Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; 186: 106278, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37683958

RESUMEN

L-DOPA-induced dyskinesia (LID) remains a major complication of Parkinson's disease management for which better therapies are necessary. The contribution of the striatonigral direct pathway to LID is widely acknowledged but whether the striatopallidal pathway is involved remains debated. Selective optogenetic stimulation of striatonigral axon terminals induces dyskinesia in mice rendered hemiparkinsonian with the toxin 6-hydroxydopamine (6-OHDA). Here we show that optogenetically-induced dyskinesia is increased by the D2-type dopamine receptor agonist quinpirole. Although the quinpirole effect may be mediated by D2 receptor stimulation in striatopallidal neurons, alternative mechanisms may be responsible as well. To selectively modulate the striatopallidal pathway, we selectively expressed channelrhodopsin-2 (ChR2) in D2 receptor expressing neurons by crossing D2-Cre and ChR2-flox mice. The animals were rendered hemiparkinsonian and implanted with an optic fiber at the ipsilateral external globus pallidus (GPe). Stimulation of ChR2 at striatopallidal axon terminals reduced LID and also general motility during the off L-DOPA state, without modifying the pro-motor effect of low doses of L-DOPA producing mild or no dyskinesia. Overall, the present study shows that D2-type dopamine receptors and the striatopallidal pathway modulate dyskinesia and suggest that targeting striatopallidal axon terminals at the GPe may have therapeutic potential in the management of LID.


Asunto(s)
Discinesias , Levodopa , Animales , Ratones , Levodopa/toxicidad , Quinpirol , Agonistas de Dopamina/farmacología , Oxidopamina/toxicidad , Receptores de Dopamina D2
2.
Neurobiol Dis ; 176: 105930, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36414182

RESUMEN

Levodopa (L-DOPA) administration remains the gold standard therapy for Parkinson's disease (PD). Despite several pharmacological advances in the use of L-DOPA, a high proportion of chronically treated patients continues to suffer disabling involuntary movements, namely, L-DOPA-induced dyskinesias (LIDs). As part of the effort to stop these unwanted side effects, the present study used a rodent model to identify and manipulate the striatal outflow circuitry responsible for LIDs. To do so, optogenetic technology was used to activate separately the striatal direct (D1R- expressing) and indirect (D2R- expressing) pathways in a mouse model of PD. Firstly, D1-cre or A2a-cre animals received unilateral injections of neurotoxin 6-hydroxydopamine (6-OHDA) to simulate the loss of dopamine observed in PD patients. The effects of independently stimulating each pathway were tested to see if experimental dyskinesias could be induced. Secondly, dopamine depleted A2a-cre animals received systemic L-DOPA to evoke dyskinetic movements. The ability of indirect pathway optogenetic stimulation to suppress pre-established LIDs was then tested. Selective manipulation of direct pathway evoked optodyskinesias both in dopamine depleted and intact animals, but optical inhibition of these neurons failed to suppress LIDs. On the other hand, selective activation of indirect striatal projection neurons produced an immediate and reliable suppression of LIDs. Thus, a functional dissociation has been found here whereby activation of D1R- and D2R-expressing projection neurons evokes and inhibits LIDs respectively, supporting the notion of tight interaction between the two striatal efferent systems in both normal and pathological conditions. This points to the importance of maintaining an equilibrium in the activity of both striatal pathways to produce normal movement. Finally, the ability of selective indirect pathway optogenetic activation to block the expression of LIDs in an animal model of PD sheds light on intrinsic mechanisms responsible for striatal-based dyskinesias and identifies a potential therapeutic target for suppressing LIDs in PD patients.


Asunto(s)
Discinesias , Enfermedad de Parkinson , Ratones , Animales , Levodopa/farmacología , Dopamina/metabolismo , Enfermedad de Parkinson/metabolismo , Cuerpo Estriado/metabolismo , Oxidopamina/toxicidad , Antiparkinsonianos/farmacología , Modelos Animales de Enfermedad
3.
Cereb Cortex ; 31(4): 2187-2204, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33264389

RESUMEN

Dopamine receptors play an important role in motivational, emotional, and motor responses. In addition, growing evidence suggests a key role of hippocampal dopamine receptors in learning and memory. It is well known that associative learning and synaptic plasticity of CA3-CA1 requires the dopamine D1 receptor (D1R). However, the specific role of the dopamine D2 receptor (D2R) on memory-related neuroplasticity processes is still undefined. Here, by using two models of D2R loss, D2R knockout mice (Drd2-/-) and mice with intrahippocampal injections of Drd2-small interfering RNA (Drd2-siRNA), we aimed to investigate how D2R is involved in learning and memory as well as in long-term potentiation of the hippocampus. Our studies revealed that the genetic inactivation of D2R impaired the spatial memory, associative learning, and the classical conditioning of eyelid responses. Similarly, deletion of D2R reduced the activity-dependent synaptic plasticity in the hippocampal CA1-CA3 synapse. Our results demonstrate the first direct evidence that D2R is essential in behaving mice for trace eye blink conditioning and associated changes in hippocampal synaptic strength. Taken together, these results indicate a key role of D2R in regulating hippocampal plasticity changes and, in consequence, acquisition and consolidation of spatial and associative forms of memory.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Región CA3 Hipocampal/metabolismo , Plasticidad Neuronal/fisiología , Receptores de Dopamina D2/deficiencia , Memoria Espacial/fisiología , Sinapsis/metabolismo , Animales , Reacción de Prevención/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/administración & dosificación , Receptores de Dopamina D2/genética , Sinapsis/genética
4.
Mov Disord ; 36(5): 1070-1085, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33219714

RESUMEN

Parkinson's disease (PD) is a progressive neurodegenerative disease where dopaminergic neurons in the substantia nigra are lost, resulting in a decrease in striatal dopamine and, consequently, motor control. Dopaminergic degeneration is associated with the appearance of Lewy bodies, which contain membrane structures and proteins, including α-synuclein (α-Syn), in surviving neurons. PD displays a multifactorial pathology and develops from interactions between multiple elements, such as age, environmental conditions, and genetics. Mutations in the GBA1 gene represent one of the major genetic risk factors for PD. This gene encodes an essential lysosomal enzyme called ß-glucocerebrosidase (GCase), which is responsible for degrading the glycolipid glucocerebroside into glucose and ceramide. GCase can generate glucosylated cholesterol via transglucosylation and can also degrade the sterol glucoside. Although the molecular mechanisms that predispose an individual to neurodegeneration remain unknown, the role of cholesterol in PD pathology deserves consideration. Disturbed cellular cholesterol metabolism, as reflected by accumulation of lysosomal cholesterol in GBA1-associated PD cellular models, could contribute to changes in lipid rafts, which are necessary for synaptic localization and vesicle cycling and modulation of synaptic integrity. α-Syn has been implicated in the regulation of neuronal cholesterol, and cholesterol facilitates interactions between α-Syn oligomers. In this review, we integrate the results of previous studies and describe the cholesterol landscape in cellular homeostasis and neuronal function. We discuss its implication in α-Syn and Lewy body pathophysiological mechanisms underlying PD, focusing on the role of GCase and cholesterol. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Colesterol , Glucosilceramidasa/genética , Humanos , Cuerpos de Lewy , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética
5.
Addict Biol ; 26(1): e12840, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-31833146

RESUMEN

Repeated cocaine exposure causes long-lasting neuroadaptations that involve alterations in cellular signaling and gene expression mediated by dopamine in different brain regions, such as the striatum. Previous studies have pointed out to the dopamine D1 receptor as one major player in psychostimulants-induced behavioral, cellular, and molecular changes. However, the role of other dopamine receptors has not been fully characterized. Here we used dopamine D2 receptor knockout (D2-/- ) mice to explore the role of D2 receptor (D2R) in behavioral sensitization and its associated gene expression after acute and chronic cocaine and amphetamine administration. We also studied the impact of D2R elimination in D1R-mediated responses. We found that cocaine- and amphetamine-induced behavioral sensitization is deficient in D2-/- mice. The expression of dynorphin, primarily regulated by D1R and a marker of direct-pathway striatal neurons, is attenuated in naïve- and in cocaine- or amphetamine-treated D2-/- mice. Moreover, c-Fos expression observed in D2-/- mice was reduced in acutely but not in chronically treated animals. Interestingly, inactivation of D2R increased c-Fos expression in neurons of the striatopallidal pathway. Finally, elimination of D2R blunted the locomotor and striatal c-Fos response to the full D1 agonist SKF81297. In conclusion, D2R is critical for the development of behavioral sensitization and the associated gene expression, after cocaine administration, and it is required for the locomotor responses promoted by D1R activation.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Cocaína/farmacología , Receptores de Dopamina D2/metabolismo , Anfetaminas/farmacología , Animales , Benzazepinas , Cuerpo Estriado/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Ratones , Ratones Noqueados , Neuronas/metabolismo , Receptores de Dopamina D1/metabolismo
6.
Neurobiol Dis ; 134: 104666, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31682992

RESUMEN

Dopaminergic afferents innervate spiny projection neurons (SPNs) in the striatum, maintaining basal ganglia activity. The loss of striatal innervation is the hallmark of Parkinson's disease (PD), which is characterized by dopaminergic denervation. A lack of dopamine in the dorsal striatum induces plasticity changes in SPNs. However, PD-associated denervation is progressive, and how plasticity is modified in partially innervated areas is poorly understood. The most studied models of PD are based on the use of neurotoxins that induce an almost complete striatal denervation. To investigate the impact of partial dopamine (DA) innervation in striatal plasticity, we use a genetic model of PD, Aphakia (Ak) mice, whose striatum presents an increasing dorso-ventral gradient of dopamine innervation. We studied SPNs in three different areas (dorsal, middle and ventral, with low, moderate and high innervation by tyrosine hydroxylase TH-positive axons, respectively) using fast scan cyclic voltammetry, microiontophoresis, immunohistochemistry and patch clamp techniques. Our data show an increasing dorso-ventral gradient of extracellular DA levels, overlapping with the gradient of TH innervation. Interestingly, spine loss in both direct (d-SPN) and indirect SPNs (i-SPN) decreases from dorsal to ventral in the parkinsonian striatum of Ak mice, following the decrease in DA levels. However, their dendritic trees and the number of nodes are only reduced in the poorly innervated dorsal areas and remain unaltered in moderate and highly innervated areas. The firing rate of direct SPNs does not change in either moderate or highly innervated areas, but increases in poorly innervated areas. In contrast, action potential frequency of indirect SPNs does not change along the dorso-ventral innervation gradient. Our findings indicate that spine density in d-SPNs and i-SPNs varies in a dopamine concentration-dependent manner, indicating that both d- and i-SPN are similarly innervated by DA.


Asunto(s)
Cuerpo Estriado , Espinas Dendríticas , Dopamina/metabolismo , Plasticidad Neuronal/fisiología , Neuronas Aferentes , Animales , Cuerpo Estriado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
7.
Neurobiol Dis ; 141: 104892, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32387338

RESUMEN

The antioxidant and CB2 receptor agonist properties of Δ9-tetrahydrocannabivarin (Δ9-THCV) afforded neuroprotection in experimental Parkinson's disease (PD), whereas its CB1 receptor antagonist profile at doses lower than 5 mg/kg caused anti-hypokinetic effects. In the present study, we investigated the anti-dyskinetic potential of Δ9-THCV (administered i.p. at 2 mg/kg for two weeks), which had not been investigated before. This objective was investigated after inducing dyskinesia by repeated administration of L-DOPA (i.p. at 10 mg/kg) in a genetic model of dopaminergic deficiency, Pitx3ak mutant mice, which serves as a useful model for testing anti-dyskinetic agents. The daily treatment of these mice with L-DOPA for two weeks progressively increased the time spent in abnormal involuntary movements (AIMs) and elevated their horizontal and vertical activities (as measured in a computer-aided actimeter), signs that reflected the dyskinetic state of these mice. Interestingly, when combined with L-DOPA from the first injection, Δ9-THCV delayed the appearance of all these signs and decreased their intensity, with a reduction in the levels of FosB protein and the histone pAcH3 (measured by immunohistochemistry), which had previously been found to be elevated in the basal ganglia in L-DOPA-induced dyskinesia. In addition to the anti-dyskinetic effects of Δ9-THCV when administered at the onset of L-DOPA treatment, Δ9-THCV was also effective in attenuating the intensity of dyskinesia when administered for three consecutive days once these signs were already present (two weeks after the onset of L-DOPA treatment). In summary, our data support the anti-dyskinetic potential of Δ9-THCV, both to delay the occurrence and to attenuate the magnitude of dyskinetic signs. Although further studies are clearly required to determine the clinical significance of these data in humans, the results nevertheless situate Δ9-THCV in a promising position for developing a cannabinoid-based therapy for patients with PD.


Asunto(s)
Antidiscinéticos/administración & dosificación , Dronabinol/análogos & derivados , Discinesia Inducida por Medicamentos/prevención & control , Levodopa/administración & dosificación , Enfermedad de Parkinson/complicaciones , Animales , Modelos Animales de Enfermedad , Dronabinol/administración & dosificación , Proteínas de Homeodominio/genética , Masculino , Factores de Transcripción/genética
8.
Mov Disord ; 35(10): 1810-1821, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32643147

RESUMEN

BACKGROUND: Dopamine transmission is involved in the maintenance of the structural plasticity of direct-pathway and indirect-pathway striatal projection neurons (d-SPNs and i-SPNs, respectively). The lack of dopamine in Parkinson's disease produces synaptic remodeling in both types of SPNs, reducing the length of the dendritic arbor and spine density and increasing the intrinsic excitability. Meanwhile, the elevation of dopamine levels by levodopa recovers these alterations selectively in i-SPNs. However, little is known about the specific role of the D1 receptor (D1R) in these alterations. METHODS: To explore the specific role of D1R in the synaptic remodeling of SPNs, we used knockout D1R mice (D1R-/- ) and wild-type mice crossed with drd2-enhanced green fluorescent protein (eGFP) to identify d-SPNs and i-SPNs. Corticostriatal slices were used for reconstruction of the dendritic arbors after Lucifer yellow intracellular injection and for whole-cell recordings in naïve and parkinsonian mice treated with saline or levodopa. RESULTS: The genetic inactivation of D1R reduces the length of the dendritic tree and the spine density in all SPNs, although more so in d-SPNs, which also increases their spiking. In parkinsonian D1R-/- mice, the spine density decreases in i-SPNs, and this spine loss recovers after chronic levodopa. CONCLUSIONS: D1R is essential for the maintenance of spine plasticity in d-SPNs but also affects i-SPNs, indicating an important crosstalk between these 2 types of neurons. © 2020 International Parkinson and Movement Disorder Society.


Asunto(s)
Cuerpo Estriado , Receptores de Dopamina D1 , Animales , Cuerpo Estriado/metabolismo , Espinas Dendríticas , Levodopa/farmacología , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo
9.
Mov Disord ; 35(9): 1636-1648, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32666590

RESUMEN

BACKGROUND: Numerous studies indicate an association between neurodegenerative and metabolic diseases. Although still a matter of debate, growing evidence from epidemiological and animal studies indicate that preexisting diabetes increases the risk to develop Parkinson's disease. However, the mechanisms of such an association are unknown. OBJECTIVES: We investigated whether diabetes alters striatal dopamine neurotransmission and assessed the vulnerability of nigrostriatal neurons to neurodegeneration. METHODS: We used streptozotocin-treated and genetically diabetic db/db mice. Expression of oxidative stress and nigrostriatal neuronal markers and levels of dopamine and its metabolites were monitored. Dopamine release and uptake were assessed using fast-scan cyclic voltammetry. 6-Hydroxydopamine was unilaterally injected into the striatum using stereotaxic surgery. Motor performance was scored using specific tests. RESULTS: Diabetes resulted in oxidative stress and decreased levels of dopamine and its metabolites in the striatum. Levels of proteins regulating dopamine release and uptake, including the dopamine transporter, the Girk2 potassium channel, the vesicular monoamine transporter 2, and the presynaptic vesicle protein synaptobrevin-2, were decreased in diabetic mice. Electrically evoked levels of extracellular dopamine in the striatum were enhanced, and altered dopamine uptake was observed. Striatal microinjections of a subthreshold dose of the neurotoxin 6-hydroxydopamine in diabetic mice, insufficient to cause motor alterations in nondiabetic animals, resulted in motor impairment, higher loss of striatal dopaminergic axons, and decreased neuronal cell bodies in the substantia nigra. CONCLUSIONS: Our results indicate that diabetes promotes striatal oxidative stress, alters dopamine neurotransmission, and increases vulnerability to neurodegenerative damage leading to motor impairment. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Diabetes Mellitus Experimental , Dopamina , Animales , Cuerpo Estriado/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ratones , Sustancia Negra/metabolismo , Transmisión Sináptica
10.
J Neurosci ; 38(15): 3619-3630, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29483281

RESUMEN

In toxin-based models of Parkinson's disease (PD), striatal projection neurons (SPNs) exhibit dendritic atrophy and spine loss concurrent with an increase in excitability. Chronic l-DOPA treatment that induces dyskinesia selectively restores spine density and excitability in indirect pathway SPNs (iSPNs), whereas spine loss and hyperexcitability persist in direct pathway SPNs (dSPNs). These alterations have only been characterized in toxin-based models of PD, raising the possibility that they are an artifact of exposure to the toxin, which may engage compensatory mechanisms independent of the PD-like pathology or due to the loss of dopaminergic afferents. To test all these, we studied the synaptic remodeling in Pitx3-/- or aphakia mice, a genetic model of PD, in which most of the dopamine neurons in the substantia nigra fail to fully differentiate and to innervate the striatum. We made 3D reconstructions of the dendritic arbor and measured excitability in identified SPNs located in dorsal striatum of BAC-Pitx3-/- mice treated with saline or l-DOPA. Both dSPNs and iSPNs from BAC-Pitx3-/- mice had shorter dendritic trees, lower spine density, and more action potentials than their counterparts from WT mice. Chronic l-DOPA treatment restored spine density and firing rate in iSPNs. By contrast, in dSPNs, spine loss and hyperexcitability persisted following l-DOPA treatment, which is similar to what happens in 6-OHDA WT mice. This indicates that dopamine-mediated synaptic remodeling and plasticity is independent of dopamine innervation during SPN development and that Pitx3-/- mice are a good model because they develop the same pathology described in the toxins-based models and in human postmortem studies of advanced PD.SIGNIFICANCE STATEMENT As the only genetic model of Parkinson's disease (PD) that develops dyskinesia, Pitx3-/- mice reproduce the behavioral effects seen in humans and are a good system for studying dopamine-induced synaptic remodeling. The studies we present here establish that the structural and functional synaptic plasticity that occur in striatal projection neurons in PD and in l-DOPA-induced dyskinesia are specifically due to modulation of the neurotransmitter dopamine and are not artifacts of the use of chemical toxins in PD models. In addition, our findings provide evidence that synaptic plasticity in the Pitx3-/- mouse is similar to that seen in toxin models despite its lack of dopaminergic innervation of the striatum during development. Pitx3-/- mice reproduced the alterations described in patients with advanced PD and in well accepted toxin-based models of PD and dyskinesia. These results further consolidate the fidelity of the Pitx3-/- mouse as a PD model in which to study the morphological and physiological remodeling of striatal projection neurons by administration of l-DOPA and other drugs.


Asunto(s)
Dendritas/efectos de los fármacos , Dopaminérgicos/farmacología , Levodopa/farmacología , Enfermedad de Parkinson/patología , Sustancia Negra/efectos de los fármacos , Sinapsis/efectos de los fármacos , Potenciales de Acción , Animales , Dendritas/patología , Dendritas/fisiología , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/fisiopatología , Sustancia Negra/patología , Sustancia Negra/fisiopatología , Sinapsis/patología , Sinapsis/fisiología , Potenciales Sinápticos , Factores de Transcripción/genética
11.
J Neural Transm (Vienna) ; 125(8): 1187-1194, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29417335

RESUMEN

The current standard treatment for Parkinson disease focuses on restoring striatal dopamine levels using L-3,4-dihydroxyphenylalanine (L-DOPA). However, disease progression and chronic treatment are associated with motor side effects such as L-DOPA-induced dyskinesia (LID). Dopamine receptor function is strongly associated with the mechanisms underlying LID. In fact, increased D1R signaling is associated with this motor side effect. Compelling evidence demonstrates that dopamine receptors in the striatum can form heteromeric complexes, and heteromerization can lead to changes in the functional and pharmacological properties of receptors compared to their monomeric subtypes. Currently, the most promising strategy for therapeutic intervention in dyskinesia originates from investigations of the D1R-D3R heteromers. Interestingly, there is a correlation between the expression of D1R-D3R heteromers and the development of LID. Moreover, D3R stimulation can potentiate the D1R signaling pathway. The aim of this review is to summarize current knowledge of the distinct roles of heteromeric dopaminergic receptor complexes in LID.


Asunto(s)
Discinesia Inducida por Medicamentos/metabolismo , Receptores de Dopamina D1/química , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D3/química , Receptores de Dopamina D3/metabolismo , Animales , Cuerpo Estriado/metabolismo , Humanos
12.
Cereb Cortex ; 27(1): 435-446, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26483399

RESUMEN

The dopamine D3 receptor (D3R) belongs to the dopamine D2-like receptor family and is principally located in the ventral striatum. However, previous studies reported D3R overexpression in the dorsal striatum following l-DOPA treatment in parkinsonian animals. This fact has drawn attention in the importance of D3R in l-DOPA-induced dyskinesia (LID). Here, we used D3R knockout mice to assess the role of D3R in LID and rotational sensitization in the hemiparkinsonian model. Mice lacking D3R presented a reduction in dyskinesia without interfering with the antiparkinsonian l-DOPA effect and were accompanied by a reduction in the l-DOPA-induced rotations. Interestingly, deleting D3R attenuated important molecular markers in the D1R-neurons such as FosB, extracellular signal-regulated kinase, and histone-3 (H3)-activation. Colocalization studies in D1R-tomato and D2R-green fluorescent protein BAC-transgenic mice indicated that l-DOPA-induced D3R overexpression principally occurs in D1R-containing neurons although it is also present in the D2R-neurons. Moreover, D3R pharmacological blockade with PG01037 reduced dyskinesia and the molecular markers expressed in D1R-neurons. In addition, this antagonist further reduced dyskinetic symptoms in D1R heterozygous mice, indicating a direct interaction between D1R and D3R. Together, our results demonstrate that D3R modulates the development of dyskinesia by targeting D1R-mediated intracellular signaling and suggest that decreasing D3R activity may help to ameliorate LID.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopaminérgicos/toxicidad , Discinesia Inducida por Medicamentos/metabolismo , Levodopa/toxicidad , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D3/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Ratones , Ratones Noqueados , Trastornos Parkinsonianos/metabolismo
13.
Neurobiol Dis ; 102: 133-139, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28315782

RESUMEN

Catechol-O-methyltransferase (COMT) degrades dopamine and its precursor l-DOPA and plays a critical role in regulating synaptic dopamine actions. We investigated the effects of heightened levels of COMT on dopamine-regulated motor behaviors and molecular alterations in a mouse model of dyskinesia. Transgenic mice overexpressing human COMT (TG) and their wildtype (WT) littermates received unilateral 6-OHDA lesions in the dorsal striatum and were treated chronically with l-DOPA for two weeks. l-DOPA-induced dyskinesia was exacerbated in TG mice without altering l-DOPA motor efficacy as determined by contralateral rotations or motor coordination. Inductions of FosB and phospho-acetylated histone 3 (molecular correlates of dyskinesia) were potentiated in the lesioned striatum of TG mice compared with their WT littermates. The TG mice had lower basal levels of dopamine in the striatum. In mice with lesions, l-DOPA induces a greater increase in the dopamine metabolite 3-methoxytyramine in the lesioned striatum of dyskinetic TG mice than in WT mice. The levels of serotonin and its metabolite were similar in TG and WT mice. Our results demonstrate that human COMT overexpression confers a heightened susceptibility to l-DOPA-induced dyskinesia and alters molecular and neurochemical responses in the lesioned striatum of mice.


Asunto(s)
Antiparkinsonianos/toxicidad , Catecol O-Metiltransferasa/metabolismo , Discinesia Inducida por Medicamentos/metabolismo , Levodopa/toxicidad , Animales , Antiparkinsonianos/farmacología , Proteínas del Dominio Armadillo/genética , Proteínas del Dominio Armadillo/metabolismo , Catecol O-Metiltransferasa/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Dopamina/metabolismo , Humanos , Levodopa/farmacología , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Oxidopamina , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Serotonina/metabolismo , Tiorredoxina Reductasa 2/genética , Tiorredoxina Reductasa 2/metabolismo
14.
Mov Disord ; 32(4): 530-537, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28256089

RESUMEN

BACKGROUND: Long-term levodopa (l-dopa) treatment is associated with the development of l-dopa-induced dyskinesias in the majority of patients with Parkinson disease (PD). The etiopathogonesis and mechanisms underlying l-dopa-induced dyskinesias are not well understood. METHODS: We used striatal optogenetic stimulation to induce dyskinesias in a hemiparkinsonian model of PD in rats. Striatal dopamine depletion was induced unilaterally by 6-hydroxydopamine injection into the medial forebrain bundle. For the optogenetic manipulation, we injected adeno-associated virus particles expressing channelrhodopsin to stimulate striatal medium spiny neurons with a laser source. RESULTS: Simultaneous optical activation of medium spiny neurons of the direct and indirect striatal pathways in the 6-hydroxydopamine lesion but l-dopa naïve rats induced involuntary movements similar to l-dopa-induced dyskinesias, labeled here as optodyskinesias. Noticeably, optodyskinesias were facilitated by l-dopa in animals that did not respond initially to the laser stimulation. In general, optodyskinesias lasted while the laser stimulus was applied, but in some instances remained ongoing for a few seconds after the laser was off. Postmortem tissue analysis revealed increased FosB expression, a molecular marker of l-dopa-induced dyskinesias, primarily in medium spiny neurons of the direct pathway in the dopamine-depleted hemisphere. CONCLUSION: Selective optogenetic activation of the dorsolateral striatum elicits dyskinesias in the 6-hydroxydopamine rat model of PD. This effect was associated with a preferential activation of the direct striato-nigral pathway. These results potentially open new avenues in the understanding of mechanisms involved in l-dopa-induced dyskinesias. © 2017 International Parkinson and Movement Disorder Society.


Asunto(s)
Adrenérgicos/toxicidad , Cuerpo Estriado/metabolismo , Discinesias/etiología , Optogenética/efectos adversos , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Animales , Antiparkinsonianos/efectos adversos , Encéfalo/metabolismo , Channelrhodopsins , Modelos Animales de Enfermedad , Dinorfinas/metabolismo , Lateralidad Funcional , Levodopa/efectos adversos , Masculino , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción Genética , Tirosina 3-Monooxigenasa/metabolismo
15.
Mov Disord ; 32(10): 1409-1422, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28779532

RESUMEN

BACKGROUND: Heterozygous mutations in the GBA1 gene, which encodes the lysosomal enzyme ß-glucocerebrosidase-1, increase the risk of developing Parkinson's disease, although the underlying mechanisms remain unclear. The aim of this study was to explore the impact of the N370S-GBA1 mutation on cellular homeostasis and vulnerability in a patient-specific cellular model of PD. METHODS: We isolated fibroblasts from 4 PD patients carrying the N370S/wild type GBA1 mutation and 6 controls to study the autophagy-lysosome pathway, endoplasmic reticulum stress, and Golgi apparatus structure by Western blot, immunofluorescence, LysoTracker and Filipin stainings, mRNA analysis, and electron microscopy. We evaluated cell vulnerability by apoptosis, reactive oxygen species and mitochondrial membrane potential with flow cytometry. RESULTS: The N370S mutation produced a significant reduction in ß-glucocerebrosidase-1 protein and enzyme activity and ß-glucocerebrosidase-1 retention within the endoplasmic reticulum, which interrupted its traffic to the lysosome. This led to endoplasmic reticulum stress activation and triggered unfolded protein response and Golgi apparatus fragmentation. Furthermore, these alterations resulted in autophagosome and p62/SQSTM1 accumulation. This impaired autophagy was a result of dysfunctional lysosomes, indicated by multilamellar body accumulation probably caused by increased cholesterol, enlarged lysosomal mass, and reduced enzyme activity. This phenotype impaired the removal of damaged mitochondria and reactive oxygen species production and enhanced cell death. CONCLUSIONS: Our results support a connection between the loss of ß-glucocerebrosidase-1 function, cholesterol accumulation, and the disruption of cellular homeostasis in GBA1-PD. Our work reveals new insights into the cellular pathways underlying PD pathogenesis, providing evidence that GBA1-PD shares common features with lipid-storage diseases. © 2017 International Parkinson and Movement Disorder Society.


Asunto(s)
Colesterol/metabolismo , Glucosilceramidasa/genética , Lisosomas/metabolismo , Mutación/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Asparagina/genética , Autofagia/genética , Beclina-1/metabolismo , Calnexina/metabolismo , Calnexina/ultraestructura , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Femenino , Fibroblastos/patología , Fibroblastos/ultraestructura , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Humanos , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Modelos Biológicos , Estrés Oxidativo/genética , Enfermedad de Parkinson/patología , Serina/genética , Serina-Treonina Quinasas TOR/metabolismo , Factor de Transcripción CHOP/metabolismo
16.
Cereb Cortex ; 26(11): 4253-4264, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27613437

RESUMEN

Dopamine depletion in Parkinson's disease (PD) produces dendritic spine loss in striatal medium spiny neurons (MSNs) and increases their excitability. However, the synaptic changes that occur in MSNs in PD, in particular those induced by chronic L-3,4-dihydroxyphenylalanine (L-DOPA) treatment, are still poorly understood. We exposed BAC-transgenic D1-tomato and D2-eGFP mice to PD and dyskinesia model paradigms, enabling cell type-specific assessment of changes in synaptic physiology and morphology. The distinct fluorescence markers allowed us to identify D1 and D2 MSNs for analysis using intracellular sharp electrode recordings, electron microscopy, and 3D reconstructions with single-cell Lucifer Yellow injections. Dopamine depletion induced spine pruning in both types of MSNs, affecting mushroom and thin spines equally. Dopamine depletion also increased firing rate in both D1- and D2-MSNs, but reduced evoked-EPSP amplitude selectively in D2-MSNs. L-DOPA treatment that produced dyskinesia differentially affected synaptic properties in D1- and D2-MSNs. In D1-MSNs, spine density remained reduced but the remaining spines were enlarged, with bigger heads and larger postsynaptic densities. These morphological changes were accompanied by facilitation of action potential firing triggered by synaptic inputs. In contrast, although L-DOPA restored the number of spines in D2-MSNs, it resulted in shortened postsynaptic densities. These changes in D2-MSNs correlated with a decrease in synaptic transmission. Our findings indicate that L-DOPA-induced dyskinesia is associated with abnormal spine morphology, modified synaptic transmission, and altered EPSP-spike coupling, with distinct effects in D1- and D2-MSNs.


Asunto(s)
Cuerpo Estriado/patología , Discinesia Inducida por Medicamentos/patología , Levodopa/farmacología , Neuronas/efectos de los fármacos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Columna Vertebral/patología , Animales , Modelos Animales de Enfermedad , Dopamina/farmacología , Dopaminérgicos/efectos adversos , Dopaminérgicos/farmacología , Discinesia Inducida por Medicamentos/etiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Levodopa/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/ultraestructura , Oxidopamina/toxicidad , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/genética , Columna Vertebral/ultraestructura , Simpaticolíticos/toxicidad
17.
Artículo en Inglés | MEDLINE | ID: mdl-26945022

RESUMEN

BACKGROUND: The C-terminal domain of the heavy chain of tetanus toxin (Hc-TeTx) is a nontoxic peptide with demonstrated in vitro and in vivo neuroprotective effects against striatal dopaminergic damage induced by 1-methyl-4-phenylpyridinium and 6-hydoxydopamine, suggesting its possible therapeutic potential in Parkinson's disease. Methamphetamine, a widely abused psychostimulant, has selective dopaminergic neurotoxicity in rodents, monkeys, and humans. This study was undertaken to determine whether Hc-TeTx might also protect against methamphetamine-induced dopaminergic neurotoxicity and the consequent motor impairment. METHODS: For this purpose, we treated mice with a toxic regimen of methamphetamine (4mg/kg, 3 consecutive i.p. injections, 3 hours apart) followed by 3 injections of 40 ug/kg of Hc-TeTx into grastrocnemius muscle at 1, 24, and 48 hours post methamphetamine treatment. RESULTS: We found that Hc-TeTx significantly reduced the loss of dopaminergic markers tyrosine hydroxylase and dopamine transporter and the increases in silver staining (a well stablished degeneration marker) induced by methamphetamine in the striatum. Moreover, Hc-TeTx prevented the increase of neuronal nitric oxide synthase but did not affect microglia activation induced by methamphetamine. Stereological neuronal count in the substantia nigra indicated loss of tyrosine hydroxylase-positive neurons after methamphetamine that was partially prevented by Hc-TeTx. Importantly, impairment in motor behaviors post methamphetamine treatment were significantly reduced by Hc-TeTx. CONCLUSIONS: Here we demonstrate that Hc-TeTx can provide significant protection against acute methamphetamine-induced neurotoxicity and motor impairment, suggesting its therapeutic potential in methamphetamine abusers.


Asunto(s)
Locomoción/efectos de los fármacos , Metanfetamina/antagonistas & inhibidores , Metanfetamina/toxicidad , Fármacos Neuroprotectores/farmacología , Síndromes de Neurotoxicidad/prevención & control , Fragmentos de Péptidos/farmacología , Toxina Tetánica/farmacología , Animales , Temperatura Corporal/efectos de los fármacos , Recuento de Células , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Masculino , Ratones , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/prevención & control , Prueba de Desempeño de Rotación con Aceleración Constante , Tirosina 3-Monooxigenasa/metabolismo
18.
Neurobiol Dis ; 73: 49-59, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25281315

RESUMEN

Nitric oxide (NO), a gaseous messenger molecule synthesized by nitric oxide synthase (NOS), plays a pivotal role in integrating dopamine transmission in the basal ganglia and has been implicated in the pathogenesis of Parkinson disease (PD). To study the role of the nitrergic system in l-DOPA-induced dyskinesia (LID), we assessed the effect of the pharmacological manipulation of NO levels and NO/cyclic guanosine monophosphate (cGMP) signaling on LID in the Pitx3(-/-) aphakia mouse, a genetic model of PD. To evaluate the effect of decreased NO signaling on the development of LID, Pitx3(-/-) mice were chronically treated with l-DOPA and 7-nitroindazole (7-NI, a neuronal NOS inhibitor). To evaluate its effect on the expression of established LID, 7-NI was administered acutely to dyskinetic mice. The chronic 7-NI treatment attenuated the development of LID in the Pitx3(-/-) mice, and the sub-acute 7-NI treatment attenuated established dyskinesia without affecting the beneficial therapeutic effect of l-DOPA. Moreover, 7-NI significantly reduced FosB and pAcH3 expression in the acutely and chronically l-DOPA-treated mice. We also examined how increasing NO/cGMP signaling affects LID expression by acutely administering molsidomine (an NO donor) or zaprinast (a cGMP phosphodiesterase 5-PDE5 inhibitor) before l-DOPA in mice with established dyskinesia. Paradoxically, the administration of either of these drugs also significantly diminished the expression of established LID; however, the effect occurred at the expense of the antiparkinsonian l-DOPA properties. We demonstrate that targeting the NO/cGMP signaling pathway reduces dyskinetic behaviors and molecular markers, but only the 7-NI treatment preserved the antiparkinsonian effect of l-DOPA, indicating that NOS inhibitors represent a potential therapy to reduce LID.


Asunto(s)
Cuerpo Estriado/metabolismo , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Indazoles/uso terapéutico , Factores de Transcripción/deficiencia , Animales , Antiparkinsonianos/toxicidad , Benserazida/uso terapéutico , Recuento de Células , Cuerpo Estriado/efectos de los fármacos , Modelos Animales de Enfermedad , Dopaminérgicos/toxicidad , Discinesia Inducida por Medicamentos/etiología , Discinesia Inducida por Medicamentos/patología , Regulación de la Expresión Génica/genética , Proteínas de Homeodominio/genética , Levodopa/toxicidad , Ratones , Ratones Noqueados , Molsidomina/administración & dosificación , Donantes de Óxido Nítrico/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Factores de Transcripción/genética
19.
Neurobiol Dis ; 74: 325-35, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25511804

RESUMEN

The dopamine (DA) transporter (DAT), a membrane glycoprotein expressed in dopaminergic neurons, clears DA from extracellular space and is regulated by diverse presynaptic proteins like protein kinases, α-synuclein, D2 and D3 autoreceptors. DAT dysfunction is implicated in Parkinson's disease and depression, which are therapeutically treated by dopaminergic D2/D3 receptor (D2/D3R) agonists. It is, then, important to improve our understanding of interactions between D3R and DAT. We show that prolonged administration of pramipexole (0.1mg/kg/day, 6 to 21 days), a preferential D3R agonist, leads to a decrease in DA uptake in mouse striatum that reflects a reduction in DAT affinity for DA in the absence of any change in DAT density or subcellular distribution. The effect of pramipexole was absent in mice with genetically-deleted D3R (D3R(-/-)), yet unaffected in mice genetically deprived of D2R (D2R(-/-)). Pramipexole treatment induced a physical interaction between D3R and DAT, as assessed by co-immunoprecipitation and in situ proximity ligation assay. Furthermore, it promoted the formation of DAT dimers and DAT association with both D2R and α-synuclein, effects that were abolished in D3R(-/-) mice, yet unaffected in D2R(-/-) mice, indicating dependence upon D3R. Collectively, these data suggest that prolonged treatment with dopaminergic D3 agonists provokes a reduction in DA reuptake by dopaminergic neurons related to a hitherto-unsuspected modification of the DAT interactome. These observations provide novel insights into the long-term antiparkinson, antidepressant and additional clinical actions of pramipexole and other D3R agonists.


Asunto(s)
Autorreceptores/metabolismo , Benzotiazoles/farmacología , Cuerpo Estriado/efectos de los fármacos , Agonistas de Dopamina/farmacología , Dopamina/metabolismo , Receptores de Dopamina D3/metabolismo , Animales , Antidepresivos/farmacología , Antiparkinsonianos/farmacología , Cuerpo Estriado/metabolismo , Dimerización , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Pramipexol , Receptores de Dopamina D3/agonistas , Receptores de Dopamina D3/genética , alfa-Sinucleína/metabolismo
20.
J Neurochem ; 130(4): 472-89, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24773031

RESUMEN

Dopamine replacement therapy in Parkinson's disease is associated with several unwanted effects, of which dyskinesia is the most disabling. The development of new therapeutic interventions to reduce the impact of dyskinesia in Parkinson's disease is therefore a priority need. This review summarizes the key molecular mechanisms that underlie dyskinesia. The role of dopamine receptors and their associated signaling mechanisms including dopamine-cAMP-regulated neuronal phosphoprotein, extracellular signal-regulated kinase, mammalian target of rapamycin, mitogen and stress-activated kinase-1 and Histone H3 are summarized, along with an evaluation of the role of cannabinoid and nicotinic acetylcholine receptors. The role of synaptic plasticity and animal behavioral results on dyskinesia are also evaluated. The most recent therapeutic advances to treat Parkinson's disease are discussed, with emphasis on the possibilities and limitations of non-pharmacological interventions such as physical activity, deep brain stimulation, transcranial magnetic field stimulation and cell replacement therapy. The review suggests new prospects for the management of Parkinson's disease-associated motor symptoms, especially the development of dyskinesia. This review aims at summarizing the key molecular mechanisms underlying dyskinesia and the most recent therapeutic advances to treat Parkinson's disease with emphasis on non-pharmacological interventions such as physical activity, deep brain stimulation (DBS), transcranial magnetic field stimulation (TMS) and cell replacement therapy. These new interventions are discussed from both the experimental and clinical point of view, describing their current strength and limitations.


Asunto(s)
Discinesias/etiología , Discinesias/terapia , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/terapia , Animales , Cannabinoides/farmacología , Trasplante de Células , Cromatina/efectos de los fármacos , Estimulación Encefálica Profunda , Dopaminérgicos/uso terapéutico , Fosfoproteína 32 Regulada por Dopamina y AMPc/fisiología , Discinesias/fisiopatología , Histonas/metabolismo , Humanos , Levodopa/farmacología , Levodopa/uso terapéutico , Actividad Motora/fisiología , Enfermedad de Parkinson/fisiopatología , Fosforilación , Desempeño Psicomotor/fisiología , Receptores de Dopamina D1/efectos de los fármacos , Receptores de Dopamina D1/metabolismo , Transducción de Señal/efectos de los fármacos , Estimulación Magnética Transcraneal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA