Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 7295, 2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37957154

RESUMO

Mutations in SNCA, the gene encoding α-synuclein (αSyn), cause familial Parkinson's disease (PD) and aberrant αSyn is a key pathological hallmark of idiopathic PD. This α-synucleinopathy leads to mitochondrial dysfunction, which may drive dopaminergic neurodegeneration. PARKIN and PINK1, mutated in autosomal recessive PD, regulate the preferential autophagic clearance of dysfunctional mitochondria ("mitophagy") by inducing ubiquitylation of mitochondrial proteins, a process counteracted by deubiquitylation via USP30. Here we show that loss of USP30 in Usp30 knockout mice protects against behavioral deficits and leads to increased mitophagy, decreased phospho-S129 αSyn, and attenuation of SN dopaminergic neuronal loss induced by αSyn. These observations were recapitulated with a potent, selective, brain-penetrant USP30 inhibitor, MTX115325, with good drug-like properties. These data strongly support further study of USP30 inhibition as a potential disease-modifying therapy for PD.


Assuntos
Doença de Parkinson , Tioléster Hidrolases , Animais , Camundongos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Neurônios Dopaminérgicos/metabolismo , Camundongos Knockout , Mitocôndrias/metabolismo , Doença de Parkinson/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Tioléster Hidrolases/genética
2.
Res Sq ; 2023 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-37886523

RESUMO

Background: In the present study we assessed the protective effects of a pharmacological approach to stabilize the retromer complex in a PD mouse model. Missense mutations in the VPS35 gene are a rare cause of familial PD. The VPS35 protein is a subunit of the retromer cargo recognition complex and has a variety of functions within neurons, many of which are potentially relevant for the pathophysiology of PD. Prior studies have revealed a role for the retromer complex in controlling accumulation and clearance of α-synuclein aggregates. We previously identified an aminoguanidine hydrazone, 1,3 phenyl bis guanylhydrazone (compound 2a), as a pharmacological stabilizer of the retromer complex that increases retromer subunit protein levels and function. Methods: Here, we validate the efficacy of 2a in protecting against αSynuclein pathology and dopaminergic neuronal degeneration in a PD mouse model generated by unilateral injection of AAV-A53T-αSynuclein in the substantia nigra. Results: Daily intraperitoneal administration of 2a at 10 mg/Kg for 100 days led to robust protection against behavioral deficits, dopaminergic neuronal loss and loss of striatal dopaminergic fibers and striatal monoamines. Treatment with 2a activated αSynuclein degradation pathways in the SN and led to significant reductions in aggregates and pathological αSynuclein. Conclusion: These data suggest retromer stabilization as a promising therapeutic strategy for Parkinson's disease leading to neuroprotection of dopaminergic neurons and rescue in the accumulation of pathological and aggregates αSynuclein. We identified 2a compound as potential clinical drug candidate for future testing in Parkinson's disease patients.

3.
Ageing Res Rev ; 91: 102077, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37742785

RESUMO

Parkinson's Disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra (SN), leading to motor and non-motor symptoms. While the exact mechanisms remain complex and multifaceted, several molecular pathways have been implicated in PD pathology, including accumulation of misfolded proteins, impaired mitochondrial function, oxidative stress, inflammation, elevated iron levels, etc. Overall, PD's molecular mechanisms involve a complex interplay between genetic, environmental, and cellular factors that disrupt cellular homeostasis, and ultimately lead to the degeneration of dopaminergic neurons. Recently, emerging evidence highlights ferroptosis, an iron-dependent non-apoptotic cell death process, as a pivotal player in the advancement of PD. Notably, oligomeric α-synuclein (α-syn) generates reactive oxygen species (ROS) and lipid peroxides within cellular membranes, potentially triggering ferroptosis. The loss of dopamine, a hallmark of PD, could predispose neurons to ferroptotic vulnerability. This unique form of cell demise unveils fresh insights into PD pathogenesis, necessitating an exploration of the molecular intricacies connecting ferroptosis and PD progression. In this review, the molecular and regulatory mechanisms of ferroptosis and their connection with the pathological processes of PD have been systematically summarized. Furthermore, the features of ferroptosis in PD animal models and clinical trials targeting ferroptosis as a therapeutic approach in PD patients' management are scrutinized.


Assuntos
Ferroptose , Doença de Parkinson , Animais , Humanos , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Substância Negra/metabolismo , Estresse Oxidativo , Ferro/metabolismo , Neurônios Dopaminérgicos/metabolismo
6.
Neurobiol Dis ; 145: 105056, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32853677

RESUMO

Mutations in VPS35 (PARK17), a key molecule in the retromer complex, are a rare cause of autosomal dominant Parkinson's disease (PD), the second most common neurodegenerative disorder. VPS35 exerts crucial functions within the cell in terms of regulating endosomal trafficking. However new data suggest its relevance also in the regulation of mitochondrial dynamics and homeostasis. Herein, we review the crosstalk between VPS35 and the mitochondria, highlighting the potential relevance to PD pathogenesis. VPS35 is not only a critical player in pathways connected to α-synuclein accumulation and clearance, but also plays a key role in ensuring mitochondrial stability and function. The genetic links of VPS35 to PD and the involvement of VPS35 in different PD related pathological mechanisms highlight the potential for targeting VPS35 as a neuroprotective strategy for PD.


Assuntos
Mitocôndrias/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/fisiopatologia , Proteínas de Transporte Vesicular/metabolismo , Animais , Humanos , Doença de Parkinson/genética , Proteínas de Transporte Vesicular/genética
7.
Nat Commun ; 11(1): 3848, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32737286

RESUMO

Amyotrophic Lateral Sclerosis (ALS) is a fatal disease characterized by the degeneration of upper and lower motor neurons (MNs). We find a significant reduction of the retromer complex subunit VPS35 in iPSCs-derived MNs from ALS patients, in MNs from ALS post mortem explants and in MNs from SOD1G93A mice. Being the retromer involved in trafficking of hydrolases, a pathological hallmark in ALS, we design, synthesize and characterize an array of retromer stabilizers based on bis-guanylhydrazones connected by a 1,3-phenyl ring linker. We select compound 2a as a potent and bioavailable interactor of VPS35-VPS29. Indeed, while increasing retromer stability in ALS mice, compound 2a attenuates locomotion impairment and increases MNs survival. Moreover, compound 2a increases VPS35 in iPSCs-derived MNs and shows brain bioavailability. Our results clearly suggest the retromer as a valuable druggable target in ALS.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Hidrazonas/farmacologia , Neurônios Motores/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Proteínas de Transporte Vesicular/genética , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Diferenciação Celular , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Hidrazonas/síntese química , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/síntese química , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica , Relação Estrutura-Atividade , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Proteínas de Transporte Vesicular/metabolismo
8.
Front Neurol ; 10: 1272, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31920908

RESUMO

Several symptomatic treatments for Parkinson's disease (PD) are currently available. Still, the challenge today is to find a therapy that might reduce degeneration and slow down disease progression. The identification of pathogenic mutations in familial Parkinsonism (fPD) associated to the monogenic forms of PD provided pathophysiological insights and highlighted novel targets for therapeutic intervention. Mutations in the VPS35 gene have been associated with autosomal dominant fPD and a clinical phenotype indistinguishable from idiopathic PD. Although VPS35 mutations are relatively rare causes of PD, their study may help understanding specific cellular and molecular alterations that lead to accumulation α-synuclein in neurons of PD patients. Interacting with such mechanisms may provide innovative therapeutic approaches. We review here the evidence on the physiological role of VPS35 as a key intracellular trafficking protein controlling relevant neuronal functions. We further analyze VPS35 activity on α-synuclein degradation pathways that control the equilibrium between α-synuclein clearance and accumulation. Finally, we highlight the strategies for increasing VPS35 levels as a potential tool to treat PD.

9.
Brain ; 139(Pt 12): 3217-3236, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27679481

RESUMO

Abnormal accumulation and propagation of the neuronal protein α-synuclein has been hypothesized to underlie the pathogenesis of Parkinson's disease, dementia with Lewy bodies and multiple system atrophy. Here we report a de novo-developed compound (NPT100-18A) that reduces α-synuclein toxicity through a novel mechanism that involves displacing α-synuclein from the membrane. This compound interacts with a domain in the C-terminus of α-synuclein. The E83R mutation reduces the compound interaction with the 80-90 amino acid region of α-synuclein and prevents the effects of NPT100-18A. In vitro studies showed that NPT100-18A reduced the formation of wild-type α-synuclein oligomers in membranes, reduced the neuronal accumulation of α-synuclein, and decreased markers of cell toxicity. In vivo studies were conducted in three different α-synuclein transgenic rodent models. Treatment with NPT100-18A ameliorated motor deficits in mThy1 wild-type α-synuclein transgenic mice in a dose-dependent manner at two independent institutions. Neuropathological examination showed that NPT100-18A decreased the accumulation of proteinase K-resistant α-synuclein aggregates in the CNS and was accompanied by the normalization of neuronal and inflammatory markers. These results were confirmed in a mutant line of α-synuclein transgenic mice that is prone to generate oligomers. In vivo imaging studies of α-synuclein-GFP transgenic mice using two-photon microscopy showed that NPT100-18A reduced the cortical synaptic accumulation of α-synuclein within 1 h post-administration. Taken together, these studies support the notion that altering the interaction of α-synuclein with the membrane might be a feasible therapeutic approach for developing new disease-modifying treatments of Parkinson's disease and other synucleinopathies.


Assuntos
Antiparkinsonianos/farmacologia , Comportamento Animal/efeitos dos fármacos , Descoberta de Drogas , Doença de Parkinson/tratamento farmacológico , alfa-Sinucleína/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Transgênicos
10.
J Neurosci ; 35(5): 1921-38, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25653352

RESUMO

Antiretroviral therapy has increased the life span of HIV+ individuals; however, HIV-associated neurocognitive disorder (HAND) occurrence is increasing in aging HIV patients. Previous studies suggest HIV infection alters autophagy function in the aging CNS and HIV-1 proteins affect autophagy in monocyte-derived cells. Despite these findings, the mechanisms leading to dysregulated autophagy in the CNS remain unclear. Here we sought to determine how HIV Tat dysregulates autophagy in neurons. Tat caused a dose-dependent decrease in autophagosome markers, microtubule-associated protein-1 light chain ß II (LC3II), and sequestosome 1(SQSTM1), in a membrane-enriched fraction, suggesting Tat increases autophagic degradation. Bafilomycin A1 increased autophagosome number, LC3II, and SQSTM1 accumulation; Tat cotreatment diminished this effect. Tat had no effect when 3-methyladenine or knockdown of beclin 1 blocked early stages of autophagy. Tat increased numbers of LC3 puncta and resulted in the formation of abnormal autophagosomes in vitro. Likewise, in vivo studies in GFAP-Tat tg mice showed increased autophagosome accumulation in neurons, altered LC3II levels, and neurodegeneration. These effects were reversed by rapamycin treatment. Tat colocalized with autophagosome and lysosomal markers and enhanced the colocalization of autophagosome with lysosome markers. Furthermore, co-IP studies showed that Tat interacts with lysosomal-associated membrane protein 2A (LAMP2A) in vitro and in vivo, and LAMP2A overexpression reduces Tat-induced neurotoxicity. Hence, Tat protein may induce autophagosome and lysosome fusion through interaction with LAMP2A leading to abnormal neuronal autophagy function and dysregulated degradation of critical intracellular components. Therapies targeting Tat-mediated autophagy alterations may decrease neurodegeneration in aging patients with HAND.


Assuntos
Autofagia , Lisossomos/metabolismo , Neurônios/metabolismo , Fagossomos/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Complexo AIDS Demência/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , HIV-1/genética , Proteína 2 de Membrana Associada ao Lisossomo/genética , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Camundongos , Ligação Proteica , Ratos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/toxicidade
11.
Neurobiol Dis ; 74: 144-57, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25173807

RESUMO

Aß accumulation plays a central role in the pathogenesis of Alzheimer's disease (AD). Recent studies suggest that the process of Aß nucleated polymerization is essential for Aß fibril formation, pathology spreading and toxicity. Therefore, targeting this process represents an effective therapeutic strategy to slow or block disease progression. To discover compounds that might interfere with the Aß seeding capacity, toxicity and pathology spreading, we screened a focused library of FDA-approved drugs in vitro using a seeding polymerization assay and identified small molecule inhibitors that specifically interfered with Aß seeding-mediated fibril growth and toxicity. Mitoxantrone, bithionol and hexachlorophene were found to be the strongest inhibitors of fibril growth and protected primary cortical neuronal cultures against Aß-induced toxicity. Next, we assessed the effects of these three inhibitors in vivo in the mThy1-APPtg mouse model of AD (8-month-old mice). We found that mitoxantrone and bithionol, but not hexachlorophene, stabilized diffuse amyloid plaques, reduced the levels of Aß42 oligomers and ameliorated synapse loss, neuronal damage and astrogliosis. Together, our findings suggest that targeting fibril growth and Aß seeding capacity constitutes a viable and effective strategy for protecting against neurodegeneration and disease progression in AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Degeneração Neural/fisiopatologia , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/efeitos dos fármacos , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Bitionol/farmacocinética , Bitionol/farmacologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Gliose/tratamento farmacológico , Gliose/patologia , Gliose/fisiopatologia , Hexaclorofeno/farmacocinética , Hexaclorofeno/farmacologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitoxantrona/farmacocinética , Mitoxantrona/farmacologia , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacocinética , Fragmentos de Peptídeos/toxicidade , Placa Amiloide/tratamento farmacológico , Placa Amiloide/patologia , Placa Amiloide/fisiopatologia , Ratos
12.
Neuron ; 85(1): 76-87, 2015 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-25533483

RESUMO

Parkinson's disease (PD) is a common neurodegenerative disorder. Functional interactions between some PD genes, like PINK1 and parkin, have been identified, but whether other ones interact remains elusive. Here we report an unexpected genetic interaction between two PD genes, VPS35 and EIF4G1. We provide evidence that EIF4G1 upregulation causes defects associated with protein misfolding. Expression of a sortilin protein rescues these defects, downstream of VPS35, suggesting a potential role for sortilins in PD. We also show interactions between VPS35, EIF4G1, and α-synuclein, a protein with a key role in PD. We extend our findings from yeast to an animal model and show that these interactions are conserved in neurons and in transgenic mice. Our studies reveal unexpected genetic and functional interactions between two seemingly unrelated PD genes and functionally connect them to α-synuclein pathobiology in yeast, worms, and mouse. Finally, we provide a resource of candidate PD genes for future interrogation.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Fator de Iniciação Eucariótico 4G/genética , Doença de Parkinson/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Transporte Vesicular/genética , alfa-Sinucleína/genética , Proteínas Adaptadoras de Transporte Vesicular , Animais , Caenorhabditis elegans , Camundongos , Camundongos Transgênicos , Saccharomyces cerevisiae
13.
Mol Ther ; 22(10): 1753-67, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25008355

RESUMO

Parkinson's disease and dementia with Lewy bodies are neurodegenerative disorders characterized by accumulation of α-synuclein (α-syn). Recently, single-chain fragment variables (scFVs) have been developed against individual conformational species of α-syn. Unlike more traditional monoclonal antibodies, these scFVs will not activate or be endocytosed by Fc receptors. For this study, we investigated an scFV directed against oligomeric α-syn fused to the LDL receptor-binding domain from apolipoprotein B (apoB). The modified scFV showed enhanced brain penetration and was imported into neuronal cells through the endosomal sorting complex required for transport (ESCRT) pathway, leading to lysosomal degradation of α-syn aggregates. Further analysis showed that the scFV was effective at ameliorating neurodegenerative pathology and behavioral deficits observed in the mouse model of dementia with Lewy bodies/Parkinson's disease. Thus, the apoB modification had the effect of both increasing accumulation of the scFV in the brain and directing scFV/α-syn complexes for degradation through the ESCRT pathway, leading to improved therapeutic potential of immunotherapy.


Assuntos
Encéfalo/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Degeneração Neural/metabolismo , Anticorpos de Cadeia Única/metabolismo , alfa-Sinucleína/metabolismo , Motivos de Aminoácidos , Animais , Apolipoproteínas B/química , Apolipoproteínas B/metabolismo , Autofagia , Comportamento Animal , Encéfalo/patologia , Linhagem Celular , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Ordem dos Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Lentivirus/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Ligação Proteica , Transporte Proteico , Proteólise , Ratos , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Transdução Genética , alfa-Sinucleína/genética , alfa-Sinucleína/imunologia
14.
J Biol Chem ; 285(20): 14941-14954, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20150427

RESUMO

Parkinson disease (PD) is the second most common neurodegenerative disorder after Alzheimer disease (AD). There is considerable consensus that the increased production and/or aggregation of alpha-synuclein (alpha-syn) plays a central role in the pathogenesis of PD and related synucleinopathies. Current therapeutic strategies for treating PD offer mainly transient symptomatic relief and aim at the restitution of dopamine levels to counterbalance the loss of dopaminergic neurons. Therefore, the identification and development of drug-like molecules that block alpha-synuclein aggregation and prevent the loss of dopaminergic neurons are desperately needed to treat or slow the progression of PD. Here, we show that entacapone and tolcapone are potent inhibitors of alpha-syn and beta-amyloid (Abeta) oligomerization and fibrillogenesis, and they also protect against extracellular toxicity induced by the aggregation of both proteins. Comparison of the anti-aggregation properties of entacapone and tolcapone with the effect of five other catechol-containing compounds, dopamine, pyrogallol, gallic acid, caffeic acid, and quercetin on the oligomerization and fibrillization of alpha-syn and Abeta, demonstrate that the catechol moiety is essential for the anti-amyloidogenic activity. Our findings present the first characterization of the anti-amyloidogenic properties of tolcapone and entacapone against both alpha-synuclein and Abeta42 and highlight the potential of this class of nitro-catechol compounds as anti-amyloidogenic agents. Their inhibitory properties, mode of action, and structural properties suggest that they constitute promising lead compounds for further optimization.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Benzofenonas/farmacologia , Inibidores de Catecol O-Metiltransferase , Catecóis/farmacologia , Inibidores Enzimáticos/farmacologia , Nitrilas/farmacologia , Nitrofenóis/farmacologia , Fragmentos de Peptídeos/biossíntese , alfa-Sinucleína/biossíntese , Peptídeos beta-Amiloides/toxicidade , Animais , Microscopia Eletrônica , Ressonância Magnética Nuclear Biomolecular , Células PC12 , Fragmentos de Peptídeos/toxicidade , Ratos , Tolcapona
15.
J Neurochem ; 110(2): 545-56, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19457129

RESUMO

Microglia, the immune cells of the CNS, play essential roles in both physiological and pathological brain states. Here we have used an in vitro model to demonstrate neuroprotection of a 48 h-microglial conditioned medium (MCM) towards cerebellar granule neurons (CGNs) challenged with the neurotoxin 6-hydroxydopamine, which induces a Parkinson-like neurodegeneration, and to identify the protective factor(s). MCM nearly completely protects CGNs from 6-hydroxydopamine neurotoxicity and at least some of the protective factor(s) are peptidic in nature. While the fraction of the medium containing molecules < 30 kDa completely protects CGNs, fractions containing molecules < 10 kDa or > 10 kDa are not neuroprotective. We further demonstrate that microglia release high amounts of transforming growth factor-beta2 (TGF-beta2) and that its exogenous addition to the fraction of the medium not containing it (< 10 kDa) fully restores the neuroprotective action. Moreover, MCM neuroprotection is significantly counteracted by an inhibitor of TGF-beta2 transduction pathway. Our results identify TGF-beta2 as an essential neuroprotective factor released by microglia in its culture medium that requires to be fully effective the concomitant presence of other factor(s) of low molecular weight.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Microglia/fisiologia , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Oxidopamina/toxicidade , Fator de Crescimento Transformador beta2/fisiologia , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Peso Molecular , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar
16.
Neurotox Res ; 15(2): 127-32, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19384575

RESUMO

Valproic acid (VPA) has been used for many years as a drug of choice for epilepsy and mood disorders. Recently, evidence has been proposed for a wide spectrum of actions of this drug, including antitumoral and neuroprotective properties. Valproic acid-mediated neuroprotection in vivo has been so far demonstrated in a limited number of experimental models. In this study, we have tested the neuroprotective potential of chronic (4 + 1 weeks) dietary administration of VPA on degeneration of cholinergic and GABAergic neurons of the rat nucleus basalis magnocellularis (NBM), injected with the excitotoxin, ibotenic acid (IBO), an animal models that is relevant for Alzheimer's disease-like neurodegeneration. We show that VPA treatment significantly protects both cholinergic and GABAergic neurons present in the injected area from the excitotoxic insult. A significant level of neuroprotection, in particular, is exerted towards the cholinergic neurons of the NBM projecting to the cortex, as demonstrated by the substantially higher levels of cholinergic markers maintained in the target cortical area of VPA-treated rats after IBO injection in the NBM. We further show that chronic VPA administration results in increased acetylation of histone H3 in brain, consistent with the histone deacetylase inhibitory action of VPA and putatively linked to a neuroprotective action of the drug mediated at the epigenetic level.


Assuntos
Núcleo Basal de Meynert/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Ibotênico/toxicidade , Fármacos Neuroprotetores/administração & dosagem , Síndromes Neurotóxicas , Ácido Valproico/administração & dosagem , Acetilcolina/metabolismo , Acetilcolinesterase/metabolismo , Animais , Núcleo Basal de Meynert/patologia , Córtex Cerebral/metabolismo , Colina O-Acetiltransferase/metabolismo , Modelos Animais de Doenças , Lateralidade Funcional , Glutamato Descarboxilase/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/prevenção & controle , Ratos , Ratos Wistar , Ácido gama-Aminobutírico/metabolismo
17.
Neurosci Lett ; 448(1): 74-8, 2008 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-18950681

RESUMO

Microglia, the immune cells of the mammalian CNS, have often been indicated as dangerous effector cells for their activation in response to traumatic CNS injuries or immunological stimuli and for their involvement in many chronic neurodegenerative diseases. Recently, several in vitro and in vivo studies have emphasized that microglial activity is essential in promoting neuronal survival. We have tested the efficacy of media directly conditioned by microglia or conditioned by microglia after having been exposed to apoptotic neurons, towards neuroprotection of rat cerebellar granule cells (CGCs) challenged with staurosporine or glutamate. Apoptotic death of CGC caused by staurosporine, as well as by a mild excitotoxic stimulus delivered through sub-chronic glutamate treatment, was significantly counteracted by microglia conditioned media. On the other hand, an acute excitotoxic insult delivered through a short pulse of glutamate exposure in the absence of magnesium and resulting in a mix of apoptotic and necrotic death was only marginally counteracted by microglia conditioned media. The present results extend the available information regarding the neuroprotective role of microglia and support the usefulness of employing the culture approach for perspective identification of neuroprotective factors released by these cells. Furthermore, the use of media previously exposed to apoptotic neurons to elicit the neuroprotective response of microglia, indicate the feasibility to re-create also in the isolated culture conditions, at least some of the elements at the basis of neuron/microglia cross-talk.


Assuntos
Apoptose/efeitos dos fármacos , Cerebelo/citologia , Meios de Cultivo Condicionados/farmacologia , Microglia/química , Neurônios/efeitos dos fármacos , Análise de Variância , Animais , Animais Recém-Nascidos , Contagem de Células/métodos , Células Cultivadas , Inibidores Enzimáticos/toxicidade , Ácido Glutâmico/toxicidade , Ratos , Ratos Wistar , Estaurosporina/toxicidade , Sais de Tetrazólio , Tiazóis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA