Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Ann Clin Transl Neurol ; 9(8): 1310-1315, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35726838

RESUMO

CAG repeat-expansion spinocerebellar ataxias (CAG-SCAs) are genetically defined multisystemic degenerative diseases, resulting in motor symptoms including dysarthria with a substantial impact on daily living. Whilst speech therapy is widely recommended in ataxia, very limited evidence exists for its use. We evaluated the efficacy of a home-delivered, ataxia-tailored biofeedback-driven speech therapy in CAG-SCA in 16 individuals with SCA1, 2, 3, or 6. Treatment was delivered intensively over 20 days. Efficacy was evaluated by blinded ratings of intelligibility (primary) and acoustic measures (secondary) leveraging an intra-individual control design. Intelligibility improved post-treatment (Z = -3.18, p = 0.004) whilst remaining stable prior to treatment (Z = 0.53, p = 1.00).


Assuntos
Disartria , Ataxias Espinocerebelares , Biorretroalimentação Psicológica , Disartria/etiologia , Disartria/terapia , Humanos , Fala , Ataxias Espinocerebelares/genética , Expansão das Repetições de Trinucleotídeos
4.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34479994

RESUMO

Patterned degeneration of Purkinje cells (PCs) can be observed in a wide range of neuropathologies, but mechanisms behind nonrandom cerebellar neurodegeneration remain unclear. Sphingolipid metabolism dyshomeostasis typically leads to PC neurodegeneration; hence, we questioned whether local sphingolipid balance underlies regional sensitivity to pathological insults. Here, we investigated the regional compartmentalization of sphingolipids and their related enzymes in the cerebellar cortex in healthy and pathological conditions. Analysis in wild-type animals revealed higher sphingosine kinase 1 (Sphk1) levels in the flocculonodular cerebellum, while sphingosine-1-phosphate (S1P) levels were higher in the anterior cerebellum. Next, we investigated a model for spinocerebellar ataxia type 1 (SCA1) driven by the transgenic expression of the expanded Ataxin 1 protein with 82 glutamine (82Q), exhibiting severe PC degeneration in the anterior cerebellum while the flocculonodular region is preserved. In Atxn1[82Q]/+ mice, we found that levels of Sphk1 and Sphk2 were region-specific decreased and S1P levels increased, particularly in the anterior cerebellum. To determine if there is a causal link between sphingolipid levels and neurodegeneration, we deleted the Sphk1 gene in Atxn1[82Q]/+ mice. Analysis of Atxn1[82Q]/+; Sphk1-/- mice confirmed a neuroprotective effect, rescuing a subset of PCs in the anterior cerebellum, in domains reminiscent of the modules defined by AldolaseC expression. Finally, we showed that Sphk1 deletion acts on the ATXN1[82Q] protein expression and prevents PC degeneration. Taken together, our results demonstrate that there are regional differences in sphingolipid metabolism and that this metabolism is directly involved in PC degeneration in Atxn1[82Q]/+ mice.


Assuntos
Ataxina-1/metabolismo , Células de Purkinje/metabolismo , Esfingolipídeos/metabolismo , Animais , Ataxina-1/genética , Encéfalo/metabolismo , Doenças Cerebelares/fisiopatologia , Cerebelo/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Proteínas Nucleares/metabolismo , Ataxias Espinocerebelares/genética
5.
Mol Pharm ; 18(2): 610-626, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32584043

RESUMO

Polyglutamine (polyQ) diseases, such as Huntington's disease and several types of spinocerebellar ataxias, are dominantly inherited progressive neurodegenerative disorders and characterized by the presence of expanded CAG trinucleotide repeats in the respective disease locus of the patient genomes. Patients with polyQ diseases currently need to rely on symptom-relieving treatments because disease-modifying therapeutic interventions remain scarce. Many disease-modifying therapeutic agents are now under clinical testing for treating polyQ diseases, but their delivery to the brain is often too invasive (e.g., intracranial injection) or inefficient, owing to in vivo degradation and clearance by physiological barriers (e.g., oral and intravenous administration). Nanoparticles provide a feasible solution for improving drug delivery to the brain, as evidenced by an increasing number of preclinical studies that document the efficacy of nanomedicines for polyQ diseases over the past 5-6 years. In this review, we present the pathogenic mechanisms of polyQ diseases, the common animal models of polyQ diseases for evaluating the efficacy of nanomedicines, and the common administration routes for delivering nanoparticles to the brain. Next, we summarize the recent preclinical applications of nanomedicines for treating polyQ diseases and improving neurological conditions in vivo, placing emphasis on antisense oligonucleotides, small peptide inhibitors, and small molecules as the disease-modifying agents. We conclude with our perspectives of the burgeoning field of "nanomedicines for polyQ diseases", including the use of inorganic nanoparticles and potential drugs as next-generation nanomedicines, development of higher-order animal models of polyQ diseases, and importance of "brain-nano" interactions.


Assuntos
Portadores de Fármacos/química , Doença de Huntington/tratamento farmacológico , Nanopartículas/química , Fármacos Neuroprotetores/administração & dosagem , Peptídeos/antagonistas & inibidores , Ataxias Espinocerebelares/tratamento farmacológico , Administração Intranasal , Administração Oral , Animais , Animais Geneticamente Modificados , Disponibilidade Biológica , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Loci Gênicos/genética , Humanos , Doença de Huntington/genética , Doença de Huntington/patologia , Injeções Intraperitoneais , Injeções Intravenosas , Injeções Intraventriculares , Injeções Espinhais , Fármacos Neuroprotetores/farmacocinética , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/farmacocinética , Peptídeos/genética , Peptídeos/metabolismo , Permeabilidade , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Distribuição Tecidual , Expansão das Repetições de Trinucleotídeos
6.
J Neurol ; 267(8): 2288-2295, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32314013

RESUMO

OBJECTIVE: To report clinical and ancillary findings in a kindred with spinocerebellar ataxia 38 (SCA38). PATIENTS AND METHODS: Five family members spanning two generations developed gait ataxia and intermittent diplopia. On examination, a cerebellar syndrome accompanied by downbeat nystagmus and a saccadic head impulse test (HIT) were found. RESULTS: Whole-exome sequencing demonstrated a heterozygous variant in ELOVL5, c.779A > G (p.Tyr260Cys), in four tested patients. Intermittent concomitant esotropia and hypertropia caused transient diplopia in one individual each. Saccadic HIT responses were found in four subjects. Sensorineural hypoacusis was present in every case. Electrophysiological studies demonstrated a sensory neuronopathy in patients from the first generation, with prolonged disease duration. Baseline serum docosahexaenoic acid (DHA) percent was diminished in four individuals. Oral 26-week dietary DHA supplementation, 650 mg/day, raised serum DHA percent and induced a statistically significant reduction in Scale for the Assessment and Rating of Ataxia (SARA) total scores, and in stance and heel-shin slide item scores. CONCLUSION: The mentioned ELOVL5 variant segregated with disease in this kindred. Downbeat nystagmus, intermittent heterotropia causing transient diplopia, vestibular impairment demonstrated by abnormal HIT, and sensory neuronopathy were part of the clinical picture in this series. DHA supplementation raised serum DHA percent in cases with diminished levels, and induced a clinical amelioration and a statistically significant reduction in SARA scores in the study group. Further studies are needed to investigate the role of these findings in SCA38, and to determine the response to prolonged DHA supplementation.


Assuntos
Nistagmo Patológico , Ataxias Espinocerebelares , Humanos , Movimentos Sacádicos , Ataxias Espinocerebelares/complicações , Ataxias Espinocerebelares/genética
7.
Parkinsonism Relat Disord ; 63: 191-194, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30862453

RESUMO

INTRODUCTION: Spinocerebellar Ataxia 38 (SCA38) is caused by ELOVL5 gene mutation, with significant reduction of serum docosahexaenoic acid (DHA) levels. DHA supplementation has been proven effective at short-term follow-up. In the present paper, we evaluated long-term safety and efficacy of 600 mg/day oral DHA in SCA38 by a 2-year open label extension study. METHODS: Nine SCA38 patients underwent standardised clinical assessment at 62 (T1), 82 (T2) and 104 (T3) weeks, and compared to pre-treatment scores (T0). Brain 18-Fluorodeoxyglucose Positron Emission Tomography and electroneurography were performed at T0 and T3. RESULTS: We found a significant maintenance of clinical symptom improvement at each follow-up time-point (p < 0.001) as compared to T0, a sustained increase of cerebellar metabolism at T3 as compared to T0 (p = 0.013), and no worsening of neurophysiological parameters. No side effect was recorded. CONCLUSIONS: Long-term DHA supplementation is an eligible treatment for SCA38.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Ataxias Espinocerebelares/tratamento farmacológico , Ataxias Espinocerebelares/fisiopatologia , Adulto , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/efeitos adversos , Estimulação Elétrica , Eletromiografia , Elongases de Ácidos Graxos/genética , Feminino , Fluordesoxiglucose F18 , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Ataxias Espinocerebelares/diagnóstico por imagem , Ataxias Espinocerebelares/genética
8.
Nat Commun ; 8(1): 1864, 2017 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-29192206

RESUMO

YAP and its neuronal isoform YAPdeltaC are implicated in various cellular functions. We found that expression of YAPdeltaC during development, but not adulthood, rescued neurodegeneration phenotypes of mutant ataxin-1 knock-in (Atxn1-KI) mice. YAP/YAPdeltaC interacted with RORα via the second WW domain and served as co-activators of its transcriptional activity. YAP/YAPdeltaC formed a transcriptional complex with RORα on cis-elements of target genes and regulated their expression. Both normal and mutant Atxn1 interacted with YAP/YAPdeltaC, but only mutant Atxn1 depleted YAP/YAPdeltaC from the RORα complex to suppress transcription on short timescales. Over longer periods, mutant Atxn1 also decreased RORα in vivo. Genetic supplementation of YAPdeltaC restored the RORα and YAP/YAPdeltaC levels, recovered YAP/YAPdeltaC in the RORα complex and normalized target gene transcription in Atxn1-KI mice in vivo. Collectively, our data suggest that functional impairment of YAP/YAPdeltaC by mutant Atxn1 during development determines the adult pathology of SCA1 by suppressing RORα-mediated transcription.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ataxina-1/genética , Cerebelo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Neurônios/metabolismo , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Fosfoproteínas/metabolismo , Ataxias Espinocerebelares/genética , Animais , Proteínas de Ciclo Celular , Cerebelo/citologia , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Masculino , Camundongos , Fenótipo , Isoformas de Proteínas , Teste de Desempenho do Rota-Rod , Ataxias Espinocerebelares/fisiopatologia , Proteínas de Sinalização YAP
9.
Ann Neurol ; 82(4): 615-621, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28976605

RESUMO

OBJECTIVE: Spinocerebellar ataxia 38 (SCA38) is caused by mutations in the ELOVL5 gene, which encodes an elongase involved in the synthesis of polyunsaturated fatty acids, including docosahexaenoic acid (DHA). As a consequence, DHA is significantly reduced in the serum of SCA38 subjects. In the present study, we evaluated the safety of DHA supplementation, its efficacy for clinical symptoms, and changes of brain functional imaging in SCA38 patients. METHODS: We enrolled 10 SCA38 patients, and carried out a double-blind randomized placebo-controlled study for 16 weeks, followed by an open-label study with overall 40-week DHA treatment. At baseline and at follow-up visit, patients underwent standardized clinical assessment, brain 18-fluorodeoxyglucose positron emission tomography, electroneurography, and ELOVL5 expression analysis. RESULTS: After 16 weeks, we showed a significant pre-post clinical improvement in the DHA group versus placebo, using the Scale for the Assessment and Rating of Ataxia (SARA; mean difference [MD] = +2.70, 95% confidence interval [CI] = +0.13 to + 5.27, p = 0.042). At 40-week treatment, clinical improvement was found significant by both SARA (MD = +2.2, 95% CI = +0.93 to + 3.46, p = 0.008) and International Cooperative Ataxia Rating Scale (MD = +3.8, 95% CI = +1.39 to + 6.41, p = 0.02) scores; clinical data were corroborated by significant improvement of cerebellar hypometabolism (statistical parametric mapping analyses, false discovery rate corrected). We also showed a decreased expression of ELOVL5 in patients' blood at 40 weeks as compared to baseline. No side effect was recorded. INTERPRETATION: DHA supplementation is a safe and effective treatment for SCA38, showing an improvement of clinical symptoms and cerebellar hypometabolism. Ann Neurol 2017;82:615-621.


Assuntos
Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/uso terapêutico , Ataxias Espinocerebelares/tratamento farmacológico , Adulto , Ataxinas/genética , Encéfalo/diagnóstico por imagem , Método Duplo-Cego , Eletromiografia , Feminino , Fluordesoxiglucose F18/farmacocinética , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Mutação/genética , Avaliação de Resultados em Cuidados de Saúde , Tomografia por Emissão de Pósitrons , Ataxias Espinocerebelares/diagnóstico por imagem , Ataxias Espinocerebelares/genética , Resultado do Tratamento
10.
Hum Mol Genet ; 25(18): 4021-4040, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27466200

RESUMO

A polyglutamine expansion within the ataxin-1 protein (ATXN1) underlies spinocerebellar ataxia type-1 (SCA1), a neurological disorder mainly characterized by ataxia and cerebellar deficits. In SCA1, both loss and gain of ATXN1 biological functions contribute to cerebellar pathogenesis. However, the critical ATXN1 functions and pathways involved remain unclear. To further investigate the early signalling pathways regulated by ATXN1, we performed an unbiased proteomic study of the Atxn1-KO 5-week-old mice cerebellum. Here, we show that lack of ATXN1 expression induces early alterations in proteins involved in glycolysis [pyruvate kinase, muscle, isoform 1 protein (PKM-i1), citrate synthase (CS), glycerol-3-phosphate dehydrogenase 2 (GPD2), glucose-6-phosphate isomerase (GPI), alpha -: enolase (ENO1)], ATP synthesis [CS, Succinate dehydrogenase complex,subunit A (SDHA), ATP synthase subunit d, mitochondrial (ATP5H)] and oxidative stress [peroxiredoxin-6 (PRDX6), aldehyde dehydrogenase family 1, subfamily A1, 10-formyltetrahydrofolate dehydrogenase]. In the SCA1 mice, several of these proteins (PKM-i1, ATP5H, PRDX6, proteome subunit A6) were down-regulated and ATP levels decreased. The underlying mechanism does not involve modulation of mitochondrial biogenesis, but dysregulation of the activity of the metabolic regulators glycogen synthase kinase 3B (GSK3ß), decreased in Atxn1-KO and increased in SCA1 mice, and mechanistic target of rapamycin (serine/threonine kinase) (mTOR), unchanged in the Atxn1-KO and decreased in SCA1 mice cerebellum before the onset of ataxic symptoms. Pharmacological inhibition of GSK3ß and activation of mTOR in a SCA1 cell model ameliorated identified ATXN1-regulated metabolic proteome and ATP alterations. Taken together, these results point to an early role of ATXN1 in the regulation of bioenergetics homeostasis in the mouse cerebellum. Moreover, data suggest GSK3ß and mTOR pathways modulate this ATXN1 function in SCA1 pathogenesis that could be targeted therapeutically prior to the onset of disease symptoms in SCA1 and other pathologies involving dysregulation of ATXN1 functions.


Assuntos
Ataxina-1/genética , Glicogênio Sintase Quinase 3 beta/genética , Ataxias Espinocerebelares/genética , Serina-Treonina Quinases TOR/genética , Trifosfato de Adenosina/metabolismo , Animais , Ataxina-1/biossíntese , Cerebelo/metabolismo , Cerebelo/patologia , Regulação da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/biossíntese , Glicólise/genética , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/patologia , Peptídeos/genética , Proteoma/biossíntese , Proteoma/genética , Transdução de Sinais , Ataxias Espinocerebelares/tratamento farmacológico , Ataxias Espinocerebelares/patologia , Serina-Treonina Quinases TOR/biossíntese
11.
Eur J Hum Genet ; 24(8): 1154-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26669662

RESUMO

Ataxia is a symptom that is often associated with syndromic inherited diseases. We previously reported the linkage of a novel syndrome, ataxia with blindness and deafness (SCAR3/SCABD, OMIM# 271250), to chromosome 6p21-p23 by linkage mapping of an Arab Israeli consanguineous family. We have now identified by whole-exome sequencing a homozygous missense mutation in the Arab Israeli family in the SLC52A2 gene located in 8qter, therefore excluding linkage of this family to 6p. We confirmed the involvement of SLC52A2 by the identification of a second mutation in an independent family with an identical syndromic presentation, which we suggest to name SCABD2. SCABD2 is therefore allelic to Brown-Vialleto-Van Laere syndrome type 2 defined by prominent motoneuronopathy and deafness, and also caused by SLC52A2 mutations. In the course of this project, we identified a clinically similar family with a homozygous missense mutation in PEX6, which is located in 6p21. Therefore, despite false linkage in the initial family, SCABD1/SCAR3 is located in 6p21 and is caused by PEX6 mutations. Both SLC52A2 and PEX6 should be included in screening panels for the diagnosis of syndromic inherited ataxias, particularly as patients with mutations in SLC52A2 can be ameliorated by riboflavin supplementation.


Assuntos
Adenosina Trifosfatases/genética , Cegueira/genética , Surdez/genética , Mutação de Sentido Incorreto , Receptores Acoplados a Proteínas G/genética , Ataxias Espinocerebelares/genética , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética , ATPases Associadas a Diversas Atividades Celulares , Adolescente , Adulto , Cegueira/diagnóstico , Células Cultivadas , Criança , Surdez/diagnóstico , Exoma , Feminino , Humanos , Masculino , Linhagem , Ataxias Espinocerebelares/diagnóstico , Síndrome
12.
Brain ; 138(Pt 12): 3555-66, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26490326

RESUMO

Spinocerebellar ataxia type 1 is one of nine polyglutamine expansion diseases and is characterized by cerebellar ataxia and neuronal degeneration in the cerebellum and brainstem. Currently, there are no effective therapies for this disease. Previously, we have shown that RNA interference mediated silencing of ATXN1 mRNA provides therapeutic benefit in mouse models of the disease. Adeno-associated viral delivery of an engineered microRNA targeting ATXN1 to the cerebella of well-established mouse models improved motor phenotypes, neuropathy, and transcriptional changes. Here, we test the translatability of this approach in adult rhesus cerebella. Nine adult male and three adult female rhesus macaque were unilaterally injected with our therapeutic vector, a recombinant adeno-associated virus type 1 (rAAV1) expressing our RNAi trigger (miS1) and co-expressing enhanced green fluorescent protein (rAAV1.miS1eGFP) into the deep cerebellar nuclei using magnetic resonance imaging guided techniques combined with a Stealth Navigation system (Medtronics Inc.). Transduction was evident in the deep cerebellar nuclei, cerebellar Purkinje cells, the brainstem and the ventral lateral thalamus. Reduction of endogenous ATXN1 messenger RNA levels were ≥30% in the deep cerebellar nuclei, the cerebellar cortex, inferior olive, and thalamus relative to the uninjected hemisphere. There were no clinical complications, and quantitative and qualitative analyses suggest that this therapeutic intervention strategy and subsequent reduction of ATXN1 is well tolerated. Collectively the data illustrate the biodistribution and tolerability of rAAV1.miS1eGFP administration to the adult rhesus cerebellum and are supportive of clinical application for spinocerebellar ataxia type 1.


Assuntos
Ataxina-1/deficiência , Núcleos Cerebelares/metabolismo , Terapia Genética/métodos , Interferência de RNA , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/terapia , Animais , Ataxina-1/genética , Tronco Encefálico/metabolismo , Dependovirus , Feminino , Macaca mulatta , Masculino , Células de Purkinje/metabolismo , Tálamo/metabolismo , Transdução Genética
13.
Biochim Biophys Acta ; 1847(4-5): 418-428, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25647692

RESUMO

Spinocerebellar ataxia type 7 (SCA7) is one of the nine neurodegenerative disorders caused by expanded polyglutamine (polyQ) domains. Common pathogenic mechanisms, including bioenergetics defects, have been suggested for these so called polyQ diseases. However, the exact molecular mechanism(s) behind the metabolic dysfunction is still unclear. In this study we identified a previously unreported mechanism, involving disruption of p53 and NADPH oxidase 1 (NOX1) activity, by which the expanded SCA7 disease protein ATXN7 causes metabolic dysregulation. The NOX1 protein is known to promote glycolytic activity, whereas the transcription factor p53 inhibits this process and instead promotes mitochondrial respiration. In a stable inducible PC12 model of SCA7, p53 and mutant ATXN7 co-aggregated and the transcriptional activity of p53 was reduced, resulting in a 50% decrease of key p53 target proteins, like AIF and TIGAR. In contrast, the expression of NOX1 was increased approximately 2 times in SCA7 cells. Together these alterations resulted in a decreased respiratory capacity, an increased reliance on glycolysis for energy production and a subsequent 20% reduction of ATP in SCA7 cells. Restoring p53 function, or suppressing NOX1 activity, both reversed the metabolic dysfunction and ameliorated mutant ATXN7 toxicity. These results hence not only enhance the understanding of the mechanisms causing metabolic dysfunction in SCA7 disease, but also identify NOX1 as a novel potential therapeutic target in SCA7 and possibly other polyQ diseases.


Assuntos
Modelos Animais de Doenças , NADH NADPH Oxirredutases/metabolismo , Proteínas do Tecido Nervoso/deficiência , Peptídeos/genética , Ataxias Espinocerebelares/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Fator de Indução de Apoptose/genética , Fator de Indução de Apoptose/metabolismo , Proteínas Reguladoras de Apoptose , Ataxina-7 , Western Blotting , Metabolismo Energético , Glucose , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Células HeLa , Humanos , Técnicas Imunoenzimáticas , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ácido Láctico , Potencial da Membrana Mitocondrial , Mutação/genética , NADH NADPH Oxirredutases/genética , NADPH Oxidase 1 , Consumo de Oxigênio , Células PC12 , Monoéster Fosfórico Hidrolases , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Proteína Supressora de Tumor p53/genética
14.
BMC Mol Biol ; 13: 6, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22397573

RESUMO

BACKGROUND: RNA interference (RNAi) and antisense strategies provide experimental therapeutic agents for numerous diseases, including polyglutamine (polyQ) disorders caused by CAG repeat expansion. We compared the potential of different oligonucleotide-based strategies for silencing the genes responsible for several polyQ diseases, including Huntington's disease and two spinocerebellar ataxias, type 1 and type 3. The strategies included nonallele-selective gene silencing, gene replacement, allele-selective SNP targeting and CAG repeat targeting. RESULTS: Using the patient-derived cell culture models of polyQ diseases, we tested various siRNAs, and antisense reagents and assessed their silencing efficiency and allele selectivity. We showed considerable allele discrimination by several SNP targeting siRNAs based on a weak G-G or G-U pairing with normal allele and strong G-C pairing with mutant allele at the site of RISC-induced cleavage. Among the CAG repeat targeting reagents the strongest allele discrimination is achieved by miRNA-like functioning reagents that bind to their targets and inhibit their translation without substantial target cleavage. Also, morpholino analog performs well in mutant and normal allele discrimination but its efficient delivery to cells at low effective concentration still remains a challenge. CONCLUSIONS: Using three cellular models of polyQ diseases and the same experimental setup we directly compared the performance of different oligonucleotide-based treatment strategies that are currently under development. Based on the results obtained by us and others we discussed the advantages and drawbacks of these strategies considering them from several different perspectives. The strategy aimed at nonallele-selective inhibiting of causative gene expression by targeting specific sequence of the implicated gene is the easiest to implement but relevant benefits are still uncertain. The gene replacement strategy that combines the nonallele-selective gene silencing with the expression of the exogenous normal allele is a logical extension of the former and it deserves to be explored further. Both allele-selective RNAi approaches challenge cellular RNA interference machinery to show its ability to discriminate between similar sequences differing in either single base substitutions or repeated sequence length. Although both approaches perform well in allele discrimination most of our efforts are focused on repeat targeting due to its potentially higher universality.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Doença de Huntington/genética , Oligonucleotídeos/uso terapêutico , Ataxias Espinocerebelares/genética , Alelos , Ataxina-3 , Linhagem Celular , Inativação Gênica , Terapia Genética , Humanos , Proteína Huntingtina , Doença de Huntington/terapia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oligonucleotídeos/genética , Oligonucleotídeos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ataxias Espinocerebelares/terapia
15.
Brain ; 133(Pt 5): 1428-37, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20400524

RESUMO

Mutations in the catalytic subunit of the mitochondrial DNA-polymerase gamma cause a wide spectrum of clinical disease ranging from infantile hepato-encephalopathy to juvenile/adult-onset spinocerebellar ataxia and late onset progressive external ophthalmoplegia. Several of these syndromes are associated with an encephalopathy that characteristically shows episodes of rapid neurological deterioration and the development of acute cerebral lesions. The purpose of this study was to investigate the nature, distribution and natural evolution of central nervous system lesions in polymerase gamma associated encephalopathy focusing particularly on lesions identified by magnetic resonance imaging. We compared radiological, electrophysiological and pathological findings where available to study potential mechanisms underlying the episodes of exacerbation and acute cerebral lesions. We studied a total of 112 magnetic resonance tomographies and 11 computed tomographies in 32 patients with polymerase gamma-encephalopathy, including multiple serial examinations performed during both the chronic and acute phases of the disease and, in several cases, magnetic resonance spectroscopy and serial diffusion weighted studies. Data from imaging, electroencephalography and post-mortem examination were compared in order to study the underlying disease process. Our findings show that magnetic resonance imaging in polymerase gamma-related encephalopathies has high sensitivity and can identify patterns that are specific for individual syndromes. One form of chronic polymerase gamma-encephalopathy, that is associated with the c.1399G > A and c.2243G > C mutations, is characterized by progressive cerebral and cerebellar atrophy and focal lesions of the thalamus, deep cerebellar structures and medulla oblongata. Acute encephalopathies, both infantile and later onset, show similar pictures with cortical stroke-like lesions occurring during episodes of exacerbation. These lesions can occur both with and without electroencephalographic evidence of concurrent epileptic activity, and have diffusion, spectroscopic and histological profiles strongly suggestive of neuronal energy failure. We suggest therefore that both infantile and later onset polymerase gamma related encephalopathies are part of a continuum.


Assuntos
Encefalopatias/genética , Encefalopatias/metabolismo , Encéfalo/metabolismo , DNA Mitocondrial/genética , DNA Polimerase Dirigida por DNA/genética , Metabolismo Energético , Mutação , Arginina , Encéfalo/patologia , Encefalopatias/complicações , Encefalopatias/diagnóstico , Cerebelo/patologia , Cisteína , DNA Polimerase gama , Esclerose Cerebral Difusa de Schilder/genética , Esclerose Cerebral Difusa de Schilder/metabolismo , Imagem de Difusão por Ressonância Magnética , Progressão da Doença , Eletroencefalografia , Epilepsia/etiologia , Epilepsia/genética , Epilepsia/metabolismo , Glicina , Humanos , Angiografia por Ressonância Magnética , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Doenças Mitocondriais/genética , Doenças Mitocondriais/metabolismo , Neocórtex/patologia , Sensibilidade e Especificidade , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/metabolismo , Acidente Vascular Cerebral/etiologia , Síndrome , Tálamo/patologia , Tomografia Computadorizada por Raios X
16.
J Neurol Sci ; 290(1-2): 146-7, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20045119

RESUMO

Autosomal dominant spinocerebellar ataxias (SCAs) are progressive neurodegenerative disorders which result in dysfunction of the neuronal systems of the spinal cord, brainstem, and cerebellum. The manifestations of daytime somnolence and abnormal sleep behavior have been described in SCA type 3 (SCA3) and SCA type 6 (SCA6), but as yet have not been described in SCA type 1 (SCA1). We report two cases of sleep disturbance, fatigue and excessive daytime somnolence in individuals with SCA1 and their progress through several therapies. These case studies are unique as they describe excessive daytime somnolence and sleep abnormalities in SCA1.


Assuntos
Distúrbios do Sono por Sonolência Excessiva/genética , Distúrbios do Sono por Sonolência Excessiva/fisiopatologia , Predisposição Genética para Doença/genética , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/fisiopatologia , Atividades Cotidianas , Amantadina/uso terapêutico , Ataxina-1 , Ataxinas , Tronco Encefálico/fisiopatologia , Cerebelo/fisiopatologia , Citalopram/uso terapêutico , Análise Mutacional de DNA , Dextroanfetamina/uso terapêutico , Progressão da Doença , Feminino , Marcadores Genéticos , Testes Genéticos , Humanos , Pessoa de Meia-Idade , Mutação/genética , Naturologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Pergolida/uso terapêutico , Polissonografia , Ataxias Espinocerebelares/complicações , Resultado do Tratamento
17.
Results Probl Cell Differ ; 44: 223-42, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17589814

RESUMO

The nicotinic acetylcholine receptors (nAChRs) are members of the large family of ligand-gated ion channels, and are constituted by the assembly of five subunits arranged pseudosymmetrically around the central axis that forms a cation-selective ion pore. They are widely distributed in both the nervous system and non-neuronal tissues, and can be activated by endogenous agonists such as acetylcholine or exogenous ligands such as nicotine. Mutations in neuronal nAChRs are found in a rare form of familial nocturnal frontal lobe epilepsy (ADNFLE), while mutations in the neuromuscular subtype of the nAChR are responsible for either congenital myasthenia syndromes (adult subtype of neuromuscular nAChR) or a form of arthrogryposis multiplex congenita type Escobar (fetal subtype of neuromuscular nAChR).


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Nicotina/metabolismo , Receptores Nicotínicos/fisiologia , Transmissão Sináptica/fisiologia , Sequência de Aminoácidos , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Epilepsia do Lobo Frontal/genética , Epilepsia do Lobo Frontal/metabolismo , Humanos , Transtornos de Enxaqueca/genética , Transtornos de Enxaqueca/metabolismo , Dados de Sequência Molecular , Mutação , Síndromes Miastênicas Congênitas/genética , Síndromes Miastênicas Congênitas/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Receptores Nicotínicos/genética , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Canais de Sódio/genética , Canais de Sódio/fisiologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/metabolismo
18.
Gene ; 380(1): 54-61, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16876337

RESUMO

A small expansion of a CAG repeat domain in exon 47 of the human CACNA1A gene, which codes for the pore-forming alpha1A subunit of P/Q-type Ca2+ channels, causes spinocerebellar ataxia type-6. Only the human alpha1A protein has been demonstrated to contain the poly(Q) tract, although this locus has also recently been detected in ape genomes. To our knowledge, no further information has been published on other mammal species. Here, we have cloned the full-length alpha1A subunit in a non-primate species, the cow. The results have made it possible to explore the exon organization of the bovine CACNA1A gene as well as the splice alpha1A isoforms expressed by bovine chromaffin cells. We found a splice variant of the protein that, as in humans, also contains a polymorphic poly(Q) tract. Based on this result and using data from different Genome Databases, we performed an interspecies comparison of exon 47 and discovered that the poly(Q) tract is present in all the species studied, with the exception of primitive fish and rodents. Our results provide insight into the evolution of the CAG repeat tract at the C-terminus coding region of the CACNA1A gene.


Assuntos
Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo Q/genética , Ataxias Espinocerebelares/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Canais de Cálcio/genética , Bovinos , Células Cultivadas , Células Cromafins/metabolismo , Clonagem Molecular , Primers do DNA/genética , DNA Complementar/genética , Evolução Molecular , Éxons , Humanos , Íntrons , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Repetições de Trinucleotídeos
19.
AJNR Am J Neuroradiol ; 27(7): 1463-6, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16908559

RESUMO

BACKGROUND AND PURPOSE: Dentatorubral-pallidoluysian atrophy (DRPLA) is an autosomal dominant spinocerebellar ataxia. Techniques for the quantitative assessment of neurodegenerative lesions remain to be established in this disease. We attempted to quantify global and region-specific neurodegeneration in DRPLA using analysis of apparent diffusion coefficient (ADC) maps. METHODS: Diffusion-weighted images (b = 1000 s/mm(2)) by echo-planar sequences were obtained with the use of a 1.5T clinical scanner. Whole-brain histogram and region of interest (ROI) analyses of ADC values as well as conventional MR imaging studies were performed in 6 patients with genetically confirmed DRPLA. RESULTS: Histograms demonstrated significantly higher mean ADC values in the patients than in age- and sex-matched control subjects (P < .01). ROI analysis revealed that the patients had significantly higher ADC values in the cerebellum and globus pallidus, preferentially affected regions (P < .05), but not in the thalamus, the region relatively spared in this disease. ADC values in the white matter were higher only in patients with adult-onset disease. Histogram analyses could more sensitively identify abnormalities than ROI analyses, because the former avoided errors associated with setting ROIs and thus had smaller P values on statistical analysis than the latter. CONCLUSIONS: Histogram ADC analyses were more sensitive for the detection of neurodegeneration in DRPLA than ROI analyses, whereas ROI analyses revealed regional alterations reflecting the distribution of pathologic changes. Thus, histogram and ROI analyses complement each other and may permit the sensitive, quantitative evaluation of neurodegeneration in DRPLA, especially that involving the globus pallidus showing normal T2 signals.


Assuntos
Encéfalo/patologia , Imageamento por Ressonância Magnética/métodos , Epilepsias Mioclônicas Progressivas/patologia , Adulto , Estudos de Casos e Controles , Cerebelo/patologia , Imagem de Difusão por Ressonância Magnética/métodos , Imagem Ecoplanar/métodos , Feminino , Globo Pálido/patologia , Humanos , Aumento da Imagem/métodos , Processamento de Imagem Assistida por Computador/métodos , Masculino , Pessoa de Meia-Idade , Epilepsias Mioclônicas Progressivas/genética , Ataxias Espinocerebelares/genética , Tálamo/patologia
20.
Brain ; 126(Pt 10): 2257-72, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12847080

RESUMO

In spite of the considerable progress in clinical and molecular research, knowledge regarding brain damage in spinocerebellar ataxia type 2 (SCA2) and type 3 (SCA3) still is limited and the extent to which the thalamus is involved in both diseases is uncertain. Accordingly, we performed a pathoanatomical analysis on serial thick sections stained for lipofuscin granules and Nissl substance through the thalami of two genetically confirmed cases: one an SCA2 patient, the other an SCA3 patient. During this systematic study, we detected severe destruction of the reticular (RT), fasciculosus (FA), ventral anterior (VA), ventral lateral (VL), ventral posterior lateral (VPL), ventral posterior medial (VPM), cucullar (CU) and mediodorsal thalamic nuclei (MD), the lateral geniculate body (LGB) and inferior nucleus of the pulvinar (PU i) in the SCA2 case, and a severe neuronal loss in the RT, FA, VA and PU i of the SCA3 case. In the SCA2 patient, additional obvious neuronal loss was observed in all nuclei of the anterior and rostral intra laminar groups, in the lateral posterior nucleus (LP), the lateral (PU l) and the medial subnuclei of the pulvinar (PU m), whereas in the SCA3 patient only two of the nuclei that belong to the anterior thalamic group, the VL, VPL, VPM, LP, LGB, PU l and PU m, displayed marked neurodegeneration. These novel findings indicate that thalamic involvement in SCA2 and SCA3 patients has been underestimated in the past. In view of what is known about the functions of the affected thalamic nuclei, the present findings provide an appropriate pathoanatomical explanation for some of the disease-related symptoms seen in both of our and other SCA2 and SCA3 patients: gait, stance, truncal and limb ataxia, dysarthria or anarthria, falls, dysdiadochokinesia and bradykinesia, problems with writing, somatosensory deficits, saccadic dysfunctions, executive dysfunctions and abnormalities of visual evoked potentials.


Assuntos
Ataxias Espinocerebelares/patologia , Tálamo/patologia , Adolescente , Idoso , Feminino , Humanos , Lipofuscina/análise , Corpos de Nissl/patologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/metabolismo , Tálamo/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA