Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 15(1): 302, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39278936

RESUMO

BACKGROUND: Cell-based strategies are being explored as a therapeutic option for muscular dystrophies, using a variety of cell types from different origin and with different characteristics. Primary pericytes are multifunctional cells found in the capillary bed that exhibit stem cell-like and myogenic regenerative properties. This unique combination allows them to be applied systemically, presenting a promising opportunity for body-wide muscle regeneration. We previously reported the successful isolation of ALP+ pericytes from skeletal muscle of patients with myotonic dystrophy type 1 (DM1). These pericytes maintained normal growth parameters and myogenic characteristics in vitro despite the presence of nuclear (CUG)n RNA foci, the cellular hallmark of DM1. Here, we examined the behaviour of DM1 pericytes during myogenic differentiation. METHODS: DMPK (CTG)n repeat lengths in patient pericytes were assessed using small pool PCR, to be able to relate variation in myogenic properties and disease hallmarks to repeat expansion. Pericytes from unaffected controls and DM1 patients were cultured under differentiating conditions in vitro. In addition, the pericytes were grown in co-cultures with myoblasts to examine their regenerative capacity by forming hybrid myotubes. Finally, the effect of pericyte fusion on DM1 disease hallmarks was investigated. RESULTS: Small pool PCR analysis revealed the presence of somatic mosaicism in pericyte cell pools. Upon differentiation to myotubes, DMPK expression was upregulated, leading to an increase in nuclear foci sequestering MBNL1 protein. Remarkably, despite the manifestation of these disease biomarkers, patient-derived pericytes demonstrated myogenic potential in co-culture experiments comparable to unaffected pericytes and myoblasts. However, only the unaffected pericytes improved the disease hallmarks in hybrid myotubes. From 20% onwards, the fraction of unaffected nuclei in myotubes positively correlated with a reduction of the number of RNA foci and an increase in the amount of free MBNL1. CONCLUSIONS: Fusion of only a limited number of unaffected myogenic precursors to DM1 myotubes already ameliorates cellular disease hallmarks, offering promise for the development of cell transplantation strategies to lower disease burden.


Assuntos
Diferenciação Celular , Fibras Musculares Esqueléticas , Distrofia Miotônica , Miotonina Proteína Quinase , Pericitos , Humanos , Distrofia Miotônica/metabolismo , Distrofia Miotônica/genética , Distrofia Miotônica/terapia , Distrofia Miotônica/patologia , Fibras Musculares Esqueléticas/metabolismo , Pericitos/metabolismo , Miotonina Proteína Quinase/genética , Miotonina Proteína Quinase/metabolismo , Mioblastos/metabolismo , Mioblastos/citologia , Desenvolvimento Muscular , Células Cultivadas , Masculino , Adulto , Feminino , Técnicas de Cocultura , Pessoa de Meia-Idade , Fusão Celular
2.
EBioMedicine ; 48: 568-580, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31607598

RESUMO

BACKGROUND: Huntington disease (HD) is caused by an unstable CAG/CAA repeat expansion encoding a toxic polyglutamine tract. Here, we tested the hypotheses that HD outcomes are impacted by somatic expansion of, and polymorphisms within, the HTT CAG/CAA glutamine-encoding repeat, and DNA repair genes. METHODS: The sequence of the glutamine-encoding repeat and the proportion of somatic CAG expansions in blood DNA from participants inheriting 40 to 50 CAG repeats within the TRACK-HD and Enroll-HD cohorts were determined using high-throughput ultra-deep-sequencing. Candidate gene polymorphisms were genotyped using kompetitive allele-specific PCR (KASP). Genotypic associations were assessed using time-to-event and regression analyses. FINDINGS: Using data from 203 TRACK-HD and 531 Enroll-HD participants, we show that individuals with higher blood DNA somatic CAG repeat expansion scores have worse HD outcomes: a one-unit increase in somatic expansion score was associated with a Cox hazard ratio for motor onset of 3·05 (95% CI = 1·94 to 4·80, p = 1·3 × 10-6). We also show that individual-specific somatic expansion scores are associated with variants in FAN1 (pFDR = 4·8 × 10-6), MLH3 (pFDR = 8·0 × 10-4), MLH1 (pFDR = 0·004) and MSH3 (pFDR = 0·009). We also show that HD outcomes are best predicted by the number of pure CAGs rather than total encoded-glutamines. INTERPRETATION: These data establish pure CAG length, rather than encoded-glutamine, as the key inherited determinant of downstream pathophysiology. These findings have implications for HD diagnostics, and support somatic expansion as a mechanistic link for genetic modifiers of clinical outcomes, a driver of disease, and potential therapeutic target in HD and related repeat expansion disorders. FUNDING: CHDI Foundation.


Assuntos
Reparo do DNA , Predisposição Genética para Doença , Proteína Huntingtina/genética , Doença de Huntington/genética , Expansão das Repetições de Trinucleotídeos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Criança , Éxons , Feminino , Genótipo , Humanos , Doença de Huntington/diagnóstico , Doença de Huntington/metabolismo , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Adulto Jovem
3.
Biochem Soc Trans ; 36(Pt 3): 522-4, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18481996

RESUMO

HPV16 (human papillomavirus type 16) is a 7.9 kb double-stranded DNA virus that infects anogenital mucosal epithelia. In some rare cases, in women, infection can progress to cervical cancer. HPV16 gene expression is regulated through use of multiple promoters and alternative splicing and polyadenylation. The virus genome can be divided into an early and a late coding region. The late coding region contains the L1 and L2 genes. These encode the virus capsid proteins L1 and L2; protein expression is confined to the upper epithelial layers and is regulated post-transcriptionally in response to epithelial differentiation. A 79 nt RNA regulatory element, the LRE (late regulatory element), involved in this regulation is sited at the 3'-end of the L1 gene and extends into the late 3'-UTR (3'-untranslated region). This element represses late gene expression in differentiated epithelial cells and may activate it in differentiated cells. The present paper describes our current knowledge of LRE RNA-protein interaction and their possible functions.


Assuntos
Diferenciação Celular , Células Epiteliais/citologia , Células Epiteliais/virologia , Regulação Viral da Expressão Gênica , Papillomavirus Humano 16/genética , Processamento Pós-Transcricional do RNA , Sequência de Bases , Humanos , Dados de Sequência Molecular , Sequências Reguladoras de Ácido Nucleico/genética
4.
Methods Mol Med ; 119: 291-315, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16350408

RESUMO

Papillomavirus late gene expression is highly dependent on host epithelial cell differentiation, such that capsid proteins are produced only in differentiating cells. Several papillomaviruses contain negative regulatory elements (NREs), that is, short regions of late transcripts that interact with host cellular RNA processing factors to prevent capsid protein synthesis in undifferentiated cells. In this chapter, the human papillomavirus (HPV)-16 NRE will be used as an example to show how cis-acting RNA regulatory elements can be identified and mapped using transient transfection of reporter gene constructs. The use of reporter gene assays is also readily applicable to the identification and characterization of novel promoters and other regulatory sequences in HPV DNA. In vitro RNA-protein binding techniques, including ultraviolet crosslinking, electrophoretic mobility shift assay, and affinity purification of RNA binding proteins, will also be described, again using the HPV-16 NRE as an example. These techniques may be used to identify cellular proteins that bind the NRE, allowing its mode of action to be deduced. They may also be used to study interactions between host cellular proteins and other protein-binding motifs on HPV mRNA. These interactions are important for the regulation of HPV gene expression, and have key roles in splicing, polyadenylation, mRNA export, stability, and translation.


Assuntos
Papillomaviridae/genética , Sequências Reguladoras de Ácido Ribonucleico/genética , Transcrição Gênica , Primers do DNA , Eletroforese em Gel de Poliacrilamida , Genes Reporter , Células HeLa , Humanos , Reação em Cadeia da Polimerase/métodos , Regiões Promotoras Genéticas , RNA Viral/genética , RNA Viral/isolamento & purificação , Proteínas de Ligação a RNA/isolamento & purificação , Proteínas de Ligação a RNA/metabolismo
5.
J Virol ; 78(19): 10598-605, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15367627

RESUMO

Pre-mRNA splicing occurs in the spliceosome, which is composed of small ribonucleoprotein particles (snRNPs) and many non-snRNP components. SR proteins, so called because of their C-terminal arginine- and serine-rich domains (RS domains), are essential members of this class. Recruitment of snRNPs to 5' and 3' splice sites is mediated and promoted by SR proteins. SR proteins also bridge splicing factors across exons to help to define these units and have a central role in alternative and enhancer-dependent splicing. Here, we show that the SR protein SF2/ASF is part of a complex that forms upon the 79-nucleotide negative regulatory element (NRE) that is thought to be pivotal in posttranscriptional regulation of late gene expression in human papillomavirus type 16 (HPV-16). However, the NRE does not contain any active splice sites, is located in the viral late 3' untranslated region, and regulates RNA-processing events other than splicing. The level of expression and extent of phosphorylation of SF2/ASF are upregulated with epithelial differentiation, as is subcellular distribution, specifically in HPV-16-infected epithelial cells, and expression levels are controlled, at least in part, by the virus transcription regulator E2.


Assuntos
Células Epiteliais/virologia , Regulação Viral da Expressão Gênica , Proteínas Nucleares/metabolismo , Papillomaviridae/genética , RNA Viral/metabolismo , Sequências Reguladoras de Ácido Nucleico , Regiões 3' não Traduzidas , Diferenciação Celular , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Ligação a DNA/fisiologia , Células Epiteliais/citologia , Humanos , Proteínas Nucleares/análise , Proteínas Nucleares/química , Proteínas Oncogênicas Virais/fisiologia , Papillomaviridae/metabolismo , Papillomaviridae/patogenicidade , Fosforilação , Ligação Proteica , Precursores de RNA/metabolismo , Splicing de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Ribonucleoproteínas/análise , Ribonucleoproteínas/metabolismo , Fatores de Processamento de Serina-Arginina , Fator de Processamento U2AF
6.
J Virol ; 77(9): 5167-77, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12692219

RESUMO

The human papillomavirus (HPV) life cycle is tightly linked to differentiation of the squamous epithelia that it infects. Capsid proteins, and hence mature virions, are produced in the outermost layer of differentiated cells. As late gene transcripts are produced in the lower layers, posttranscriptional mechanisms likely prevent capsid protein production in less differentiated cells. For HPV type 16 (HPV-16), a 79-nucleotide (nt) negative regulatory element (NRE) inhibits gene expression in basal epithelial cells. To identify key NRE sequences, we carried out transient transfection in basal epithelial cells with reporter constructs containing the HPV-16 late 3' untranslated region with deletions and mutations of the NRE. Reporter gene expression was increased over 40-fold by deletion of the entire element, 10-fold by deletion of the 5' portion of the NRE that contains four weak consensus 5' splice sites, and only 3-fold by deletion of the 3' GU-rich region. Both portions of the element appear to be necessary for full repression. Inactivating mutations in the 5' splice sites in the 5' NRE partially alleviated repression in the context of the 79-nt NRE but caused full derepression when assayed in a construct with the 3' NRE deleted. All four contribute to the inhibitory effect, though the second splice site is most inhibitory. Sm proteins, U1A and U1 snRNA, but not U1 70K, could be affinity purified with the wild-type NRE but not with the NRE containing mutations in the 5' splice sites, indicating that a U1 snRNP-like complex forms upon the element.


Assuntos
Regiões 5' não Traduzidas/química , Papillomaviridae/genética , Splicing de RNA , RNA Nuclear Pequeno/metabolismo , Sequências Reguladoras de Ácido Nucleico/fisiologia , Regiões 5' não Traduzidas/genética , Regiões 5' não Traduzidas/metabolismo , Sequência de Bases , Deleção de Genes , Regulação Viral da Expressão Gênica , Células HeLa , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Papillomaviridae/metabolismo , Processamento Pós-Transcricional do RNA , Proteínas de Ligação a RNA/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética
7.
J Virol ; 76(12): 5993-6003, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021332

RESUMO

The papillomavirus life cycle is tightly linked to epithelial cell differentiation. Production of virus capsid proteins is restricted to the most terminally differentiated keratinocytes in the upper layers of the epithelium. However, mRNAs encoding the capsid proteins can be detected in less-differentiated cells, suggesting that late gene expression is controlled posttranscriptionally. Short sequence elements (less than 80 nucleotides in length) that inhibit gene expression in undifferentiated epithelial cells have been identified in the late 3' untranslated regions (UTRs) of several papillomaviruses, including the high-risk mucosal type human papillomavirus type 16 (HPV-16). Here we show that closely related high-risk mucosal type HPV-31 also contains elements that can act to repress gene expression in undifferentiated epithelial cells. However, the HPV-31 negative regulatory element is surprisingly complex, comprising a major inhibitory element of approximately 130 nucleotides upstream of the late polyadenylation site and a minor element of approximately 110 nucleotides mapping downstream. The first 60 nucleotides of the major element have 68% identity to the negative regulatory element of HPV-16, and these elements bind the same cellular proteins, CstF-64, U2AF(65), and HuR. The minor inhibitory element binds some cellular proteins in common with the major inhibitory element, though it also binds certain proteins that do not bind the upstream element.


Assuntos
Regiões 3' não Traduzidas , Antígenos de Superfície , Regulação da Expressão Gênica , Proteínas Nucleares , Papillomaviridae/genética , Sequências Reguladoras de Ácido Nucleico , Sequência de Bases , Diferenciação Celular , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Células HeLa , Humanos , Dados de Sequência Molecular , Papillomaviridae/metabolismo , Poli A , Processamento Pós-Transcricional do RNA , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Fator de Processamento U2AF
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA