Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
ACS Chem Neurosci ; 15(2): 278-289, 2024 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-38154144

RESUMO

Spinocerebellar ataxia type 3 (SCA3) is a neurodegenerative disorder characterized by ataxia and other neurological manifestations, with a poor prognosis and a lack of effective therapies. The amyloid aggregation of the ataxin-3 protein is a hallmark of SCA3 and one of the main biochemical events prompting its onset, making it a prominent target for the development of preventive and therapeutic interventions. Here, we tested the efficacy of an aqueous Lavado cocoa extract and its polyphenolic components against ataxin-3 aggregation and neurotoxicity. The combination of biochemical assays and atomic force microscopy morphological analysis provided clear evidence of cocoa flavanols' ability to hinder ATX3 amyloid aggregation through direct physical interaction, as assessed by NMR spectroscopy. The chemical identity of the flavanols was investigated by ultraperformance liquid chromatography-high-resolution mass spectrometry. The use of the preclinical model Caenorhabditis elegans allowed us to demonstrate cocoa flavanols' ability to ameliorate ataxic phenotypes in vivo. To the best of our knowledge, Lavado cocoa is the first natural source whose extract is able to directly interfere with ATX3 aggregation, leading to the formation of off-pathway species.


Assuntos
Doença de Machado-Joseph , Animais , Ataxina-3/genética , Ataxina-3/metabolismo , Doença de Machado-Joseph/tratamento farmacológico , Doença de Machado-Joseph/genética , Doença de Machado-Joseph/metabolismo , Proteínas Amiloidogênicas/metabolismo , Amiloide/metabolismo , Caenorhabditis elegans , Polifenóis/uso terapêutico , Extratos Vegetais/farmacologia
2.
Mol Brain ; 14(1): 128, 2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34416891

RESUMO

Machado-Joseph disease (MJD, also known as spinocerebellar ataxia type 3) is a fatal neurodegenerative disease that impairs control and coordination of movement. Here we tested whether treatment with the histone deacetylase inhibitor sodium valproate (valproate) prevented a movement phenotype that develops in larvae of a transgenic zebrafish model of the disease. We found that treatment with valproate improved the swimming of the MJD zebrafish, affected levels of acetylated histones 3 and 4, but also increased expression of polyglutamine expanded human ataxin-3. Proteomic analysis of protein lysates generated from the treated and untreated MJD zebrafish also predicted that valproate treatment had activated the sirtuin longevity signaling pathway and this was confirmed by findings of increased SIRT1 protein levels and sirtuin activity in valproate treated MJD zebrafish and HEK293 cells expressing ataxin-3 84Q, respectively. Treatment with resveratrol (another compound known to activate the sirtuin pathway), also improved swimming in the MJD zebrafish. Co-treatment with valproate alongside EX527, a SIRT1 activity inhibitor, prevented induction of autophagy by valproate and the beneficial effects of valproate on the movement in the MJD zebrafish, supporting that they were both dependent on sirtuin activity. These findings provide the first evidence of sodium valproate inducing activation of the sirtuin pathway. Further, they indicate that drugs that target the sirtuin pathway, including sodium valproate and resveratrol, warrant further investigation for the treatment of MJD and related neurodegenerative diseases.


Assuntos
Inibidores de Histona Desacetilases/uso terapêutico , Doença de Machado-Joseph/tratamento farmacológico , Sirtuínas/efeitos dos fármacos , Ácido Valproico/uso terapêutico , Acetilação , Animais , Animais Geneticamente Modificados , Ataxina-3/antagonistas & inibidores , Ataxina-3/genética , Ataxina-3/metabolismo , Autofagia/efeitos dos fármacos , Carbazóis/farmacologia , Carbazóis/uso terapêutico , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Genes Reporter , Células HEK293 , Inibidores de Histona Desacetilases/farmacologia , Histonas/metabolismo , Humanos , Peptídeos/genética , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Resveratrol/farmacologia , Resveratrol/uso terapêutico , Transdução de Sinais , Sirtuína 1/fisiologia , Sirtuínas/fisiologia , Natação , Expansão das Repetições de Trinucleotídeos , Ácido Valproico/farmacologia , Peixe-Zebra , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
3.
Neurotherapeutics ; 18(2): 1151-1165, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33782863

RESUMO

Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease (MJD), is a polyglutamine expansion disease arising from a trinucleotide CAG repeat expansion in exon 10 of the gene ATXN3. There are no effective pharmacological treatments for MJD, thus the identification of new pathogenic mechanisms, and the development of novel therapeutics is urgently needed. In this study, we performed a comprehensive, blind drug screen of 3942 compounds (many FDA approved) and identified small molecules that rescued the motor-deficient phenotype in transgenic ATXN3 Caenorhabditis elegans strain. Out of this screen, five lead compounds restoring motility, protecting against neurodegeneration, and increasing the lifespan in ATXN3-CAG89 mutant worms were identified. These compounds were alfacalcidol, chenodiol, cyclophosphamide, fenbufen, and sulfaphenazole. We then investigated how these molecules might exert their neuroprotective properties. We found that three of these compounds, chenodiol, fenbufen, and sulfaphenazole, act as modulators for TFEB/HLH-30, a key transcriptional regulator of the autophagy process, and require this gene for their neuroprotective activities. These genetic-chemical approaches, using genetic C. elegans models for MJD and the screening, are promising tools to understand the mechanisms and pathways causing neurodegeneration, leading to MJD. Positively acting compounds may be promising candidates for investigation in mammalian models of MJD and preclinical applications in the treatment of this disease.


Assuntos
Ataxina-3/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Caenorhabditis elegans/genética , Ácido Quenodesoxicólico/administração & dosagem , Fenilbutiratos/administração & dosagem , Sulfafenazol/administração & dosagem , Animais , Animais Geneticamente Modificados , Ataxina-3/toxicidade , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/toxicidade , Avaliação Pré-Clínica de Medicamentos/métodos , Longevidade/efeitos dos fármacos , Longevidade/fisiologia , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/genética
4.
Neurochem Int ; 144: 104979, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33535071

RESUMO

Polyglutamine (polyQ)-mediated mitochondria damage is one of the prime causes of polyQ toxicity, which leads to the loss of neurons and the injury of non-neuronal cells. With the discovery of the crucial role of the gut-brain axis and gut microbes in neurological diseases, the relationship between visceral damage and neurological disorders has also received extensive attention. This study successfully simulated the polyQ mitochondrial damage model by expressing 78 or 84 polyglutamine-containing Ataxin3 proteins in Drosophila intestinal enterocytes. In vivo, polyQ expression can reduce mitochondrial membrane potential, mitochondrial DNA damage, abnormal mitochondrial morphology, and loose mitochondrial cristae. Expression profiles evaluated by RNA-seq showed that mitochondrial structural genes and functional genes (oxidative phosphorylation and tricarboxylic acid cycle-related) were significantly down-regulated. More importantly, Bioinformatic analyses demonstrated that pathological polyQ expression induced vitamin B6 metabolic pathways abnormality. Active vitamin B6 participates in hundreds of enzymatic reactions and is very important for maintaining mitochondria's activities. In the SCA3 Drosophila model, Vitamin B6 supplementation significantly suppressed ECs mitochondria damage in guts and inhibited cellular polyQ aggregates in fat bodies, indicating a promising therapeutic strategy for the treatment of polyQ. Taken together, our results reveal a crucial role for the Vitamin B6-mediated mitochondrial protection in polyQ-induced cellular toxicity, which provides strong evidence for this process as a drug target in polyQ diseases treatment.


Assuntos
Ataxina-3/genética , Modelos Animais de Doenças , Doença de Machado-Joseph/genética , Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Vitamina B 6/farmacologia , Animais , Animais Geneticamente Modificados , Ataxina-3/metabolismo , Drosophila , Redes Reguladoras de Genes/fisiologia , Humanos , Doença de Machado-Joseph/tratamento farmacológico , Doença de Machado-Joseph/metabolismo , Mitocôndrias/genética , Fármacos Neuroprotetores/uso terapêutico , Vitamina B 6/uso terapêutico
5.
J Mol Diagn ; 22(6): 782-793, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32205289

RESUMO

Polyglutamine spinocerebellar ataxias (SCAs) constitute a group of autosomal dominantly inherited neurodegenerative disorders with considerable phenotypic overlap. Definitive diagnoses rely on the detection of a mutation in each associated locus, comprising the abnormal expansion of the trinucleotide cytosine-adenine-guanine (CAG) in coding exons. Assessment of single nucleotide polymorphisms associated with the CAG expansion in the context of SCAs is also relevant for improving molecular diagnosis and for generating novel therapeutic strategies. The current study is focused on Machado-Joseph disease/SCA type 3, with the aim of developing a protocol for the accurate determination of the CAG length in exon 10 of the human ATXN3 gene and to characterize flanking polymorphisms. A single pair of primers was designed and validated, and two complementary PCR-based methods were established. In method I, PCR amplicons were cloned and sequenced, allowing the assessment of three single nucleotide polymorphisms in the vicinity of the CAG repeat (C987GG/G987GG, TAA1118/TAC1118, and C1178/A1178), which can constitute potential targets for personalized gene-based therapies. Method II combines PCR, capillary electrophoresis, and a size correction formula, enabling a time and cost-effective determination of the number of CAGs. The established protocol paves the way to overcome technical difficulties related to the molecular characterization of the CAG motif and intragenic polymorphisms in the context of Machado-Joseph disease/SCA type 3 and may prove useful when applied to other polyglutamine SCAs.


Assuntos
Adenina , Ataxina-3/genética , Citosina , Guanina , Doença de Machado-Joseph/diagnóstico , Doença de Machado-Joseph/genética , Polimorfismo de Nucleotídeo Único , Proteínas Repressoras/genética , Expansão das Repetições de Trinucleotídeos/genética , Alelos , Estudos de Casos e Controles , Éxons , Humanos , Doença de Machado-Joseph/sangue , Reação em Cadeia da Polimerase
6.
Nature ; 575(7781): 203-209, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31666698

RESUMO

Accumulation of mutant proteins is a major cause of many diseases (collectively called proteopathies), and lowering the level of these proteins can be useful for treatment of these diseases. We hypothesized that compounds that interact with both the autophagosome protein microtubule-associated protein 1A/1B light chain 3 (LC3)1 and the disease-causing protein may target the latter for autophagic clearance. Mutant huntingtin protein (mHTT) contains an expanded polyglutamine (polyQ) tract and causes Huntington's disease, an incurable neurodegenerative disorder2. Here, using small-molecule-microarray-based screening, we identified four compounds that interact with both LC3 and mHTT, but not with the wild-type HTT protein. Some of these compounds targeted mHTT to autophagosomes, reduced mHTT levels in an allele-selective manner, and rescued disease-relevant phenotypes in cells and in vivo in fly and mouse models of Huntington's disease. We further show that these compounds interact with the expanded polyQ stretch and could lower the level of mutant ataxin-3 (ATXN3), another disease-causing protein with an expanded polyQ tract3. This study presents candidate compounds for lowering mHTT and potentially other disease-causing proteins with polyQ expansions, demonstrating the concept of lowering levels of disease-causing proteins using autophagosome-tethering compounds.


Assuntos
Alelos , Avaliação Pré-Clínica de Medicamentos/métodos , Proteína Huntingtina/antagonistas & inibidores , Proteína Huntingtina/genética , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/genética , Mutação/genética , Animais , Ataxina-3/genética , Autofagossomos/metabolismo , Autofagia , Modelos Animais de Doenças , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Feminino , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação/efeitos dos fármacos , Neurônios/citologia , Peptídeos/genética , Fenótipo , Reprodutibilidade dos Testes
7.
Hum Mol Genet ; 28(9): 1463-1473, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30576445

RESUMO

Spinocerebellar ataxia type 3 (SCA3) is caused by the expansion of CAG repeats in the ATXN3 gene leading to an elongated polyglutamine tract in the ataxin-3 protein. Previously, we demonstrated that symptoms of SCA3 are reversible in the first conditional mouse model for SCA3 directing ataxin-3 predominantly to the hindbrain. Here, we report on the effects of transgenic ataxin-3 expression in forebrain regions. Employing the Tet-off CamKII-promoter mouse line and our previously published SCA3 responder line, we generated double transgenic mice (CamKII/MJD77), which develop a neurological phenotype characterized by impairment in rotarod performance, and deficits in learning new motor tasks as well as hyperactivity. Ataxin-3 and ubiquitin-positive inclusions are detected in brains of double transgenic CamKII/MJD77 mice. After turning off the expression of pathologically expanded ataxin-3, these inclusions disappear. However, the observed phenotype could not be reversed, very likely due to pronounced apoptotic cell death in the frontal brain. Our data demonstrate that cerebellar expression is not required to induce a neurological phenotype using expanded ATXN3 as well as the pronounced sensibility of forebrain neurons for toxic ataxin-3.


Assuntos
Ataxina-3/genética , Lobo Frontal/metabolismo , Doença de Machado-Joseph/genética , Doença de Machado-Joseph/metabolismo , Neurônios/metabolismo , Expansão das Repetições de Trinucleotídeos , Animais , Ataxina-3/metabolismo , Comportamento Animal , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Modelos Animais de Doenças , Lobo Frontal/patologia , Expressão Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Imuno-Histoquímica , Doença de Machado-Joseph/patologia , Camundongos , Camundongos Transgênicos , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Especificidade de Órgãos/genética , Agregados Proteicos , Agregação Patológica de Proteínas , Desempenho Psicomotor
8.
Proteomics ; 17(13-14)2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28544614

RESUMO

Narcolepsy is a disabling neurological disorder of sleepiness linked to the loss of neurons producing orexin neuropeptides in the hypothalamus. Two well-characterized phenotypic mouse models of narcolepsy, loss-of-function (orexin-knockout), and progressive loss of orexin (orexin/ataxin-3) exist. The open question is whether the proteomics signatures of the hypothalamus would be different between the two models. To address this gap, we utilized a label-free proteomics approach and conducted a hypothalamic proteome analysis by comparing each disease model to that of wild type. Following data processing and statistical analysis, 14 484 peptides mapping to 2282 nonredundant proteins were identified, of which 39 proteins showed significant differences in protein expression across groups. Altered proteins in both models showed commonalties in pathways for mitochondrial dysfunction and neuronal degeneration, as well as altered proteins related to inflammatory demyelination, insulin resistance, metabolic responses, and the dopaminergic and monoaminergic systems. Model-specific alterations in insulin degraded enzyme (IDE) and synaptosomal-associated protein-25 were unique to orexin-KO and orexin/ataxin-3, respectively. For both models, proteomics not only identified clinically suspected consequences of orexin loss on energy homeostasis and neurotransmitter systems, but also identified commonalities in inflammation and degeneration despite the entirely different genetic basis of the two mouse models.


Assuntos
Modelos Animais de Doenças , Hipotálamo/metabolismo , Narcolepsia/metabolismo , Proteoma/metabolismo , Animais , Ataxina-3/fisiologia , Camundongos , Camundongos Knockout , Orexinas/fisiologia , Proteoma/análise , Proteômica
9.
Sleep ; 40(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-28364459

RESUMO

Study Objectives: Recent findings showed that 16%-26% of narcolepsy patients were positive for anti-tribbles pseudokinase 2 (TRIB2) antibody, and the intracerebroventricular administration of immunoglobulin-G purified from anti-TRIB2 positive narcolepsy patients caused hypocretin/orexin neuron loss. We investigated the pathophysiological role of TRIB2 antibody using TRIB2-immunized rats and hypocretin/ataxin-3 transgenic (ataxin-3) mice. Methods: Plasma, cerebrospinal fluid (CSF), and hypothalamic tissues from TRIB2-immunized rats were collected. Anti-TRIB2 titers, hypocretin contents, mRNA expressions, the cell count of hypocretin neurons, and immunoreactivity of anti-TRIB2 antibodies on hypocretin neurons were investigated. The plasma from ataxin-3 mice was also used to determine the anti-TRIB2 antibody titer changes following the loss of hypocretin neurons. Results: TRIB2 antibody titers increased in the plasma and CSF of TRIB2-immunized rats. The hypothalamic tissue immunostained with the sera from TRIB2-immunized rats revealed positive signals in the cytoplasm of hypcretin neurons. While no changes were found regarding hypothalamic hypocretin contents or cell counts, but there were significant decreases of the hypocretin mRNA level and release into the CSF. The plasma from over 26-week-old ataxin-3 mice, at the advanced stage of hypocretin cell destruction, showed positive reactions against TRIB2 antigen, and positive plasma also reacted with murine hypothalamic hypocretin neurons. Conclusions: Our results suggest that the general activation of the immune system modulates the functions of hypocretin neurons. The absence of a change in hypocretin cell populations suggested that factors other than anti-TRIB2 antibody play a part in the loss of hypocretin neurons in narcolepsy. The increased anti-TRIB2 antibody after the destruction of hypocretin neurons suggest that anti-TRIB2 antibody in narcolepsy patients is the consequence rather than the inciting cause of hypocretin cell destruction.


Assuntos
Autoanticorpos/metabolismo , Autoantígenos/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Narcolepsia/imunologia , Neurônios/imunologia , Orexinas/metabolismo , Animais , Animais Geneticamente Modificados , Ataxina-3/metabolismo , Biomarcadores/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Feminino , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Narcolepsia/metabolismo , Narcolepsia/fisiopatologia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Vacinação
10.
Brain ; 139(11): 2891-2908, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27645800

RESUMO

No disease-modifying treatment exists for the fatal neurodegenerative polyglutamine disease known both as Machado-Joseph disease and spinocerebellar ataxia type 3. As a potential route to therapy, we identified small molecules that reduce levels of the mutant disease protein, ATXN3. Screens of a small molecule collection, including 1250 Food and Drug Administration-approved drugs, in a novel cell-based assay, followed by secondary screens in brain slice cultures from transgenic mice expressing the human disease gene, identified the atypical antipsychotic aripiprazole as one of the hits. Aripiprazole increased longevity in a Drosophila model of Machado-Joseph disease and effectively reduced aggregated ATXN3 species in flies and in brains of transgenic mice treated for 10 days. The aripiprazole-mediated decrease in ATXN3 abundance may reflect a complex response culminating in the modulation of specific components of cellular protein homeostasis. Aripiprazole represents a potentially promising therapeutic drug for Machado-Joseph disease and possibly other neurological proteinopathies.


Assuntos
Antipsicóticos/uso terapêutico , Aripiprazol/uso terapêutico , Ataxina-3/metabolismo , Doença de Machado-Joseph/tratamento farmacológico , Doença de Machado-Joseph/metabolismo , Proteínas Mutantes/efeitos dos fármacos , Animais , Animais Geneticamente Modificados , Ataxina-3/genética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Modelos Animais de Doenças , Drosophila , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Células HEK293/efeitos dos fármacos , Células HEK293/metabolismo , Células HEK293/ultraestrutura , Humanos , Doença de Machado-Joseph/genética , Camundongos , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Cultura de Órgãos , Peptídeos/genética , Piperidinas/farmacologia , Piranos/farmacologia , Pirazóis/farmacologia
11.
Neuropharmacology ; 110(Pt A): 268-276, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27474349

RESUMO

Prostaglandin (PG)D2 is an endogenous sleep substance, and a series of animal studies reported that PGD2 or PGD2 receptor (DP1) agonists promote sleep, while DP1 antagonists promote wakefulness. This suggests the possibility of use of PG DP1 antagonists as wake-promoting compounds. We therefore evaluated the wake-promoting effects of ONO-4127Na, a DP1 antagonist, in a mouse model of narcolepsy (i.e., orexin/ataxin-3 transgenic mice) and compared those to effects of modafinil. ONO-4127Na perfused in the basal forebrain (BF) area potently promoted wakefulness in both wild type and narcoleptic mice, and the wake-promoting effects of ONO-4127Na at 2.93 × 10(-4) M roughly corresponded to those of modafinil at 100 mg/kg (p.o.). The wake promoting effects of ONO-4127Na was observed both during light and dark periods, and much larger effects were seen during the light period when mice slept most of the time. ONO-4127Na, when perfused in the hypothalamic area, had no effects on sleep. We further demonstrated that wake-promoting effects of ONO-4127Na were abolished in DP1 KO mice, confirming that the wake-promoting effect of ONO-4127Na is mediated by blockade of the PG DP1 receptors located in the BF area. ONO-4127Na reduced DREM, an EEG/EMG assessment of behavioral cataplexy in narcoleptic mice, suggesting that ONO-4127Na is likely to have anticataplectic effects. DP1 antagonists may be a new class of compounds for the treatment of narcolepsy-cataplexy, and further studies are warranted.


Assuntos
Ataxina-3/deficiência , Narcolepsia/tratamento farmacológico , Orexinas/deficiência , Antagonistas de Prostaglandina/farmacologia , Promotores da Vigília/farmacologia , Animais , Ataxina-3/genética , Compostos Benzidrílicos/farmacologia , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Modelos Animais de Doenças , Eletroencefalografia , Eletromiografia , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modafinila , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Narcolepsia/fisiopatologia , Orexinas/genética , Fotoperíodo , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/fisiopatologia , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Fases do Sono/efeitos dos fármacos , Fases do Sono/fisiologia , Vigília/efeitos dos fármacos , Vigília/fisiologia
12.
Brain Behav Immun ; 57: 58-67, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27318095

RESUMO

Hypocretin, also known as orexin, maintains the vigilance state and regulates various physiological processes, such as arousal, sleep, food intake, energy expenditure, and reward. Previously, we found that when wild-type mice and hypocretin/ataxin-3 littermates (which are depleted of hypothalamic hypocretin-expressing neurons postnatally) were administered lipopolysaccharide (LPS), the two genotypes exhibited significant differences in their sleep/wake cycle, including differences in the degree of increase in sleep periods and in recovery from sickness behaviour. In the present study, we examined changes in the hypothalamic vigilance system and in the hypothalamic expression of inflammatory factors in response to LPS in hypocretin/ataxin-3 mice. Peripheral immune challenge with LPS affected the hypothalamic immune response and vigilance states. This response was altered by the loss of hypocretin. Hypocretin expression was inhibited after LPS injection in both hypocretin/ataxin-3 mice and their wild-type littermates, but expression was completely abolished only in hypocretin/ataxin-3 mice. Increases in the number of histidine decarboxylase (HDC)-positive cells and in Hdc mRNA expression were found in hypocretin/ataxin-3 mice, and this increase was suppressed by LPS. Hypocretin loss did not impact the change in expression of hypothalamic inflammatory factors in response to LPS, except for interferon gamma and colony stimulating factor 3. The number of c-Fos-positive/HDC-positive cells in hypocretin/ataxin-3 mice administered LPS injections was elevated, even during the rest period, in all areas, suggesting that there is an increase in the activity of histaminergic neurons in hypocretin/ataxin-3 mice following LPS injection. Taken together, our results suggest a novel role for hypocretin in the hypothalamic response to peripheral immune challenge. Our findings contribute to the understanding of the pathophysiology of narcolepsy.


Assuntos
Hipotálamo/imunologia , Hipotálamo/metabolismo , Inflamação , Lipopolissacarídeos/farmacologia , Orexinas/metabolismo , Sono/imunologia , Vigília , Animais , Ataxina-3/metabolismo , Expressão Gênica , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Lipopolissacarídeos/administração & dosagem , Masculino , Camundongos , Camundongos Transgênicos
13.
Neuroscience ; 313: 162-73, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26601773

RESUMO

A major pathological hallmark in several neurodegenerative disorders, like polyglutamine disorders (polyQ), including Machado-Joseph disease (MJD), is the formation of protein aggregates. MJD is caused by a CAG repeat expansion in the ATXN3 gene, resulting in an abnormal protein, which is prone to misfolding and forms cytoplasmic and nuclear aggregates within neurons, ultimately inducing neurodegeneration. Treatment of proteinopathies with drugs that up-regulate autophagy has shown promising results in models of polyQ diseases. Temsirolimus (CCI-779) inhibits the mammalian target of rapamycin (m-TOR), while lithium chloride (LiCl) acts by inhibiting inositol monophosphatase, both being able to induce autophagy. We have previously shown that chronic treatment with LiCl (10.4 mg/kg) had limited effects in a transgenic MJD mouse model. Also, others have shown that CCI-779 had mild positive effects in a different mouse model of the disease. It has been suggested that the combination of mTOR-dependent and -independent autophagy inducers could be a more effective therapeutic approach. To further explore this avenue toward therapy, we treated CMVMJD135 transgenic mice with a conjugation of CCI-779 and LiCl, both at concentrations known to induce autophagy and not to be toxic. Surprisingly, this combined treatment proved to be deleterious to both wild-type (wt) and transgenic animals, failing to rescue their neurological symptoms and actually exerting neurotoxic effects. These results highlight the possible dangers of manipulating autophagy in the nervous system and suggest that a better understanding of the potential disruption in the autophagy pathway in MJD is required before successful long-term autophagy modulating therapies can be developed.


Assuntos
Autofagia/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/toxicidade , Compostos de Lítio/toxicidade , Doença de Machado-Joseph/tratamento farmacológico , Síndromes Neurotóxicas/fisiopatologia , Sirolimo/análogos & derivados , Animais , Animais Geneticamente Modificados , Ataxina-3/metabolismo , Autofagia/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Caenorhabditis elegans , Fármacos do Sistema Nervoso Central/administração & dosagem , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Compostos de Lítio/administração & dosagem , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Doença de Machado-Joseph/patologia , Doença de Machado-Joseph/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Síndromes Neurotóxicas/patologia , Sirolimo/administração & dosagem , Sirolimo/toxicidade , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo
14.
Parkinsonism Relat Disord ; 21(12): 1441-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26552869

RESUMO

INTRODUCTION: Dystonia is frequent in Machado-Joseph disease, but several important aspects are not yet defined, such as the detailed clinical profile, response to treatment and anatomical substrate. METHODS: We screened 75 consecutive patients and identified those with dystonia. The Burke-Marsden-Fahn Dystonia Rating Scale was employed to quantify dystonia severity. Patients with dystonia received levodopa 600 mg/day for 2 months and were videotaped before and after treatment. A blinded evaluator rated dystonia in the videos. Patients with disabling dystonia who failed to respond to levodopa treatment received botulinum toxin. Finally, volumetric T1 and diffusion tensor imaging sequences were obtained in the dystonic group using a 3T-MRI scanner to identify areas of gray and white matter that were selectively damaged. RESULTS: There were 21 patients with dystonia (28%): 9 classified as generalized and 12 as focal/segmental. Patients with dystonia had earlier onset and larger (CAG) expansions (28.9 ± 11.7 vs 40.6 ± 11.4; p < 0.001 and 75 vs 70; p < 0.001, respectively). Although group analyses failed to show benefit on levodopa (p = 0.07), some patients had objective improvement. In addition, ten patients received botulinum toxin resulting in a significant change in dystonia scores after 4 weeks (p = 0.03). Patients with dystonia had atrophy at pre- and paracentral cortices; whereas, non-dystonic patients had occipital atrophy. Basal ganglia volume was reduced in both groups, but atrophy at the thalami, cerebellar white matter and ventral diencephali was disproportionately higher in the dystonic group. CONCLUSION: Dystonia in Machado-Joseph disease is frequent and often disabling, but may respond to levodopa. It is associated predominantly with structural abnormalities around the motor cortices and in the thalami.


Assuntos
Encéfalo/patologia , Distúrbios Distônicos/etiologia , Doença de Machado-Joseph/complicações , Adulto , Ataxina-3/genética , Atrofia , Toxinas Botulínicas Tipo A/uso terapêutico , Imagem de Tensor de Difusão , Distúrbios Distônicos/tratamento farmacológico , Distúrbios Distônicos/genética , Distúrbios Distônicos/patologia , Feminino , Substância Cinzenta/patologia , Humanos , Levodopa/uso terapêutico , Doença de Machado-Joseph/genética , Doença de Machado-Joseph/patologia , Doença de Machado-Joseph/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Córtex Motor/patologia , Proteínas Repressoras/genética , Índice de Gravidade de Doença , Método Simples-Cego , Tálamo/patologia , Repetições de Trinucleotídeos , Gravação de Videoteipe , Substância Branca/patologia
15.
Neuropharmacology ; 99: 308-17, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26254860

RESUMO

More studies are required to develop therapeutic agents for treating spinocerebellar ataxia type 3 (SCA3), which is caused by mutant polyglutamine-expanded ataxin-3 and is the most prevalent subtype of spinocerebellar ataxias. T1-11 [N6-(4-Hydroxybenzyl) adenosine], isolated from a Chinese medicinal herb Gastordia elata, is an adenosine A2A receptor agonist. SCA3 and Huntington's disease (HD) belong to a family of polyglutamine neurodegenerative diseases. T1-11 exerted a therapeutic effect on HD transgenic mouse by decreasing protein level of polyglutamine-expanded huntingtin in the striatum. In the present study, we test the possibility that T1-11 or JMF1907 [N6-(3-Indolylethyl) adenosine], a synthetic analog of T1-11, alleviates pontine neuronal death, cerebellar transcriptional downregulation and ataxic symptom in the SCA3 transgenic mouse expressing HA-tagged polyglutamine-expanded ataxin-3-Q79 (ataxin-3-Q79HA). Daily oral administration of T1-11 or JMF1907 prevented neuronal death of pontine nuclei in the SCA3 mouse with a dose-dependent manner. Oral application of T1-11 or JMF1907 reversed mutant ataxin-3-Q79-induced cerebellar transcriptional repression in the SCA3 transgenic mouse. T1-11 or JMF1907 ameliorated the symptom of motor incoordination displayed by SCA3 mouse. Oral administration of T1-11 or JMF1907 significantly decreased protein level of ataxin-3-Q79HA in the pontine nuclei or cerebellum of SCA3 mouse. T1-11 or JMF1907 significantly augmented the chymotrypsin-like activity of proteasome in the pontine nuclei or cerebellum of SCA3 mouse. Our results suggests that T1-11 and JMF1907 alleviate pontine neuronal death, cerebellar transcriptional downregulation and ataxic symptom of SCA3 transgenic mouse by augmenting the proteasome activity and reducing the protein level of polyglutamine-expanded ataxin-3-Q79 in the pontine nuclei and cerebellum.


Assuntos
Adenosina/análogos & derivados , Indóis/farmacologia , Doença de Machado-Joseph/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Adenosina/farmacologia , Administração Oral , Animais , Ataxina-3/genética , Ataxina-3/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/patologia , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Humanos , Doença de Machado-Joseph/patologia , Doença de Machado-Joseph/fisiopatologia , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ponte/efeitos dos fármacos , Ponte/metabolismo , Ponte/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
16.
Hippocampus ; 25(11): 1361-73, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25808129

RESUMO

The cognitive role of melanin-concentrating hormone (MCH) neurons, a neuronal population located in the mammalian postero-lateral hypothalamus sending projections to all cortical areas, remains poorly understood. Mainly activated during paradoxical sleep (PS), MCH neurons have been implicated in sleep regulation. The genetic deletion of the only known MCH receptor in rodent leads to an impairment of hippocampal dependent forms of memory and to an alteration of hippocampal long-term synaptic plasticity. By using MCH/ataxin3 mice, a genetic model characterized by a selective deletion of MCH neurons in the adult, we investigated the role of MCH neurons in hippocampal synaptic plasticity and hippocampal-dependent forms of memory. MCH/ataxin3 mice exhibited a deficit in the early part of both long-term potentiation and depression in the CA1 area of the hippocampus. Post-tetanic potentiation (PTP) was diminished while synaptic depression induced by repetitive stimulation was enhanced suggesting an alteration of pre-synaptic forms of short-term plasticity in these mice. Behaviorally, MCH/ataxin3 mice spent more time and showed a higher level of hesitation as compared to their controls in performing a short-term memory T-maze task, displayed retardation in acquiring a reference memory task in a Morris water maze, and showed a habituation deficit in an open field task. Deletion of MCH neurons could thus alter spatial short-term memory by impairing short-term plasticity in the hippocampus. Altogether, these findings could provide a cellular mechanism by which PS may facilitate memory encoding. Via MCH neuron activation, PS could prepare the day's learning by increasing and modulating short-term synaptic plasticity in the hippocampus.


Assuntos
Comportamento Animal/fisiologia , Região CA1 Hipocampal/fisiologia , Hormônios Hipotalâmicos/fisiologia , Hipotálamo/citologia , Melaninas/fisiologia , Memória de Curto Prazo/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Hormônios Hipofisários/fisiologia , Sono REM/fisiologia , Animais , Ataxina-3/genética , Hormônios Hipotalâmicos/genética , Hipotálamo/metabolismo , Melaninas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hormônios Hipofisários/genética
17.
Am J Physiol Regul Integr Comp Physiol ; 307(8): R978-89, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25324552

RESUMO

Thermogenesis in brown adipose tissue (BAT) contributes to substantial increases in body temperature evoked by threatening or emotional stimuli. BAT thermogenesis also contributes to increases in body temperature that occur during active phases of the basic rest-activity cycle (BRAC), as part of normal daily life. Hypothalamic orexin-synthesizing neurons influence many physiological and behavioral variables, including BAT and body temperature. In conscious unrestrained animals maintained for 3 days in a quiet environment (24-26°C) with ad libitum food and water, we compared temperatures in transgenic rats with ablation of orexin neurons induced by expression of ataxin-3 (Orx_Ab) with wild-type (WT) rats. Both baseline BAT temperature and baseline body temperature, measured at the onset of BRAC episodes, were similar in Orx_Ab and WT rats. The time interval between BRAC episodes was also similar in the two groups. However, the initial slopes and amplitudes of BRAC-related increases in BAT and body temperature were reduced in Orx_Ab rats. Similarly, the initial slopes and amplitudes of the increases in BAT temperatures induced by sudden exposure to an intruder rat (freely moving or confined to a small cage) or by sudden exposure to live cockroaches were reduced in resident Orx_Ab rats. Constriction of the tail artery induced by salient alerting stimuli was also reduced in Orx_Ab rats. Our results suggest that orexin-synthesizing neurons contribute to the intensity with which rats interact with the external environment, both when the interaction is "spontaneous" and when the interaction is provoked by threatening or salient environmental events.


Assuntos
Tecido Adiposo Marrom/fisiologia , Meio Ambiente , Hipotálamo/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Neuropeptídeos/fisiologia , Termogênese/fisiologia , Animais , Ataxina-3 , Comportamento Animal/fisiologia , Temperatura Corporal/fisiologia , Baratas , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Modelos Animais , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Orexinas , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Fluxo Sanguíneo Regional/fisiologia , Descanso/fisiologia , Cauda/irrigação sanguínea
18.
Chemistry ; 20(42): 13793-800, 2014 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-25179684

RESUMO

By combining NMR spectroscopy, transmission electron microscopy, and circular dichroism we have identified the structural determinants involved in the interaction of green tea catechins with Aß1-42, PrP106-126, and ataxin-3 oligomers. The data allow the elucidation of their mechanism of action, showing that the flavan-3-ol unit of catechins is essential for interaction. At the same time, the gallate moiety, when present, seems to increase the affinity for the target proteins. These results provide important information for the rational design of new compounds with anti-amyloidogenic activity and/or molecular tools for the specific targeting of amyloid aggregates in vivo.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Catequina/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/prevenção & controle , Proteínas Nucleares/metabolismo , Fragmentos de Peptídeos/metabolismo , Príons/metabolismo , Agregação Patológica de Proteínas/prevenção & controle , Proteínas Repressoras/metabolismo , Chá/química , Sequência de Aminoácidos , Peptídeos beta-Amiloides/química , Ataxina-3 , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Catequina/química , Flavonoides/química , Flavonoides/farmacologia , Humanos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Doenças Neurodegenerativas/metabolismo , Ressonância Magnética Nuclear Biomolecular , Proteínas Nucleares/química , Fragmentos de Peptídeos/química , Príons/química , Agregação Patológica de Proteínas/metabolismo , Proteínas Repressoras/química
19.
Neuropharmacology ; 81: 166-75, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24486383

RESUMO

Spinocerebellar ataxias (SCAs), caused by expanded CAG repeats encoding a long polyglutamine (polyQ) tract in the respective proteins, are characterized by the accumulation of intranuclear and cytoplasmic misfolded polyQ aggregation that leads to cell death. Suppression of aggregate formation can inhibit a wide range of downstream pathogenic events and is expected to be a therapeutic strategy for SCAs. Here we show the anti-aggregation potential of Gardenia jasminoides (G. jasminoides) and its components/metabolite geniposide, crocin, and genipin, in ATXN3/Q75-GFP 293 cells, a putative SCA3 cell model. We found the aggregation can be significantly prohibited by G. jasminoides, genipin, geniposide and crocin. Meanwhile, G. jasminoides, genipin, geniposide, and crocin up-regulated anti-oxidative markers NFE2L2, NQO1, GCLC and GSTP1, and reduced the production of reactive oxidative species (ROS) in the same cell models. All of them further inhibited the aggregation in neurally differentiated SH-SY5Y ATXN3/Q75-GFP cells. Our results demonstrate that G. jasminoides, genipin, geniposide and crocin work on polyQ-aggregation reduction by suppressing ROS. These findings indicate the therapeutic applications of G. jasminoides in treating SCAs. Furthermore, oxidative stress inhibition could be a good target for drug development of anti-polyQ aggregation.


Assuntos
Antioxidantes/farmacologia , Monoterpenos/farmacologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Estresse Oxidativo/efeitos dos fármacos , Peptídeos/metabolismo , Extratos Vegetais/farmacologia , Proteínas Repressoras/genética , Ataxina-3 , Compostos de Bifenilo/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Concentração Inibidora 50 , Monoterpenos/química , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuroblastoma/patologia , Proteínas Nucleares/metabolismo , Peptídeos/genética , Picratos/metabolismo , Extratos Vegetais/química , Espécies Reativas de Oxigênio/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Tempo , Transfecção
20.
Sleep ; 36(8): 1129-38, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23904672

RESUMO

STUDY OBJECTIVES: Narcolepsy is caused by selective loss of the orexin/hypocretin-producing neurons of the hypothalamus. For patients with narcolepsy, chronic sleepiness is often the most disabling symptom, but current therapies rarely normalize alertness and do not address the underlying orexin deficiency. We hypothesized that the sleepiness of narcolepsy would substantially improve if orexin signaling were restored in specific brain regions at appropriate times of day. DESIGN: We used gene therapy to restore orexin signaling in a mouse model of narcolepsy. In these Atx mice, expression of a toxic protein (ataxin-3) selectively kills the orexin neurons. INTERVENTIONS: To induce ectopic expression of the orexin neuropeptides, we microinjected an adeno-associated viral vector coding for prepro-orexin plus a red fluorescence protein (AAV-orexin) into the mediobasal hypothalamus of Atx and wild-type mice. Control mice received an AAV coding only for red fluorescence protein. Two weeks later, we recorded sleep/wake behavior, locomotor activity, and body temperature and examined the patterns of orexin expression. MEASUREMENTS AND RESULTS: Atx mice rescued with AAV-orexin produced long bouts of wakefulness and had a normal diurnal pattern of arousal, with the longest bouts of wake and the highest amounts of locomotor activity in the first hours of the night. In addition, AAV-orexin improved the timing of rapid eye movement sleep and the consolidation of nonrapid eye movement sleep in Atx mice. CONCLUSIONS: These substantial improvements in sleepiness and other symptoms of narcolepsy demonstrate the effectiveness of orexin gene therapy in a mouse model of narcolepsy. Additional work is needed to optimize this approach, but in time, AAV-orexin could become a useful therapeutic option for patients with narcolepsy.


Assuntos
Terapia Genética/métodos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Narcolepsia/terapia , Neuropeptídeos/genética , Vigília/genética , Animais , Ataxina-3 , Modelos Animais de Doenças , Hipotálamo/citologia , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Masculino , Camundongos , Camundongos Transgênicos/genética , Camundongos Transgênicos/fisiologia , Narcolepsia/genética , Neuropeptídeos/biossíntese , Neuropeptídeos/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Orexinas , Polissonografia , Sono/genética , Sono/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Vigília/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA