Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
EMBO J ; 37(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30206144

RESUMO

Spinocerebellar ataxia type 8 (SCA8) is caused by a bidirectionally transcribed CTG·CAG expansion that results in the in vivo accumulation of CUG RNA foci, an ATG-initiated polyGln and a polyAla protein expressed by repeat-associated non-ATG (RAN) translation. Although RAN proteins have been reported in a growing number of diseases, the mechanisms and role of RAN translation in disease are poorly understood. We report a novel toxic SCA8 polySer protein which accumulates in white matter (WM) regions as aggregates that increase with age and disease severity. WM regions with polySer aggregates show demyelination and axonal degeneration in SCA8 human and mouse brains. Additionally, knockdown of the eukaryotic translation initiation factor eIF3F in cells reduces steady-state levels of SCA8 polySer and other RAN proteins. Taken together, these data show polySer and WM abnormalities contribute to SCA8 and identify eIF3F as a novel modulator of RAN protein accumulation.


Assuntos
Envelhecimento/metabolismo , Fator de Iniciação 3 em Eucariotos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Degenerações Espinocerebelares/metabolismo , Substância Branca/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Animais , Fator de Iniciação 3 em Eucariotos/genética , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Degenerações Espinocerebelares/genética , Degenerações Espinocerebelares/patologia , Substância Branca/patologia
2.
Proc Natl Acad Sci U S A ; 115(16): 4234-4239, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610297

RESUMO

Expansions of simple sequence repeats, or microsatellites, have been linked to ∼30 neurological-neuromuscular diseases. While these expansions occur in coding and noncoding regions, microsatellite sequence and repeat length diversity is more prominent in introns with eight different trinucleotide to hexanucleotide repeats, causing hereditary diseases such as myotonic dystrophy type 2 (DM2), Fuchs endothelial corneal dystrophy (FECD), and C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). Here, we test the hypothesis that these GC-rich intronic microsatellite expansions selectively trigger host intron retention (IR). Using DM2, FECD, and C9-ALS/FTD as examples, we demonstrate that retention is readily detectable in affected tissues and peripheral blood lymphocytes and conclude that IR screening constitutes a rapid and inexpensive biomarker for intronic repeat expansion disease.


Assuntos
Esclerose Lateral Amiotrófica/genética , Expansão das Repetições de DNA/genética , Demência Frontotemporal/genética , Distrofia Endotelial de Fuchs/genética , Íntrons/genética , Distrofia Miotônica/genética , Composição de Bases , Biomarcadores , Humanos , Linfócitos/química , Músculo Esquelético/química , Miocárdio/química , Especificidade de Órgãos , Polimorfismo de Nucleotídeo Único , Splicing de RNA , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Análise Serial de Tecidos
3.
Neurobiol Dis ; 112: 35-48, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29331264

RESUMO

Myotonic dystrophy (DM) is a progressive, multisystem disorder affecting skeletal muscle, heart, and central nervous system. In both DM1 and DM2, microsatellite expansions of CUG and CCUG RNA repeats, respectively, accumulate and disrupt functions of alternative splicing factors, including muscleblind (MBNL) proteins. Grey matter loss and white matter changes, including the corpus callosum, likely underlie cognitive and executive function deficits in DM patients. However, little is known how cerebral cortical circuitry changes in DM. Here, flavoprotein optical imaging was used to assess local and contralateral responses to intracortical motor cortex stimulation in DM-related mouse models. In control mice, brief train stimulation generated ipsilateral and contralateral homotopic fluorescence increases, the latter mediated by the corpus callosum. Single pulse stimulation produced an excitatory response with an inhibitory-like surround response mediated by GABAA receptors. In a mouse model of DM2 (Mbnl2 KO), we observed prolonged and increased responsiveness to train stimulation and loss of the inhibition from single pulse stimulation. Conversely, mice overexpressing human MBNL1 (MBNL1-OE) exhibited decreased contralateral response to train stimulation and reduction of inhibitory-like surround to single pulse stimulation. Therefore, altering levels of two key DM-associated splicing factors modifies functions of local cortical circuits and contralateral responses mediated through the corpus callosum.


Assuntos
Processamento Alternativo/fisiologia , Modelos Animais de Doenças , Córtex Motor/metabolismo , Distrofia Miotônica/metabolismo , Proteínas de Ligação a RNA/biossíntese , Animais , Estimulação Elétrica/métodos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Córtex Motor/fisiopatologia , Distrofia Miotônica/genética , Distrofia Miotônica/fisiopatologia , Splicing de RNA/fisiologia , Proteínas de Ligação a RNA/genética
4.
Mol Ther ; 24(8): 1378-87, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27203440

RESUMO

Myotonic dystrophy type 1 (DM1) is caused by expanded Cytosine-Thymine-Guanine (CTG) repeats in the 3'-untranslated region (3' UTR) of the Dystrophia myotonica protein kinase (DMPK) gene, for which there is no effective therapy. The objective of this study is to develop genome therapy in human DM1 induced pluripotent stem (iPS) cells to eliminate mutant transcripts and reverse the phenotypes for developing autologous stem cell therapy. The general approach involves targeted insertion of polyA signals (PASs) upstream of DMPK CTG repeats, which will lead to premature termination of transcription and elimination of toxic mutant transcripts. Insertion of PASs was mediated by homologous recombination triggered by site-specific transcription activator-like effector nuclease (TALEN)-induced double-strand break. We found genome-treated DM1 iPS cells continue to maintain pluripotency. The insertion of PASs led to elimination of mutant transcripts and complete disappearance of nuclear RNA foci and reversal of aberrant splicing in linear-differentiated neural stem cells, cardiomyocytes, and teratoma tissues. In conclusion, genome therapy by insertion of PASs upstream of the expanded DMPK CTG repeats prevented the production of toxic mutant transcripts and reversal of phenotypes in DM1 iPS cells and their progeny. These genetically-treated iPS cells will have broad clinical application in developing autologous stem cell therapy for DM1.


Assuntos
Terapia Genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Transplante de Células-Tronco , Animais , Diferenciação Celular , Núcleo Celular/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Marcação de Genes , Loci Gênicos , Humanos , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Distrofia Miotônica/terapia , Miotonina Proteína Quinase/genética , Poli A , Ligação Proteica , Splicing de RNA , Teratoma/genética , Teratoma/metabolismo , Teratoma/patologia , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição , Transplante Autólogo , Repetições de Trinucleotídeos
5.
Proc Natl Acad Sci U S A ; 110(51): E4968-77, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24248382

RESUMO

The finding that a GGGGCC (G4C2) hexanucleotide repeat expansion in the chromosome 9 ORF 72 (C9ORF72) gene is a common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) links ALS/FTD to a large group of unstable microsatellite diseases. Previously, we showed that microsatellite expansion mutations can be bidirectionally transcribed and that these mutations express unexpected proteins by a unique mechanism, repeat-associated non-ATG (RAN) translation. In this study, we show that C9ORF72 antisense transcripts are elevated in the brains of C9ORF72 expansion-positive [C9(+)] patients, and antisense GGCCCC (G2C4) repeat-expansion RNAs accumulate in nuclear foci in brain. Additionally, sense and antisense foci accumulate in blood and are potential biomarkers of the disease. Furthermore, we show that RAN translation occurs from both sense and antisense expansion transcripts, resulting in the expression of six RAN proteins (antisense: Pro-Arg, Pro-Ala, Gly-Pro; and sense: Gly-Ala, Gly-Arg, Gly-Pro). These proteins accumulate in cytoplasmic aggregates in affected brain regions, including the frontal and motor cortex, hippocampus, and spinal cord neurons, with some brain regions showing dramatic RAN protein accumulation and clustering. The finding that unique antisense G2C4 RNA foci and three unique antisense RAN proteins accumulate in patient tissues indicates that bidirectional transcription of expanded alleles is a fundamental pathologic feature of C9ORF72 ALS/FTD. Additionally, these findings suggest the need to test therapeutic strategies that target both sense and antisense RNAs and RAN proteins in C9ORF72 ALS/FTD, and to more broadly consider the role of antisense expression and RAN translation across microsatellite expansion diseases.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Encéfalo/metabolismo , Demência Frontotemporal/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas/metabolismo , RNA Antissenso/biossíntese , Proteína ran de Ligação ao GTP/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Encéfalo/patologia , Proteína C9orf72 , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Feminino , Demência Frontotemporal/genética , Demência Frontotemporal/patologia , Células HEK293 , Humanos , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas/genética , RNA Antissenso/genética , Sequências Repetitivas de Ácido Nucleico , Proteína ran de Ligação ao GTP/genética
6.
EMBO Mol Med ; 13(11): e14095, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34632710

RESUMO

Spinocerebellar ataxia type 8 (SCA8), a dominantly inherited neurodegenerative disorder caused by a CTG•CAG expansion, is unusual because most individuals that carry the mutation do not develop ataxia. To understand the variable penetrance of SCA8, we studied the molecular differences between highly penetrant families and more common sporadic cases (82%) using a large cohort of SCA8 families (n = 77). We show that repeat expansion mutations from individuals with multiple affected family members have CCG•CGG interruptions at a higher frequency than sporadic SCA8 cases and that the number of CCG•CGG interruptions correlates with age at onset. At the molecular level, CCG•CGG interruptions increase RNA hairpin stability, and in cell culture experiments, increase p-eIF2α and polyAla and polySer RAN protein levels. Additionally, CCG•CGG interruptions, which encode arginine interruptions in the polyGln frame, increase toxicity of the resulting proteins. In summary, SCA8 CCG•CGG interruptions increase polyAla and polySer RAN protein levels, polyGln protein toxicity, and disease penetrance and provide novel insight into the molecular differences between SCA8 families with high vs. low disease penetrance.


Assuntos
Degenerações Espinocerebelares , Expansão das Repetições de Trinucleotídeos , Ataxia , Humanos , Proteínas do Tecido Nervoso/genética , Penetrância , Proteínas , RNA Longo não Codificante/genética , Degenerações Espinocerebelares/genética
7.
Methods Enzymol ; 420: 136-62, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17161697

RESUMO

Gene trapping in embryonic stem cells (ESCs) generates random, sequence-tagged insertional mutations, which can often report the gene expression pattern of the mutated gene. This mutagenesis strategy has often been coupled to expression or function-based assays in gene discovery screens. The availability of the mouse genome sequence has shifted gene trapping from a gene discovery platform to a high-throughput mutagenesis platform. At present, a concerted worldwide effort is underway to develop a library of loss-of-function mutations in all mouse genes. The International Gene Trap Consortium (IGTC) is leading the way by making a first pass of the genome by random mutagenesis before a high-throughput gene targeting program takes over. In this chapter, we provide a methods guidebook to exploring and using the IGTC resource, explain the different kinds of vectors and insertions that reside in the different libraries, and provide advice and methods for investigators to design novel expression-based "cottage industry" screens.


Assuntos
Células-Tronco Embrionárias , Mutagênese Insercional , Animais , Técnicas de Cultura de Células , Células Clonais , Biologia Computacional , Células-Tronco Embrionárias/classificação , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Etiquetas de Sequências Expressas/metabolismo , Vetores Genéticos , Humanos , Camundongos
8.
Nucleic Acids Res ; 32(Database issue): D557-9, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14681480

RESUMO

Gene trap mutagenesis of mouse embryonic stem cells generates random loss-of-function mutations, which can be identified by a sequence tag and can often report the endogenous expression of the mutated gene. The Centre for Modeling Human Disease is performing expression- and sequence-based screens of gene trap insertions to generate new mouse mutations as a resource for the scientific community. The gene trap insertions are screened using multiplexed in vitro differentiation and induction assays, and sequence tags are generated to complement expression profiles. Researchers may search for insertions in genes expressed in target cell lineages, under specific in vitro conditions, or based upon sequence identity via an online searchable database (http://www.cmhd.ca/sub/genetrap.asp). The clones are available as a resource to researchers worldwide to help to functionally annotate the mammalian genome and will serve as a source to test candidate loci identified by phenotype-driven mutagenesis screens.


Assuntos
Bases de Dados Genéticas , Modelos Animais de Doenças , Doença , Perfilação da Expressão Gênica , Mutagênese , Animais , Linhagem da Célula , Clonagem Molecular , Teste de Complementação Genética , Genômica , Humanos , Armazenamento e Recuperação da Informação , Internet , Camundongos , Especificidade de Órgãos , Proteômica , Interface Usuário-Computador
9.
Neuron ; 90(3): 521-34, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27112499

RESUMO

To define how the C9orf72 GGGGCC expansion mutation causes ALS/FTD and to facilitate therapy development, a mouse model that recapitulates the molecular and phenotypic features of the disease is urgently needed. Two groups recently reported BAC mouse models that produce RNA foci and RAN proteins but, surprisingly, do not develop the neurodegenerative or behavioral features of ALS/FTD. We now report a BAC mouse model of C9orf72 ALS/FTD that shows decreased survival, paralysis, muscle denervation, motor neuron loss, anxiety-like behavior, and cortical and hippocampal neurodegeneration. These mice express C9orf72 sense transcripts and upregulated antisense transcripts. In contrast to sense RNA foci, antisense foci preferentially accumulate in ALS/FTD-vulnerable cell populations. RAN protein accumulation increases with age and disease, and TDP-43 inclusions are found in degenerating brain regions in end-stage animals. The ALS/FTD phenotypes in our mice provide a unique tool that will facilitate developing therapies targeting pathways that prevent neurodegeneration and increase survival.


Assuntos
Esclerose Lateral Amiotrófica/genética , Encéfalo/metabolismo , Demência Frontotemporal/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Neurônios Motores/fisiologia , Animais , Proteína C9orf72 , Expansão das Repetições de DNA/genética , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Camundongos Transgênicos , Mutação/genética
10.
Cell Rep ; 12(7): 1159-68, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26257173

RESUMO

For some neurological disorders, disease is primarily RNA mediated due to expression of non-coding microsatellite expansion RNAs (RNA(exp)). Toxicity is thought to result from enhanced binding of proteins to these expansions and depletion from their normal cellular targets. However, experimental evidence for this sequestration model is lacking. Here, we use HITS-CLIP and pre-mRNA processing analysis of human control versus myotonic dystrophy (DM) brains to provide compelling evidence for this RNA toxicity model. MBNL2 binds directly to DM repeat expansions in the brain, resulting in depletion from its normal RNA targets with downstream effects on alternative splicing and polyadenylation. Similar RNA processing defects were detected in Mbnl compound-knockout mice, highlighted by dysregulation of Mapt splicing and fetal tau isoform expression in adults. These results demonstrate that MBNL proteins are directly sequestered by RNA(exp) in the DM brain and introduce a powerful experimental tool to evaluate RNA-mediated toxicity in other expansion diseases.


Assuntos
Encéfalo/metabolismo , Proteínas de Ligação a DNA/metabolismo , Distrofia Miotônica/genética , Splicing de RNA , RNA não Traduzido/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Expansão das Repetições de DNA , Proteínas de Ligação a DNA/genética , Humanos , Camundongos , Repetições de Microssatélites , Distrofia Miotônica/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas tau/genética , Proteínas tau/metabolismo
11.
Neurobiol Aging ; 35(10): 2419.e17-21, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24819148

RESUMO

Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are the main syndromes of the chromosome 9 ORF72 (C9ORF72) hexanucleotide repeat expansion, but studies have shown a substantial phenotypic diversity that includes psychiatric presentations. This study describes hippocampal sclerosis dementia (HSD) in carriers of the C9ORF72 mutation. We compared clinical and neuropathological features of HSD in carriers and noncarriers autopsied at Johns Hopkins. Carriers presented with amnesia, agitation, dissocial behavior, and impaired self-care, whereas noncarriers showed little agitation. The groups were not dissimilar in cognitive or motor dysfunction. Neuropathological examination of carriers showed cerebellar neuronal inclusions positive for ubiquitin, p62, and ubiquilin-2, and negative for TAR DNA-binding protein 43. Noncarriers did not have cerebellar inclusions. C9ORF72 repeat-associated non-ATG translation was confirmed by immunohistochemistry. These observations broaden the C9ORF72 phenotype and place HSD in the FTD spectrum. The amnesic phenotype of HSD, which is consistent with the focal hippocampal atrophy, should be included in clinical categorizations of FTD.


Assuntos
Expansão das Repetições de DNA/genética , Demência Frontotemporal/genética , Demência Frontotemporal/patologia , Estudos de Associação Genética , Hipocampo/patologia , Mutação , Proteínas/genética , Idoso , Idoso de 80 Anos ou mais , Proteína C9orf72 , Feminino , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose
12.
PLoS One ; 5(6): e11260, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20582321

RESUMO

Genetically modified mouse strains derived from embryonic stem (ES) cells have become essential tools for functional genomics and biomedical research. Large scale mutagenesis projects are producing libraries of mutant C57BL/6 (B6) ES cells to enable the functional annotation of every gene of the mouse genome. To realize the utility of these resources, efficient and accessible methods of generating mutant mice from these ES cells are necessary. Here, we describe a combination of ICR morula aggregation and a chemically-defined culture medium with widely available and accessible components for the high efficiency generation of germline transmitting chimeras from C57BL/6N ES cells. Together these methods will ease the access of the broader biomedical research community to the publicly available B6 ES cell resources.


Assuntos
Quimera , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/citologia , Células Germinativas , Animais , Meios de Cultura , Camundongos , Camundongos Endogâmicos C57BL
13.
Genes Dev ; 17(3): 380-93, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12569129

RESUMO

Mouse embryos mutant for the VEGF receptor, VEGFR2, Flk-1, or Kdr, fail to form both endothelial and hematopoietic cells, suggesting a possible role in a common progenitor to both lineages. The transcription factor Tal1 (Scl), is not expressed in Flk1(-/-) embryos, consistent with a downstream role in the Flk1 pathway. We tested whether expression of Tal1 under the Flk1 promoter was sufficient to rescue the loss of endothelial and hematopoietic cells in Flk1 mutants. Only partial rescue of hematopoiesis and endothelial development was observed in vivo. However, Flk1(-/Tal1) embryonic stem (ES) cells were capable of blast colony formation in vitro at levels equivalent to Flk1(+/-) heterozygotes. Ectopic expression of Tal1 under the Flk1 promoter in Flk1(+/-) mouse embryos or ES cells caused no obvious pathology but increased the number of blast colony forming cells (BL-CFCs) and enhanced their hematopoietic potential. These single-cell-derived BL-CFCs also produced smooth muscle cells in vitro. Increased Tal1 expression inhibited smooth muscle differentiation in this assay, whereas loss of Tal1 promoted smooth muscle formation. We propose a model in which the combinatorial effects of Flk1 and Tal1 act to regulate cell fate choice in early development into hematopoietic, endothelial, and smooth muscle lineages.


Assuntos
Sistema Cardiovascular/embriologia , Proteínas de Ligação a DNA/metabolismo , Hematopoese/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diferenciação Celular/fisiologia , Endotélio/fisiologia , Camundongos , Células-Tronco , Proteína 1 de Leucemia Linfocítica Aguda de Células T
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA