutic benefit. To the contrary, we report that mGlu7 is not functionally increased in mice overexpressing MeCP2 and that neither genetic nor pharmacological reduction of mGlu7 activity impacts phenotypes that are antiparallel to those observed in Rett syndrome model mice. These data expand our understanding of how mGlu7 expression and function is affected by changes in MeCP2 dosage and have important implications for the therapeutic development of mGlu7 modulators.


Asunto(s)
Ansiedad/metabolismo , Miedo/fisiología , Aprendizaje/fisiología , Discapacidad Intelectual Ligada al Cromosoma X/genética , Receptores de Glutamato Metabotrópico/genética , Regulación Alostérica , Animales , Ansiedad/fisiopatología , Ansiedad/psicología , Benzoxazoles/farmacología , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/farmacología , Miedo/efectos de los fármacos , Miedo/psicología , Aprendizaje/efectos de los fármacos , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Discapacidad Intelectual Ligada al Cromosoma X/psicología , Ratones , Fenotipo , Ácidos Fosfínicos/farmacología , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Receptores de Glutamato Metabotrópico/metabolismo
9.
Cereb Cortex ; 28(12): 4291-4304, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29136107

RESUMEN

Dysregulated fear memory can lead to a broad spectrum of anxiety disorders. The brain systems underlying fear memory are manifold, with the hippocampus being prominently involved by housing fear-related spatial memories as engrams, which are created and stored through neural changes such as synaptic plasticity. Although metabotropic glutamate (mGlu) receptors contribute significantly to both fear behavior and hippocampal synaptic plasticity, the relationship between these two phenomena has not been fully elucidated. Here, we report that contextual fear extinction induces a novel form of metaplasticity mediated by mGlu5 at the hippocampal SC-CA1 synapse. Further, blockade of mGlu5 prevents both contextual fear extinction and expression of this metaplasticity. This form of metaplasticity was absent in a mouse model of MECP2-duplication syndrome, corresponding to a complete deficit in extinction learning. These findings suggest that mGlu5-dependent metaplasticity within the hippocampus may play a critical role in extinction of contextual fear.


Asunto(s)
Extinción Psicológica/fisiología , Miedo/fisiología , Hipocampo/fisiología , Potenciación a Largo Plazo , Depresión Sináptica a Largo Plazo , Receptores de Ácido Kaínico/fisiología , Animales , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor Cannabinoide CB1/fisiología , Receptores de N-Metil-D-Aspartato/fisiología
10.
ACS Med Chem Lett ; 8(12): 1326-1330, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29259756

RESUMEN

Herein, we report the structure-activity relationships within a series of mGlu7 NAMs based on an N-(2-(1H-1,2,4-triazol-1-yl)-5-(trifluoromethoxy)phenyl)benzamide core with excellent CNS penetration (Kp 1.9-5.8 and Kp,uu 0.4-1.4). Analogues in this series displayed steep SAR. Of these, VU6010608 (11a) emerged with robust efficacy in blocking high frequency stimulated long-term potentiation in electrophysiology studies.

11.
Neuropsychopharmacology ; 42(13): 2553-2566, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28664928

RESUMEN

Activation of ß-adrenergic receptors (ßARs) enhances both the induction of long-term potentiation (LTP) in hippocampal CA1 pyramidal cells and hippocampal-dependent cognitive function. Interestingly, previous studies reveal that coincident activation of group II metabotropic glutamate (mGlu) receptors with ßARs in the hippocampal astrocytes induces a large increase in cyclic-AMP (cAMP) accumulation and release of adenosine. Adenosine then acts on A1 adenosine receptors at neighboring excitatory Schaffer collateral terminals, which could counteract effects of activation of neuronal ßARs on excitatory transmission. On the basis of this, we postulated that activation of the specific mGlu receptor subtype that mediates this response could inhibit ßAR-mediated effects on hippocampal synaptic plasticity and cognitive function. Using novel mGlu receptor subtype-selective allosteric modulators along with knockout mice we now report that the effects of mGlu2/3 agonists on ßAR-mediated increases in cAMP accumulation are exclusively mediated by mGlu3. Furthermore, mGlu3 activation inhibits the ability of the ßAR agonist isoproterenol to enhance hippocampal LTP, and this effect is absent in slices treated with either a glial toxin or an adenosine A1 receptor antagonist. Finally, systemic administration of the mGlu2/3 agonist LY379268 disrupted contextual fear memory in a manner similar to the effect of the ßAR antagonist propranolol, and this effect was reversed by the mGlu3-negative allosteric modulator VU0650786. Taken together, these data suggest that mGlu3 can influence astrocytic signaling and modulate ßAR-mediated effects on hippocampal synaptic plasticity and cognitive function.


Asunto(s)
AMP Cíclico/metabolismo , Potenciación a Largo Plazo/fisiología , Consolidación de la Memoria/fisiología , Receptores Adrenérgicos beta/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Consolidación de la Memoria/efectos de los fármacos , Ratones Endogámicos ICR , Ratones Noqueados , Neurotransmisores/farmacología , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/genética , Técnicas de Cultivo de Tejidos
12.
Psychopharmacology (Berl) ; 232(17): 3203-13, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26037945

RESUMEN

RATIONALE: L-dopa, the main therapeutic for Parkinson's disease (PD), has been shown to increase brain dopamine concentrations that are necessary for proper motor control; however, PD patients experience non-motor symptoms that are not improved or could be exacerbated by L-dopa. OBJECTIVES: The purpose of this study is to determine the effects of L-dopa treatment on cognitive and affective behavioral responses of rats, as well as their corresponding monoamine brain concentrations. METHODS: Rats were treated with L-dopa (6 mg/kg; twice daily) for 10 consecutive days. Sodium ascorbate (400 mg/kg) was co-administered with L-dopa to investigate the effects of antioxidant co-treatment on behavior and monoamine concentrations. Rats underwent cognitive and affective behavioral testing. Monoamine concentrations of several brain regions were analyzed. RESULTS: L-dopa treatment resulted in significant impairment in the performance in the Barnes maze and improvement in conditioned fear stress paradigms. Specifically, L-dopa caused an increase in latency to find the goal box during Barnes maze testing and increased freezing behavior in context-induced conditioned fear testing. Furthermore, the rats in the conditioned fear stress experiments showed corresponding depletions in serotonin (5-HT) and its metabolite, 5-HIAA, in the dorsal raphe nucleus (DRN) and the mPFC. The behavioral impairments as well as monoamine depletions were blocked by ascorbate co-treatment. CONCLUSIONS: Chronic L-dopa may contribute to non-motor symptoms related to spatial memory and fear. These effects may be attributable to a dysregulation of brain 5-HT caused by L-dopa treatment. The results presented here provide further rationale for investigating adjunctive therapeutics to L-dopa for PD, such as antioxidants.


Asunto(s)
Conducta Animal/efectos de los fármacos , Dopaminérgicos/farmacología , Levodopa/farmacología , Antagonistas de la Serotonina/farmacología , Serotonina/metabolismo , Afecto/efectos de los fármacos , Animales , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Monoaminas Biogénicas/metabolismo , Química Encefálica/efectos de los fármacos , Cognición/efectos de los fármacos , Miedo/psicología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Desempeño Psicomotor/efectos de los fármacos , Núcleos del Rafe/efectos de los fármacos , Núcleos del Rafe/metabolismo , Ratas , Ratas Sprague-Dawley
13.
Toxics ; 3(1): 75-88, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-29056652

RESUMEN

L-dopa is used to treat the motor symptoms associated with Parkinson's disease, a neurodegenerative movement disorder characterized by a loss of dopamine neurons. L-dopa is the precursor to dopamine and crosses the blood-brain barrier to increase dopamine neurotransmission. This review will focus on the findings that dopamine produced from L-dopa is mediated in part by serotonin neurons. Direct evidence will be provided that increases in dopamine cause oxidative stress and damage serotonin neurons. Similarly, chronic L-dopa produces deficits in serotonin neurotransmission, including decreases in both serotonin cell bodies within the dorsal raphe and serotonin neurotransmitter concentrations in several forebrain regions. Since serotonin is involved in many important physiological processes including mood and cognition, L-dopa induced serotonin deficits may play a role in the side-effect symptoms observed in Parkinson's disease patients treated with L-dopa.

14.
J Pharmacol Exp Ther ; 351(2): 440-7, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25212217

RESUMEN

L-Dopa (l-3,4-dihydroxyphenylalanine) is the precursor to dopamine and has become the mainstay therapeutic treatment for Parkinson's disease. Chronic L-dopa is administered to recover motor function in Parkinson's disease patients. However, drug efficacy decreases over time, and debilitating side effects occur, such as dyskinesia and mood disturbances. The therapeutic effect and some of the side effects of L-dopa have been credited to its effect on serotonin (5-HT) neurons. Given these findings, it was hypothesized that chronic L-dopa treatment decreases 5-HT neurons in the dorsal raphe nucleus (DRN) and the content of 5-HT in forebrain regions in a manner that is mediated by oxidative stress. Rats were treated chronically with l-dopa (6 mg/kg; twice daily) for 10 days. Results indicated that the number of 5-HT neurons was significantly decreased in the DRN after l-dopa treatment compared with vehicle. This effect was more pronounced in the caudal-extent of the dorsal DRN, a subregion found to have a significantly higher increase in the 3,4-dihydroxyphenylacetic acid/dopamine ratio in response to acute L-dopa treatment. Furthermore, pretreatment with ascorbic acid (400 mg/kg) or deprenyl (2 mg/kg) prevented the l-dopa-induced decreases in 5-HT neurons. In addition, 5-HT content was decreased significantly in the DRN and prefrontal cortex by l-dopa treatment, effects that were prevented by ascorbic acid pretreatment. Taken together, these data illustrate that chronic L-dopa causes a 5-HT neuron loss and the depletion of 5-HT content in a subregion of the DRN as well as in the frontal cortex through an oxidative-stress mechanism.


Asunto(s)
Levodopa/farmacología , Neuronas/efectos de los fármacos , Núcleos del Rafe/efectos de los fármacos , Serotonina/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Dopamina/metabolismo , Masculino , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Núcleos del Rafe/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Neuropharmacology ; 67: 243-51, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23196068

RESUMEN

L-dopa is a precursor for dopamine synthesis and a mainstay treatment for Parkinson's disease. However, l-dopa therapy is not without side effects that may be attributed to non-dopaminergic mechanisms. Synthesized dopamine can be neurotoxic through its enzymatic degradation by monoamine oxidase (MAO) to form the reactive byproduct, hydrogen peroxide and hydroxyl radicals or through auto-oxidation to form highly reactive quinones that can bind proteins and render them non-functional. Since l-dopa could be decarboxylated by aromatic amino acid decarboxylase (AADC) present within both dopamine and serotonin neurons, it was hypothesized that serotonin neurons convert l-dopa into dopamine to generate excessive reactive oxygen species and quinoproteins that ultimately lead to serotonin neuron death. To examine the effects of l-dopa on serotonin neurons, the RN46A-B14 cell line was used. These immortalized serotonergic cell cultures were terminally differentiated and then incubated with varying concentrations of l-dopa. Results show that RN46A-B14 cells contain AADC and can synthesize dopamine after incubation with l-dopa. Furthermore, l-dopa dose-dependently increased intracellular reactive oxygen species (ROS) and cell death. Dopamine, ROS production and cell death were attenuated by co-incubation with the AADC inhibitor, NSD-1015. The MAO inhibitor, pargyline, also attenuated cell death and ROS after l-dopa treatment. Lastly, quinoprotein formation was enhanced significantly by incubation with l-dopa. Taken together, these data illustrate that serotonergic cells can produce dopamine and that the accumulation of dopamine after l-dopa and its subsequent degradation can lead to ROS production and death of RN46A-B14 serotonergic cells.


Asunto(s)
Dopamina/biosíntesis , Levodopa/farmacología , Estrés Oxidativo/fisiología , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Línea Celular Transformada , Humanos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...