Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Neurobiol Dis ; 190: 106367, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38042508

RESUMO

X-linked dystonia-parkinsonism (XDP) is a rare neurodegenerative disease endemic to the Philippines. The genetic cause for XDP is an insertion of a SINE-VNTR-Alu (SVA)-type retrotransposon within intron 32 of TATA-binding protein associated factor 1 (TAF1) that causes an alteration of TAF1 splicing, partial intron retention, and decreased transcription. Although TAF1 is expressed in all organs, medium spiny neurons (MSNs) within the striatum are one of the cell types most affected in XDP. To define how mutations in the TAF1 gene lead to MSN vulnerability, we carried out a proteomic analysis of human XDP patient-derived neural stem cells (NSCs) and MSNs derived from induced pluripotent stem cells. NSCs and MSNs were grown in parallel and subjected to quantitative proteomic analysis in data-independent acquisition mode on the Orbitrap Eclipse Tribrid mass spectrometer. Subsequent functional enrichment analysis demonstrated that neurodegenerative disease-related pathways, such as Huntington's disease, spinocerebellar ataxia, cellular senescence, mitochondrial function and RNA binding metabolism, were highly represented. We used weighted coexpression network analysis (WGCNA) of the NSC and MSN proteomic data set to uncover disease-driving network modules. Three of the modules significantly correlated with XDP genotype when compared to the non-affected control and were enriched for DNA helicase and nuclear chromatin assembly, mitochondrial disassembly, RNA location and mRNA processing. Consistent with aberrant mRNA processing, we found splicing and intron retention of TAF1 intron 32 in XDP MSN. We also identified TAF1 as one of the top enriched transcription factors, along with YY1, ATF2, USF1 and MYC. Notably, YY1 has been implicated in genetic forms of dystonia. Overall, our proteomic data set constitutes a valuable resource to understand mechanisms relevant to TAF1 dysregulation and to identify new therapeutic targets for XDP.


Assuntos
Distonia , Distúrbios Distônicos , Doenças Neurodegenerativas , Transtornos Parkinsonianos , Humanos , Distonia/genética , Distonia/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteômica , Fator de Transcrição TFIID/genética , Distúrbios Distônicos/genética , Distúrbios Distônicos/metabolismo , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo
2.
J Neurochem ; 166(1): 87-106, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37328918

RESUMO

Ischemic stroke is a leading cause of disability worldwide. There is no simple treatment to alleviate ischemic brain injury, as thrombolytic therapy is applicable within a narrow time window. During the last years, the ketogenic diet (KD) and the exogenous administration of the ketone body ß-hydroxybutyrate (BHB) have been proposed as therapeutic tools for acute neurological disorders and both can reduce ischemic brain injury. However, the mechanisms involved are not completely clear. We have previously shown that the D enantiomer of BHB stimulates the autophagic flux in cultured neurons exposed to glucose deprivation (GD) and in the brain of hypoglycemic rats. Here, we have investigated the effect of the systemic administration of D-BHB, followed by its continuous infusion after middle cerebral artery occlusion (MCAO), on the autophagy-lysosomal pathway and the activation of the unfolded protein response (UPR). Results show for the first time that the protective effect of BHB against MCAO injury is enantiomer selective as only D-BHB, the physiologic enantiomer of BHB, significantly reduced brain injury. D-BHB treatment prevented the cleavage of the lysosomal membrane protein LAMP2 and stimulated the autophagic flux in the ischemic core and the penumbra. In addition, D-BHB notably reduced the activation of the PERK/eIF2α/ATF4 pathway of the UPR and inhibited IRE1α phosphorylation. L-BHB showed no significant effect relative to ischemic animals. In cortical cultures under GD, D-BHB prevented LAMP2 cleavage and decreased lysosomal number. It also abated the activation of the PERK/eIF2α/ATF4 pathway, partially sustained protein synthesis, and reduced pIRE1α. In contrast, L-BHB showed no significant effects. Results suggest that protection elicited by D-BHB treatment post-ischemia prevents lysosomal rupture allowing functional autophagy, preventing the loss of proteostasis and UPR activation.


Assuntos
Lesões Encefálicas , Acidente Vascular Cerebral , Ratos , Animais , Corpos Cetônicos/farmacologia , Corpos Cetônicos/metabolismo , Endorribonucleases/farmacologia , Proteínas Serina-Treonina Quinases , Estresse do Retículo Endoplasmático , Ácido 3-Hidroxibutírico/metabolismo , Ácido 3-Hidroxibutírico/farmacologia , Glucose/metabolismo , Autofagia , Infarto da Artéria Cerebral Média , Modelos Teóricos , Acidente Vascular Cerebral/tratamento farmacológico
3.
Neurochem Res ; 41(3): 600-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26303508

RESUMO

Glucose is the major energy substrate in brain, however, during ketogenesis induced by starvation or prolonged hypoglycemia, the ketone bodies (KB), acetoacetate and ß-hydroxybutyrate (BHB) can substitute for glucose. KB improve neuronal survival in diverse injury models, but the mechanisms by which KB prevent neuronal damage are still not well understood. In the present study we have investigated whether protection by the D isomer of BHB (D-BHB) against neuronal death induced by glucose deprivation (GD), is related to autophagy. Autophagy is a lysosomal-dependent degradation process activated during nutritional stress, which leads to the digestion of damaged proteins and organelles providing energy for cell survival. Results show that autophagy is activated in cortical cultured neurons during GD, as indicated by the increase in the levels of the lipidated form of the microtubule associated protein light chain 3 (LC3-II), and the number of autophagic vesicles. At early phases of glucose reintroduction (GR), the levels of p62 declined suggesting that the degradation of the autophagolysosomal content takes place at this time. In cultures exposed to GD and GR in the presence of D-BHB, the levels of LC3-II and p62 rapidly declined and remained low during GR, suggesting that the KB stimulates the autophagic flux preventing autophagosome accumulation and improving neuronal survival.


Assuntos
Ácido 3-Hidroxibutírico/metabolismo , Autofagia , Córtex Cerebral/citologia , Glucose/metabolismo , Neurônios/citologia , Ácido 3-Hidroxibutírico/química , Ácido 3-Hidroxibutírico/farmacologia , Animais , Sobrevivência Celular , Células Cultivadas , Córtex Cerebral/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos Wistar , Proteína Sequestossoma-1 , Estereoisomerismo
4.
EMBO Mol Med ; 15(12): e17907, 2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-37860842

RESUMO

Following peripheral nerve injury, successful axonal growth and functional recovery require Schwann cell (SC) reprogramming into a reparative phenotype, a process dependent upon c-Jun transcription factor activation. Unfortunately, axonal regeneration is greatly impaired in aged organisms and following chronic denervation, which can lead to poor clinical outcomes. While diminished c-Jun expression in SCs has been associated with regenerative failure, it is unclear whether the inability to maintain a repair state is associated with the transition into an axonal growth inhibition phenotype. We here find that reparative SCs transition into a senescent phenotype, characterized by diminished c-Jun expression and secretion of inhibitory factors for axonal regeneration in aging and chronic denervation. In both conditions, the elimination of senescent SCs by systemic senolytic drug treatment or genetic targeting improved nerve regeneration and functional recovery, increased c-Jun expression and decreased nerve inflammation. This work provides the first characterization of senescent SCs and their influence on axonal regeneration in aging and chronic denervation, opening new avenues for enhancing regeneration and functional recovery after peripheral nerve injuries.


Assuntos
Traumatismos dos Nervos Periféricos , Humanos , Idoso , Traumatismos dos Nervos Periféricos/terapia , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/metabolismo , Células de Schwann/metabolismo , Envelhecimento , Regulação da Expressão Gênica , Denervação
5.
Cell Death Discov ; 7(1): 131, 2021 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-34083523

RESUMO

Altered protein homeostasis is associated with neurodegenerative diseases and acute brain injury induced under energy depletion conditions such as ischemia. The accumulation of damaged or unfolded proteins triggers the unfolded protein response (UPR), which can act as a homeostatic response or lead to cell death. However, the factors involved in turning and adaptive response into a cell death mechanism are still not well understood. Several mechanisms leading to brain injury induced by severe hypoglycemia have been described but the contribution of the UPR has been poorly studied. Cell responses triggered during both the hypoglycemia and the glucose reinfusion periods can contribute to neuronal death. Therefore, we have investigated the activation dynamics of the PERK and the IRE1α branches of the UPR and their contribution to neuronal death in a model of glucose deprivation (GD) and glucose reintroduction (GR) in cortical neurons. Results show a rapid activation of the PERK/p-eIF2α/ATF4 pathway leading to protein synthesis inhibition during GD, which contributes to neuronal adaptation, however, sustained blockade of protein synthesis during GR promotes neuronal death. On the other hand, IRE1α activation occurs early during GD due to its interaction with BAK/BAX, while ASK1 is recruited to IRE1α activation complex during GR promoting the nuclear translocation of JNK and the upregulation of Chop. Most importantly, results show that IRE1α RNase activity towards its splicing target Xbp1 mRNA occurs late after GR, precluding a homeostatic role. Instead, IRE1α activity during GR drives neuronal death by positively regulating ASK1/JNK activity through the degradation of 14-3-3 θ mRNA, a negative regulator of ASK and an adaptor protein highly expressed in brain, implicated in neuroprotection. Collectively, results describe a novel regulatory mechanism of cell death in neurons, triggered by the downregulation of 14-3-3 θ mRNA induced by the IRE1α branch of the UPR.

6.
Oxid Med Cell Longev ; 2021: 6682336, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34434486

RESUMO

Brain aging is characterized by dysfunctional autophagy and cellular senescence, among other features. While autophagy can either promote or suppress cellular senescence in proliferating cells, in postmitotic cells, such as neurons, autophagy impairment promotes cellular senescence. CRM1 (exportin-1/XPO1) exports hundreds of nuclear proteins into the cytoplasm, including the transcription factors TFEB (the main inducer of autophagy and lysosomal biogenesis genes) and STAT3, another autophagy modulator. It appears that CRM1 is a modulator of aging-associated senescence and autophagy, because pharmacological inhibition of CRM1 improved autophagic degradation in flies, by increasing nuclear TFEB levels, and because enhanced CRM1 activity is mechanistically linked to senescence in fibroblasts from Hutchinson-Gilford progeria syndrome patients and old healthy individuals; furthermore, the exogenous overexpression of CRM1 induced senescence in normal fibroblasts. In this work, we tested the hypothesis that impaired autophagic flux during brain aging occurs due to CRM1 accumulation in the brain. We found that CRM1 levels and activity increased in the hippocampus and cortex during physiological aging, which resulted in a decrease of nuclear TFEB and STAT3. Consistent with an autophagic flux impairment, we observed accumulation of the autophagic receptor p62/SQSTM1 in neurons of old mice, which correlated with increased neuronal senescence. Using an in vitro model of neuronal senescence, we demonstrate that CRM1 inhibition improved autophagy flux and reduced SA-ß-gal activity by restoring TFEB nuclear localization. Collectively, our data suggest that enhanced CRM1-mediated export of proteins during brain aging perturbs neuronal homeostasis, contributing to autophagy impairment, and neuronal senescence.


Assuntos
Envelhecimento/metabolismo , Autofagia , Encéfalo/metabolismo , Senescência Celular , Carioferinas/metabolismo , Neurônios/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Envelhecimento/patologia , Animais , Encéfalo/patologia , Camundongos , Camundongos Transgênicos , Neurônios/patologia , Ratos , Ratos Wistar , Proteína Exportina 1
7.
Front Aging Neurosci ; 12: 581767, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33192476

RESUMO

Alzheimer's disease (AD) represents the most common age-related neurodegenerative disorder, affecting around 35 million people worldwide. Despite enormous efforts dedicated to AD research over decades, there is still no cure for the disease. Misfolding and accumulation of Aß and tau proteins in the brain constitute a defining signature of AD neuropathology, and mounting evidence has documented a link between aggregation of these proteins and neuronal dysfunction. In this context, progressive axonal degeneration has been associated with early stages of AD and linked to Aß and tau accumulation. As the axonal degeneration mechanism has been starting to be unveiled, it constitutes a promising target for neuroprotection in AD. A comprehensive understanding of the mechanism of axonal destruction in neurodegenerative conditions is therefore critical for the development of new therapies aimed to prevent axonal loss before irreversible neuronal death occurs in AD. Here, we review current evidence of the involvement of Aß and tau pathologies in the activation of signaling cascades that can promote axonal demise.

8.
Mol Neurobiol ; 56(9): 6594-6608, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30905004

RESUMO

Autophagy is considered a major bulk degradation system that helps cells to counteract different intracellular and extracellular stress signals. Several protein complexes integrate multiple signals in order to activate autophagy, which sequesters damaged cellular components and carries them to lysosomes for degradation. This active mechanism is essential to maintain cell homeostasis and particularly in neurons to sustain their viability. Because of their polarized morphology, neurons face special challenges to recycle cellular components through autophagy in dendrites and distal regions of axons. Thus, autophagy is critical in the remodeling of pre- and post-synaptic constituents to sustain neuronal functionality. Under stress conditions, autophagy may play either a cytotoxic or a cytoprotective role. This discrepancy is partly due to the lack of a full characterization of the autophagic process and conclusive evidence to support whether basal autophagy is stimulated or impaired in a particular condition. Moreover, in many studies, only pharmacologic tools have been used to modulate autophagy. Throughout the present review, we go over the literature revealing autophagy induction in the nervous system under diverse stressful conditions, the signaling pathways involved, and its consequences for neuronal homeostasis and survival. We have focused on five particular stress conditions that alter neuronal homeostasis and can induce neuronal death including, starvation, oxidative stress, endoplasmic reticulum (ER) stress, proteotoxic stress, and aging.


Assuntos
Autofagia , Sistema Nervoso Central/patologia , Homeostase , Estresse Fisiológico , Envelhecimento/patologia , Animais , Humanos , Estresse Oxidativo
9.
Aging (Albany NY) ; 11(16): 6175-6198, 2019 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-31469660

RESUMO

Senescent cells accumulate in various tissues and organs with aging altering surrounding tissue due to an active secretome, and at least in mice their elimination extends healthy lifespan and ameliorates several chronic diseases. Whether all cell types senesce, including post-mitotic cells, has been poorly described mainly because cellular senescence was defined as a permanent cell cycle arrest. Nevertheless, neurons with features of senescence have been described in old rodent and human brains. In this study we characterized an in vitro model useful to study the molecular basis of senescence of primary rat cortical cells that recapitulates senescent features described in brain aging. We found that in long-term cultures, rat primary cortical neurons displayed features of cellular senescence before glial cells did, and developed a functional senescence-associated secretory phenotype able to induce paracrine premature senescence of mouse embryonic fibroblasts but proliferation of rat glial cells. Functional autophagy seems to prevent neuronal senescence, as we observed an autophagic flux reduction in senescent neurons both in vitro and in vivo, and autophagy impairment induced cortical cell senescence while autophagy stimulation inhibited it. Our findings suggest that aging-associated dysfunctional autophagy contributes to senescence transition also in neuronal cells.


Assuntos
Autofagia/fisiologia , Senescência Celular/fisiologia , Córtex Cerebral/citologia , Neurônios/fisiologia , Envelhecimento , Animais , Proliferação de Células , Sobrevivência Celular , Masculino , Ratos , Ratos Wistar
10.
Behav Brain Res ; 366: 108-117, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-30898683

RESUMO

Nitric oxide (NO) plays a leading role in learning and memory processes. Previously, we showed its ability to modify the deleterious effect of immunotoxin 192 IgG-saporin (192-IgG-SAP) in the cholinergic system. The aim of this study was to analyze the potential of a NO donor (molsidomine, MOLS) to prevent the recognition memory deficits resulting from the septal cholinergic denervation by 192 IgG-SAP in rats. Quantification of neuronal and endothelial nitric oxide synthase (nNOS and eNOS, respectively) expression was evaluated in striatum, prefrontal cortex, and hippocampus. In addition, a choline acetyltransferase immunohistochemical analysis was performed in medial septum and assessed the effect of MOLS treatment on the spatial working memory of rats through a recognition memory test. Results showed that 192-IgG-SAP reduced the immunoreactivity of cholinergic septal neurons (41%), compared with PBS-receiving control rats (p < 0.05). Treatment with MOLS alone failed to antagonize the septal neuron population loss but prevented the progressive abnormal morphological changes of neurons. Those animals exposed to 192-IgG-SAP immunotoxin exhibited a reduction of cortical nNOS expression against the control group, whereas expression was enhanced in the 192-IgG-SAP + MOLS group. The most relevant finding was the recovering of the discrimination index exhibited by the 192-IgG-SAP + MOLS group. When compared with the rats exposed to the 192-IgG-SAP immunotoxin, they reached values similar to those observed in the PBS group. Our results show that although MOLS failed to block the cholinergic neurons loss induced by 192-IgG-SAP, it avoided the neuronal damage progression.


Assuntos
Transtornos da Memória/tratamento farmacológico , Molsidomina/farmacologia , Reconhecimento Psicológico/efeitos dos fármacos , Acetilcolina/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Colina O-Acetiltransferase/metabolismo , Neurônios Colinérgicos/efeitos dos fármacos , Cognição/efeitos dos fármacos , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Hipocampo/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Molsidomina/metabolismo , Doadores de Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Ratos , Ratos Wistar , Saporinas/farmacologia , Percepção Visual/efeitos dos fármacos
11.
Cell Death Dis ; 8(6): e2911, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28661473

RESUMO

Autophagy is triggered during nutrient and energy deprivation in a variety of cells as a homeostatic response to metabolic stress. In the CNS, deficient autophagy has been implicated in neurodegenerative diseases and ischemic brain injury. However, its role in hypoglycemic damage is poorly understood and the dynamics of autophagy during the hypoglycemic and the glucose reperfusion periods, has not been fully described. In the present study, we analyzed the changes in the content of the autophagy proteins BECN1, LC3-II and p62/SQSTM1 by western blot, and autophagosome formation was followed through time-lapse experiments, during glucose deprivation (GD) and glucose reintroduction (GR) in cortical cultures. According to the results, autophagosome formation rapidly increased during GD, and was followed by an active autophagic flux early after glucose replenishment. However, cells progressively died during GR and autophagy inhibition reduced neuronal death. Neurons undergoing apoptosis during GR did not form autophagosomes, while those surviving up to late GR showed autophagosomes. Calpain activity strongly increased during GR and remained elevated during progressive neuronal death. Its activation led to the cleavage of LAMP2 resulting in lysosome membrane permeabilization (LMP) and release of cathepsin B to the cytosol. Calpain inhibition prevented LMP and increased the number of neurons containing lysosomes and autophagosomes increasing cell viability. Taken together, the present results suggest that calpain-mediated lysosome dysfunction during GR turns an adaptive autophagy response to energy stress into a defective autophagy pathway, which contributes to neuronal death. In these conditions, autophagy inhibition results in the improvement of cell survival.


Assuntos
Autofagia/genética , Calpaína/genética , Glucose/metabolismo , Doenças Neurodegenerativas/genética , Neurônios/metabolismo , Animais , Apoptose/genética , Autofagossomos/metabolismo , Autofagossomos/patologia , Calpaína/metabolismo , Sobrevivência Celular/genética , Glucose/efeitos adversos , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/genética , Lisossomos/genética , Lisossomos/metabolismo , Lisossomos/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/patologia , Ratos
12.
J Cereb Blood Flow Metab ; 35(5): 851-60, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25649993

RESUMO

Glucose is the main energy substrate in brain but in certain circumstances such as prolonged fasting and the suckling period alternative substrates can be used such as the ketone bodies (KB), beta-hydroxybutyrate (BHB), and acetoacetate. It has been shown that KB prevent neuronal death induced during energy limiting conditions and excitotoxicity. The protective effect of KB has been mainly attributed to the improvement of mitochondrial function. In the present study, we have investigated the protective effect of D-BHB against neuronal death induced by severe noncoma hypoglycemia in the rat in vivo and by glucose deprivation (GD) in cortical cultures. Results show that systemic administration of D-BHB reduces reactive oxygen species (ROS) production in distinct cortical areas and subregions of the hippocampus and efficiently prevents neuronal death in the cortex of hypoglycemic animals. In vitro results show that D-BHB stimulates ATP production and reduces ROS levels, while the nonphysiologic isomer of BHB, L-BHB, has no effect on energy production but reduces ROS levels. Data suggest that protection by BHB, not only results from its metabolic action but is also related to its capability to reduce ROS, rendering this KB as a suitable candidate for the treatment of ischemic and traumatic injury.


Assuntos
Ácido 3-Hidroxibutírico/farmacologia , Córtex Cerebral , Metabolismo Energético/efeitos dos fármacos , Hipoglicemia , Neurônios , Espécies Reativas de Oxigênio/metabolismo , Acetoacetatos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Glucose/metabolismo , Hipoglicemia/tratamento farmacológico , Hipoglicemia/metabolismo , Hipoglicemia/patologia , Corpos Cetônicos/metabolismo , Masculino , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Wistar
13.
Methods Mol Biol ; 1010: 177-200, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23754226

RESUMO

Huntington's disease (HD) is a hereditary neurodegenerative disorder, characterized by motor, psychiatric, and cognitive symptoms. The genetic defect responsible for the onset of the disease, expansion of CAG repeats in exon 1 of the gene that codes for huntingtin, has been unambiguously identified. The mechanisms by which the mutation causes the disease are not completely understood yet. However, defects in the energy metabolism of affected cells, which may cause oxidative damage, have been proposed as underlying molecular mechanisms that participate in the etiology of the disease. In this chapter, we describe biochemical methods that allow us to determine striatal oxidative damage in transgenic mice and in the quinolinic acid-induced excitotoxicity model in rat, and establish the status of protective cellular systems. The excitotoxic model is acute, easier and faster to perform than the transgenic model, and can within a short period provide valuable data to try new therapeutic strategies. The methods described in this chapter permit us to link the kynurenine pathway with the cascade of toxic and harmful reactions that cause the damage observed in HD. We consider that determining the mechanisms inducing oxidative damage in two different models of HD will allow the testing of drugs or other therapeutic strategies with antioxidant activities.


Assuntos
Antioxidantes/metabolismo , Doença de Huntington/metabolismo , Estresse Oxidativo , Animais , Comportamento Animal , DNA/genética , DNA/isolamento & purificação , Modelos Animais de Doenças , Genótipo , Glutationa/metabolismo , Humanos , Doença de Huntington/genética , Peroxidação de Lipídeos , Camundongos , Neostriado/metabolismo , Óxido Nítrico Sintase/metabolismo , Reação em Cadeia da Polimerase , Ratos , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA