Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3270, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38627364

RESUMEN

Epigenetic defects caused by hereditary or de novo mutations are implicated in various human diseases. It remains uncertain whether correcting the underlying mutation can reverse these defects in patient cells. Here we show by the analysis of myotonic dystrophy type 1 (DM1)-related locus that in mutant human embryonic stem cells (hESCs), DNA methylation and H3K9me3 enrichments are completely abolished by repeat excision (CTG2000 expansion), whereas in patient myoblasts (CTG2600 expansion), repeat deletion fails to do so. This distinction between undifferentiated and differentiated cells arises during cell differentiation, and can be reversed by reprogramming of gene-edited myoblasts. We demonstrate that abnormal methylation in DM1 is distinctively maintained in the undifferentiated state by the activity of the de novo DNMTs (DNMT3b in tandem with DNMT3a). Overall, the findings highlight a crucial difference in heterochromatin maintenance between undifferentiated (sequence-dependent) and differentiated (sequence-independent) cells, thus underscoring the role of differentiation as a locking mechanism for repressive epigenetic modifications at the DM1 locus.


Asunto(s)
Distrofia Miotónica , Humanos , Distrofia Miotónica/genética , Heterocromatina/genética , Diferenciación Celular/genética , Metilación de ADN , Epigénesis Genética
2.
Mol Ther ; 25(1): 24-43, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28129118

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by (CTG⋅CAG)n-repeat expansion within the DMPK gene and thought to be mediated by a toxic RNA gain of function. Current attempts to develop therapy for this disease mainly aim at destroying or blocking abnormal properties of mutant DMPK (CUG)n RNA. Here, we explored a DNA-directed strategy and demonstrate that single clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-cleavage in either its 5' or 3' unique flank promotes uncontrollable deletion of large segments from the expanded trinucleotide repeat, rather than formation of short indels usually seen after double-strand break repair. Complete and precise excision of the repeat tract from normal and large expanded DMPK alleles in myoblasts from unaffected individuals, DM1 patients, and a DM1 mouse model could be achieved at high frequency by dual CRISPR/Cas9-cleavage at either side of the (CTG⋅CAG)n sequence. Importantly, removal of the repeat appeared to have no detrimental effects on the expression of genes in the DM1 locus. Moreover, myogenic capacity, nucleocytoplasmic distribution, and abnormal RNP-binding behavior of transcripts from the edited DMPK gene were normalized. Dual sgRNA-guided excision of the (CTG⋅CAG)n tract by CRISPR/Cas9 technology is applicable for developing isogenic cell lines for research and may provide new therapeutic opportunities for patients with DM1.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Inestabilidad Genómica , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/genética , Expansión de Repetición de Trinucleótido , Repeticiones de Trinucleótidos , Animales , Proteínas Bacterianas/genética , Secuencia de Bases , Proteína 9 Asociada a CRISPR , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Codón , Modelos Animales de Enfermedad , Endonucleasas/genética , Fibroblastos/metabolismo , Expresión Génica , Orden Génico , Sitios Genéticos , Humanos , Ratones , ARN Guía de Kinetoplastida , ARN Mensajero/genética , ARN Mensajero/metabolismo , Eliminación de Secuencia
3.
Hum Mol Genet ; 25(8): 1648-62, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26908607

RESUMEN

Muscular manifestation of myotonic dystrophy type 1 (DM1), a common inheritable degenerative multisystem disorder, is mainly caused by expression of RNA from a (CTG·CAG)n-expanded DM1 locus. Here, we report on comparative profiling of expression of normal and expanded endogenous or transgenic transcripts in skeletal muscle cells and biopsies from DM1 mouse models and patients in order to help us in understanding the role of this RNA-mediated toxicity. In tissue of HSA(LR) mice, the most intensely used 'muscle-only' model in the DM1 field, RNA from the α-actin (CTG)250 transgene was at least 1000-fold more abundant than that from the Dmpk gene, or the DMPK gene in humans. Conversely, the DMPK transgene in another line, DM500/DMSXL mice, was expressed ∼10-fold lower than the endogenous gene. Temporal regulation of expanded RNA expression differed between models. Onset of expression occurred remarkably late in HSA(LR) myoblasts during in vitro myogenesis whereas Dmpk or DMPK (trans)genes were expressed throughout proliferation and differentiation phases. Importantly, quantification of absolute transcript numbers revealed that normal and expanded Dmpk/DMPK transcripts in mouse models and DM1 patients are low-abundance RNA species. Northern blotting, reverse transcriptase-quantitative polymerase chain reaction, RNA-sequencing and fluorescent in situ hybridization analyses showed that they occur at an absolute number between one and a few dozen molecules per cell. Our findings refine the current RNA dominance theory for DM1 pathophysiology, as anomalous factor binding to expanded transcripts and formation of soluble or insoluble ribonucleoprotein aggregates must be nucleated by only few expanded DMPK transcripts and therefore be a small numbers game.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Músculo Esquelético/citología , Distrofia Miotónica/genética , ARN Mensajero/genética , Expansión de Repetición de Trinucleótido , Actinas/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Desarrollo de Músculos , Músculo Esquelético/patología , Distrofia Miotónica/patología , Proteína Quinasa de Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/metabolismo
4.
PLoS One ; 10(3): e0121556, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25799359

RESUMEN

Myotonic Dystrophy type 1 (DM1) is a multisystemic disease caused by toxic RNA from a DMPK gene carrying an expanded (CTG•CAG)n repeat. Promising strategies for treatment of DM1 patients are currently being tested. These include antisense oligonucleotides and drugs for elimination of expanded RNA or prevention of aberrant binding to RNP proteins. A significant hurdle for preclinical development along these lines is efficient systemic delivery of compounds across endothelial and target cell membranes. It has been reported that DM1 patients show elevated levels of markers of muscle damage or loss of sarcolemmal integrity in their serum and that splicing of dystrophin, an essential protein for muscle membrane structure, is abnormal. Therefore, we studied cell membrane integrity in DM1 mouse models commonly used for preclinical testing. We found that membranes in skeletal muscle, heart and brain were impermeable to Evans Blue Dye. Creatine kinase levels in serum were similar to those in wild type mice and expression of dystrophin protein was unaffected. Also in patient muscle biopsies cell surface expression of dystrophin was normal and calcium-positive fibers, indicating elevated intracellular calcium levels, were only rarely seen. Combined, our findings indicate that cells in DM1 tissues do not display compromised membrane integrity. Hence, the cell membrane is a barrier that must be overcome in future work towards effective drug delivery in DM1 therapy.


Asunto(s)
Permeabilidad de la Membrana Celular , Membrana Celular/metabolismo , Distrofia Miotónica/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Calcio/metabolismo , Niño , Distrofina/genética , Distrofina/metabolismo , Azul de Evans/farmacocinética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Distrofia Miotónica/tratamiento farmacológico
5.
Proc Natl Acad Sci U S A ; 106(33): 13915-20, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19667189

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by toxicity of an expanded, noncoding (CUG)n tract in DM protein kinase (DMPK) transcripts. According to current evidence the long (CUG)n segment is involved in entrapment of muscleblind (Mbnl) proteins in ribonuclear aggregates and stabilized expression of CUG binding protein 1 (CUGBP1), causing aberrant premRNA splicing and associated pathogenesis in DM1 patients. Here, we report on the use of antisense oligonucleotides (AONs) in a therapeutic strategy for reversal of RNA-gain-of-function toxicity. Using a previously undescribed mouse DM1 myoblast-myotube cell model and DM1 patient cells as screening tools, we have identified a fully 2'-O-methyl-phosphorothioate-modified (CAG)7 AON that silences mutant DMPK RNA expression and reduces the number of ribonuclear aggregates in a selective and (CUG)n-length-dependent manner. Direct administration of this AON in muscle of DM1 mouse models in vivo caused a significant reduction in the level of toxic (CUG)n RNA and a normalizing effect on aberrant premRNA splicing. Our data demonstrate proof of principle for therapeutic use of simple sequence AONs in DM1 and potentially other unstable microsatellite diseases.


Asunto(s)
Distrofia Miotónica/genética , Oligonucleótidos/genética , ARN/genética , Alelos , Animales , Proteínas CELF1 , Silenciador del Gen , Ratones , Modelos Genéticos , Músculo Esquelético/metabolismo , Mutación , Mioblastos/metabolismo , Distrofia Miotónica/terapia , Oligonucleótidos/química , Oligonucleótidos Antisentido/genética , Interferencia de ARN , Empalme del ARN , Proteínas de Unión al ARN/genética
6.
BMC Mol Biol ; 8: 61, 2007 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-17645799

RESUMEN

BACKGROUND: Trinucleotide instability is a hallmark of degenerative neurological diseases like Huntington's disease, some forms of spinocerebellar ataxia and myotonic dystrophy type 1 (DM1). To investigate the effect of cell type and cell state on the behavior of the DM1 CTG*CAG repeat, we studied a knock-in mouse model for DM1 at different time points during ageing and followed how repeat fate in cells from liver and pancreas is associated with polyploidization and changes in nuclearity after the onset of terminal differentiation. RESULTS: After separation of liver hepatocytes and pancreatic acinar cells in pools with 2n, 4n or 8n DNA, we analyzed CTG*CAG repeat length variation by resolving PCR products on an automated PAGE system. We observed that somatic CTG*CAG repeat expansion in our DM1 mouse model occurred almost uniquely in the fraction of cells with high cell nuclearity and DNA ploidy and aggravated with aging. CONCLUSION: Our findings suggest that post-replicative and terminal-differentiation events, coupled to changes in cellular DNA content, form a preconditional state that influences the control of DNA repair or recombination events involved in trinucleotide expansion in liver hepatocytes and pancreatic acinar cells.


Asunto(s)
Inestabilidad Genómica/genética , Distrofia Miotónica/genética , Ploidias , Expansión de Repetición de Trinucleótido/genética , Envejecimiento/fisiología , Animales , Secuencia de Bases , Diferenciación Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Citometría de Flujo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/metabolismo , Ratones , Ratones Transgénicos , Miocardio/citología , Miocardio/metabolismo , Distrofia Miotónica/patología , Proteína Quinasa de Distrofia Miotónica , Páncreas/citología , Páncreas/metabolismo , Proteínas Serina-Treonina Quinasas/genética
7.
FEBS Lett ; 580(22): 5208-14, 2006 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-16978612

RESUMEN

The mechanism of trinucleotide repeat expansion, an important cause of neuromuscular and neurodegenerative diseases, is poorly understood. We report here on the study of the role of flap endonuclease 1 (Fen1), a structure-specific nuclease with both 5' flap endonuclease and 5'-3' exonuclease activity, in the somatic hypermutability of the (CTG)(n)*(CAG)(n) repeat of the DMPK gene in a mouse model for myotonic dystrophy type 1 (DM1). By intercrossing mice with Fen1 deficiency with transgenics with a DM1 (CTG)(n)*(CAG)(n) repeat (where 104n110), we demonstrate that Fen1 is not essential for faithful maintenance of this repeat in early embryonic cleavage divisions until the blastocyst stage. Additionally, we found that the frequency of somatic DM1 (CTG)(n)*(CAG)(n) repeat instability was essentially unaltered in mice with Fen1 haploinsufficiency up to 1.5 years of age. Based on these findings, we propose that Fen1, despite its role in DNA repair and replication, is not primarily involved in maintaining stability at the DM1 locus.


Asunto(s)
Reparación del ADN/genética , Replicación del ADN/genética , Endonucleasas de ADN Solapado/genética , Distrofia Miotónica/genética , Sitios de Carácter Cuantitativo/genética , Expansión de Repetición de Trinucleótido/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Blastocisto/enzimología , Modelos Animales de Enfermedad , Endonucleasas de ADN Solapado/metabolismo , Humanos , Ratones , Ratones Transgénicos , Distrofia Miotónica/enzimología
8.
Hum Mol Genet ; 11(2): 191-8, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11809728

RESUMEN

The mechanism of expansion of the (CTG)n repeat in myotonic dystrophy (DM1) patients and the cause of its pathobiological effects are still largely unknown. Most likely, long repeats exert toxicity at the level of nuclear RNA transport or splicing. Here, we analyse cis- and trans-acting parameters that determine repeat behaviour in novel mouse models for DM1. Our mice carry 'humanized' myotonic dystrophy protein kinase (Dmpk) allele(s) with either a (CTG)84 or a (CTG)11 repeat, inserted at the correct position into the endogenous DM locus. Unlike in the human situation, the (CTG)84 repeat in the syntenic mouse environment was relatively stable during intergenerational segregation. However, somatic tissues showed substantial repeat expansions which were progressive upon aging and prominent in kidney, and in stomach and small intestine, where it was cell-type restricted. Other tissues examined showed only marginal size changes. The (CTG)11 allele was completely stable, as anticipated. Introducing the (CTG)84 allele into an Msh3-deficient background completely blocked the somatic repeat instability. In contrast, Msh6 deficiency resulted in a significant increase in the frequency of somatic expansions. Competition of Msh3 and Msh6 for binding to Msh2 in functional complexes with different DNA mismatch-recognition specificity may explain why the somatic (CTG)n expansion rate is differentially affected by ablation of Msh3 and Msh6.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Distrofia Miotónica/genética , Repeticiones de Trinucleótidos/genética , Animales , Disparidad de Par Base , ADN Ligasas/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Masculino , Ratones , Datos de Secuencia Molecular , Proteína 3 Homóloga de MutS , Distrofia Miotónica/etiología , Expansión de Repetición de Trinucleótido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...