Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mov Disord ; 2024 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-38798069

RESUMO

BACKGROUND: Spinocerebellar ataxia type 12 (SCA12) is a neurodegenerative disease caused by a CAG/CTG repeat expansion at the PPP2R2B locus. OBJECTIVE: We investigated how the CAG repeat expansion within the PPP2R2B 7B7D transcript influences the expression of Bß1 and a potential protein containing a long polyserine tract. METHODS: Transcript and protein expression were measured using quantitative PCR (qPCR) Role of Bß1 overexpression in the pathogenesis of SCA12 and Western blot, respectively, in an SK-N-MC cell model that overexpresses the full-length PPP2R2B 7B7D transcript. The apoptotic effect of a protein containing a long polyserine tract on SK-N-MC cells was evaluated using caspase 3/7 activity. RESULTS: The CAG repeat expansion increases the expression of the PPP2R2B 7B7D transcript, as well as Bß1 protein, in an SK-N-MC cell model in which the full-length PPP2R2B 7B7D transcript is overexpressed. The CAG repeat expansion within the 7B7D transcript is translated into a long polyserine tract that triggers apoptosis in SK-N-MC cells. CONCLUSIONS: The SCA12 mutation leads to overexpression of PPP2R2B Bß1 and to expression of a protein containing a long polyserine tract; both these effects potentially contribute to SCA12 pathogenesis. © 2024 International Parkinson and Movement Disorder Society.

2.
Stem Cell Res ; 77: 103441, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38759410

RESUMO

Spinocerebellar ataxia type 12 (SCA12) is caused by a CAG expansion mutation in PPP2R2B, a gene encoding brain-specific regulatory units of protein phosphatase 2A (PP2A); while normal alleles carry 4 to 31 triplets, the disease alleles carry 43 to 78 triplets. Here, by CRISPR/Cas9n genome editing, we have generated a human heterozygous SCA12 iPSC line with 73 triplets for the mutant allele. The heterozygous SCA12 iPSCs have normal karyotype, express pluripotency markers and are able to differentiate into the three germ layers.


Assuntos
Edição de Genes , Heterozigoto , Células-Tronco Pluripotentes Induzidas , Mutação , Ataxias Espinocerebelares , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Edição de Genes/métodos , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Linhagem Celular , Sistemas CRISPR-Cas/genética , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Proteínas do Tecido Nervoso
3.
Nat Rev Neurol ; 20(1): 36-49, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38114648

RESUMO

Huntington disease (HD)-like 2 (HDL2) is a rare genetic disease caused by an expanded trinucleotide repeat in the JPH3 gene (encoding junctophilin 3) that shows remarkable clinical similarity to HD. To date, HDL2 has been reported only in patients with definite or probable African ancestry. A single haplotype background is shared by patients with HDL2 from different populations, supporting a common African origin for the expansion mutation. Nevertheless, outside South Africa, reports of patients with HDL2 in Africa are scarce, probably owing to limited clinical services across the continent. Systematic comparisons of HDL2 and HD have revealed closely overlapping motor, cognitive and psychiatric features and similar patterns of cerebral and striatal atrophy. The pathogenesis of HDL2 remains unclear but it is proposed to occur through several mechanisms, including loss of protein function and RNA and/or protein toxicity. This Review summarizes our current knowledge of this African-specific HD phenocopy and highlights key areas of overlap between HDL2 and HD. Given the aforementioned similarities in clinical phenotype and pathology, an improved understanding of HDL2 could provide novel insights into HD and other neurodegenerative and/or trinucleotide repeat expansion disorders.


Assuntos
Coreia , Transtornos Cognitivos , Demência , Doença de Huntington , Humanos , Doença de Huntington/metabolismo , Coreia/complicações , Coreia/genética , Coreia/patologia , Demência/genética , Transtornos Cognitivos/patologia
4.
medRxiv ; 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37961425

RESUMO

INTRODUCTION: The APOE gene is the strongest genetic risk factor for late-onset Alzheimer's Disease (LOAD). However, the gene regulatory mechanisms at this locus have not been fully characterized. METHODS: To identify novel AD-linked functional elements within the APOE locus, we integrated SNP variants with RNA-seq, DNA methylation, and ChIP-seq data from human postmortem brains. RESULTS: We identified an AD-linked APOE transcript (jxn1.2.2) observed in the dorsolateral prefrontal cortex (DLPFC). The APOE jxn1.2.2 transcript is associated with brain neuropathological features in DLPFC. We prioritized an independent functional SNP, rs157580, significantly associated with jxn1.2.2 transcript abundance and DNA methylation levels. rs157580 is located within active chromatin regions and predicted to affect brain-related transcriptional factors binding affinity. rs157580 shared the effects on the jxn1.2.2 transcript between European and African ethnic groups. DISCUSSION: The novel APOE functional elements provide potential therapeutic targets with mechanistic insight into the disease's etiology.

5.
Mov Disord ; 38(12): 2230-2240, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37735923

RESUMO

BACKGROUND: Spinocerebellar ataxia type 12 (SCA12) is a neurodegenerative disease caused by expansion of a CAG repeat in the PPP2R2B gene. OBJECTIVE: In this study, we tested the hypothesis that the PPP2R2B antisense (PPP2R2B-AS1) transcript containing a CUG repeat is expressed and contributes to SCA12 pathogenesis. METHODS: Expression of PPP2R2B-AS1 transcript was detected in SCA12 human induced pluripotent stem cells (iPSCs), iPSC-derived NGN2 neurons, and SCA12 knock-in mouse brains using strand-specific reverse transcription polymerase chain reaction. The tendency of expanded PPP2R2B-AS1 (expPPP2R2B-AS1) RNA to form foci, a marker of toxic processes involving mutant RNAs, was examined in SCA12 cell models by fluorescence in situ hybridization. The apoptotic effect of expPPP2R2B-AS1 transcripts on SK-N-MC neuroblastoma cells was evaluated by caspase 3/7 activity. Western blot was used to examine the expression of repeat associated non-ATG-initiated translation of expPPP2R2B-AS1 transcript in SK-N-MC cells. RESULTS: The repeat region in the PPP2R2B gene locus is bidirectionally transcribed in SCA12 iPSCs, iPSC-derived NGN2 neurons, and SCA12 mouse brains. Transfected expPPP2R2B-AS1 transcripts induce apoptosis in SK-N-MC cells, and the apoptotic effect may be mediated, at least in part, by the RNA secondary structure. The expPPP2R2B-AS1 transcripts form CUG RNA foci in SK-N-MC cells. expPPP2R2B-AS1 transcript is translated in the alanine open reading frame (ORF) via repeat-associated non-ATG translation, which is diminished by single-nucleotide interruptions within the CUG repeat and MBNL1 overexpression. CONCLUSIONS: These findings suggest that PPP2R2B-AS1 contributes to SCA12 pathogenesis and may therefore provide a novel therapeutic target for the disease. © 2023 International Parkinson and Movement Disorder Society.


Assuntos
Sequências Repetitivas de Aminoácidos , Ataxias Espinocerebelares , Transcrição Gênica , Células-Tronco Pluripotentes Induzidas , Neurônios/patologia , Apoptose/genética , Linhagem Celular , Sequências Repetitivas de Aminoácidos/genética , Proteínas de Ligação a RNA/metabolismo , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Técnicas de Introdução de Genes , Humanos , Animais , Camundongos , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/fisiopatologia , RNA Antissenso/genética
6.
bioRxiv ; 2023 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-37066173

RESUMO

OBJECTIVE: Spinocerebellar ataxia type 12 (SCA12) is a neurodegenerative disease caused by expansion of a CAG repeat in the PPP2R2B gene . Here we tested the hypothesis that the PPP2R2B antisense ( PPP2R2B-AS1 ) transcript containing a CUG repeat is expressed and contributes to SCA12 pathogenesis. METHODS: Expression of PPP2R2B-AS1 transcript was detected in SCA12 human induced pluripotent stem cells (iPSCs), iPSC-derived NGN2 neurons, and SCA12 knock-in mouse brains using strand-specific RT-PCR (SS-RT-PCR). The tendency of expanded PPP2R2B-AS1 ( expPPP2R2B-AS1 ) RNA to form foci, a marker of toxic processes involving mutant RNAs, was examined in SCA12 cell models by fluorescence in situ hybridization. The toxic effect of expPPP2R2B-AS1 transcripts on SK-N-MC neuroblastoma cells was evaluated by caspase 3/7 activity. Western blot was used to examine the expression of repeat associated non-ATG-initiated (RAN) translation of expPPP2R2B-AS1 transcript in SK-N-MC cells. RESULTS: The repeat region in PPP2R2B gene locus is bidirectionally transcribed in SCA12 iPSCs, iPSC-derived NGN2 neurons, and SCA12 mouse brains. Transfected expPPP2R2B-AS1 transcripts are toxic to SK-N-MC cells, and the toxicity may be mediated, at least in part, by the RNA secondary structure. The expPPP2R2B-AS1 transcripts form CUG RNA foci in SK-N-MC cells. expPPP2R2B-AS1 transcript is translated in the Alanine ORF via repeat-associated non-ATG (RAN) translation, which is diminished by single nucleotide interruptions within the CUG repeat, and MBNL1 overexpression. INTERPRETATION: These findings suggest that PPP2R2B-AS1 contributes to SCA12 pathogenesis, and may therefore provide a novel therapeutic target for the disease.

7.
Mov Disord ; 36(11): 2519-2529, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34390268

RESUMO

BACKGROUND: Spinocerebellar ataxia type 2 (SCA2) is a neurodegenerative disease caused by expansion of a CAG repeat in Ataxin-2 (ATXN2) gene. The mutant ATXN2 protein with a polyglutamine tract is known to be toxic and contributes to the SCA2 pathogenesis. OBJECTIVE: Here, we tested the hypothesis that the mutant ATXN2 transcript with an expanded CAG repeat (expATXN2) is also toxic and contributes to SCA2 pathogenesis. METHODS: The toxic effect of expATXN2 transcripts on SK-N-MC neuroblastoma cells and primary mouse cortical neurons was evaluated by caspase 3/7 activity and nuclear condensation assay, respectively. RNA immunoprecipitation assay was performed to identify RNA binding proteins (RBPs) that bind to expATXN2 RNA. Quantitative PCR was used to examine if ribosomal RNA (rRNA) processing is disrupted in SCA2 and Huntington's disease (HD) human brain tissue. RESULTS: expATXN2 RNA induces neuronal cell death, and aberrantly interacts with RBPs involved in RNA metabolism. One of the RBPs, transducin ß-like protein 3 (TBL3), involved in rRNA processing, binds to both expATXN2 and expanded huntingtin (expHTT) RNA in vitro. rRNA processing is disrupted in both SCA2 and HD human brain tissue. CONCLUSION: These findings provide the first evidence of a contributory role of expATXN2 transcripts in SCA2 pathogenesis, and further support the role of expHTT transcripts in HD pathogenesis. The disruption of rRNA processing, mediated by aberrant interaction of RBPs with expATXN2 and expHTT transcripts, suggest a point of convergence in the pathogeneses of repeat expansion diseases with potential therapeutic implications. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
RNA , Ataxias Espinocerebelares , Animais , Ataxinas/metabolismo , Encéfalo/patologia , Camundongos , Neurônios/metabolismo , RNA/metabolismo , Proteínas de Ligação a RNA/genética , Ataxias Espinocerebelares/patologia
8.
Stem Cell Res ; 53: 102346, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34087983

RESUMO

Spinocerebellar ataxia type 12 (SCA12) is caused by a CAG expansion mutation in PPP2R2B, a gene encoding a brain-specific regulatory unit of protein phosphatase 2A (PP2A); while normal alleles carry 4 to 31 triplets, the disease alleles carry 43 to 78 triplets. Here, by CRISPR/Cas9 genome editing, we have generated a human homozygous SCA12 iPSC line with 69 and 72 triplets for each allele. The homozygous SCA12 iPSCs have normal karyotype, express pluripotency markers and are able to differentiate into the three germ layers.


Assuntos
Células-Tronco Pluripotentes Induzidas , Ataxias Espinocerebelares , Sistemas CRISPR-Cas/genética , Edição de Genes , Homozigoto , Humanos , Mutação/genética , Ataxias Espinocerebelares/genética
9.
Sci Rep ; 11(1): 9865, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33972655

RESUMO

Cas9 nucleases permit rapid and efficient generation of gene-edited cell lines. However, in typical protocols, mutations are intentionally introduced into the donor template to avoid the cleavage of donor template or re-cleavage of the successfully edited allele, compromising the fidelity of the isogenic lines generated. In addition, the double-stranded breaks (DSBs) used for editing can introduce undesirable "on-target" indels within the second allele of successfully modified cells via non-homologous end joining (NHEJ). To address these problems, we present an optimized protocol for precise genome editing in human iPSCs that employs (1) single guided Cas9 nickase to generate single-stranded breaks (SSBs), (2) transient overexpression of BCL-XL to enhance survival post electroporation, and (3) the PiggyBac transposon system for seamless removal of dual selection markers. We have used this method to modify the length of the CAG repeat contained in exon 7 of PPP2R2B. When longer than 43 triplets, this repeat causes the neurodegenerative disorder spinocerebellar ataxia type 12 (SCA12); our goal was to seamlessly introduce the SCA12 mutation into a human control iPSC line. With our protocol, ~ 15% of iPSC clones selected had the desired gene editing without "on target" indels or off-target changes, and without the deliberate introduction of mutations via the donor template. This method will allow for the precise and efficient editing of human iPSCs for disease modeling and other purposes.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Desoxirribonuclease I/metabolismo , Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas/transplante , Ataxias Espinocerebelares/terapia , Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas , Linhagem Celular , Quebras de DNA de Cadeia Simples , Reparo do DNA por Junção de Extremidades , Desoxirribonuclease I/genética , Eletroporação/métodos , Éxons/genética , Terapia Genética/métodos , Genoma Humano , Humanos , Cariotipagem , Mutação , Proteínas do Tecido Nervoso/genética , Proteína Fosfatase 2/genética , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , Ataxias Espinocerebelares/genética
10.
Sci Rep ; 11(1): 6157, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33731741

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in exon 1 of huntingtin (HTT). While there are currently no disease-modifying treatments for HD, recent efforts have focused on the development of nucleotide-based therapeutics to lower HTT expression. As an alternative to siRNA or oligonucleotide methods, we hypothesized that suppression of HTT expression might be accomplished by small molecules that either (1) directly decrease HTT expression by suppressing HTT promoter activity or (2) indirectly decrease HTT expression by increasing the promoter activity of HTT-AS, the gene antisense to HTT that appears to inhibit expression of HTT. We developed and employed a high-throughput screen for modifiers of HTT and HTT-AS promoter activity using luminescent reporter HEK293 cells; of the 52,041 compounds tested, we identified 898 replicable hits. We used a rigorous stepwise approach to assess compound toxicity and the capacity of the compounds to specifically lower huntingtin protein in 5 different cell lines, including HEK293 cells, HD lymphoblastoid cells, mouse primary neurons, HD iPSCs differentiated into cortical-like neurons, and HD hESCs. We found no compounds which were able to lower huntingtin without lowering cell viability in all assays, though the potential efficacy of a few compounds at non-toxic doses could not be excluded. Our results suggest that more specific targets may facilitate a small molecule approach to HTT suppression.


Assuntos
Proteína Huntingtina/genética , Doença de Huntington/genética , Proteínas do Tecido Nervoso/genética , Animais , Linhagem Celular , Humanos , Camundongos , Regiões Promotoras Genéticas , Expansão das Repetições de Trinucleotídeos
11.
Ann Neurol ; 80(4): 600-15, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27531668

RESUMO

OBJECTIVE: Spinocerebellar ataxia type 2 (SCA2) is a neurodegenerative disease caused by a CAG repeat expansion in the gene ataxin-2 (ATXN2). ATXN2 intermediate-length CAG expansions were identified as a risk factor for amyotrophic lateral sclerosis (ALS). The ATXN2 CAG repeat is translated into polyglutamine, and SCA2 pathogenesis has been thought to derive from ATXN2 protein containing an expanded polyglutamine tract. However, recent evidence of bidirectional transcription at multiple CAG/CTG disease loci has led us to test whether additional mechanisms of pathogenesis may contribute to SCA2. METHODS: In this work, using human postmortem tissue, various cell models, and animal models, we provide the first evidence that an antisense transcript at the SCA2 locus contributes to SCA2 pathogenesis. RESULTS: We demonstrate the expression of a transcript, containing the repeat as a CUG tract, derived from a gene (ATXN2-AS) directly antisense to ATXN2. ATXN2-AS transcripts with normal and expanded CUG repeats are expressed in human postmortem SCA2 brains, human SCA2 fibroblasts, induced SCA2 pluripotent stem cells, SCA2 neural stem cells, and lymphoblastoid lines containing an expanded ATXN2 allele associated with ALS. ATXN2-AS transcripts with a CUG repeat expansion are toxic in an SCA2 cell model and form RNA foci in SCA2 cerebellar Purkinje cells. Finally, we detected missplicing of amyloid beta precursor protein and N-methyl-D-aspartate receptor 1 in SCA2 brains, consistent with findings in other diseases characterized by RNA-mediated pathogenesis. INTERPRETATION: These results suggest that ATXN2-AS has a role in SCA2 and possibly ALS pathogenesis, and may therefore provide a novel therapeutic target for these diseases. Ann Neurol 2016;80:600-615.


Assuntos
Esclerose Lateral Amiotrófica/genética , Ataxina-2/genética , Ataxias Espinocerebelares/genética , Expansão das Repetições de Trinucleotídeos/genética , Adulto , Animais , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos , Humanos , Células-Tronco Pluripotentes Induzidas , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais , Adulto Jovem
12.
Dis Model Mech ; 9(3): 321-34, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26839389

RESUMO

Polyglutamine (polyQ) diseases represent a group of progressive neurodegenerative disorders that are caused by abnormal expansion of CAG triplet nucleotides in disease genes. Recent evidence indicates that not only mutant polyQ proteins, but also their corresponding mutant RNAs, contribute to the pathogenesis of polyQ diseases. Here, we describe the identification of a 13-amino-acid peptide, P3, which binds directly and preferentially to long-CAG RNA within the pathogenic range. When administered to cell and Drosophila disease models, as well as to patient-derived fibroblasts, P3 inhibited expanded-CAG-RNA-induced nucleolar stress and suppressed neurotoxicity. We further examined the combined therapeutic effect of P3 and polyQ-binding peptide 1 (QBP1), a well-characterized polyQ protein toxicity inhibitor, on neurodegeneration. When P3 and QBP1 were co-administered to disease models, both RNA and protein toxicities were effectively mitigated, resulting in a notable improvement of neurotoxicity suppression compared with the P3 and QBP1 single-treatment controls. Our findings indicate that targeting toxic RNAs and/or simultaneous targeting of toxic RNAs and their corresponding proteins could open up a new therapeutic strategy for treating polyQ degeneration.


Assuntos
Drosophila melanogaster/metabolismo , Peptídeos/farmacologia , RNA/toxicidade , Sequência de Aminoácidos , Animais , Morte Celular/efeitos dos fármacos , Drosophila melanogaster/efeitos dos fármacos , Células HEK293 , Humanos , Modelos Biológicos , Degeneração Neural/patologia , Peptídeos/administração & dosagem , Peptídeos/química , Peptídeos/toxicidade , Fosfoproteínas/metabolismo , RNA Ribossômico/genética , Proteínas de Ligação a RNA/metabolismo , Estresse Fisiológico , Relação Estrutura-Atividade , Transcrição Gênica/efeitos dos fármacos , Transfecção , Expansão das Repetições de Trinucleotídeos/genética , Nucleolina
13.
Sci Rep ; 5: 12521, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26218986

RESUMO

Huntington's disease (HD) is caused by a CAG repeat expansion in the huntingtin (HTT) gene. Recent evidence suggests that HD is a consequence of multimodal, non-mutually exclusive mechanisms of pathogenesis that involve both HTT protein- and HTT RNA-triggered mechanisms. Here we provide further evidence for the role of expanded HTT (expHTT) RNA in HD by demonstrating that a fragment of expHTT is cytotoxic in the absence of any translation and that the extent of cytotoxicity is similar to the cytotoxicity of an expHTT protein fragment encoded by a transcript of similar length and with a similar repeat size. In addition, full-length (FL) expHTT is retained in the nucleus. Overexpression of the splicing factor muscleblind-like 1 (MBNL1) increases nuclear retention of expHTT and decreases the expression of expHTT protein in the cytosol. The splicing and nuclear export factor U2AF65 has the opposite effect, decreasing expHTT nuclear retention and increasing expression of expHTT protein. This suggests that MBNL1 and U2AF65 play a role in nuclear export of expHTT RNA.


Assuntos
Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/metabolismo , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Transporte Ativo do Núcleo Celular , Linhagem Celular Tumoral , Códon de Iniciação , Regulação da Expressão Gênica , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Biossíntese de Proteínas/genética , RNA Mensageiro/metabolismo , Fator de Processamento U2AF , Expansão das Repetições de Trinucleotídeos
14.
Hum Mol Genet ; 23(23): 6302-17, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25035419

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the huntingtin (HTT) gene. Disease pathogenesis derives, at least in part, from the long polyglutamine tract encoded by mutant HTT. Therefore, considerable effort has been dedicated to the development of therapeutic strategies that significantly reduce the expression of the mutant HTT protein. Antisense oligonucleotides (ASOs) targeted to the CAG repeat region of HTT transcripts have been of particular interest due to their potential capacity to discriminate between normal and mutant HTT transcripts. Here, we focus on phosphorodiamidate morpholino oligomers (PMOs), ASOs that are especially stable, highly soluble and non-toxic. We designed three PMOs to selectively target expanded CAG repeat tracts (CTG22, CTG25 and CTG28), and two PMOs to selectively target sequences flanking the HTT CAG repeat (HTTex1a and HTTex1b). In HD patient-derived fibroblasts with expanded alleles containing 44, 77 or 109 CAG repeats, HTTex1a and HTTex1b were effective in suppressing the expression of mutant and non-mutant transcripts. CTGn PMOs also suppressed HTT expression, with the extent of suppression and the specificity for mutant transcripts dependent on the length of the targeted CAG repeat and on the CTG repeat length and concentration of the PMO. PMO CTG25 reduced HTT-induced cytotoxicity in vitro and suppressed mutant HTT expression in vivo in the N171-82Q transgenic mouse model. Finally, CTG28 reduced mutant HTT expression and improved the phenotype of Hdh(Q7/Q150) knock-in HD mice. These data demonstrate the potential of PMOs as an approach to suppressing the expression of mutant HTT.


Assuntos
Morfolinos/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Animais , Sequência de Bases , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica , Técnicas de Introdução de Genes , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Camundongos , Camundongos Transgênicos , Morfolinos/química , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Oligonucleotídeos Antissenso/química , RNA Mensageiro/metabolismo , Expansão das Repetições de Trinucleotídeos
15.
PLoS One ; 7(9): e44759, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22957106

RESUMO

BACKGROUND: The assembly of the vertebrate neuromuscular junction (NMJ) is initiated when nerve and muscle first contact each other by filopodial processes which are thought to enable close interactions between the synaptic partners and facilitate synaptogenesis. We recently reported that embryonic Xenopus spinal neurons preferentially extended filopodia towards cocultured muscle cells and that basic fibroblast growth factor (bFGF) produced by muscle activated neuronal FGF receptor 1 (FGFR1) to induce filopodia and favor synaptogenesis. Intriguingly, in an earlier study we found that neurotrophins (NTs), a different set of target-derived factors that act through Trk receptor tyrosine kinases, promoted neuronal growth but hindered presynaptic differentiation and NMJ formation. Thus, here we investigated how bFGF- and NT-signals in neurons jointly elicit presynaptic changes during the earliest stages of NMJ development. METHODOLOGY/PRINCIPAL FINDINGS: Whereas forced expression of wild-type TrkB in neurons reduced filopodial extension and triggered axonal outgrowth, expression of a mutant TrkB lacking the intracellular kinase domain enhanced filopodial growth and slowed axonal advance. Neurons overexpressing wild-type FGFR1 also displayed more filopodia than control neurons, in accord with our previous findings, and, notably, this elevation in filopodial density was suppressed when neurons were chronically treated from the beginning of the culture period with BDNF, the NT that specifically activates TrkB. Conversely, inhibition by BDNF of NMJ formation in nerve-muscle cocultures was partly reversed by the overexpression of bFGF in muscle. CONCLUSIONS: Our results suggest that the balance between neuronal FGFR1- and TrkB-dependent filopodial assembly and axonal outgrowth regulates the establishment of incipient NMJs.


Assuntos
Axônios/metabolismo , Regulação da Expressão Gênica , Junção Neuromuscular/metabolismo , Pseudópodes/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor trkB/metabolismo , Xenopus laevis/metabolismo , Animais , Proliferação de Células , DNA Complementar/metabolismo , Células HEK293 , Humanos , Músculos/metabolismo , Junção Neuromuscular/embriologia , Neurônios/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Vesículas Sinápticas/metabolismo
16.
Mol Biol Cell ; 23(20): 4109-17, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22918949

RESUMO

During the development of the vertebrate neuromuscular junction (NMJ), motor axon tips stop growing after contacting muscle and transform into presynaptic terminals that secrete the neurotransmitter acetylcholine and activate postsynaptic ACh receptors (AChRs) to trigger muscle contraction. The neuron-intrinsic signaling that retards axonal growth to facilitate stable nerve-muscle interaction and synaptogenesis is poorly understood. In this paper, we report a novel function of presynaptic signaling by phosphatase and tensin homologue (PTEN) in mediating a growth-to-synaptogenesis transition in neurons. In Xenopus nerve-muscle cocultures, axonal growth speed was halved after contact with muscle, when compared with before contact, but when cultures were exposed to the PTEN blocker bisperoxo (1,10-phenanthroline) oxovanadate, axons touching muscle grew ~50% faster than their counterparts in control cultures. Suppression of neuronal PTEN expression using morpholinos or the forced expression of catalytically inactive PTEN in neurons also resulted in faster than normal axonal advance after contact with muscle cells. Significantly, interference with PTEN by each of these methods also led to reduced AChR clustering at innervation sites in muscle, indicating that disruption of neuronal PTEN signaling inhibited NMJ assembly. We thus propose that PTEN-dependent slowing of axonal growth enables the establishment of stable nerve-muscle contacts that develop into NMJs.


Assuntos
Axônios/enzimologia , Junção Neuromuscular/enzimologia , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais , Proteínas de Xenopus/metabolismo , Xenopus/metabolismo , Animais , Axônios/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/enzimologia , Músculos/efeitos dos fármacos , Músculos/metabolismo , Junção Neuromuscular/efeitos dos fármacos , Compostos Organometálicos/farmacologia , PTEN Fosfo-Hidrolase/antagonistas & inibidores , Fenantrolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Xenopus/embriologia
17.
Dev Dyn ; 241(10): 1562-74, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22911543

RESUMO

BACKGROUND: During vertebrate neuromuscular junction (NMJ) development, contact between motor axons and muscle fibers is followed by pre- and post-synaptic specialization. Using Xenopus nerve-muscle cocultures, we recently showed that spinal neurons initially contacted muscle cells by means of filopodial processes, and that muscle-derived basic fibroblast growth factor induced axonal filopodia and slowed axonal advance to promote nerve-muscle interaction and NMJ establishment. In contrast, neurotrophins enhanced axonal growth but suppressed the extension of axonal filopodia and blocked NMJ formation. RESULTS: Here we report that hepatocyte growth factor (HGF), which also supports motor neuron survival, was expressed by Xenopus muscle cells, and that forced expression of HGF in Xenopus spinal neurons inhibited the extension of axonal filopodia. Overexpression of the HGF-receptor c-Met in neurons also blocked the formation of axonal filopodia and furthermore sped up axonal growth, but a kinase-dead form of c-Met was unable to effect these changes. Importantly, treatment of nerve-muscle cocultures with recombinant HGF or the expression of HGF or active c-Met in neurons, or that of excess HGF in muscle, inhibited nerve-induced AChR clustering in muscle. CONCLUSIONS: Our results suggest that HGF/c-Met signaling in neurons promotes axonal growth but suppresses filopodial assembly in neurons and hinders NMJ establishment.


Assuntos
Axônios/fisiologia , Fator de Crescimento de Hepatócito/metabolismo , Neurônios Motores/citologia , Junção Neuromuscular/crescimento & desenvolvimento , Pseudópodes/fisiologia , Transdução de Sinais/fisiologia , Animais , Axônios/metabolismo , Técnicas de Cultura de Células , Primers do DNA/genética , Immunoblotting , Imunoprecipitação , Microscopia , Neurônios Motores/metabolismo , Músculo Esquelético/metabolismo , Fatores de Crescimento Neural/metabolismo , Junção Neuromuscular/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Pseudópodes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinapses/metabolismo , Sinapses/fisiologia , Xenopus
18.
Mol Biol Cell ; 23(14): 2680-91, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22648172

RESUMO

At the developing neuromuscular junction (NMJ), physical contact between motor axons and muscle cells initiates presynaptic and postsynaptic differentiation. Using Xenopus nerve-muscle cocultures, we previously showed that innervating axons induced muscle filopodia (myopodia), which facilitated interactions between the synaptic partners and promoted NMJ formation. The myopodia were generated by nerve-released signals through muscle p120 catenin (p120ctn), a protein of the cadherin complex that modulates the activity of Rho GTPases. Because axons also extend filopodia that mediate early nerve-muscle interactions, here we test p120ctn's function in the assembly of these presynaptic processes. Overexpression of wild-type p120ctn in Xenopus spinal neurons leads to an increase in filopodial growth and synaptic vesicle (SV) clustering along axons, whereas the development of these specializations is inhibited following the expression of a p120ctn mutant lacking sequences important for regulating Rho GTPases. The p120ctn mutant also inhibits the induction of axonal filopodia and SV clusters by basic fibroblast growth factor, a muscle-derived molecule that triggers presynaptic differentiation. Of importance, introduction of the p120ctn mutant into neurons hinders NMJ formation, which is observed as a reduction in the accumulation of acetylcholine receptors at innervation sites in muscle. Our results suggest that p120ctn signaling in motor neurons promotes nerve-muscle interaction and NMJ assembly.


Assuntos
Neurônios/fisiologia , Pseudópodes/fisiologia , Vesículas Sinápticas/fisiologia , Proteína p120 Ativadora de GTPase/metabolismo , Animais , Axônios/metabolismo , Cateninas/metabolismo , Comunicação Celular , Diferenciação Celular , Células Cultivadas , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/metabolismo , Junção Neuromuscular/metabolismo , Junção Neuromuscular/fisiologia , Neurônios/metabolismo , Neurônios/ultraestrutura , Pseudópodes/metabolismo , Receptores Colinérgicos/metabolismo , Transdução de Sinais , Vesículas Sinápticas/metabolismo , Xenopus laevis , Proteína p120 Ativadora de GTPase/genética
19.
Mol Biol Cell ; 22(14): 2480-90, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21613540

RESUMO

During vertebrate neuromuscular junction (NMJ) assembly, motor axons and their muscle targets exchange short-range signals that regulate the subsequent steps of presynaptic and postsynaptic specialization. We report here that this interaction is in part mediated by axonal filopodia extended preferentially by cultured Xenopus spinal neurons toward their muscle targets. Immunoblotting and labeling experiments showed that basic fibroblast growth factor (bFGF) was expressed by muscle and associated with the cell surface, and treatment of cultured spinal neurons with recombinant bFGF nearly doubled the normal density of filopodia in neurites. This effect of bFGF was abolished by SU5402, a selective inhibitor of FGF-receptor 1 (FGFR1), and forced expression of wild-type or dominant-negative FGFR1 in neurons enhanced or suppressed the assembly of filopodia, respectively. Significantly, in nerve-muscle cocultures, knocking down bFGF in muscle decreased both the asymmetric extension of filopodia by axons toward muscle and the assembly of NMJs. In addition, neurons expressing dominant-negative FGFR1 less effectively triggered the aggregation of muscle acetylcholine receptors at innervation sites than did control neurons. These results suggest that bFGF activation of neuronal FGFR1 generates filopodial processes in neurons that promote nerve-muscle interaction and facilitate NMJ establishment.


Assuntos
Axônios/metabolismo , Axônios/fisiologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Junção Neuromuscular/crescimento & desenvolvimento , Pseudópodes/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Células Cultivadas , Células Musculares/metabolismo , Células Musculares/fisiologia , Neuritos/metabolismo , Neuritos/fisiologia , Junção Neuromuscular/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia , Terminações Pré-Sinápticas/metabolismo , Pseudópodes/metabolismo , Transdução de Sinais , Técnicas de Cultura de Tecidos , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA