Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Neurosci ; 40(2): 459-477, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31748376

RESUMO

α-Synuclein (α-Syn) accumulation is a pathological hallmark of Parkinson's disease. Duplications and triplications of SNCA, the gene coding for α-Syn, cause genetic forms of the disease, which suggests that increased α-Syn dosage can drive PD. To identify the proteins that regulate α-Syn, we previously performed a screen of potentially druggable genes that led to the identification of 60 modifiers. Among them, Doublecortin-like kinase 1 (DCLK1), a microtubule binding serine threonine kinase, emerged as a promising target due to its potent effect on α-Syn and potential druggability as a neuron-expressed kinase. In this study, we explore the relationship between DCLK1 and α-Syn in human cellular and mouse models of PD. First, we show that DCLK1 regulates α-Syn levels post-transcriptionally. Second, we demonstrate that knockdown of Dclk1 reduces phosphorylated species of α-Syn and α-Syn-induced neurotoxicity in the SNc in two distinct mouse models of synucleinopathy. Last, silencing DCLK1 in human neurons derived from individuals with SNCA triplications reduces phosphorylated and total α-Syn, thereby highlighting DCLK1 as a potential therapeutic target to reduce pathological α-Syn in disease.SIGNIFICANCE STATEMENT DCLK1 regulates α-Syn protein levels, and Dclk1 knockdown rescues α-Syn toxicity in mice. This study provides evidence for a novel function for DCLK1 in the mature brain, and for its potential as a new therapeutic target for synucleinopathies.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Doença de Parkinson/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , alfa-Sinucleína/metabolismo , Animais , Quinases Semelhantes a Duplacortina , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo
2.
Mol Psychiatry ; 25(10): 2534-2555, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-30610205

RESUMO

Genome sequencing has revealed an increasing number of genetic variations that are associated with neuropsychiatric disorders. Frequently, studies limit their focus to likely gene-disrupting mutations because they are relatively easy to interpret. Missense variants, instead, have often been undervalued. However, some missense variants can be informative for developing a more profound understanding of disease pathogenesis and ultimately targeted therapies. Here we present an example of this by studying a missense variant in a well-known autism spectrum disorder (ASD) causing gene SHANK3. We analyzed Shank3's in vivo phosphorylation profile and identified S685 as one phosphorylation site where one ASD-linked variant has been reported. Detailed analysis of this variant revealed a novel function of Shank3 in recruiting Abelson interactor 1 (ABI1) and the WAVE complex to the post-synaptic density (PSD), which is critical for synapse and dendritic spine development. This function was found to be independent of Shank3's other functions such as binding to GKAP and Homer. Introduction of this human ASD mutation into mice resulted in a small subset of phenotypes seen previously in constitutive Shank3 knockout mice, including increased allogrooming, increased social dominance, and reduced pup USV. Together, these findings demonstrate the modularity of Shank3 function in vivo. This modularity further indicates that there is more than one independent pathogenic pathway downstream of Shank3 and correcting a single downstream pathway is unlikely to be sufficient for clear clinical improvement. In addition, this study illustrates the value of deep biological analysis of select missense mutations in elucidating the pathogenesis of neuropsychiatric phenotypes.


Assuntos
Transtorno do Espectro Autista/genética , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Mutação de Sentido Incorreto/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Transtorno Autístico/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Humanos , Masculino , Camundongos , Densidade Pós-Sináptica/metabolismo , Ratos
3.
Nature ; 498(7454): 325-331, 2013 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-23719381

RESUMO

Many neurodegenerative disorders, such as Alzheimer's, Parkinson's and polyglutamine diseases, share a common pathogenic mechanism: the abnormal accumulation of disease-causing proteins, due to either the mutant protein's resistance to degradation or overexpression of the wild-type protein. We have developed a strategy to identify therapeutic entry points for such neurodegenerative disorders by screening for genetic networks that influence the levels of disease-driving proteins. We applied this approach, which integrates parallel cell-based and Drosophila genetic screens, to spinocerebellar ataxia type 1 (SCA1), a disease caused by expansion of a polyglutamine tract in ataxin 1 (ATXN1). Our approach revealed that downregulation of several components of the RAS-MAPK-MSK1 pathway decreases ATXN1 levels and suppresses neurodegeneration in Drosophila and mice. Importantly, pharmacological inhibitors of components of this pathway also decrease ATXN1 levels, suggesting that these components represent new therapeutic targets in mitigating SCA1. Collectively, these data reveal new therapeutic entry points for SCA1 and provide a proof-of-principle for tackling other classes of intractable neurodegenerative diseases.


Assuntos
Drosophila melanogaster/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/toxicidade , Proteínas Nucleares/metabolismo , Proteínas Nucleares/toxicidade , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/patologia , Proteínas ras/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Ataxina-1 , Ataxinas , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Drosophila melanogaster/genética , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Dados de Sequência Molecular , Terapia de Alvo Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosforilação , Estabilidade Proteica/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas 90-kDa/deficiência , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Transgenes
4.
Hum Mol Genet ; 24(7): 1813-23, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25432536

RESUMO

Silencing of fragile X mental retardation 1 (FMR1) gene and loss of fragile X mental retardation protein (FMRP) cause fragile X syndrome (FXS), a genetic disorder characterized by intellectual disability and autistic behaviors. FMRP is an mRNA-binding protein regulating neuronal translation of target mRNAs. Abnormalities in actin-rich dendritic spines are major neuronal features in FXS, but the molecular mechanism and identity of FMRP targets mediating this phenotype remain largely unknown. Cytoplasmic FMR1-interacting protein 2 (Cyfip2) was identified as an interactor of FMRP, and its mRNA is a highly ranked FMRP target in mouse brain. Importantly, Cyfip2 is a component of WAVE regulatory complex, a key regulator of actin cytoskeleton, suggesting that Cyfip2 could be implicated in the dendritic spine phenotype of FXS. Here, we generated and characterized Cyfip2-mutant (Cyfip2(+/-)) mice. We found that Cyfip2(+/-) mice exhibited behavioral phenotypes similar to Fmr1-null (Fmr1(-/y)) mice, an animal model of FXS. Synaptic plasticity and dendritic spines were normal in Cyfip2(+/-) hippocampus. However, dendritic spines were altered in Cyfip2(+/-) cortex, and the dendritic spine phenotype of Fmr1(-/y) cortex was aggravated in Fmr1(-/y); Cyfip2(+/-) double-mutant mice. In addition to the spine changes at basal state, metabotropic glutamate receptor (mGluR)-induced dendritic spine regulation was impaired in both Fmr1(-/y) and Cyfip2(+/-) cortical neurons. Mechanistically, mGluR activation induced mRNA translation-dependent increase of Cyfip2 in wild-type cortical neurons, but not in Fmr1(-/y) or Cyfip2(+/-) neurons. These results suggest that misregulation of Cyfip2 function and its mGluR-induced expression contribute to the neurobehavioral phenotypes of FXS.


Assuntos
Córtex Cerebral/metabolismo , Citoplasma/metabolismo , Espinhas Dendríticas/metabolismo , Síndrome do Cromossomo X Frágil/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Comportamento Animal , Córtex Cerebral/anormalidades , Citoplasma/genética , Modelos Animais de Doenças , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/fisiopatologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência
5.
Nat Genet ; 39(3): 373-9, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17322884

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disease caused by expansion of a glutamine tract in ataxin-1 (ATXN1). SCA1 pathogenesis studies support a model in which the expanded glutamine tract causes toxicity by modulating the normal activities of ATXN1. To explore native interactions that modify the toxicity of ATXN1, we generated a targeted duplication of the mouse ataxin-1-like (Atxn1l, also known as Boat) locus, a highly conserved paralog of SCA1, and tested the role of this protein in SCA1 pathology. Using a knock-in mouse model of SCA1 that recapitulates the selective neurodegeneration seen in affected individuals, we found that elevated Atxn1l levels suppress neuropathology by displacing mutant Atxn1 from its native complex with Capicua (CIC). Our results provide genetic evidence that the selective neuropathology of SCA1 arises from modulation of a core functional activity of ATXN1, and they underscore the importance of studying the paralogs of genes mutated in neurodegenerative diseases to gain insight into mechanisms of pathogenesis.


Assuntos
Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Ataxias Espinocerebelares/genética , Animais , Ataxina-1 , Ataxinas , Células Cultivadas , Cerebelo/metabolismo , Expansão das Repetições de DNA , Células-Tronco Embrionárias/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Modelos Genéticos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/análise , Células de Purkinje/metabolismo , Proteínas Repressoras/metabolismo , Ataxias Espinocerebelares/patologia
6.
J Neurosci ; 33(22): 9328-36, 2013 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-23719801

RESUMO

Polyglutamine diseases are dominantly inherited neurodegenerative diseases caused by an expansion of a CAG trinucleotide repeat encoding a glutamine tract in the respective disease-causing proteins. Extensive studies have been performed to unravel disease pathogenesis and to develop therapeutics. Here, we report on several lines of evidence demonstrating that Nemo-like kinase (NLK) is a key molecule modulating disease toxicity in spinocerebellar ataxia type 1 (SCA1), a disease caused by a polyglutamine expansion in the protein ATAXIN1 (ATXN1). Specifically, we show that NLK, a serine/threonine kinase that interacts with ATXN1, modulates disease phenotypes of polyglutamine-expanded ATXN1 in a Drosophila model of SCA1. Importantly, the effect of NLK on SCA1 pathology is dependent upon NLK's enzymatic activity. Consistent with this, reduced Nlk expression suppresses the behavioral and neuropathological phenotypes in SCA1 knock-in mice. These data clearly indicate that either reducing NLK enzymatic activity or decreasing NLK expression levels can have beneficial effects against the toxicity induced by polyglutamine-expanded ATXN1.


Assuntos
Drosophila melanogaster/fisiologia , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Transtornos Heredodegenerativos do Sistema Nervoso/patologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Peptídeos/fisiologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Animais , Animais Geneticamente Modificados , Ataxina-1 , Ataxinas , Comportamento Animal/fisiologia , Western Blotting , Encéfalo/anatomia & histologia , Cerebelo/patologia , Cromatografia em Gel , Feminino , Expressão Gênica , Células HEK293 , Transtornos Heredodegenerativos do Sistema Nervoso/psicologia , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Fosforilação , Proteínas Serina-Treonina Quinases , Ataxias Espinocerebelares/psicologia
7.
Nature ; 452(7188): 713-8, 2008 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-18337722

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disease caused by expansion of a glutamine-encoding repeat in ataxin 1 (ATXN1). In all known polyglutamine diseases, the glutamine expansion confers toxic functions onto the protein; however, the mechanism by which this occurs remains enigmatic, in light of the fact that the mutant protein apparently maintains interactions with its usual partners. Here we show that the expanded polyglutamine tract differentially affects the function of the host protein in the context of different endogenous protein complexes. Polyglutamine expansion in ATXN1 favours the formation of a particular protein complex containing RBM17, contributing to SCA1 neuropathology by means of a gain-of-function mechanism. Concomitantly, polyglutamine expansion attenuates the formation and function of another protein complex containing ATXN1 and capicua, contributing to SCA1 through a partial loss-of-function mechanism. This model provides mechanistic insight into the molecular pathogenesis of SCA1 as well as other polyglutamine diseases.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Ataxias Espinocerebelares/metabolismo , Expansão das Repetições de Trinucleotídeos , Alelos , Animais , Ataxina-1 , Ataxinas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Humanos , Camundongos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fases de Leitura Aberta/genética , Peptídeos/genética , Ligação Proteica , Estrutura Quaternária de Proteína , Células de Purkinje/citologia , Células de Purkinje/metabolismo , Fatores de Processamento de RNA , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Ribonucleoproteína Nuclear Pequena U2/genética , Ribonucleoproteína Nuclear Pequena U2/metabolismo , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Expansão das Repetições de Trinucleotídeos/genética , Técnicas do Sistema de Duplo-Híbrido
8.
Proc Natl Acad Sci U S A ; 108(5): 2142-7, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21245341

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by the expansion of a CAG repeat encoding a polyglutamine tract in Ataxin-1 (ATXN1). Both WT and mutant ATXN1 interact with 14-3-3 proteins, and 14-3-3 overexpression stabilizes ATXN1 levels in cells and increases ATXN1 toxicity in flies. To determine whether reducing 14-3-3 levels might mitigate SCA1 pathogenesis, we bred Sca1(154Q/+) mice to mice lacking one allele of 14-3-3ε. 14-3-3ε haploinsufficiency rescued cerebellar pathology and motor phenotypes but, surprisingly, not weight loss, respiratory dysfunction, or premature lethality. Biochemical studies revealed that reducing 14-3-3ε levels exerted different effects in two brain regions especially vulnerable in SCA1: Although diminishing levels of both WT and mutant ATXN1 in the cerebellum, 14-3-3ε haploinsufficiency did not alter ATXN1 levels in the brainstem. Furthermore, 14-3-3ε haploinsufficiency decreased the incorporation of expanded ATXN1 into its large toxic complexes in the cerebellum but not in the brainstem, and the distribution of ATXN1's small and large native complexes differed significantly between the two regions. These data suggest that distinct pathogenic mechanisms operate in different vulnerable brain regions, adding another level of complexity to SCA1 pathogenesis.


Assuntos
Proteínas 14-3-3/genética , Haploinsuficiência , Ataxias Espinocerebelares/genética , Alelos , Animais , Ataxina-1 , Ataxinas , Encéfalo/patologia , Linhagem Celular , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Fenótipo
9.
J Clin Invest ; 132(9)2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35499073

RESUMO

Many neurodegenerative disorders are caused by abnormal accumulation of misfolded proteins. In spinocerebellar ataxia type 1 (SCA1), accumulation of polyglutamine-expanded (polyQ-expanded) ataxin-1 (ATXN1) causes neuronal toxicity. Lowering total ATXN1, especially the polyQ-expanded form, alleviates disease phenotypes in mice, but the molecular mechanism by which the mutant ATXN1 is specifically modulated is not understood. Here, we identified 22 mutant ATXN1 regulators by performing a cross-species screen of 7787 and 2144 genes in human cells and Drosophila eyes, respectively. Among them, transglutaminase 5 (TG5) preferentially regulated mutant ATXN1 over the WT protein. TG enzymes catalyzed cross-linking of ATXN1 in a polyQ-length-dependent manner, thereby preferentially modulating mutant ATXN1 stability and oligomerization. Perturbing Tg in Drosophila SCA1 models modulated mutant ATXN1 toxicity. Moreover, TG5 was enriched in the nuclei of SCA1-affected neurons and colocalized with nuclear ATXN1 inclusions in brain tissue from patients with SCA1. Our work provides a molecular insight into SCA1 pathogenesis and an opportunity for allele-specific targeting for neurodegenerative disorders.


Assuntos
Cerebelo , Ataxias Espinocerebelares , Animais , Ataxina-1/genética , Ataxina-1/metabolismo , Cerebelo/metabolismo , Drosophila/genética , Drosophila/metabolismo , Humanos , Camundongos , Peptídeos , Ataxias Espinocerebelares/metabolismo , Transglutaminases
10.
Dis Model Mech ; 14(6)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34165550

RESUMO

Spinocerebellar ataxias (SCAs) are a group of genetically heterogeneous inherited neurodegenerative disorders characterized by progressive ataxia and cerebellar degeneration. Here, we used a mouse model to test a possible connection between SCA and Ronin (Thap11), a polyglutamine-containing transcriptional regulator encoded in a region of human chromosome 16q22.1 that has been genetically linked to SCA type 4. We report that transgenic expression of Ronin in mouse cerebellar Purkinje cells leads to detrimental loss of these cells and the development of severe ataxia as early as 10 weeks after birth. Mechanistically, we find that several SCA-causing genes harbor Ronin DNA-binding motifs and are transcriptionally deregulated in transgenic animals. In addition, ectopic expression of Ronin in embryonic stem cells significantly increases the protein level of Ataxin-1, the protein encoded by Atxn1, alterations of which cause SCA type 1. This increase is also seen in the cerebellum of transgenic animals, although the latter was not statistically significant. Hence, our data provide evidence for a link between Ronin and SCAs, and suggest that Ronin may be involved in the development of other neurodegenerative diseases.


Assuntos
Ataxia/metabolismo , Proteínas Repressoras/metabolismo , Ataxias Espinocerebelares/metabolismo , Animais , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Células de Purkinje/metabolismo
11.
Ann Neurol ; 66(6): 771-82, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20035514

RESUMO

OBJECTIVE: There have been no objective assessments to determine whether boys with MECP2 duplication have autism or whether female carriers manifest phenotypes. This study characterizes the clinical and neuropsychiatric phenotypes of affected boys and carrier females. METHODS: Eight families (9 males and 9 females) with MECP2 duplication participated. A detailed history, physical examination, electroencephalogram, developmental evaluation, Autism Diagnostic Observation Schedule, and Autism Diagnostic Interview-Revised were performed for each boy. Carrier females completed the Symptom Checklist-90-R, Wechsler Abbreviated Scale of Intelligence, Broad Autism Phenotype Questionnaire, and detailed medical and mental health histories. Size and gene content of each duplication were determined by array comparative genome hybridization. X-chromosome inactivation patterns were analyzed using leukocyte DNA. MECP2 and IRAK1 RNA levels were quantified from lymphoblast cell lines, and western blots were performed to assess MeCP2 protein levels. RESULTS: All of the boys demonstrated mental retardation and autism. Poor expressive language, gaze avoidance, repetitive behaviors, anxiety, and atypical socialization were prevalent. Female carriers had psychiatric symptoms, including generalized anxiety, depression, and compulsions that preceded the birth of their children. The majority exhibited features of the broad autism phenotype and had higher nonverbal compared to verbal reasoning skills. INTERPRETATION: Autism is a defining feature of the MECP2 duplication syndrome in boys. Females manifest phenotypes despite 100% skewing of X-inactivation and normal MECP2 RNA levels in peripheral blood. Analysis of the duplication size, MECP2 and IRAK1 RNA levels, and MeCP2 protein levels revealed that most of the traits in affected boys are likely due to the genomic region spanning of MECP2 and IRAK1. The phenotypes observed in carrier females may be secondary to tissue-specific dosage alterations and require further study. Ann Neurol 2009;66:771-782.


Assuntos
Transtorno Autístico/genética , Duplicação Gênica , Predisposição Genética para Doença , Deficiência Intelectual/genética , Proteína 2 de Ligação a Metil-CpG/genética , Adolescente , Transtorno Autístico/metabolismo , Transtorno Autístico/fisiopatologia , Criança , Pré-Escolar , Cromossomos Humanos X , Saúde da Família , Feminino , Humanos , Deficiência Intelectual/metabolismo , Deficiência Intelectual/fisiopatologia , Quinases Associadas a Receptores de Interleucina-1/genética , Masculino , Proteína 2 de Ligação a Metil-CpG/metabolismo , Testes Neuropsicológicos , RNA Mensageiro/genética , Caracteres Sexuais , Inativação do Cromossomo X/genética
12.
Cell Rep ; 25(3): 726-736.e7, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30332651

RESUMO

RNA splicing entails the coordinated interaction of more than 150 proteins in the spliceosome, one of the most complex of the cell's molecular machines. We previously discovered that the RNA-binding motif protein 17 (RBM17), a component of the spliceosome, is essential for survival and cell maintenance. Here, we find that it interacts with the spliceosomal factors U2SURP and CHERP and that they reciprocally regulate each other's stability, both in mouse and in human cells. Individual knockdown of each of the three proteins induces overlapping changes in splicing and gene expression of transcripts enriched for RNA-processing factors. Our results elucidate the function of RBM17, U2SURP, and CHERP and link the activity of the spliceosome to the regulation of downstream RNA-binding proteins. These data support the hypothesis that, beyond driving constitutive splicing, spliceosomal factors can regulate alternative splicing of specific targets.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Fatores de Processamento de RNA/metabolismo , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Animais , Sistemas CRISPR-Cas , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Feminino , Células HEK293 , Humanos , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Processamento de RNA/antagonistas & inibidores , Fatores de Processamento de RNA/genética , Fatores de Processamento de RNA/fisiologia , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Spliceossomos
13.
PLoS Med ; 4(5): e182, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17535104

RESUMO

BACKGROUND: Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disorder characterized by progressive motor and cognitive dysfunction. Caused by an expanded polyglutamine tract in ataxin 1 (ATXN1), SCA1 pathogenesis involves a multifactorial process that likely begins with misfolding of ATXN1, which has functional consequences on its interactions, leading to transcriptional dysregulation. Because lithium has been shown to exert neuroprotective effects in a variety of conditions, possibly by affecting gene expression, we tested the efficacy of lithium treatment in a knock-in mouse model of SCA1 (Sca1(154Q/2Q) mice) that replicates many features of the human disease. METHODS AND FINDINGS: Sca1(154Q/2Q) mice and their wild-type littermates were fed either regular chow or chow that contained 0.2% lithium carbonate. Dietary lithium carbonate supplementation resulted in improvement of motor coordination, learning, and memory in Sca1(154Q/2Q) mice. Importantly, motor improvement was seen when treatment was initiated both presymptomatically and after symptom onset. Neuropathologically, lithium treatment attenuated the reduction of dendritic branching in mutant hippocampal pyramidal neurons. We also report that lithium treatment restored the levels of isoprenylcysteine carboxyl methyltransferase (Icmt; alternatively, Pccmt), down-regulation of which is an early marker of mutant ATXN1 toxicity. CONCLUSIONS: The effect of lithium on a marker altered early in the course of SCA1 pathogenesis, coupled with its positive effect on multiple behavioral measures and hippocampal neuropathology in an authentic disease model, make it an excellent candidate treatment for human SCA1 patients.


Assuntos
Antimaníacos/farmacologia , Carbonato de Lítio/farmacologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Ataxias Espinocerebelares/tratamento farmacológico , Ataxias Espinocerebelares/patologia , Animais , Ataxina-1 , Ataxinas , Dendritos/enzimologia , Dendritos/patologia , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hipocampo/patologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Atividade Motora/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Metiltransferases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Ataxias Espinocerebelares/genética
14.
Nat Genet ; 49(4): 527-536, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28288114

RESUMO

Gain-of-function mutations in some genes underlie neurodegenerative conditions, whereas loss-of-function mutations in the same genes have distinct phenotypes. This appears to be the case with the protein ataxin 1 (ATXN1), which forms a transcriptional repressor complex with capicua (CIC). Gain of function of the complex leads to neurodegeneration, but ATXN1-CIC is also essential for survival. We set out to understand the functions of the ATXN1-CIC complex in the developing forebrain and found that losing this complex results in hyperactivity, impaired learning and memory, and abnormal maturation and maintenance of upper-layer cortical neurons. We also found that CIC activity in the hypothalamus and medial amygdala modulates social interactions. Informed by these neurobehavioral features in mouse mutants, we identified five individuals with de novo heterozygous truncating mutations in CIC who share similar clinical features, including intellectual disability, attention deficit/hyperactivity disorder (ADHD), and autism spectrum disorder. Our study demonstrates that loss of ATXN1-CIC complexes causes a spectrum of neurobehavioral phenotypes.


Assuntos
Ataxina-1/genética , Transtorno do Espectro Autista/genética , Doenças Neurodegenerativas/genética , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Animais , Cerebelo/patologia , Feminino , Humanos , Deficiência Intelectual/genética , Relações Interpessoais , Masculino , Camundongos , Proteínas do Tecido Nervoso/genética , Fenótipo
15.
Elife ; 42015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26673892

RESUMO

Previously, we reported that ATXN1 oligomers are the primary drivers of toxicity in Spinocerebellar ataxia type 1 (SCA1; Lasagna-Reeves et al., 2015). Here we report that polyQ ATXN1 oligomers can propagate locally in vivo in mice predisposed to SCA1 following intracerebral oligomeric tissue inoculation. Our data also show that targeting these oligomers with passive immunotherapy leads to some improvement in motor coordination in SCA1 mice and to a modest increase in their life span. These findings provide evidence that oligomer propagation is regionally limited in SCA1 and that immunotherapy targeting extracellular oligomers can mildly modify disease phenotypes.


Assuntos
Ataxina-1/toxicidade , Imunização Passiva , Ataxias Espinocerebelares/patologia , Ataxias Espinocerebelares/terapia , Animais , Ataxina-1/antagonistas & inibidores , Modelos Animais de Doenças , Camundongos , Resultado do Tratamento
16.
Elife ; 42015 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-25988806

RESUMO

Recent studies indicate that soluble oligomers drive pathogenesis in several neurodegenerative proteinopathies, including Alzheimer and Parkinson disease. Curiously, the same conformational antibody recognizes different disease-related oligomers, despite the variations in clinical presentation and brain regions affected, suggesting that the oligomer structure might be responsible for toxicity. We investigated whether polyglutamine-expanded ATAXIN-1, the protein that underlies spinocerebellar ataxia type 1, forms toxic oligomers and, if so, what underlies their toxicity. We found that mutant ATXN1 does form oligomers and that oligomer levels correlate with disease progression in the Atxn1(154Q/+) mice. Moreover, oligomeric toxicity, stabilization and seeding require interaction with Capicua, which is expressed at greater ratios with respect to ATXN1 in the cerebellum than in less vulnerable brain regions. Thus, specific interactors, not merely oligomeric structure, drive pathogenesis and contribute to regional vulnerability. Identifying interactors that stabilize toxic oligomeric complexes could answer longstanding questions about the pathogenesis of other proteinopathies.


Assuntos
Ataxina-1/química , Ataxina-1/toxicidade , Cerebelo/metabolismo , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/fisiopatologia , Análise de Variância , Animais , Western Blotting , Cromatografia em Gel , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Peptídeos/análise , Proteínas Repressoras/metabolismo , Teste de Desempenho do Rota-Rod , Testes de Toxicidade
17.
Cell ; 127(7): 1335-47, 2006 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-17190598

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is one of several neurodegenerative diseases caused by expansion of a polyglutamine tract in the disease protein, in this case, ATAXIN-1 (ATXN1). A key question in the field is whether neurotoxicity is mediated by aberrant, novel interactions with the expanded protein or whether its wild-type functions are augmented to a deleterious degree. We examined soluble protein complexes from mouse cerebellum and found that the majority of wild-type and expanded ATXN1 assembles into large stable complexes containing the transcriptional repressor Capicua. ATXN1 directly binds Capicua and modulates Capicua repressor activity in Drosophila and mammalian cells, and its loss decreases the steady-state level of Capicua. Interestingly, the S776A mutation, which abrogates the neurotoxicity of expanded ATXN1, substantially reduces the association of mutant ATXN1 with Capicua in vivo. These data provide insight into the function of ATXN1 and suggest that SCA1 neuropathology depends on native, not novel, protein interactions.


Assuntos
Cerebelo/metabolismo , Drosophila/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Ataxias Espinocerebelares/etiologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Ataxina-1 , Ataxinas , Encéfalo/metabolismo , Sequência Conservada , Drosophila/embriologia , Anormalidades do Olho/etiologia , Humanos , Camundongos , Dados de Sequência Molecular , Mutação , Peptídeos/metabolismo , Homologia de Sequência de Aminoácidos , Ataxias Espinocerebelares/genética , Transcrição Gênica , Asas de Animais/anormalidades
18.
Wilehm Roux Arch Dev Biol ; 194(1): 53-55, 1984 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28305194

RESUMO

In the present study the effect of histones H1o and H5, and the nonhistone chromatin proteins HMG 1, 2, 14 and 17 (the high mobility group proteins), as well as the acidic peptide fragments of HMG 1 and 2 and polyglutamate, on cell division and differentation of cultured murine erythroleukemia (Friend) cells has been investigated. It was found that histones H1o and H5, the acidic peptide fragments of HMG 1 and 2, HMG 14 and 17 and sodium polyglutamate stimulated cell division at a concentration of 10 µg/ml. None of the H1o, H5 or HMG protein preparations induced hemoglobin synthesis, as judged by benzidine staining.

19.
Hum Mol Genet ; 13(21): 2679-89, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15351775

RESUMO

Mutations in the X-linked methyl-CpG-binding protein 2 (MECP2), encoding a transcriptional repressor, cause Rett syndrome and a variety of related neurodevelopmental disorders. The vast majority of mutations associated with human disease are loss-of-function mutations, but precisely what aspect of MeCP2 function is responsible for these phenotypes remains unknown. We overexpressed wild-type human protein in transgenic mice using a large genomic clone containing the entire human MECP2 locus. Detailed neurobehavioral and electrophysiological studies in transgenic line MeCP2(Tg1), which expresses MeCP2 at approximately 2-fold wild-type levels, demonstrated onset of phenotypes around 10 weeks of age. Surprisingly, these mice displayed enhanced motor and contextual learning and enhanced synaptic plasticity in the hippocampus. After 20 weeks of age, however, these mice developed seizures, became hypoactive and approximately 30% of them died by 1 year of age. These data demonstrate that MeCP2 levels must be tightly regulated in vivo, and that even mild overexpression of this protein is detrimental. Furthermore, these results support the possibility that duplications or gain-of-function mutations in MECP2 might underlie some cases of X-linked delayed-onset neurobehavioral disorders.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/patologia , Proteínas Repressoras/metabolismo , Síndrome de Rett/genética , Animais , Comportamento Animal , Western Blotting , Modelos Animais de Doenças , Eletroencefalografia , Eletrofisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Proteína 2 de Ligação a Metil-CpG , Camundongos , Camundongos Transgênicos , Mutação , Fatores de Tempo , Cromossomo X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA