Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Prog Neurobiol ; 231: 102536, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37805096

RESUMEN

Excessive daytime sleepiness (EDS) and sleep fragmentation are often observed in Parkinson's disease (PD) patients and are poorly understood despite their considerable impact on quality of life. We examined the ability of a neurotoxin-based mouse model of PD to reproduce these disorders and tested the potential counteracting effects of dopamine replacement therapy. Experiments were conducted in female mice with a unilateral 6-hydroxydopamine lesion of the medial forebrain bundle, leading to the loss of dopamine neurons projecting to the dorsal and ventral striatum. Sham-operated mice were used as control. Electroencephalographic and electromyographic recording was used to identify and quantify awaken, rapid eye movement (REM) and non-REM (NREM) sleep states. PD mice displayed enhanced NREM sleep and reduced wakefulness during the active period of the 24-hour circadian cycle, indicative of EDS. In addition, they also showed fragmentation of NREM sleep and increased slow-wave activity, a marker of sleep pressure. Electroencephalographic analysis of the PD model also revealed decreased density and increased length of burst-like thalamocortical oscillations (spindles). Treatment of PD mice with the dopamine receptor agonist, pramipexole, but not with L-DOPA, counteracted EDS by reducing the number, but not the length, of NREM sleep episodes during the first half of the active period. The present model recapitulates some prominent PD-related anomalies affecting sleep macro- and micro-structure. Based on the pharmacological profile of pramipexole these results also indicate the involvement of impaired dopamine D2/D3 receptor transmission in EDS.


Asunto(s)
Enfermedad de Parkinson , Trastornos del Sueño-Vigilia , Humanos , Femenino , Ratones , Animales , Enfermedad de Parkinson/tratamiento farmacológico , Dopamina , Pramipexol/farmacología , Pramipexol/uso terapéutico , Calidad de Vida , Sueño , Trastornos del Sueño-Vigilia/tratamiento farmacológico , Trastornos del Sueño-Vigilia/etiología , Modelos Animales de Enfermedad
2.
Neurobiol Dis ; 144: 105044, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32798726

RESUMEN

Acetylcholine muscarinic receptors (mAChRs) contribute to both the facilitation and inhibition of levodopa-induced dyskinesia operated by striatal cholinergic interneurons, although the receptor subtypes involved remain elusive. Cholinergic afferents from the midbrain also innervate the substantia nigra reticulata, although the role of nigral mAChRs in levodopa-induced dyskinesia is unknown. Here, we investigate whether striatal and nigral M1 and/or M4 mAChRs modulate dyskinesia and the underlying striato-nigral GABAergic pathway activation in 6-hydroxydopamine hemilesioned rats. Reverse microdialysis allowed to deliver the mAChR antagonists telenzepine (M1 subtype preferring), PD-102807 and tropicamide (M4 subtype preferring), as well as the selective M4 mAChR positive allosteric modulator VU0152100 in striatum or substantia nigra, while levodopa was administered systemically. Dyskinetic movements were monitored along with nigral GABA (and glutamate) and striatal glutamate dialysate levels, taken as neurochemical correlates of striato-nigral pathway and cortico-basal ganglia-thalamo-cortical loop activation. We observed that intrastriatal telenzepine, PD-102807 and tropicamide alleviated dyskinesia and inhibited nigral GABA and striatal glutamate release. This was partially replicated by intrastriatal VU0152100. The M2 subtype preferring antagonist AFDX-116, used to elevate striatal acetylcholine levels, blocked the behavioral and neurochemical effects of PD-102807. Intranigral VU0152100 prevented levodopa-induced dyskinesia and its neurochemical correlates whereas PD-102807 was ineffective. These results suggest that striatal, likely postsynaptic, M1 mAChRs facilitate dyskinesia and striato-nigral pathway activation in vivo. Conversely, striatal M4 mAChRs can both facilitate and inhibit dyskinesia, possibly depending on their localization. Potentiation of striatal and nigral M4 mAChR transmission leads to powerful multilevel inhibition of striato-nigral pathway and attenuation of dyskinesia.


Asunto(s)
Dopaminérgicos/efectos adversos , Discinesia Inducida por Medicamentos/metabolismo , Levodopa/efectos adversos , Neostriado/metabolismo , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M4/metabolismo , Sustancia Negra/metabolismo , Regulación Alostérica , Animales , Discinesia Inducida por Medicamentos/etiología , Discinesia Inducida por Medicamentos/fisiopatología , Ácido Glutámico/efectos de los fármacos , Ácido Glutámico/metabolismo , Microdiálisis , Antagonistas Muscarínicos/farmacología , Neostriado/efectos de los fármacos , Vías Nerviosas , Oxidopamina/toxicidad , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/etiología , Trastornos Parkinsonianos/metabolismo , Ratas , Receptor Muscarínico M1/antagonistas & inhibidores , Receptor Muscarínico M4/antagonistas & inhibidores , Sustancia Negra/efectos de los fármacos , Simpaticolíticos/toxicidad , Ácido gamma-Aminobutírico/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
3.
Br J Pharmacol ; 177(17): 3957-3974, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32464686

RESUMEN

BACKGROUND AND PURPOSE: l-DOPA-induced dyskinesia (LID) is considered a major complication in the treatment of Parkinson's disease (PD). Buspirone (5-HT1A partial agonist) have shown promising results in the treatment of PD and LID, however no 5-HT-based treatment has been approved in PD. The present study was aimed to investigate how the substantia nigra pars reticulata (SNr) is affected by buspirone and whether it is a good target to study 5-HT antidyskinetic treatments. EXPERIMENTAL APPROACH: Buspirone was studied using in vivo single-unit, electrocorticogram, local field potential recordings along with microdialysis and immunohistochemistry in naïve/sham, 6-hydroxydopamine (6-OHDA)-lesioned or 6-OHDA-lesioned and l-DOPA-treated (6-OHDA/l-DOPA) rats. KEY RESULTS: Local buspirone inhibited SNr neuron activity in all groups. However, systemic buspirone reduced burst activity in 6-OHDA-lesioned rats (with or without l-DOPA treatment), whereas 8-OH-DPAT, a full 5-HT1A agonist induced larger inhibitory effects in sham animals. Neither buspirone nor 8-OH-DPAT markedly modified the low-frequency oscillatory activity in the SNr or synchronization within the SNr with the cortex. In addition, local perfusion of buspirone increased GABA and glutamate release in the SNr of naïve and 6-OHDA-lesioned rats but no effect in 6-OHDA/l-DOPA rats. In the 6-OHDA/l-DOPA group, increased 5-HT transporter and decreased 5-HT1A receptor expression was found. CONCLUSIONS AND IMPLICATIONS: The effects of buspirone in SNr are influenced by dopamine loss and l-DOPA treatment. The present results suggest that the regulation of burst activity of the SNr induced by DA loss may be a good target to test new drugs for the treatment of PD and LID.


Asunto(s)
Levodopa , Porción Reticular de la Sustancia Negra , Animales , Antiparkinsonianos/farmacología , Buspirona/farmacología , Dopamina , Oxidopamina , Ratas , Sustancia Negra
4.
J Med Chem ; 63(5): 2688-2704, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-31951130

RESUMEN

A novel series of C(3)-substituted piperdinylindoles were developed as nociceptin opioid receptor (NOP) partial agonists to explore a pharmacological hypothesis that NOP partial agonists would afford a dual pharmacological action of attenuating Parkinson's disease (PD) motor symptoms and development of levodopa-induced dyskinesias. SAR around the C-3 substituents investigated effects on NOP binding, intrinsic activity, and selectivity and showed that while the C(3)-substituted indoles are selective, high affinity NOP ligands, the steric, polar, and cationic nature of the C-3 substituents affected intrinsic activity to afford partial agonists with a range of efficacies. Compounds 4, 5, and 9 with agonist efficacies between 25% and 35% significantly attenuated motor deficits in the 6-OHDA-hemilesioned rat model of PD. Further, unlike NOP antagonists, which appear to worsen dyskinesia expression, these NOP partial agonists did not attenuate or worsen dyskinesia expression. The NOP partial agonists and their SAR reported here may be useful to develop nondopaminergic treatments for PD.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Indoles/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Receptores Opioides/agonistas , Animales , Antiparkinsonianos/química , Antiparkinsonianos/farmacocinética , Células CACO-2 , Modelos Animales de Enfermedad , Humanos , Indoles/química , Indoles/farmacocinética , Masculino , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/fisiopatología , Piperidinas/química , Piperidinas/farmacocinética , Piperidinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Relación Estructura-Actividad , Receptor de Nociceptina
5.
Handb Exp Pharmacol ; 254: 213-232, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30689087

RESUMEN

Nociceptin/Orphanin FQ (N/OFQ) and its NOP receptor are highly expressed in motor areas of the rodent, nonhuman, and human primate brain, such as primary motor cortex, thalamus, globus pallidus, striatum, and substantia nigra. Endogenous N/OFQ negatively regulates motor behavior and dopamine transmission through NOP receptors expressed by dopaminergic neurons of the substantia nigra compacta. Consistent with the existence of an N/OFQ tone over dopaminergic transmission, blockade of NOP receptor antagonists increases striatal dopamine release. In this chapter, we will review the evidence linking the N/OFQ-NOP receptor system to Parkinson's disease (PD). We will first discuss data showing that the central N/OFQ-NOP receptor system undergoes plastic changes in different basal ganglia nuclei following dopamine depletion. Then we will show that NOP receptor antagonists relieve motor deficits in different rodent and nonhuman primate models of PD. Mechanistically, NOP receptor blockade in substantia nigra reticulata results in rebalancing of the inhibitory GABAergic and excitatory glutamatergic inputs impinging on nigro-thalamic GABAergic neurons, leading to thalamic disinhibition. We will also present data showing that, in addition to motor symptoms, N/OFQ also plays a role in the parkinsonian neurodegeneration. In fact, NOP receptor antagonists possess neuroprotective/neurorescue properties in in vitro and in vivo models of PD.


Asunto(s)
Péptidos Opioides/farmacología , Enfermedad de Parkinson , Sustancia Negra/metabolismo , Animales , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Péptidos Opioides/química , Péptidos Opioides/metabolismo , Enfermedad de Parkinson/fisiopatología , Ratas , Ratas Sprague-Dawley , Sustancia Negra/química , Nociceptina
6.
Br J Pharmacol ; 175(5): 782-796, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29232769

RESUMEN

BACKGROUND AND PURPOSE: We previously showed that nociceptin/orphanin FQ opioid peptide (NOP) receptor agonists attenuate the expression of levodopa-induced dyskinesia in animal models of Parkinson's disease. We now investigate the efficacy of two novel, potent and chemically distinct NOP receptor agonists, AT-390 and AT-403, to improve Parkinsonian disabilities and attenuate dyskinesia development and expression. EXPERIMENTAL APPROACH: Binding affinity and functional efficacy of AT-390 and AT-403 at the opioid receptors were determined in radioligand displacement assays and in GTPγS binding assays respectively, conducted in CHO cells. Their anti-Parkinsonian activity was evaluated in 6-hydroxydopamine hemi-lesioned rats whereas the anti-dyskinetic properties were assessed in 6-hydroxydopamine hemi-lesioned rats chronically treated with levodopa. The ability of AT-403 to inhibit the D1 receptor-induced phosphorylation of striatal ERK was investigated. KEY RESULTS: AT-390 and AT-403 selectively improved akinesia at low doses and disrupted global motor activity at higher doses. AT-403 palliated dyskinesia expression without causing sedation in a narrow therapeutic window, whereas AT-390 delayed the appearance of abnormal involuntary movements and increased their duration at doses causing sedation. AT-403 did not prevent the priming to levodopa, although it significantly inhibited dyskinesia on the first day of administration. AT-403 reduced the ERK phosphorylation induced by SKF38393 in vitro and by levodopa in vivo. CONCLUSIONS AND IMPLICATIONS: NOP receptor stimulation can provide significant albeit mild anti-dyskinetic effect at doses not causing sedation. The therapeutic window, however, varies across compounds. AT-403 could be a potent and selective tool to investigate the role of NOP receptors in vivo.


Asunto(s)
Acetamidas/farmacología , Antiparkinsonianos/farmacología , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Piperidinas/farmacología , Receptores Opioides/agonistas , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/antagonistas & inhibidores , Acetamidas/uso terapéutico , Animales , Antiparkinsonianos/uso terapéutico , Cuerpo Estriado/metabolismo , Cricetinae , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Levodopa/antagonistas & inhibidores , Masculino , Oxidopamina , Fosforilación/efectos de los fármacos , Piperidinas/uso terapéutico , Ensayo de Unión Radioligante , Ratas , Receptor de Nociceptina
7.
J Pharmacol Exp Ther ; 364(2): 198-206, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29167350

RESUMEN

Safinamide has been recently approved as an add-on to levodopa therapy for Parkinson disease. In addition to inhibiting monoamine oxidase type B, it blocks sodium channels and modulates glutamate (Glu) release in vitro. Since this property might contribute to the therapeutic action of the drug, we undertook the present study to investigate whether safinamide inhibits Glu release also in vivo and whether this effect is consistent across different brain areas and is selective for glutamatergic neurons. To this aim, in vivo microdialysis was used to monitor the spontaneous and veratridine-induced Glu and GABA release in the hippocampus and basal ganglia of naive, awake rats. Brain levels of safinamide were measured as well. To shed light on the mechanisms underlying the effect of safinamide, sodium currents were measured by patch-clamp recording in rat cortical neurons. Safinamide maximally inhibited the veratridine-induced Glu and GABA release in hippocampus at 15 mg/kg, which reached free brain concentrations of 1.89-1.37 µM. This dose attenuated veratridine-stimulated Glu (but not GABA) release in subthalamic nucleus, globus pallidus, and substantia nigra reticulata, but not in striatum. Safinamide was ineffective on spontaneous neurotransmitter release. In vitro, safinamide inhibited sodium channels, showing a greater affinity at depolarized (IC50 = 8 µM) than at resting (IC50 = 262 µM) potentials. We conclude that safinamide inhibits in vivo Glu release from stimulated nerve terminals, likely via blockade of sodium channels at subpopulations of neurons with specific firing patterns. These data are consistent with the anticonvulsant and antiparkinsonian actions of safinamide and provide support for the nondopaminergic mechanism of its action.


Asunto(s)
Alanina/análogos & derivados , Ganglios Basales/efectos de los fármacos , Ganglios Basales/metabolismo , Bencilaminas/farmacología , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Alanina/farmacología , Animales , Ganglios Basales/citología , Hipocampo/citología , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA