Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Neurosci Methods ; 405: 110102, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38432304

RESUMEN

Cell reprogramming holds enormous potential to revolutionize our understanding of neurological and neurodevelopmental disorders, as well as enhance drug discovery and regenerative medicine. We have developed a direct cell reprogramming technology that allows us to generate lineage-specific neural cells. To extend our technology, we have investigated the incorporation of directly reprogrammed human lateral ganglionic eminence precursor cells (hiLGEPs) in a 3-dimensional (3D) matrix. Hydrogels are one of the most promising bio-scaffolds for 3D cell culture, providing cells with a supportive environment to adhere, proliferate, and differentiate. In particular, gelatin methacryloyl (GelMA) hydrogels have been used for a variety of 3D biomedical applications due to their biocompatibility, enzymatic cleavage, cell adhesion and tunable physical characteristics. This study therefore investigated the effect of GelMA hydrogel encapsulation on the survival and differentiation of hiLGEPs, both in vitro and following ex vivo transplantation into a quinolinic acid (QA) lesion rat organotypic slice culture model. We demonstrate, for the first time, that the encapsulation of hiLGEPs in GelMA hydrogel significantly enhances the survival and generation of DARPP32+ striatal neurons both in vitro and following ex vivo transplant. Furthermore, GelMA-encapsulated hiLGEPs were predominantly located away from the reactive astrocyte network that forms following QA lesioning, suggesting GelMA provides a protective barrier for cells in regions of inflammatory activation. Overall, these results indicate that GelMA hydrogel has the potential to act as a 3D bio-scaffold to augment the viability and differentiation of hiLGEPs for research and translation of pharmaceutical development and regenerative medicine.


Asunto(s)
Eminencia Ganglionar , Hidrogeles , Humanos , Ratas , Animales , Gelatina/farmacología , Metacrilatos , Andamios del Tejido
2.
Neurosci Bull ; 39(10): 1544-1560, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37253985

RESUMEN

The secondary motor cortex (M2) encodes choice-related information and plays an important role in cue-guided actions. M2 neurons innervate the dorsal striatum (DS), which also contributes to decision-making behavior, yet how M2 modulates signals in the DS to influence perceptual decision-making is unclear. Using mice performing a visual Go/No-Go task, we showed that inactivating M2 projections to the DS impaired performance by increasing the false alarm (FA) rate to the reward-irrelevant No-Go stimulus. The choice signal of M2 neurons correlated with behavioral performance, and the inactivation of M2 neurons projecting to the DS reduced the choice signal in the DS. By measuring and manipulating the responses of direct or indirect pathway striatal neurons defined by M2 inputs, we found that the indirect pathway neurons exhibited a shorter response latency to the No-Go stimulus, and inactivating their early responses increased the FA rate. These results demonstrate that the M2-to-DS pathway is crucial for suppressing inappropriate responses in perceptual decision behavior.


Asunto(s)
Corteza Motora , Ratones , Animales , Cuerpo Estriado/fisiología , Neostriado , Neuronas/fisiología , Tiempo de Reacción
3.
Trends Neurosci ; 46(5): 365-376, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36907678

RESUMEN

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by a trinucleotide repeat expansion in the huntingtin gene resulting in long stretches of polyglutamine repeats in the huntingtin protein. The disease involves progressive degeneration of neurons in the striatum and cerebral cortex resulting in loss of control of motor function, psychiatric problems, and cognitive deficits. There are as yet no treatments that can slow disease progression in HD. Recent advances in gene editing using clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) systems and demonstrations of their ability to correct gene mutations in animal models of a range of diseases suggest that gene editing may prove effective in preventing or ameliorating HD. Here we describe (i) potential CRISPR-Cas designs and cellular delivery methods for the correction of mutant genes that cause inherited diseases, and (ii) recent preclinical findings demonstrating the efficacy of such gene-editing approaches in animal models, with a focus on HD.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Animales , Edición Génica/métodos , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Sistemas CRISPR-Cas/genética , Enfermedades Neurodegenerativas/genética , Mutación , Modelos Animales de Enfermedad
4.
Neuroscience Bulletin ; (6): 1544-1560, 2023.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-1010633

RESUMEN

The secondary motor cortex (M2) encodes choice-related information and plays an important role in cue-guided actions. M2 neurons innervate the dorsal striatum (DS), which also contributes to decision-making behavior, yet how M2 modulates signals in the DS to influence perceptual decision-making is unclear. Using mice performing a visual Go/No-Go task, we showed that inactivating M2 projections to the DS impaired performance by increasing the false alarm (FA) rate to the reward-irrelevant No-Go stimulus. The choice signal of M2 neurons correlated with behavioral performance, and the inactivation of M2 neurons projecting to the DS reduced the choice signal in the DS. By measuring and manipulating the responses of direct or indirect pathway striatal neurons defined by M2 inputs, we found that the indirect pathway neurons exhibited a shorter response latency to the No-Go stimulus, and inactivating their early responses increased the FA rate. These results demonstrate that the M2-to-DS pathway is crucial for suppressing inappropriate responses in perceptual decision behavior.


Asunto(s)
Ratones , Animales , Corteza Motora , Cuerpo Estriado/fisiología , Neostriado , Neuronas/fisiología , Tiempo de Reacción
5.
Front Mol Neurosci ; 15: 1014497, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36385759

RESUMEN

Gintonin, a glycolipid protein conjugated with lysophosphatidic acid (LPA), is a newly identified compound extracted from Korean ginseng. LPA receptor isotypes exhibit high affinity for gintonin and mediate intracellular calcium signaling in various animal cell models. In this study, we found that gintonin induced the activation of Akt and cAMP-response element binding protein (CREB) in mouse striatal neurons, and chronic treatment with gintonin potently induced dendritic growth and filopodia formation. Gintonin-induced Akt/CREB activation and dendritic development were significantly impaired by LPA receptor (LPAR1/3) inhibition with Ki16425. Intriguingly, prolonged treatment with gintonin ameliorated the reduction in dendritic formation caused by Shank3 and Slitrk5 deficiency in the striatal neurons. In addition, gintonin and brain-derived neurotrophic factor (BDNF) had a synergistic effect on AKT/CREB activation and dendritic growth at suboptimal concentrations. These findings imply that gintonin-stimulated LPA receptors play a role in dendritic growth in striatal neurons and that they may act synergistically with BDNF, which is known to play a role in dendritogenesis.

6.
eNeuro ; 9(6)2022.
Artículo en Inglés | MEDLINE | ID: mdl-36446572

RESUMEN

Autoantibodies against central nervous system proteins are increasingly being recognized in association with neurologic disorders. Although a growing number of neural autoantibodies have been identified, a causal link between specific autoantibodies and disease symptoms remains unclear, as most studies use patient-derived CSF-containing mixtures of autoantibodies. This raises questions concerning mechanism of action and which autoantibodies truly contribute to disease progression. To address this issue, monoclonal autoantibodies were isolated from a young girl with a range of neurologic symptoms, some of which reacted with specific GABAA receptor (GABAAR) subunits, α1-subunit and α1γ2-subunit, which in this study we have characterized in detail using a combination of cellular imaging and electrophysiological techniques. These studies in neurons from wild-type mice (C57BL/6J; RRID:IMSR_JAX:000664) of mixed-sex revealed that the α1 and α1γ2 subunit-specific antibodies have differential effects on the GABAA receptor. Namely, the α1-antibody was found to directly affect GABAA receptor function on a short time scale that diminished GABA currents, leading to increased network excitability. On longer time scales those antibodies also triggered a redistribution of the GABAA receptor away from synapses. In contrast, the α1γ2-antibody had no direct effect on GABAA receptor function and could possibly mediate its effect through other actors of the immune system. Taken together, these data highlight the complexity underlying autoimmune disorders and show that antibodies can exert their effect through many mechanisms within the same disease.


Asunto(s)
Encefalitis , Receptores de GABA-A , Animales , Ratones , Receptores de GABA-A/metabolismo , Autoanticuerpos/metabolismo , Ratones Endogámicos C57BL , Ácido gamma-Aminobutírico
7.
Neurobiol Dis ; 174: 105878, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36183947

RESUMEN

The striatum receives abundant glutamatergic afferents from the cortex and thalamus. These inputs play a major role in the functions of the striatal neurons in normal conditions, and are significantly altered in pathological states, such as Parkinson's disease. This review summarizes the current knowledge of the connectivity of the corticostriatal and thalamostriatal pathways, with emphasis on the most recent advances in the field. We also discuss novel findings regarding structural changes in cortico- and thalamostriatal connections that occur in these connections as a consequence of striatal loss of dopamine in parkinsonism.


Asunto(s)
Enfermedad de Parkinson , Tálamo , Humanos , Tálamo/patología , Cuerpo Estriado/patología , Corteza Cerebral/patología , Neuronas/patología , Enfermedad de Parkinson/patología , Vías Nerviosas/patología
8.
Elife ; 112022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-35467530

RESUMEN

Dopamine is a key catecholamine in the brain and kidney, where it is involved in a number of physiological functions such as locomotion, cognition, emotion, endocrine regulation, and renal function. As a membrane-impermeant hormone and neurotransmitter, dopamine is thought to signal by binding and activating dopamine receptors, members of the G protein coupled receptor (GPCR) family, only on the plasma membrane. Here, using novel nanobody-based biosensors, we demonstrate for the first time that the dopamine D1 receptor (D1DR), the primary mediator of dopaminergic signaling in the brain and kidney, not only functions on the plasma membrane but becomes activated at the Golgi apparatus in the presence of its ligand. We present evidence that activation of the Golgi pool of D1DR is dependent on organic cation transporter 2 (OCT2), a dopamine transporter, providing an explanation for how the membrane-impermeant dopamine accesses subcellular pools of D1DR. We further demonstrate that dopamine activates Golgi-D1DR in murine striatal medium spiny neurons, and this activity depends on OCT2 function. We also introduce a new approach to selectively interrogate compartmentalized D1DR signaling by inhibiting Gαs coupling using a nanobody-based chemical recruitment system. Using this strategy, we show that Golgi-localized D1DRs regulate cAMP production and mediate local protein kinase A activation. Together, our data suggest that spatially compartmentalized signaling hubs are previously unappreciated regulatory aspects of D1DR signaling. Our data provide further evidence for the role of transporters in regulating subcellular GPCR activity.


Asunto(s)
Aparato de Golgi , Transportador 2 de Cátion Orgánico , Receptores de Dopamina D1 , Animales , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Aparato de Golgi/metabolismo , Ratones , Transportador 2 de Cátion Orgánico/metabolismo , Receptores de Dopamina D1/metabolismo
9.
J Pharmacol Sci ; 147(4): 367-375, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34663519

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by CAG repeat expansion in the huntingtin (HTT) gene. Here, we examined the effects of antioxidants on 3-nitropropionic acid (3-NP; a mitochondrial complex II inhibitor)-induced mitochondrial dysfunction and cell death in STHdhQ111 striatal cells carrying homozygous mutant HTT with extended CAG repeats compared with those in STHdhQ7 striatal cells. 3-NP reduced cell viability and increased cell death both in STHdhQ111 and STHdhQ7, and the cytotoxicity was markedly attenuated by antioxidants (N-acetyl-l-cysteine and edaravone). Furthermore, 3-NP increased intracellular reactive oxygen species (ROS) production in both cell lines, and this increase was inhibited by antioxidants. Mitochondrial ROS was also increased by 3-NP in STHdhQ111 but not in STHdhQ7, and this increase was significantly inhibited by edaravone. Mitochondrial membrane potential (MMP) was lower in STHdhQ111 than that in STHdhQ7, and antioxidants prevented 3-NP-induced MMP decrease in STHdhQ111.3-NP enhanced oligomerization of dynamin-related protein 1 (Drp1), a protein that promotes mitochondrial fission in both cells, and both antioxidants prevented the increase in oligomerization. These results suggest that reduced mitochondrial complex II activity enhances cell death via intracellular ROS production and Drp1 oligomerization in striatal cells with mutant HTT and antioxidants may reduce striatal cell death.


Asunto(s)
Antioxidantes/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Complejo II de Transporte de Electrones/metabolismo , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación/genética , Especies Reactivas de Oxígeno/metabolismo , Expansión de Repetición de Trinucleótido/genética , Animales , Edaravona/farmacología , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Transgénicos , Dinámicas Mitocondriales/efectos de los fármacos , Nitrocompuestos/efectos adversos , Nitrocompuestos/antagonistas & inhibidores , Propionatos/efectos adversos , Propionatos/antagonistas & inhibidores
10.
Front Cell Neurosci ; 15: 742763, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34658796

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene (HTT). Disease progression is characterized by the loss of vulnerable neuronal populations within the striatum. A consistent phenotype across HD models is disruption of nucleocytoplasmic transport and nuclear pore complex (NPC) function. Here we demonstrate that high content imaging is a suitable method for detecting mislocalization of lamin-B1, RAN and RANGAP1 in striatal neuronal cultures thus allowing a robust, unbiased, highly powered approach to assay nuclear pore deficits. Furthermore, nuclear pore deficits extended to the selectively vulnerable DARPP32 + subpopulation neurons, but not to astrocytes. Striatal neuron cultures are further affected by changes in gene and protein expression of RAN, RANGAP1 and lamin-B1. Lowering total HTT using HTT-targeted anti-sense oligonucleotides partially restored gene expression, as well as subtly reducing mislocalization of proteins involved in nucleocytoplasmic transport. This suggests that mislocalization of RAN, RANGAP1 and lamin-B1 cannot be normalized by simply reducing expression of CAG-expanded HTT in the absence of healthy HTT protein.

11.
Stem Cell Res ; 55: 102486, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34385043

RESUMEN

Human induced pluripotent stem cells (hiPSCs) are powerful tools for modeling human brain development and treating neurodegenerative diseases. Here we established a robust protocol with high scalability for generating striatal medium spiny neurons (MSNs) from hiPSCs using small molecules under two- and three-dimensional culture conditions. Using this protocol, GSH2+ lateral ganglionic eminence (LGE) progenitors were generated in two-dimensional culture by Sonic hedgehog signaling activation using purmorphamine, WNT signaling inhibition using XAV939, and dual-SMAD inhibition using LDN193189 and A83-01. Quantitative PCR analysis revealed sequential expression of LGE and striatal genes during differentiation. These LGE progenitors subsequently gave rise to DARPP32+ MSNs exhibiting spontaneous and evoked monophasic spiking activity. Applying this protocol in three-dimensional culture, we generated striatal neurospheres with gene expression profiles and cell layer organization resembling that of the developing striatum, including distinct ventricular and subventricular zones and DARPP32+ neurons at the surface. This protocol provides a useful experimental model for studying striatal development and yields cells potentially applicable for regenerative medicine to treat striatum-related disorders such as Huntington's disease.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular , Cuerpo Estriado/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Proteínas Hedgehog/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo
12.
Stem Cells ; 39(10): 1410-1422, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34028139

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by the progressive loss of striatal medium spiny neurons. Using a highly efficient protocol for direct reprogramming of adult human fibroblasts with chemically modified mRNA, we report the first generation of HD induced neural precursor cells (iNPs) expressing striatal lineage markers that differentiated into DARPP32+ neurons from individuals with adult-onset HD (41-57 CAG). While no transcriptional differences between normal and HD reprogrammed neurons were detected by NanoString nCounter analysis, a subpopulation of HD reprogrammed neurons contained ubiquitinated polyglutamine aggregates. Importantly, reprogrammed HD neurons exhibited impaired neuronal maturation, displaying altered neurite morphology and more depolarized resting membrane potentials. Reduced BDNF protein expression in reprogrammed HD neurons correlated with increased CAG repeat lengths and earlier symptom onset. This model represents a platform for investigating impaired neuronal maturation and screening for neuronal maturation modifiers to treat HD.


Asunto(s)
Enfermedad de Huntington , Células-Madre Neurales , Cuerpo Estriado , Humanos , Enfermedad de Huntington/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/metabolismo
13.
Int J Mol Sci ; 22(5)2021 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-33803075

RESUMEN

Methamphetamine is, worldwide, one of the most consumed drugs of abuse. One important side effect is neurodegeneration leading to a decrease in life expectancy. The aim of this paper was to check whether the drug affects one of the receptors involved in neurodegeneration/neuroprotection events, namely the adenosine A2A receptor (A2AR). First, we noticed that methamphetamine does not affect A2A functionality if the receptor is expressed in a heterologous system. However, A2AR becomes sensitive to the drug upon complexes formation with the cannabinoid CB1 receptor (CB1R) and the sigma 1 receptor (σ1R). Signaling via both adenosine A2AR and cannabinoid CB1R was affected by methamphetamine in cells co-expressing the two receptors. In striatal primary cultures, the A2AR-CB1R heteromer complex was detected and methamphetamine not only altered its expression but completely blocked the A2AR- and the CB1R-mediated activation of the mitogen activated protein kinase (MAPK) pathway. In conclusion, methamphetamine, with the participation of σ1R, alters the expression and function of two interacting receptors, A2AR, which is a therapeutic target for neuroprotection, and CB1R, which is the most abundant G protein-coupled receptor (GPCR) in the brain.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Cuerpo Estriado/metabolismo , Metanfetamina/farmacología , Neuronas/metabolismo , Receptor de Adenosina A2A/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptores sigma/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Receptor Sigma-1
14.
J Neuroinflammation ; 18(1): 94, 2021 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-33874957

RESUMEN

BACKGROUND: Neuroinflammation may contribute to the pathogenesis of Huntington's disease, given evidence of activated microglia and elevated levels of inflammatory molecules in disease gene carriers, even those many years from symptom onset. We have shown previously that monocytes from Huntington's disease patients are hyper-reactive to stimulation in a manner dependent on their autonomous expression of the disease-causing mutant HTT protein. To date, however, whether human microglia are similarly hyper-responsive in a cell-autonomous manner has not been determined. METHODS: Microglial-like cells were derived from human pluripotent stem cells (PSCs) expressing mutant HTT containing varying polyglutamine lengths. These included lines that are otherwise isogenic, such that any observed differences can be attributed with certainty to the disease mutation itself. Analyses by quantitative PCR and immunofluorescence microscopy respectively of key genes and protein markers were undertaken to determine whether Huntington's disease PSCs differentiated normally to a microglial fate. The resultant cultures and their supernatants were then assessed by various biochemical assays and multiplex ELISAs for viability and responses to stimulation, including the release of pro-inflammatory cytokines and reactive oxygen species. Conditioned media were applied to PSC-derived striatal neurons, and vice versa, to determine the effects that the secretomes of each cell type might have on the other. RESULTS: Human PSCs generated microglia successfully irrespective of the expression of mutant HTT. These cells, however, were hyper-reactive to stimulation in the production of pro-inflammatory cytokines such as IL-6 and TNFα. They also released elevated levels of reactive oxygen species that have neurotoxic potential. Accompanying such phenotypes, human Huntington's disease PSC-derived microglia showed increased levels of apoptosis and were more susceptible to exogenous stress. Such stress appeared to be induced by supernatants from human PSC-derived striatal neurons expressing mutant HTT with a long polyglutamine tract. CONCLUSIONS: These studies show, for the first time, that human Huntington's disease PSC-derived microglia are hyper-reactive due to their autonomous expression of mutant HTT. This provides a cellular basis for the contribution that neuroinflammation might make to Huntington's disease pathogenesis.


Asunto(s)
Enfermedad de Huntington , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Microglía/metabolismo , Microglía/patología , Especies Reactivas de Oxígeno/metabolismo , Diferenciación Celular , Línea Celular , Cuerpo Estriado/metabolismo , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Mutación , Neuronas/metabolismo
15.
Neuroscience ; 458: 153-165, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33428968

RESUMEN

Differences in the intrinsic properties of intralaminar thalamo-striatal neurons such as expressing low-threshold-spikes (LTS) or after hyperpolarizing potentials (AHPs) of different duration have been attributed to different maturation stages. However, two morphological types: "diffuse" and "bushy" have been described. Therefore, we explored whether electrophysiological differences persist in adult mice using whole cell recordings. Some recorded neurons were identified by intracellular labeling with biocytin and double labeling with retrograde or anterograde tracings using Cre-mice. We classified these neurons by their AHPs during spontaneous firing. Neurons with long duration AHPs, with fast and slow components, were mostly found in the parafascicular (Pf) nucleus. Neurons with brief AHPs were mainly found in the central lateral (CL) nucleus. However, neurons with both AHPs were found in both nuclei in different proportions. Firing frequency adaptation differed between these neuron classes: those with prolonged AHPs exhibited firing frequency adaptation with fast and slow time constants whereas those with brief AHPs were slow adapters. Neurons with more prolonged AHPs had significant higher input resistances than neurons with brief AHPs. Both cell classes could fire in two modes: trains of single action potentials at depolarized potentials or high frequency bursts on top of LTS at more hyperpolarized potentials. LTS were probably generated by Cav3 calcium channels since they were blocked by the selective antagonist TTA-P2. About 11% of neurons with brief AHPs and 55% of neurons with prolonged AHPs do not show LTS and bursts, even when potassium currents are blocked.


Asunto(s)
Cuerpo Estriado , Neuronas , Potenciales de Acción , Animales , Canales de Calcio , Ratones
16.
J Neuroinflammation ; 17(1): 290, 2020 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-33023623

RESUMEN

BACKGROUND: Evidence shows significant heterogeneity in astrocyte gene expression and function. We previously demonstrated that brain-derived neurotrophic factor (BDNF) exerts protective effects on whole brain primary cultured rat astrocytes treated with 3-nitropropionic acid (3NP), a mitochondrial toxin widely used as an in vitro model of Huntington's disease (HD). Therefore, we now investigated 3NP and BDNF effects on astrocytes from two areas involved in HD: the striatum and the entire cortex, and their involvement in neuron survival. METHODS: We prepared primary cultured rat cortical or striatal astrocytes and treated them with BDNF and/or 3NP for 24 h. In these cells, we assessed expression of astrocyte markers, BDNF receptor, and glutamate transporters, and cytokine release. We prepared astrocyte-conditioned medium (ACM) from cortical and striatal astrocytes and tested its effect on a cellular model of HD. RESULTS: BDNF protected astrocytes from 3NP-induced death, increased expression of its own receptor, and activation of ERK in both cortical and striatal astrocytes. However, BDNF modulated glutamate transporter expression differently by increasing GLT1 and GLAST expression in cortical astrocytes but only GLT1 expression in striatal astrocytes. Striatal astrocytes released higher amounts of tumor necrosis factor-α than cortical astrocytes in response to 3NP but BDNF decreased this effect in both populations. 3NP decreased transforming growth factor-ß release only in cortical astrocytes, whereas BDNF treatment increased its release only in striatal astrocytes. Finally, we evaluated ACM effect on a cellular model of HD: the rat striatal neuron cell line ST14A expressing mutant human huntingtin (Q120) or in ST14A cells expressing normal human huntingtin (Q15). Neither striatal nor cortical ACM modified the viability of Q15 cells. Only ACM from striatal astrocytes treated with BDNF and ACM from 3NP + BDNF-treated striatal astrocytes protected Q120 cells, whereas ACM from cortical astrocytes did not. CONCLUSIONS: Data suggest that cortical and striatal astrocytes respond differently to mitochondrial toxin 3NP and BDNF. Moreover, striatal astrocytes secrete soluble neuroprotective factors in response to BDNF that selectively protect neurons expressing mutant huntingtin implicating that BDNF modulation of striatal astrocyte function has therapeutic potential against neurodegeneration.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/toxicidad , Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Proteína Huntingtina/biosíntesis , Nitrocompuestos/toxicidad , Propionatos/toxicidad , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Femenino , Expresión Génica , Humanos , Proteína Huntingtina/genética , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación/efectos de los fármacos , Mutación/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuroprotección/efectos de los fármacos , Neuroprotección/fisiología , Ratas , Ratas Wistar
17.
Curr Pharm Des ; 26(37): 4738-4746, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32065086

RESUMEN

Parkinson's disease is a progressive neurodegenerative disorder of dopaminergic striatal neurons in basal ganglia. Treatment of Parkinson's disease (PD) through dopamine replacement strategies may provide improvement in early stages and this treatment response is related to dopaminergic neuronal mass which decreases in advanced stages. This treatment failure was revealed by many studies and levodopa treatment became ineffective or toxic in chronic stages of PD. Early diagnosis and neuroprotective agents may be a suitable approach for the treatment of PD. The essentials required for early diagnosis are biomarkers. Characterising the striatal neurons, understanding the status of dopaminergic pathways in different PD stages may reveal the effects of the drugs used in the treatment. This review updates on characterisation of striatal neurons, electrophysiology of dopaminergic pathways in PD, biomarkers of PD, approaches for success of neuroprotective agents in clinical trials. The literature was collected from the articles in database of PubMed, MedLine and other available literature resources.


Asunto(s)
Fármacos Neuroprotectores , Enfermedad de Parkinson , Neuronas Dopaminérgicas , Humanos , Levodopa , Neuroprotección , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico
18.
Bio Protoc ; 8(8): e2823, 2018 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-34286033

RESUMEN

Primary cultures of murine striatal neurons are widely used to explore cellular mechanisms in neurobiology, including brain diseases. Here we describe a detailed and standardized protocol to dissect and culture embryonic murine striatal neurons GABA-positive/DARPP-32-positive for 12 days in vitro, when they show good neuronal cell connectivity and the presence of dendritic spines, which reflects the maturation of the network.

19.
Mol Neurodegener ; 12(1): 65, 2017 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-28923065

RESUMEN

Protein inclusions are a predominant molecular pathology found in numerous neurodegenerative diseases, including amyotrophic lateral sclerosis and Huntington's disease. Protein inclusions form in discrete areas of the brain characteristic to the type of neurodegenerative disease, and coincide with the death of neurons in that region (e.g. spinal cord motor neurons in amyotrophic lateral sclerosis). This suggests that the process of protein misfolding leading to inclusion formation is neurotoxic, and that cell-autonomous and non-cell autonomous mechanisms that maintain protein homeostasis (proteostasis) can, at times, be insufficient to prevent protein inclusion formation in the central nervous system. The heat shock response is a pro-survival pathway induced under conditions of cellular stress that acts to maintain proteostasis through the up-regulation of heat shock proteins, a superfamily of molecular chaperones, other co-chaperones and mitotic regulators. The kinetics and magnitude of the heat shock response varies in a stress- and cell-type dependent manner. It remains to be determined if and/or how the heat shock response is activated in the different cell-types that comprise the central nervous system (e.g. neurons and astroglia) in response to protein misfolding events that precede cellular dysfunctions in neurodegenerative diseases. This is particularly relevant considering emerging evidence demonstrating the non-cell autonomous nature of amyotrophic lateral sclerosis and Huntington's disease (and other neurodegenerative diseases) and the destructive role of astroglia in disease progression. This review highlights the complexity of heat shock response activation and addresses whether neurons and glia sense and respond to protein misfolding and aggregation associated with neurodegenerative diseases, in particular Huntington's disease and amyotrophic lateral sclerosis, by inducing a pro-survival heat shock response.


Asunto(s)
Astrocitos/metabolismo , Respuesta al Choque Térmico/fisiología , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Animales , Astrocitos/patología , Humanos , Enfermedades Neurodegenerativas/patología , Neuronas/patología
20.
J Cereb Blood Flow Metab ; 37(3): 837-847, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27055780

RESUMEN

Striatal neurons regulate the activity of neural progenitor cells in the subventricular zone, but the effect of striatal neuronal activity on neurogenesis after ischemic stroke is unclear. In this study, we used optogenetic tools to investigate the impact of striatal neuronal activity on the neurogenesis and functional recovery after cerebral ischemia. We transfected striatal neurons with channelrhodopsin-2 or halorhodopsin from Natronomonas so that they can be excited by 473 nm laser or inhibited by 594 nm laser, respectively. Neural inhibition but not excitation at 4-7 days after middle cerebral artery occlusion resulted in reduced atrophy volume (6.8 ± 0.7 vs 8.5 ± 1.2 mm3, p < 0.05) and better performance represented by longer sustaining time on rotarod (99.3 ± 9 vs 80.1 ± 11 s, p < 0.01) and faster moving speed (7.7 ± 2 vs 5.7 ± 1.1 cm/s, p < 0.05) in open field tests. Furthermore, neural inhibition increased the number of nestin+, BrdU+/doublecortin+ and BrdU+/NeuN+ cells ( p < 0.001) in the subventricular zone and peri-focal region, and the expression level of axon guidance factor Netrin-1 (0.39 ± 0.16 vs 0.16 ± 0.02, p < 0.05) in the peri-focal region. These data suggest that striatal neuronal activity plays an important role in regulating neurogenesis and neural-behavioral outcomes, and that inhibiting striatal neurons by optogenetics promotes the recovery after ischemic stroke in mice.


Asunto(s)
Isquemia Encefálica/terapia , Neostriado/patología , Neurogénesis , Neuronas/fisiología , Animales , Channelrhodopsins , Halorrodopsinas/genética , Halorrodopsinas/efectos de la radiación , Infarto de la Arteria Cerebral Media , Ratones , Neuronas/efectos de la radiación , Recuperación de la Función , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA