Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 134: 104564, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31381977

RESUMO

Expression of mutant Ataxin-1 with an abnormally expanded polyglutamine domain is necessary for the onset and progression of spinocerebellar ataxia type 1 (SCA1). Understanding how Ataxin-1 expression is regulated in the human brain could inspire novel molecular therapies for this fatal, dominantly inherited neurodegenerative disease. Previous studies have shown that the ATXN1 3'UTR plays a key role in regulating the Ataxin-1 cellular pool via diverse post-transcriptional mechanisms. Here we show that elements within the ATXN1 5'UTR also participate in the regulation of Ataxin-1 expression. PCR and PacBio sequencing analysis of cDNA obtained from control and SCA1 human brain samples revealed the presence of three major, alternatively spliced ATXN1 5'UTR variants. In cell-based assays, fusion of these variants upstream of an EGFP reporter construct revealed significant and differential impacts on total EGFP protein output, uncovering a type of genetic rheostat-like function of the ATXN1 5'UTR. We identified ribosomal scanning of upstream AUG codons and increased transcript instability as potential mechanisms of regulation. Importantly, transcript-based analyses revealed significant differences in the expression pattern of ATXN1 5'UTR variants between control and SCA1 cerebellum. Together, the data presented here shed light into a previously unknown role for the ATXN1 5'UTR in the regulation of Ataxin-1 and provide new opportunities for the development of SCA1 therapeutics.


Assuntos
Regiões 5' não Traduzidas/fisiologia , Ataxina-1/genética , Ataxina-1/metabolismo , Regulação da Expressão Gênica/fisiologia , Ataxias Espinocerebelares , Cerebelo , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
2.
Lab Invest ; 99(7): 1030-1040, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31019288

RESUMO

RNA binding proteins associated with amyotrophic lateral sclerosis (ALS) and muscle myopathy possess sequence elements that are low in complexity, or bear resemblance to yeast prion domains. These sequence elements appear to mediate phase separation into liquid-like membraneless organelles. Using fusion proteins of matrin 3 (MATR3) to yellow fluorescent protein (YFP), we recently observed that deletion of the second RNA recognition motif (RRM2) caused the protein to phase separate and form intranuclear liquid-like droplets. Here, we use fusion constructs of MATR3, TARDBP43 (TDP43) and FUS with YFP or mCherry to examine phase separation and protein colocalization in mouse C2C12 myoblast cells. We observed that the N-terminal 397 amino acids of MATR3 (tagged with a nuclear localization signal and expressed as a fusion protein with YFP) formed droplet-like structures within nuclei. Introduction of the myopathic S85C mutation into NLS-N397 MATR3:YFP, but not ALS mutations F115C or P154S, inhibited droplet formation. Further, we analyzed interactions between variants of MATR3 lacking RRM2 (ΔRRM2) and variants of TDP43 with disabling mutations in its RRM1 domain (deletion or mutation). We observed that MATR3:YFP ΔRRM2 formed droplets that appeared to recruit the TDP43 RRM1 mutants. Further, coexpression of the NLS-397 MATR3:YFP construct with a construct that encodes the prion-like domain of TDBP43 produced intranuclear droplet-like structures containing both proteins. Collectively, our studies show that N-terminal sequences in MATR3 can mediate phase separation into intranuclear droplet-like structures that can recruit TDP43 under conditions of low RNA binding.


Assuntos
Esclerose Lateral Amiotrófica/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Ligação a RNA/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos , Doenças Musculares/genética , Proteínas Associadas à Matriz Nuclear/genética , Proteínas de Ligação a RNA/genética
3.
J Exp Med ; 216(3): 539-555, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30770411

RESUMO

It has been challenging to produce ex vivo models of the inclusion pathologies that are hallmark pathologies of many neurodegenerative diseases. Using three-dimensional mouse brain slice cultures (BSCs), we have developed a paradigm that rapidly and robustly recapitulates mature neurofibrillary inclusion and Lewy body formation found in Alzheimer's and Parkinson's disease, respectively. This was achieved by transducing the BSCs with recombinant adeno-associated viruses (rAAVs) that express α-synuclein or variants of tau. Notably, the tauopathy BSC model enables screening of small molecule therapeutics and tracking of neurodegeneration. More generally, the rAAV BSC "toolkit" enables efficient transduction and transgene expression from neurons, microglia, astrocytes, and oligodendrocytes, alone or in combination, with transgene expression lasting for many months. These rAAV-based BSC models provide a cost-effective and facile alternative to in vivo studies, and in the future can become a widely adopted methodology to explore physiological and pathological mechanisms related to brain function and dysfunction.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Dependovirus/genética , Doença de Parkinson/patologia , Doença de Alzheimer/virologia , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Avaliação Pré-Clínica de Medicamentos/métodos , Expressão Gênica , Humanos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Microrganismos Geneticamente Modificados , Mutação , Neurônios/patologia , Técnicas de Cultura de Órgãos , Doença de Parkinson/virologia , Transdução Genética , Transgenes , alfa-Sinucleína/genética , Proteínas tau/genética
4.
Sci Rep ; 8(1): 4049, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29511296

RESUMO

To understand how mutations in Matrin 3 (MATR3) cause amyotrophic lateral sclerosis (ALS) and distal myopathy, we used transcriptome and interactome analysis, coupled with microscopy. Over-expression of wild-type (WT) or F115C mutant MATR3 had little impact on gene expression in neuroglia cells. Only 23 genes, expressed at levels of >100 transcripts showed ≥1.6-fold changes in expression by transfection with WT or mutant MATR3:YFP vectors. We identified ~123 proteins that bound MATR3, with proteins associated with stress granules and RNA processing/splicing being prominent. The interactome of myopathic S85C and ALS-variant F115C MATR3 were virtually identical to WT protein. Deletion of RNA recognition motif (RRM1) or Zn finger motifs (ZnF1 or ZnF2) diminished the binding of a subset of MATR3 interacting proteins. Remarkably, deletion of the RRM2 motif caused enhanced binding of >100 hundred proteins. In live cells, MATR3 lacking RRM2 (ΔRRM2) formed intranuclear spherical structures that fused over time into large structures. Our findings in the cell models used here suggest that MATR3 with disease-causing mutations is not dramatically different from WT protein in modulating gene regulation or in binding to normal interacting partners. The intra-nuclear localization and interaction network of MATR3 is strongly modulated by its RRM2 domain.


Assuntos
Esclerose Lateral Amiotrófica/genética , Regulação da Expressão Gênica , Doenças Musculares/genética , Mutação de Sentido Incorreto , Proteínas Associadas à Matriz Nuclear/genética , Mapas de Interação de Proteínas , Proteínas de Ligação a RNA/genética , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Modelos Biológicos , Ligação Proteica , Domínios Proteicos , Mapeamento de Interação de Proteínas , Deleção de Sequência
5.
Mol Ther ; 21(10): 1909-18, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23820820

RESUMO

Spinocerebellar ataxia type 3 (SCA3) is a neurodegenerative disease caused by a polyglutamine expansion in the deubiquitinating enzyme, Ataxin-3. Currently, there are no effective treatments for this fatal disorder but studies support the hypothesis that reducing mutant Ataxin-3 protein levels might reverse or halt the progression of disease in SCA3. Here, we sought to modulate ATXN3 expression in vivo using RNA interference. We developed artificial microRNA mimics targeting the 3'-untranslated region (3'UTR) of human ATXN3 and then used recombinant adeno-associated virus to deliver them to the cerebellum of transgenic mice expressing the full human disease gene (SCA3/MJD84.2 mice). Anti-ATXN3 microRNA mimics effectively suppressed human ATXN3 expression in SCA3/MJD84.2 mice. Short-term treatment cleared the abnormal nuclear accumulation of mutant Ataxin-3 throughout the transduced SCA3/MJD84.2 cerebellum. Analysis also revealed changes in the steady-state levels of specific microRNAs in the cerebellum of SCA3/MJD84.2 mice, a previously uncharacterized molecular phenotype of SCA3 that appears to be dependent on mutant Ataxin-3 expression. Our findings support the preclinical development of molecular therapies aimed at halting the expression of ATXN3 as a viable approach to SCA3 and point to microRNA deregulation as a potential surrogate marker of SCA3 pathogenesis.


Assuntos
Doença de Machado-Joseph/patologia , MicroRNAs/efeitos adversos , Proteínas Mutantes/efeitos dos fármacos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas Nucleares/efeitos dos fármacos , Fenótipo , Proteínas Repressoras/efeitos dos fármacos , Regiões 3' não Traduzidas , Animais , Ataxina-3 , Cerebelo/patologia , Dependovirus/efeitos dos fármacos , Dependovirus/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Inativação Gênica , Vetores Genéticos/efeitos dos fármacos , Vetores Genéticos/genética , Células HEK293 , Humanos , Doença de Machado-Joseph/genética , Camundongos , Camundongos Transgênicos , MicroRNAs/farmacologia , Mimetismo Molecular , Terapia de Alvo Molecular , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução Genética/métodos
6.
Mol Ther ; 21(10): 1898-908, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23765441

RESUMO

Machado-Joseph disease (MJD) is a dominantly inherited ataxia caused by a polyglutamine-coding expansion in the ATXN3 gene. Suppressing expression of the toxic gene product represents a promising approach to therapy for MJD and other polyglutamine diseases. We performed an extended therapeutic trial of RNA interference (RNAi) targeting ATXN3 in a mouse model expressing the full human disease gene and recapitulating key disease features. Adeno-associated virus (AAV) encoding a microRNA (miRNA)-like molecule, miRATXN3, was delivered bilaterally into the cerebellum of 6- to 8-week-old MJD mice, which were then followed up to end-stage disease to assess the safety and efficacy of anti-ATXN3 RNAi. Despite effective, lifelong suppression of ATXN3 in the cerebellum and the apparent safety of miRATXN3, motor impairment was not ameliorated in treated MJD mice and survival was not prolonged. These results with an otherwise effective RNAi agent suggest that targeting a large extent of the cerebellum alone may not be sufficient for effective human therapy. Artificial miRNAs or other nucleotide-based suppression strategies targeting ATXN3 more widely in the brain should be considered in future preclinical tests.


Assuntos
Doença de Machado-Joseph/terapia , MicroRNAs/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Interferência de RNA , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Animais , Ataxina-3 , Cerebelo/metabolismo , Cerebelo/patologia , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Vetores Genéticos , Humanos , Doença de Machado-Joseph/metabolismo , Doença de Machado-Joseph/patologia , Doença de Machado-Joseph/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Transdução Genética
7.
Neurobiol Dis ; 54: 456-63, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23376683

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disorder caused by polyglutamine repeat expansions in Ataxin-1. Recent evidence supports a role for microRNAs (miRNAs) deregulation in SCA1 pathogenesis. However, the extent to which miRNAs may modulate the onset, progression or severity of SCA1 remains largely unknown. In this study, we used a mouse model of SCA1 to determine if miRNAs are misregulated in pre- and post-symptomatic SCA1 cerebellum. We found a significant alteration in the steady-state levels of numerous miRNAs prior to and following phenotypic onset. In addition, we provide evidence that increased miR-150 levels in SCA1 Purkinje neurons may modulate disease pathogenesis by targeting the expression of Rgs8 and Vegfa.


Assuntos
MicroRNAs/genética , Células de Purkinje/metabolismo , Ataxias Espinocerebelares/genética , Animais , Ataxina-1 , Ataxinas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Reação em Cadeia da Polimerase , Proteínas RGS/genética , Proteínas RGS/metabolismo , Ataxias Espinocerebelares/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Mol Cell ; 43(4): 599-612, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21855799

RESUMO

The mechanisms by which ubiquitin ligases are regulated remain poorly understood. Here we describe a series of molecular events that coordinately regulate CHIP, a neuroprotective E3 implicated in protein quality control. Through their opposing activities, the initiator E2, Ube2w, and the specialized deubiquitinating enzyme (DUB), ataxin-3, participate in initiating, regulating, and terminating the CHIP ubiquitination cycle. Monoubiquitination of CHIP by Ube2w stabilizes the interaction between CHIP and ataxin-3, which through its DUB activity limits the length of chains attached to CHIP substrates. Upon completion of substrate ubiquitination, ataxin-3 deubiquitinates CHIP, effectively terminating the reaction. Our results suggest that functional pairing of E3s with ataxin-3 or similar DUBs represents an important point of regulation in ubiquitin-dependent protein quality control. In addition, the results shed light on disease pathogenesis in SCA3, a neurodegenerative disorder caused by polyglutamine expansion in ataxin-3.


Assuntos
Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Enzimas de Conjugação de Ubiquitina/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Ataxina-3 , Humanos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Estresse Fisiológico , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação
9.
Neurobiol Dis ; 43(3): 533-42, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21550405

RESUMO

Spinocerebellar ataxia type 6 (SCA6) is an inherited neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the Ca(V)2.1 voltage-gated calcium channel subunit (CACNA1A). There is currently no treatment for this debilitating disorder and thus a pressing need to develop preventative therapies. RNA interference (RNAi) has proven effective at halting disease progression in several models of spinocerebellar ataxia (SCA), including SCA types 1 and 3. However, in SCA6 and other dominantly inherited neurodegenerative disorders, RNAi-based strategies that selectively suppress expression of mutant alleles may be required. Using a Ca(V)2.1 mini-gene reporter system, we found that pathogenic CAG expansions in Ca(V)2.1 enhance splicing activity at the 3'end of the transcript, leading to a CAG repeat length-dependent increase in the levels of a polyQ-encoding Ca(V)2.1 mRNA splice isoform and the resultant disease protein. Taking advantage of this molecular phenomenon, we developed a novel splice isoform-specific (SIS)-RNAi strategy that selectively targets the polyQ-encoding Ca(V)2.1 splice variant. Selective suppression of transiently expressed and endogenous polyQ-encoding Ca(V)2.1 splice variants was achieved in a variety of cell-based models including a human neuronal cell line, using a new artificial miRNA-like delivery system. Moreover, the efficacy of gene silencing correlated with effective intracellular recognition and processing of SIS-RNAi miRNA mimics. These results lend support to the preclinical development of SIS-RNAi as a potential therapy for SCA6 and other dominantly inherited diseases.


Assuntos
Processamento Alternativo/genética , Canais de Cálcio Tipo N/genética , Canais de Cálcio/genética , Terapia Genética/métodos , Variação Genética/genética , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/terapia , Linhagem Celular Tumoral , Humanos , Neuroblastoma , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Splicing de RNA/genética , RNA Mensageiro/biossíntese , Sequências Repetitivas de Ácido Nucleico/genética , Transcrição Gênica/genética
10.
Hum Mol Genet ; 20(R1): R21-7, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21459775

RESUMO

RNAi interference (RNAi) is a powerful gene silencing technology that has immense potential for treating a vast array of human ailments, for which suppressing disease-associated genes may provide clinical benefit. Here, we review the development of RNAi as a therapeutic modality for neurodegenerative diseases affecting the central nervous system (CNS). We overview promising preclinical data for the application of RNAi in the CNS and discuss key challenges (e.g. delivery and specificity) that remain as these approaches transition to the clinic.


Assuntos
Terapia Genética/métodos , Doenças Neurodegenerativas/terapia , Interferência de RNA , Encéfalo/patologia , Sistema Nervoso Central , Inativação Gênica , Humanos , Modelos Biológicos , Doenças Neurodegenerativas/genética , Peptídeos/genética
11.
Mol Ther ; 17(9): 1563-73, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19532137

RESUMO

Despite recent advances suggesting new therapeutic targets, Alzheimer's disease (AD) remains incurable. Aberrant production and accumulation of the Abeta peptide resulting from altered processing of the amyloid precursor protein (APP) is central to the pathogenesis of disease, particularly in dominantly inherited forms of AD. Thus, modulating the production of APP is a potential route to effective AD therapy. Here, we describe the successful use of an allele-specific RNA interference (RNAi) approach targeting the Swedish variant of APP (APPsw) in a transgenic mouse model of AD. Using recombinant adeno-associated virus (rAAV), we delivered an anti-APPsw short-hairpin RNA (shRNA) to the hippocampus of AD transgenic mice (APP/PS1). In short- and long-term transduction experiments, reduced levels of APPsw transprotein were observed throughout targeted regions of the hippocampus while levels of wild-type murine APP remained unaltered. Moreover, intracellular production of transfer RNA (tRNA)-valine promoter-driven shRNAs did not lead to detectable neuronal toxicity. Finally, long-term bilateral hippocampal expression of anti-APPsw shRNA mitigated abnormal behaviors in this mouse model of AD. The difference in phenotype progression was associated with reduced levels of soluble Abeta but not with a reduced number of amyloid plaques. Our results support the development of allele-specific RNAi strategies to treat familial AD and other dominantly inherited neurodegenerative diseases.


Assuntos
Alelos , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/genética , Dependovirus/genética , Interferência de RNA/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Camundongos , Camundongos Transgênicos
12.
J Neurosci Res ; 86(8): 1748-57, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18293418

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by an elongation of CAG repeats in the HD gene, which encodes a mutant copy of huntingtin with an expanded polyglutatmine repeat. Individuals who are affected by the disease suffer from motor, cognitive, and emotional impairments. Levels of certain striatal-enriched mRNAs decrease in both HD patients and transgenic HD mice prior to the development of motor symptoms and neuronal cell death. Ciliary neurotrophic factor (CNTF) has been shown to protect neurons against chemically induced toxic insults in vitro and in vivo. To test the hypothesis that CNTF might protect neurons from the negative effects of the mutant huntingtin protein in vivo, CNTF was continuously expressed following transduction of the striatum by recombinant adeno-associated viral vectors (rAAV2). Wild-type and R6/1 HD transgenic (R6/1) mice that received bilateral or unilateral intrastriatal injections of rAAV2-CNTF experienced weight loss. The CNTF-treated R6/1 HD transgenic mice experienced motor impairments at an earlier age than expected compared with age-matched control R6/1 HD transgenic animals. CNTF also caused abnormal behavior in WT mice. In addition to behavioral impairments, in situ hybridization showed that, in both WT and R6/1 mice, CNTF expression caused a significant decrease in the levels of striatal-enriched transcripts. Overall, continuous expression of striatal CNTF at the dose mediated by the expression cassette used in this study was detrimental to HD and wild-type mice.


Assuntos
Fator Neurotrófico Ciliar/biossíntese , Corpo Estriado/metabolismo , Regulação para Baixo/fisiologia , Transtornos das Habilidades Motoras/metabolismo , Animais , Comportamento Animal/fisiologia , Fator Neurotrófico Ciliar/genética , Fator Neurotrófico Ciliar/fisiologia , Regulação para Baixo/genética , Humanos , Doença de Huntington/complicações , Doença de Huntington/genética , Doença de Huntington/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Transtornos das Habilidades Motoras/etiologia , Transtornos das Habilidades Motoras/genética , Fatores de Tempo
13.
Neurobiol Dis ; 29(3): 446-55, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18166484

RESUMO

Gene transfer strategies to reduce levels of mutant huntingtin (mHtt) mRNA and protein by targeting human Htt have shown therapeutic promise in vivo. Previously, we have reported that a specific, adeno-associated viral vector (rAAV)-delivered short-hairpin RNA (siHUNT-2) targeting human Htt mRNA unexpectedly decreased levels of striatal-specific transcripts in both wild-type and R6/1 transgenic HD mice. The goal of this study was to determine whether the siHUNT-2-mediated effect was due to adverse effects of RNA interference (RNAi) expression in the brain. To this end, we designed two catalytically active hammerhead ribozymes directed against the same region of human Htt mRNA targeted by siHUNT-2 and delivered them to wild-type and R6/1 transgenic HD mice. After 10 weeks of continuous expression, these ribozymes, like siHUNT-2, negatively impacted the expression of a subset of genes in the striatum. This effect was independent of rAAV transduction and specific to the targeting of a unique sequence in human Htt mRNA. After consideration of the known potential RNAi-specific toxic mechanisms, only cleavage of an unintended RNA target can account for the data reported herein. Thus, long-term rAAV-mediated RNAi in the brain does not, in and of itself, negatively affect striatal gene expression. These findings have important implications in the development of therapeutic RNAi for the treatment of neurological disease.


Assuntos
Marcação de Genes/métodos , Doença de Huntington/genética , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , RNA Catalítico/fisiologia , RNA Interferente Pequeno/fisiologia , Animais , Sequência de Bases , Encéfalo/patologia , Encéfalo/fisiologia , Linhagem Celular , Galinhas , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , RNA Catalítico/biossíntese
14.
Expert Rev Neurother ; 6(2): 223-33, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16466302

RESUMO

RNA interference (RNAi) is a recently described conserved biological pathway where non-coding RNAs suppress the expression of specific genes. Research efforts in the RNAi field aim to gain a better understanding of how its underlying machinery is orchestrated, to define the biological role of this conserved pathway, determine how to effectively manipulate RNAi in the laboratory and to integrate all this knowledge to develop novel therapies for human disease. This review summarizes the advances in the design of therapeutic RNAi for neurodegenerative diseases and discusses some of the experimental steps required to bring this therapy to human clinical trials.


Assuntos
Doenças Neurodegenerativas/terapia , Interferência de RNA/fisiologia , RNA Interferente Pequeno/administração & dosagem , Animais , Ataxina-1 , Ataxinas , Sistemas de Liberação de Medicamentos , Prova Pericial , Humanos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/uso terapêutico , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/uso terapêutico , RNA Interferente Pequeno/química
15.
J Neurosci ; 25(40): 9152-61, 2005 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-16207874

RESUMO

Huntington's disease (HD) and other polyglutamine (polyQ) neurodegenerative diseases are characterized by neuronal accumulation of the disease protein, suggesting that the cellular ability to handle abnormal proteins is compromised. As both a cochaperone and ubiquitin ligase, the C-terminal Hsp70 (heat shock protein 70)-interacting protein (CHIP) links the two major arms of protein quality control, molecular chaperones, and the ubiquitin-proteasome system. Here, we demonstrate that CHIP suppresses polyQ aggregation and toxicity in transfected cell lines, primary neurons, and a novel zebrafish model of disease. Suppression by CHIP requires its cochaperone function, suggesting that CHIP acts to facilitate the solubility of mutant polyQ proteins through its interactions with chaperones. Conversely, HD transgenic mice that are haploinsufficient for CHIP display a markedly accelerated disease phenotype. We conclude that CHIP is a critical mediator of the neuronal response to misfolded polyQ protein and represents a potential therapeutic target in this important class of neurodegenerative diseases.


Assuntos
Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Peptídeos/metabolismo , Ubiquitina-Proteína Ligases/farmacologia , Animais , Western Blotting/métodos , Células Cultivadas , Córtex Cerebral/citologia , Chlorocebus aethiops , Modelos Animais de Doenças , Embrião de Mamíferos , Embrião não Mamífero , Imunofluorescência/métodos , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/metabolismo , Doença de Huntington/tratamento farmacológico , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Neurônios/citologia , Peptídeos/genética , Radioimunoensaio/métodos , Ratos , Transfecção/métodos , Peixe-Zebra
16.
Mol Ther ; 12(4): 618-33, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16019264

RESUMO

Huntington's disease (HD) is a fatal neurodegenerative disorder caused by the presence of an abnormally expanded polyglutamine domain in the N-terminus of huntingtin. We developed a recombinant adeno-associated viral serotype 5 (rAAV5) gene transfer strategy to posttranscriptionally suppress the levels of striatal mutant huntingtin (mHtt) in the R6/1 HD transgenic mouse via RNA interference. Transient cotransfection of HEK293 cells with plasmids expressing a portion of human mHtt derived from R6/1 transgenic HD mice and a short-hairpin RNA directed against the 5' UTR of the mHtt mRNA (siHUNT-1) resulted in reduction in the levels of mHtt mRNA (-75%) and protein (-60%). Long-term in vivo rAAV5-mediated expression of siHUNT-1 in the striatum of R6/1 mice reduced the levels of mHtt mRNA (-78%) and protein (-28%) as determined by quantitative RT-PCR and Western blot analysis, respectively. The reduction in mHtt was concomitant with a reduction in the size and number of neuronal intranuclear inclusions and a small but significant normalization of the steady-state levels of preproenkephalin and dopamine- and cAMP-responsive phosphoprotein 32 kDa mRNA. Finally, bilateral expression of rAAV5-siHUNT-1 resulted in delayed onset of the rear paw clasping phenotype exhibited by the R6/1 mice. These results suggest that a reduction in the levels of striatal mHtt can ameliorate the HD phenotype of R6/1 mice.


Assuntos
Dependovirus/genética , Terapia Genética , Doença de Huntington/terapia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Interferência de RNA , Animais , Modelos Animais de Doenças , Progressão da Doença , Expressão Gênica , Vetores Genéticos , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Corpos de Inclusão Intranuclear , Camundongos , Camundongos Transgênicos/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fenótipo , Plasmídeos/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...