Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurobiol Dis ; 191: 106398, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38182075

RESUMEN

Parkinson's disease (PD) is characterized by the progressive and asymmetrical degeneration of the nigrostriatal dopamine neurons and the unilateral presentation of the motor symptoms at onset, contralateral to the most impaired hemisphere. We previously developed a rat PD model that mimics these typical features, based on unilateral injection of a substrate inhibitor of excitatory amino acid transporters, L-trans-pyrrolidine-2,4-dicarboxylate (PDC), in the substantia nigra (SN). Here, we used this progressive model in a multilevel study (behavioral testing, in vivo 1H-magnetic resonance spectroscopy, slice electrophysiology, immunocytochemistry and in situ hybridization) to characterize the functional changes occurring in the cortico-basal ganglia-cortical network in an evolving asymmetrical neurodegeneration context and their possible contribution to the cell death progression. We focused on the corticostriatal input and the subthalamic nucleus (STN), two glutamate components with major implications in PD pathophysiology. In the striatum, glutamate and glutamine levels increased from presymptomatic stages in the PDC-injected hemisphere only, which also showed enhanced glutamatergic transmission and loss of plasticity at corticostriatal synapses assessed at symptomatic stage. Surprisingly, the contralateral STN showed earlier and stronger reactivity than the ipsilateral side (increased intraneuronal cytochrome oxidase subunit I mRNA levels; enhanced glutamate and glutamine concentrations). Moreover, its lesion at early presymptomatic stage halted the ongoing neurodegeneration in the PDC-injected SN and prevented the expression of motor asymmetry. These findings reveal the existence of endogenous interhemispheric processes linking the primary injured SN and the contralateral STN that could sustain progressive dopamine neuron loss, opening new perspectives for disease-modifying treatment of PD.


Asunto(s)
Enfermedad de Parkinson , Trastornos Parkinsonianos , Núcleo Subtalámico , Ratas , Animales , Neuronas Dopaminérgicas/metabolismo , Dopamina/metabolismo , Glutamina/metabolismo , Trastornos Parkinsonianos/metabolismo , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , Glutamatos/metabolismo , Oxidopamina/farmacología
3.
J Neurosci Res ; 100(6): 1370-1385, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35355316

RESUMEN

Accumulating evidence implicates the parafascicular nucleus of the thalamus (Pf) in basal ganglia (BG)-related functions and pathologies. Despite Pf connectivity with all BG components, most attention is focused on the thalamostriatal system and an integrated view of thalamic information processing in this network is still lacking. Here, we addressed this question by recording the responses elicited by Pf activation in single neurons of the substantia nigra pars reticulata (SNr), the main BG output structure in rodents, in anesthetized mice. We performed optogenetic activation of Pf neurons innervating the striatum, the subthalamic nucleus (STN), or the SNr using virally mediated transcellular delivery of Cre from injection in either target in Rosa26-LoxP-stop-ChR2-EYFP mice to drive channelrhodopsin expression. Photoactivation of Pf neurons connecting the striatum evoked an inhibition often followed by an excitation, likely resulting from the activation of the trans-striatal direct and indirect pathways, respectively. Photoactivation of Pf neurons connecting the SNr or the STN triggered one or two early excitations, suggesting partial functional overlap of trans-subthalamic and direct thalamonigral projections. Excitations were followed in about half of the cases by an inhibition that might reflect recruitment of intranigral inhibitory loops. Finally, global Pf stimulation, electrical or optogenetic, elicited similar complex responses comprising up to four components: one or two short-latency excitations, an inhibition, and a late excitation. These data provide evidence for functional connections between the Pf and different BG components and for convergence of the information processed through these pathways in single SNr neurons, stressing their importance in regulating BG outflow.


Asunto(s)
Núcleos Talámicos Intralaminares , Núcleo Subtalámico , Animales , Ganglios Basales/fisiología , Cuerpo Estriado/fisiología , Núcleos Talámicos Intralaminares/fisiología , Ratones , Vías Nerviosas/fisiología , Tálamo/fisiología
4.
Transl Psychiatry ; 12(1): 106, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35292625

RESUMEN

We previously linked TSHZ3 haploinsufficiency to autism spectrum disorder (ASD) and showed that embryonic or postnatal Tshz3 deletion in mice results in behavioral traits relevant to the two core domains of ASD, namely social interaction deficits and repetitive behaviors. Here, we provide evidence that cortical projection neurons (CPNs) and striatal cholinergic interneurons (SCINs) are two main and complementary players in the TSHZ3-linked ASD syndrome. In the cerebral cortex, TSHZ3 is expressed in CPNs and in a proportion of GABAergic interneurons, but not in cholinergic interneurons or glial cells. In the striatum, TSHZ3 is expressed in all SCINs, while its expression is absent or partial in the other main brain cholinergic systems. We then characterized two new conditional knockout (cKO) models generated by crossing Tshz3flox/flox with Emx1-Cre (Emx1-cKO) or Chat-Cre (Chat-cKO) mice to decipher the respective role of CPNs and SCINs. Emx1-cKO mice show altered excitatory synaptic transmission onto CPNs and impaired plasticity at corticostriatal synapses, with neither cortical neuron loss nor abnormal layer distribution. These animals present social interaction deficits but no repetitive patterns of behavior. Chat-cKO mice exhibit no loss of SCINs but changes in the electrophysiological properties of these interneurons, associated with repetitive patterns of behavior without social interaction deficits. Therefore, dysfunction in either CPNs or SCINs segregates with a distinct ASD behavioral trait. These findings provide novel insights onto the implication of the corticostriatal circuitry in ASD by revealing an unexpected neuronal dichotomy in the biological background of the two core behavioral domains of this disorder.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Animales , Trastorno del Espectro Autista/genética , Trastorno Autístico/genética , Haploinsuficiencia , Interneuronas , Ratones , Sinapsis
5.
Cell Death Dis ; 12(5): 460, 2021 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-33966044

RESUMEN

TP53INP1 is a stress-induced protein, which acts as a dual positive regulator of transcription and of autophagy and whose deficiency has been linked with cancer and metabolic syndrome. Here, we addressed the unexplored role of TP53INP1 and of its Drosophila homolog dDOR in the maintenance of neuronal homeostasis under chronic stress, focusing on dopamine (DA) neurons under normal ageing- and Parkinson's disease (PD)-related context. Trp53inp1-/- mice displayed additional loss of DA neurons in the substantia nigra compared to wild-type (WT) mice, both with ageing and in a PD model based on targeted overexpression of α-synuclein. Nigral Trp53inp1 expression of WT mice was not significantly modified with ageing but was markedly increased in the PD model. Trp53inp2 expression showed similar evolution and did not differ between WT and Trp53inp1-/- mice. In Drosophila, pan-neuronal dDOR overexpression improved survival under paraquat exposure and mitigated the progressive locomotor decline and the loss of DA neurons caused by the human α-synuclein A30P variant. dDOR overexpression in DA neurons also rescued the locomotor deficit in flies with RNAi-induced downregulation of dPINK1 or dParkin. Live imaging, confocal and electron microscopy in fat bodies, neurons, and indirect flight muscles showed that dDOR acts as a positive regulator of basal autophagy and mitophagy independently of the PINK1-mediated pathway. Analyses in a mammalian cell model confirmed that modulating TP53INP1 levels does not impact mitochondrial stress-induced PINK1/Parkin-dependent mitophagy. These data provide the first evidence for a neuroprotective role of TP53INP1/dDOR and highlight its involvement in the regulation of autophagy and mitophagy in neurons.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Choque Térmico/metabolismo , Neuroprotección/genética , Enfermedad de Parkinson/genética , Estrés Fisiológico/genética , Factores de Edad , Animales , Humanos , Ratones
6.
Stem Cells Transl Med ; 10(5): 725-742, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33528918

RESUMEN

Enhancing the differentiation potential of human induced pluripotent stem cells (hiPSC) into disease-relevant cell types is instrumental for their widespread application in medicine. Here, we show that hiPSCs downregulated for the signaling modulator GLYPICAN-4 (GPC4) acquire a new biological state characterized by increased hiPSC differentiation capabilities toward ventral midbrain dopaminergic (VMDA) neuron progenitors. This biological trait emerges both in vitro, upon exposing cells to VMDA neuronal differentiation signals, and in vivo, even when transplanting hiPSCs at the extreme conditions of floor-plate stage in rat brains. Moreover, it is compatible with the overall neuronal maturation process toward acquisition of substantia nigra neuron identity. HiPSCs with downregulated GPC4 also retain self-renewal and pluripotency in stemness conditions, in vitro, while losing tumorigenesis in vivo as assessed by flank xenografts. In conclusion, our results highlight GPC4 downregulation as a powerful approach to enhance generation of VMDA neurons. Outcomes may contribute to establish hiPSC lines suitable for translational applications.


Asunto(s)
Diferenciación Celular , Neuronas Dopaminérgicas , Glipicanos , Células Madre Pluripotentes Inducidas , Animales , Células Cultivadas , Neuronas Dopaminérgicas/citología , Regulación hacia Abajo , Glipicanos/genética , Xenoinjertos , Humanos , Células Madre Pluripotentes Inducidas/citología , Mesencéfalo , Células-Madre Neurales/citología , Ratas
8.
Glia ; 68(10): 2028-2039, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32170887

RESUMEN

Glial cells have a major role in protecting neurons against various forms of stress. Especially, astrocytes mediate the bulk of glutamate clearance in the brain via specific membrane transporters (GLAST and GLT1), thereby preventing the occurrence of excitotoxic events. Although glutamate-mediated mechanisms are thought to contribute to nigral dopaminergic neuron degeneration in Parkinson's disease, detailed information on the organization of glia in the substantia nigra is still lacking. The present study was performed to provide quantitative information on the organization of astroglia and on the relationships between astrocytes and excitatory synapses in the rat substantia nigra. Using immunolabeling of GLT1 and confocal imaging, we found that the substantia nigra was filled with a dense meshwork of immunoreactive astrocyte processes. Stereological analysis performed on electron microscope images revealed that the density of immunoreactive astrocyte plasma membranes was substantial, close to 1 µm2 /µm3 , in the substantia nigra neuropil, both in the pars compacta and the pars reticulata. Excitatory synapses had on average two thirds of their perimeters free from glia, a disposition that may favor transmitter spillover. The density of glutamatergic synapses, as quantified on confocal images by the simultaneous detection of bassoon and of vesicular glutamate transporter 1 or 2, was very low (0.01 and 0.025 per µm3 in the reticulata and compacta subdivisions, respectively). Thus the ratio of GLT1-expressing glial membrane surface to glutamatergic synapses was very high (40-100 µm2 ), suggesting an efficient regulation of extracellular glutamate concentrations.


Asunto(s)
Transportador 2 de Aminoácidos Excitadores/biosíntesis , Neuroglía/metabolismo , Sustancia Negra/metabolismo , Sinapsis/metabolismo , Animales , Transportador 2 de Aminoácidos Excitadores/ultraestructura , Masculino , Neuroglía/ultraestructura , Ratas , Ratas Wistar , Sustancia Negra/ultraestructura , Sinapsis/ultraestructura
9.
Mov Disord ; 35(4): 616-628, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31930749

RESUMEN

BACKGROUND: Apathy is one of the most disabling neuropsychiatric symptoms in Parkinson's disease (PD) patients and has a higher prevalence in patients under subthalamic nucleus deep brain stimulation. Indeed, despite its effectiveness for alleviating PD motor symptoms, its neuropsychiatric repercussions have not yet been fully uncovered. Because it can be alleviated by dopaminergic therapies, especially D2 and D3 dopaminergic receptor agonists, the commonest explanation proposed for apathy after subthalamic nucleus deep brain stimulation is a too-strong reduction in dopaminergic treatments. The objective of this study was to determine whether subthalamic nucleus deep brain stimulation can induce apathetic behaviors, which remains an important matter of concern. We aimed to unambiguously address this question of the motivational effects of chronic subthalamic nucleus deep brain stimulation. METHODS: We longitudinally assessed the motivational effects of chronic subthalamic nucleus deep brain stimulation by using innovative wireless microstimulators, allowing continuous stimulation of the subthalamic nucleus in freely moving rats and a pharmacological therapeutic approach. RESULTS: We showed for the first time that subthalamic nucleus deep brain stimulation induces a motivational deficit in naive rats and intensifies those existing in a rodent model of PD neuropsychiatric symptoms. As reported from clinical studies, this loss of motivation was fully reversed by chronic treatment with pramipexole, a D2 and D3 dopaminergic receptor agonist. CONCLUSIONS: Taken together, these data provide experimental evidence that chronic subthalamic nucleus deep brain stimulation by itself can induce loss of motivation, reminiscent of apathy, independently of the dopaminergic neurodegenerative process or reduction in dopamine replacement therapy, presumably reflecting a dopaminergic-driven deficit. Therefore, our data help to clarify and reconcile conflicting clinical observations by highlighting some of the mechanisms of the neuropsychiatric side effects induced by chronic subthalamic nucleus deep brain stimulation. © 2020 International Parkinson and Movement Disorder Society.


Asunto(s)
Apatía , Estimulación Encefálica Profunda , Enfermedad de Parkinson , Núcleo Subtalámico , Animales , Agonistas de Dopamina/farmacología , Humanos , Enfermedad de Parkinson/terapia , Ratas
10.
Mol Cell Neurosci ; 100: 103397, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31454665

RESUMEN

The corpus callosum is the largest bundle of commissural fibres that transfer information between the two cerebral hemispheres. Callosal projection neurons (CPNs) are a diverse population of pyramidal neurons within the neocortex that mainly interconnect homotopic regions of the opposite cortices. Nevertheless, some CPNs are involved in heterotopic projections between distinct cortical areas or to subcortical regions such as the striatum. In this study, we showed that the axon guidance receptor PlexinD1 is expressed by a large proportion of heterotopically projecting CPNs in layer 5A of the primary somatosensory (S1) and motor (M1) areas. Retrograde tracing of M1 CPNs projecting to the contralateral striatum revealed the presence of ectopic neurons aberrantly located in layers 2/3 of Plxnd1 and Sema3e mutant cortices. These results showed that Sema3E/PlexinD1 signalling controls the laminar distribution of heterotopically projecting CPNs.


Asunto(s)
Cuerpo Calloso/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Glicoproteínas de Membrana/metabolismo , Neuronas/metabolismo , Semaforinas/metabolismo , Animales , Cuerpo Calloso/metabolismo , Femenino , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Corteza Motora/citología , Corteza Motora/metabolismo , Técnicas de Trazados de Vías Neuroanatómicas , Neuronas/citología , Neuronas/fisiología , Semaforinas/genética , Corteza Somatosensorial/citología , Corteza Somatosensorial/metabolismo
11.
Biol Psychiatry ; 86(4): 274-285, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31060802

RESUMEN

BACKGROUND: Heterozygous deletion of the TSHZ3 gene, encoding for the teashirt zinc-finger homeobox family member 3 (TSHZ3) transcription factor that is highly expressed in cortical projection neurons (CPNs), has been linked to an autism spectrum disorder (ASD) syndrome. Similarly, mice with Tshz3 haploinsufficiency show ASD-like behavior, paralleled by molecular changes in CPNs and corticostriatal synaptic dysfunctions. Here, we aimed at gaining more insight into "when" and "where" TSHZ3 is required for the proper development of the brain, and its deficiency crucial for developing this ASD syndrome. METHODS: We generated and characterized a novel mouse model of conditional Tshz3 deletion, obtained by crossing Tshz3flox/flox with CaMKIIalpha-Cre mice, in which Tshz3 is deleted in CPNs from postnatal day 2 to 3 onward. We characterized these mice by a multilevel approach combining genetics, cell biology, electrophysiology, behavioral testing, and bioinformatics. RESULTS: These conditional Tshz3 knockout mice exhibit altered cortical expression of more than 1000 genes, ∼50% of which have their human orthologue involved in ASD, in particular genes encoding for glutamatergic synapse components. Consistently, we detected electrophysiological and synaptic changes in CPNs and impaired corticostriatal transmission and plasticity. Furthermore, these mice showed strong ASD-like behavioral deficits. CONCLUSIONS: Our study reveals a crucial postnatal role of TSHZ3 in the development and functioning of the corticostriatal circuitry and provides evidence that dysfunction in these circuits might be determinant for ASD pathogenesis. Our conditional Tshz3 knockout mouse constitutes a novel ASD model, opening the possibility for an early postnatal therapeutic window for the syndrome linked to TSHZ3 haploinsufficiency.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas de Homeodominio/genética , Sinapsis/genética , Factores de Transcripción/genética , Animales , Trastorno del Espectro Autista/patología , Conducta Animal , Deleción Cromosómica , Cromosomas Humanos Par 19 , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Haploinsuficiencia , Heterocigoto , Humanos , Masculino , Ratones , Ratones Noqueados
12.
Nat Genet ; 48(11): 1359-1369, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27668656

RESUMEN

TSHZ3, which encodes a zinc-finger transcription factor, was recently positioned as a hub gene in a module of the genes with the highest expression in the developing human neocortex, but its functions remained unknown. Here we identify TSHZ3 as the critical region for a syndrome associated with heterozygous deletions at 19q12-q13.11, which includes autism spectrum disorder (ASD). In Tshz3-null mice, differentially expressed genes include layer-specific markers of cerebral cortical projection neurons (CPNs), and the human orthologs of these genes are strongly associated with ASD. Furthermore, mice heterozygous for Tshz3 show functional changes at synapses established by CPNs and exhibit core ASD-like behavioral abnormalities. These findings highlight essential roles for Tshz3 in CPN development and function, whose alterations can account for ASD in the newly defined TSHZ3 deletion syndrome.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas de Homeodominio/genética , Neocórtex/patología , Neuronas/patología , Factores de Transcripción/genética , Animales , Trastorno del Espectro Autista/patología , Deleción Cromosómica , Cromosomas Humanos Par 19 , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Haploinsuficiencia , Heterocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos CBA , Neocórtex/embriología , Neurogénesis/genética , Sinapsis/genética
13.
J Neurochem ; 136(5): 1004-16, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26576509

RESUMEN

The long-term effects and action mechanisms of subthalamic nucleus (STN) high-frequency stimulation (HFS) for Parkinson's disease still remain poorly characterized, mainly due to the lack of experimental models relevant to clinical application. To address this issue, we performed a multilevel study in freely moving hemiparkinsonian rats undergoing 5-week chronic STN HFS, using a portable constant-current microstimulator. In vivo metabolic neuroimaging by (1) H-magnetic resonance spectroscopy (11.7 T) showed that STN HFS normalized the tissue levels of the neurotransmission-related metabolites glutamate, glutamine and GABA in both the striatum and substantia nigra reticulata (SNr), which were significantly increased in hemiparkinsonian rats, but further decreased nigral GABA levels below control values; taurine levels, which were not affected in hemiparkinsonian rats, were significantly reduced. Slice electrophysiological recordings revealed that STN HFS was, uniquely among antiparkinsonian treatments, able to restore both forms of corticostriatal synaptic plasticity, i.e. long-term depression and potentiation, which were impaired in hemiparkinsonian rats. Behavior analysis (staircase test) showed a progressive recovery of motor skill during the stimulation period. Altogether, these data show that chronic STN HFS efficiently counteracts metabolic and synaptic defects due to dopaminergic lesion in both the striatum and SNr. Comparison of chronic STN HFS with acute and subchronic treatment further suggests that the long-term benefits of this treatment rely both on the maintenance of acute effects and on delayed actions on the basal ganglia network. We studied the effects of chronic (5 weeks) continuous subthalamic nucleus (STN) high-frequency stimulation (HFS) in hemiparkinsonian rats. The levels of glutamate and GABA in the striatum () and substantia nigra reticulata (SNr) (), measured by in vivo proton magnetic resonance spectroscopy ((1) H-MRS), were increased by 6-hydroxydopamine (6-OHDA) lesion, which also disrupted corticostriatal synaptic plasticity () and impaired forepaw skill () in the staircase test. Five-week STN HFS normalized glutamate and GABA levels and restored both synaptic plasticity and motor function. A partial behavioral recovery was observed at 2-week STN HFS.


Asunto(s)
Ganglios Basales/metabolismo , Conducta Animal/efectos de los fármacos , Estimulación Encefálica Profunda , Plasticidad Neuronal/efectos de los fármacos , Sustancia Negra/metabolismo , Núcleo Subtalámico/metabolismo , Animales , Ganglios Basales/fisiopatología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , Estimulación Encefálica Profunda/métodos , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Oxidopamina/farmacología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/terapia , Ratas , Sustancia Negra/fisiopatología , Núcleo Subtalámico/fisiopatología , Tiempo
14.
Front Syst Neurosci ; 9: 51, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25926776

RESUMEN

In prior studies, we described the differential organization of corticostriatal and thalamostriatal inputs to the spines of direct pathway (dSPNs) and indirect pathway striatal projection neurons (iSPNs) of the matrix compartment. In the present electron microscopic (EM) analysis, we have refined understanding of the relative amounts of cortical axospinous vs. axodendritic input to the two types of SPNs. Of note, we found that individual dSPNs receive about twice as many axospinous synaptic terminals from IT-type (intratelencephalically projecting) cortical neurons as they do from PT-type (pyramidal tract projecting) cortical neurons. We also found that PT-type axospinous synaptic terminals were about 1.5 times as common on individual iSPNs as IT-type axospinous synaptic terminals. Overall, a higher percentage of IT-type terminals contacted dSPN than iSPN spines, while a higher percentage of PT-type terminals contacted iSPN than dSPN spines. Notably, IT-type axospinous synaptic terminals were significantly larger on iSPN spines than on dSPN spines. By contrast to axospinous input, the axodendritic PT-type input to dSPNs was more substantial than that to iSPNs, and the axodendritic IT-type input appeared to be meager and comparable for both SPN types. The prominent axodendritic PT-type input to dSPNs may accentuate their PT-type responsiveness, and the large size of axospinous IT-type terminals on iSPNs may accentuate their IT-type responsiveness. Using transneuronal labeling with rabies virus to selectively label the cortical neurons with direct input to the dSPNs projecting to the substantia nigra pars reticulata, we found that the input predominantly arose from neurons in the upper layers of motor cortices, in which IT-type perikarya predominate. The differential cortical input to SPNs is likely to play key roles in motor control and motor learning.

15.
J Neurochem ; 132(6): 703-12, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25533782

RESUMEN

Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is an efficient neurosurgical treatment for advanced Parkinson's disease. Non-invasive metabolic neuroimaging during the course of DBS in animal models may contribute to our understanding of its action mechanisms. Here, DBS was adapted to in vivo proton magnetic resonance spectroscopy at 11.7 T in the rat to follow metabolic changes in main basal ganglia structures, the striatum, and the substantia nigra pars reticulata (SNr). Measurements were repeated OFF and ON acute and subchronic (7 days) STN-DBS in control and parkinsonian (6-hydroxydopamine lesion) conditions. Acute DBS reversed the increases in glutamate, glutamine, and GABA levels induced by the dopamine lesion in the striatum but not in the SNr. Subchronic DBS normalized GABA in both the striatum and SNr, and glutamate in the striatum. Taurine levels were markedly decreased under subchronic DBS in the striatum and SNr in both lesioned and unlesioned rats. Microdialysis in the striatum further showed that extracellular taurine was increased. These data reveal that STN-DBS has duration-dependent metabolic effects in the basal ganglia, consistent with development of adaptive mechanisms. In addition to counteracting defects induced by the dopamine lesion, prolonged DBS has proper effects independent of the pathological condition. Non-invasive metabolic neuroimaging might be useful to understand the physiological mechanisms of deep brain stimulation (DBS). Here, we demonstrate the feasibility of repeated high-field proton magnetic resonance spectroscopy of basal ganglia structures under subthalamic nucleus DBS in control and parkinsonian rats. Results show that DBS has both rapid and delayed effects either dependent or independent of disease state.


Asunto(s)
Ganglios Basales/metabolismo , Estimulación Encefálica Profunda/tendencias , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/terapia , Núcleo Subtalámico/metabolismo , Núcleo Subtalámico/cirugía , Animales , Encéfalo/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Masculino , Ratas , Ratas Wistar , Factores de Tiempo
16.
Neuropsychopharmacology ; 39(11): 2662-72, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24903652

RESUMEN

The striatum is the input structure of the basal ganglia network that contains heterogeneous neuronal populations, including two populations of projecting neurons called the medium spiny neurons (MSNs), and different types of interneurons. We developed a transgenic mouse model enabling inducible ablation of the striatonigral MSNs constituting the direct pathway by expressing the human diphtheria toxin (DT) receptor under the control of the Slc35d3 gene promoter, a gene enriched in striatonigral MSNs. DT injection into the striatum triggered selective elimination of the majority of striatonigral MSNs. DT-mediated ablation of striatonigral MSNs caused selective loss of cholinergic interneurons in the dorsal striatum but not in the ventral striatum (nucleus accumbens), suggesting a region-specific critical role of the direct pathway in striatal cholinergic neuron homeostasis. Mice with DT injection into the dorsal striatum showed altered basal and cocaine-induced locomotion and dramatic reduction of L-DOPA-induced dyskinesia in the parkinsonian condition. In addition, these mice exhibited reduced anxiety, revealing a role of the dorsal striatum in the modulation of behaviors involving an emotional component, behaviors generally associated with limbic structures. Altogether, these results highlight the implication of the direct striatonigral pathway in the regulation of heterogeneous functions from cell survival to regulation of motor and emotion-associated behaviors.


Asunto(s)
Ansiedad/fisiopatología , Neuronas Colinérgicas/fisiología , Cuerpo Estriado/fisiología , Interneuronas/fisiología , Movimiento/fisiología , Sustancia Negra/fisiología , Animales , Antiparkinsonianos/toxicidad , Neuronas Colinérgicas/efectos de los fármacos , Cocaína/farmacología , Cuerpo Estriado/efectos de los fármacos , Toxina Diftérica/toxicidad , Inhibidores de Captación de Dopamina/farmacología , Discinesia Inducida por Medicamentos/fisiopatología , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Humanos , Interneuronas/efectos de los fármacos , Levodopa/toxicidad , Ratones Transgénicos , Proteínas de Transporte de Monosacáridos/genética , Movimiento/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Neurotoxinas/toxicidad , Regiones Promotoras Genéticas , Sustancia Negra/efectos de los fármacos
17.
Neurobiol Dis ; 65: 69-81, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24480091

RESUMEN

Parkinson's disease (PD) is characterized by the progressive degeneration of substantia nigra (SN) dopamine neurons, involving a multifactorial cascade of pathogenic events. Here we explored the hypothesis that dysfunction of excitatory amino acid transporters (EAATs) might be involved. Acutely-induced dysfunction of EAATs in the rat SN, by single unilateral injection of their substrate inhibitor l-trans-pyrrolidine-2,4-dicarboxylate (PDC), triggers a neurodegenerative process mimicking several PD features. Dopamine neurons are selectively affected, consistent with their sustained excitation by PDC measured by slice electrophysiology. The anti-oxidant N-acetylcysteine and the NMDA receptor antagonists ifenprodil and memantine provide neuroprotection. Besides oxidative stress and NMDA receptor-mediated excitotoxicity, glutathione depletion and neuroinflammation characterize the primary insult. Most interestingly, the degeneration progresses overtime with unilateral to bilateral and caudo-rostral evolution. Transient adaptive changes in dopamine function markers in SN and striatum accompany cell loss and axonal dystrophy, respectively. Motor deficits appear when neuron loss exceeds 50% in the most affected SN and striatal dopamine tone is dramatically reduced. These findings outline a functional link between EAAT dysfunction and several PD pathogenic mechanisms/pathological hallmarks, and provide a novel acutely-triggered model of progressive Parkinsonism.


Asunto(s)
Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Sustancia Negra/metabolismo , Acetilcisteína/uso terapéutico , Potenciales de Acción/efectos de los fármacos , Animales , Ácidos Dicarboxílicos/toxicidad , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Miembro Anterior/fisiopatología , Depuradores de Radicales Libres/uso terapéutico , Lateralidad Funcional , Glutamato Descarboxilasa/metabolismo , Técnicas In Vitro , Masculino , Actividad Motora/efectos de los fármacos , Neuroglía/patología , Inhibidores de la Captación de Neurotransmisores/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/tratamiento farmacológico , Pirrolidinas/toxicidad , Ratas , Ratas Wistar , Sustancia Negra/efectos de los fármacos , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
18.
PLoS One ; 8(4): e60447, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23593219

RESUMEN

This study addresses the molecular mechanisms underlying the action of subthalamic nucleus high frequency stimulation (STN-HFS) in the treatment of Parkinson's disease and its interaction with levodopa (L-DOPA), focusing on the striatum. Striatal gene expression profile was assessed in rats with nigral dopamine neuron lesion, either treated or not, using agilent microarrays and qPCR verification. The treatments consisted in anti-akinetic STN-HFS (5 days), chronic L-DOPA treatment inducing dyskinesia (LIDs) or the combination of the two treatments that exacerbated LIDs. STN-HFS modulated 71 striatal genes. The main biological processes associated with the differentially expressed gene products include regulation of growth, of apoptosis and of synaptic transmission, and extracellular region is a major cellular component implicated. In particular, several of these genes have been shown to support survival or differentiation of striatal or of dopaminergic neurons. These results indicate that STN HFS may induce widespread anatomo-functional rearrangements in the striatum and create a molecular environment favorable for neuroprotection and neuroplasticity. STN-HFS and L-DOPA treatment share very few common gene regulation features indicating that the molecular substrates underlying their striatal action are mostly different; among the common effects is the down-regulation of Adrb1, which encodes the adrenergic beta-1-receptor, supporting a major role of this receptor in Parkinson's disease. In addition to genes already reported to be associated with LIDs (preprodynorphin, thyrotropin-releasing hormone, metabotropic glutamate receptor 4, cannabinoid receptor 1), the comparison between DOPA and DOPA/HFS identifies immunity-related genes as potential players in L-DOPA side effects.


Asunto(s)
Cuerpo Estriado/metabolismo , Estimulación Encefálica Profunda , Enfermedad de Parkinson/genética , Núcleo Subtalámico/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Análisis por Conglomerados , Desnervación , Modelos Animales de Enfermedad , Estimulación Eléctrica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Levodopa/farmacología , Redes y Vías Metabólicas/genética , Enfermedad de Parkinson/metabolismo , Ratas
19.
J Neurosci Methods ; 209(1): 50-7, 2012 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-22659685

RESUMEN

In the last decades, deep brain stimulation (DBS) has been widely used as a functional surgical strategy for the treatment of a variety of neurological and psychiatric disorders, including Parkinson's disease (PD), dystonia, epilepsy, depression or obsessive-compulsive disorder. While the therapeutic benefits of DBS are now recognized, experimental data on its mechanisms and impact at long term remain poor. This is mainly due to the lack of a microstimulation system adapted for chronic DBS in small laboratory animals. In this context, we have developed a microstimulator for DBS adapted to rat. This device, which has a size and weight compatible for use in freely moving rat, can be clipped to a support fixed on the animal's head. This easy "removal" property is crucial because it enables removing or even switching the microstimulator during the experiments without having to anaesthetize or to operate the animal, thus minimizing stress. The design of the microstimulator allows to set the DBS parameters easily (intensity, frequency and pulse width) and to replace the battery for long-term DBS. To validate our device, we performed continuous DBS of the subthalamic nucleus (known to improve motor deficits in clinic) in a classical rat model of PD during 5 weeks. We show that this long duration stimulation reduces significantly PD-induced akinesia without inducing animal discomfort and tissue damage. These first data demonstrated that long term DBS procedure in behaving rat is now workable.


Asunto(s)
Estimulación Encefálica Profunda/instrumentación , Modelos Animales de Enfermedad , Microelectrodos , Animales , Masculino , Movimiento/fisiología , Trastornos Parkinsonianos/terapia , Ratas , Ratas Wistar
20.
Neurobiol Dis ; 42(3): 284-91, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21296669

RESUMEN

Chronic high frequency stimulation of the subthalamic nucleus (STN-HFS) efficiently alleviates motor symptoms of advanced Parkinson's disease (PD). Here, we looked for possible STN-HFS-induced changes on adult brain neurogenesis in the hippocampus and olfactory bulb that may be related to non-motor deficits associated to PD, such as mood disorders and olfaction deficits. Cell proliferation (Ki-67 immuno-positive-cells) and survival (bromodeoxyuridine (BrdU)-immuno-positive cells) were assessed in the subventricular zone-olfactory bulb continuum and the dentate gyrus of the hippocampus of hemiparkinsonian rats with or without continuous STN-HFS for 8 days. Dopamine lesion impaired cell proliferation and survival through different mechanisms, the effect on proliferation being correlated to the level of dopamine depletion whereas the effect on survival was not. Prolonged STN-HFS did not affect cell proliferation, but increased cell survival bilaterally. In these regions of constitutive neurogenesis, the percentage of new neuroblasts (BrdU-doublecortin-positive cells) was unchanged, suggesting that STN-HFS can lead to a net increase in newly formed neurons later on. STN-HFS also increased new cell survival in the striatum and promoted dopamine system recovery detected by tyrosine hydroxylase immunostaining. These data provide the first evidence that prolonged STN-HFS has a neurorestorative action and support the view that the action of this neurosurgical treatment can bypass the cortico-basal ganglia-thalamocortical loop circuits and largely impinge neuroplasticity and brain function.


Asunto(s)
Neurogénesis/fisiología , Enfermedad de Parkinson/fisiopatología , Núcleo Subtalámico/fisiopatología , Análisis de Varianza , Animales , Estimulación Encefálica Profunda , Modelos Animales de Enfermedad , Dopamina/metabolismo , Proteína Doblecortina , Electrodos Implantados , Inmunohistoquímica , Masculino , Enfermedad de Parkinson/metabolismo , Ratas , Ratas Wistar , Núcleo Subtalámico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...