Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
N Engl J Med ; 380(15): 1433-1441, 2019 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-30970188

RESUMEN

We report an inborn error of metabolism caused by an expansion of a GCA-repeat tract in the 5' untranslated region of the gene encoding glutaminase (GLS) that was identified through detailed clinical and biochemical phenotyping, combined with whole-genome sequencing. The expansion was observed in three unrelated patients who presented with an early-onset delay in overall development, progressive ataxia, and elevated levels of glutamine. In addition to ataxia, one patient also showed cerebellar atrophy. The expansion was associated with a relative deficiency of GLS messenger RNA transcribed from the expanded allele, which probably resulted from repeat-mediated chromatin changes upstream of the GLS repeat. Our discovery underscores the importance of careful examination of regions of the genome that are typically excluded from or poorly captured by exome sequencing.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Ataxia/genética , Discapacidades del Desarrollo/genética , Glutaminasa/deficiencia , Glutaminasa/genética , Glutamina/metabolismo , Repeticiones de Microsatélite , Mutación , Atrofia/genética , Cerebelo/patología , Preescolar , Femenino , Genotipo , Glutamina/análisis , Humanos , Masculino , Fenotipo , Reacción en Cadena de la Polimerasa , Secuenciación Completa del Genoma
2.
Hum Mol Genet ; 25(17): 3689-3698, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27378697

RESUMEN

Expansion of a CGG-repeat tract in the 5'-untranslated region of the FMR1 gene to >200 repeats results in epigenetic silencing of the gene by a mechanism that is still unknown. FMR1 gene silencing results in fragile X syndrome (FXS), the most common heritable cause of intellectual disability. We have previously shown that reactivation of the FMR1 gene in FXS cells with 5-azadeoxycytidine (AZA) leads to the transient recruitment of EZH2, the polycomb repressive complex 2 (PRC2) component responsible for H3K27 trimethylation, and that this recruitment depends on the presence of the FMR1 transcript. However, whether H3K27 trimethylation was essential for FMR1 re-silencing was not known. We show here that EZH2 inhibitors increased FMR1 expression and significantly delayed re-silencing of the FMR1 gene in AZA-treated FXS cells. This delay occurred despite the fact that EZH2 inhibition did not prevent the return of DNA methylation. Treatment with compound 1a, a small molecule that targets CGG-repeats in the FMR1 mRNA, also resulted in sustained expression of the FMR1 gene in AZA-treated cells. This effect of 1a was also associated with a decrease in the levels of H3K27 trimethylation but not DNA methylation. Thus, our data show that EZH2 plays a critical role in the FMR1 gene silencing process and that its inhibition can prolong expression of the FMR1 gene even in the presence of its transcript.


Asunto(s)
Azacitidina/análogos & derivados , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Histonas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Azacitidina/farmacología , Línea Celular Tumoral , Metilación de ADN , Decitabina , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metilación/efectos de los fármacos , Repeticiones de Microsatélite/efectos de los fármacos , Regulación hacia Arriba
3.
Hum Mol Genet ; 24(24): 7087-96, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26420841

RESUMEN

Fragile X-associated disorders are Repeat Expansion Diseases that result from expansion of a CGG/CCG-repeat in the FMR1 gene. Contractions of the repeat tract also occur, albeit at lower frequency. However, these contractions can potentially modulate disease symptoms or generate an allele with repeat numbers in the normal range. Little is known about the expansion mechanism and even less about contractions. We have previously demonstrated that the mismatch repair (MMR) protein MSH2 is required for expansions in a mouse model of these disorders. Here, we show that MSH3, the MSH2-binding partner in the MutSß complex, is required for 98% of germ line expansions and all somatic expansions in this model. In addition, we provide evidence for two different contraction mechanisms that operate in the mouse model, a MutSß-independent one that generates small contractions and a MutSß-dependent one that generates larger ones. We also show that MutSß complexes formed with the repeats have altered kinetics of ATP hydrolysis relative to complexes with bona fide MMR substrates and that MutSß increases the stability of the CCG-hairpins at physiological temperatures. These data may have important implications for our understanding of the mechanism(s) of repeat instability and for the role of MMR proteins in this process.


Asunto(s)
Síndrome del Cromosoma X Frágil/genética , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/fisiología , Proteínas/fisiología , Expansión de Repetición de Trinucleótido , Animales , Línea Celular , Inestabilidad Cromosómica , Modelos Animales de Enfermedad , Femenino , Síndrome del Cromosoma X Frágil/fisiopatología , Mutación de Línea Germinal , Masculino , Ratones , Ratones Mutantes , Proteína 3 Homóloga de MutS , Conformación de Ácido Nucleico , Unión Proteica , Proteínas/genética
4.
Hum Genet ; 136(10): 1313-1327, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28866801

RESUMEN

The fragile X-related disorders are a group of three clinical conditions resulting from the instability of a CGG-repeat tract at the 5' end of the FMR1 transcript. Fragile X-associated tremor/ataxia syndrome (FXTAS) and fragile X-associated primary ovarian insufficiency (FXPOI) are disorders seen in carriers of FMR1 alleles with 55-200 repeats. Female carriers of these premutation (PM) alleles are also at risk of having a child who has an FMR1 allele with >200 repeats. Most of these full mutation (FM) alleles are epigenetically silenced resulting in a deficit of the FMR1 gene product, FMRP. This results in fragile X Syndrome (FXS), the most common heritable cause of intellectual disability and autism. The diagnosis and study of these disorders is challenging, in part because the detection of alleles with large repeat numbers has, until recently, been either time-consuming or unreliable. This problem is compounded by the mosaicism for repeat length and/or DNA methylation that is frequently seen in PM and FM carriers. Furthermore, since AGG interruptions in the repeat tract affect the risk that a FM allele will be maternally transmitted, the ability to accurately detect these interruptions in female PM carriers is an additional challenge that must be met. This review will discuss some of the pros and cons of some recently described assays for these disorders, including those that detect FMRP levels directly, as well as emerging technologies that promise to improve the diagnosis of these conditions and to be useful in both basic and translational research settings.


Asunto(s)
Ataxia , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil , Silenciador del Gen , Insuficiencia Ovárica Primaria , Estabilidad del ARN , Temblor , Ataxia/genética , Ataxia/metabolismo , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Humanos , Masculino , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/metabolismo , Temblor/genética , Temblor/metabolismo
5.
Hum Mol Genet ; 23(24): 6575-83, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25055869

RESUMEN

The FMR1 gene is subject to repeat mediated-gene silencing when the CGG-repeat tract in the 5' UTR exceeds 200 repeat units. This results in Fragile X syndrome, the most common heritable cause of intellectual disability and a major cause of autism spectrum disorders. The mechanism of gene silencing is not fully understood, and efforts to reverse this gene silencing have had limited success. Here, we show that the level of trimethylation of histone H3 on lysine 27, a hallmark of the activity of EZH2, a component of repressive Polycomb Group (PcG) complexes like PRC2, is increased on reactivation of the silenced allele by either the DNA demethylating agent 5-azadeoxycytidine or the SIRT1 inhibitor splitomicin. The level of H3K27me3 increases and decreases in parallel with the FMR1 mRNA level. Furthermore, reducing the levels of the FMR1 mRNA reduces the accumulation of H3K27me3. This suggests a model for FMR1 gene silencing in which the FMR1 mRNA generated from the reactivated allele acts in cis to repress its own transcription via the recruitment of PcG complexes to the FMR1 locus.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Histonas/genética , Linfocitos/metabolismo , Complejo Represivo Polycomb 2/genética , ARN Mensajero/genética , Transcripción Genética , Alelos , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular , Decitabina , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos/farmacología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/antagonistas & inhibidores , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Regulación de la Expresión Génica , Histonas/metabolismo , Humanos , Linfocitos/efectos de los fármacos , Linfocitos/patología , Masculino , Naftalenos/farmacología , Complejo Represivo Polycomb 2/metabolismo , Pironas/farmacología , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Sirtuina 1/metabolismo
6.
Hum Mol Genet ; 23(11): 2940-52, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24419320

RESUMEN

Fragile X Syndrome (FXS) is a learning disability seen in individuals who have >200 CGG•CCG repeats in the 5' untranslated region of the X-linked FMR1 gene. Such alleles are associated with a fragile site, FRAXA, a gap or constriction in the chromosome that is coincident with the repeat and is induced by folate stress or thymidylate synthase inhibitors like fluorodeoxyuridine (FdU). The molecular basis of the chromosome fragility is unknown. Previous work has suggested that the stable intrastrand structures formed by the repeat may be responsible, perhaps via their ability to block DNA synthesis. We have examined the replication dynamics of normal and FXS cells with and without FdU. We show here that an intrinsic problem with DNA replication exists in the FMR1 gene of individuals with FXS even in the absence of FdU. Our data suggest a model for chromosome fragility in FXS in which the repeat impairs replication from an origin of replication (ORI) immediately adjacent to the repeat. The fact that the replication problem occurs even in the absence of FdU suggests that this phenomenon may have in vivo consequences, including perhaps accounting for the loss of the X chromosome containing the fragile site that causes Turner syndrome (45, X0) in female carriers of such alleles. Our data on FRAXA may also be germane for the other FdU-inducible fragile sites in humans, that we show here share many common features with FRAXA.


Asunto(s)
Fragilidad Cromosómica , Replicación del ADN , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Línea Celular , Sitios Frágiles del Cromosoma , Cromosomas Humanos X/química , Cromosomas Humanos X/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Heterocigoto , Humanos , Repeticiones de Trinucleótidos
7.
Hum Mutat ; 35(12): 1485-94, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25224527

RESUMEN

Fragile X syndrome (FXS) is the most frequent cause of inherited intellectual disability and autism. It is caused by the absence of the fragile X mental retardation 1 (FMR1) gene product, fragile X mental retardation protein (FMRP), an RNA-binding protein involved in the regulation of translation of a subset of brain mRNAs. In Fmr1 knockout mice, the absence of FMRP results in elevated protein synthesis in the brain as well as increased signaling of many translational regulators. Whether protein synthesis is also dysregulated in FXS patients is not firmly established. Here, we demonstrate that fibroblasts from FXS patients have significantly elevated rates of basal protein synthesis along with increased levels of phosphorylated mechanistic target of rapamycin (p-mTOR), phosphorylated extracellular signal regulated kinase 1/2, and phosphorylated p70 ribosomal S6 kinase 1 (p-S6K1). The treatment with small molecules that inhibit S6K1 and a known FMRP target, phosphoinositide 3-kinase (PI3K) catalytic subunit p110ß, lowered the rates of protein synthesis in both control and patient fibroblasts. Our data thus demonstrate that fibroblasts from FXS patients may be a useful in vitro model to test the efficacy and toxicity of potential therapeutics prior to clinical trials, as well as for drug screening and designing personalized treatment approaches.


Asunto(s)
Biomarcadores/metabolismo , Síndrome del Cromosoma X Frágil/genética , Animales , Estudios de Casos y Controles , Células Cultivadas , Evaluación Preclínica de Medicamentos , Fibroblastos/citología , Fibroblastos/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Humanos , Leucina/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo
8.
iScience ; 27(2): 108814, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38303711

RESUMEN

A long CGG-repeat tract in the FMR1 gene induces the epigenetic silencing that causes fragile X syndrome (FXS). Epigenetic changes include H4K20 trimethylation, a heterochromatic modification frequently implicated in transcriptional silencing. Here, we report that treatment with A-196, an inhibitor of SUV420H1/H2, the enzymes responsible for H4K20 di-/trimethylation, does not affect FMR1 transcription, but does result in increased chromosomal duplications. Increased duplications were also seen in FXS cells treated with SCR7, an inhibitor of Lig4, a ligase essential for NHEJ. Our study suggests that the fragile X (FX) locus is prone to spontaneous DNA damage that is normally repaired by NHEJ. We suggest that heterochromatinization of the FX allele may be triggered, at least in part, in response to this DNA damage.

9.
Sci Rep ; 14(1): 13772, 2024 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877099

RESUMEN

The Repeat Expansion Diseases (REDs) arise from the expansion of a disease-specific short tandem repeat (STR). Different REDs differ with respect to the repeat involved, the cells that are most expansion prone and the extent of expansion. Furthermore, whether these diseases share a common expansion mechanism is unclear. To date, expansion has only been studied in a limited number of REDs. Here we report the first studies of the expansion mechanism in induced pluripotent stem cells derived from a patient with a form of the glutaminase deficiency disorder known as Global Developmental Delay, Progressive Ataxia, And Elevated Glutamine (GDPAG; OMIM# 618412) caused by the expansion of a CAG-STR in the 5' UTR of the glutaminase (GLS) gene. We show that alleles with as few as ~ 120 repeats show detectable expansions in culture despite relatively low levels of R-loops formed at this locus. Additionally, using a CRISPR-Cas9 knockout approach we show that PMS2 and MLH3, the constituents of MutLα and MutLγ, the 2 mammalian MutL complexes known to be involved in mismatch repair (MMR), are essential for expansion. Furthermore, PMS1, a component of a less well understood MutL complex, MutLß, is also important, if not essential, for repeat expansion in these cells. Our results provide insights into the factors important for expansion and lend weight to the idea that, despite some differences, the same mechanism is responsible for expansion in many, if not all, REDs.


Asunto(s)
Glutaminasa , Células Madre Pluripotentes Inducidas , Expansión de Repetición de Trinucleótido , Humanos , Glutaminasa/genética , Glutaminasa/metabolismo , Expansión de Repetición de Trinucleótido/genética , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas MutL/genética , Proteínas MutL/metabolismo , Sistemas CRISPR-Cas
10.
bioRxiv ; 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38260514

RESUMEN

The Repeat Expansion Diseases (REDs) arise from the expansion of a disease-specific short tandem repeat (STR). Different REDs differ with respect to the repeat involved, the cells that are most expansion prone and the extent of expansion. Furthermore, whether these diseases share a common expansion mechanism is unclear. To date, expansion has only been studied in a limited number of REDs. Here we report the first studies of the expansion mechanism in induced pluripotent stem cells derived from a patient with a form of the glutaminase deficiency disorder known as Global Developmental Delay, Progressive Ataxia, And Elevated Glutamine (GDPAG; OMIM# 618412) caused by the expansion of a CAG-STR in the 5' UTR of the glutaminase (GLS) gene. We show that alleles with as few as ~120 repeats show detectable expansions in culture despite relatively low levels of R-loops formed at this locus. Additionally, using a CRISPR-Cas9 knockout approach we show that PMS2 and MLH3, the constituents of MutLα and MutLγ, the 2 mammalian MutL complexes known to be involved in mismatch repair (MMR), are essential for expansion. Furthermore, PMS1, a component of a less well understood MutL complex, MutLß, is also important, if not essential, for repeat expansion in these cells. Our results provide insights into the factors important for expansion and lend weight to the idea that, despite some differences, the same mechanism is responsible for expansion in many, if not all, REDs.

11.
Hum Mutat ; 34(1): 157-66, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22887750

RESUMEN

Repeat expansion diseases result from expansion of a specific tandem repeat. The three fragile X-related disorders (FXDs) arise from germline expansions of a CGG•CCG repeat tract in the 5' UTR (untranslated region) of the fragile X mental retardation 1 (FMR1) gene. We show here that in addition to germline expansion, expansion also occurs in the somatic cells of both mice and humans carriers of premutation alleles. Expansion in mice primarily affects brain, testis, and liver with very little expansion in heart or blood. Our data would be consistent with a simple two-factor model for the organ specificity. Somatic expansion in humans may contribute to the mosaicism often seen in individuals with one of the FXDs. Because expansion risk and disease severity are related to repeat number, somatic expansion may exacerbate disease severity and contribute to the age-related increased risk of expansion seen on paternal transmission in humans. As little somatic expansion occurs in murine lymphocytes, our data also raise the possibility that there may be discordance in humans between repeat numbers measured in blood and that present in brain. This could explain, at least in part, the variable penetrance seen in some of these disorders.


Asunto(s)
Regiones no Traducidas 5'/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Expansión de Repetición de Trinucleótido , Alelos , Animales , Western Blotting , Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Perfilación de la Expresión Génica , Heterocigoto , Humanos , Hígado/metabolismo , Masculino , Ratones , Proteína 2 Homóloga a MutS/metabolismo , Proteína 3 Homóloga de MutS , Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/metabolismo
12.
Biochim Biophys Acta ; 1819(7): 802-10, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22245581

RESUMEN

The Fragile X-associated disorders (FXDs) and Friedreich ataxia (FRDA) are genetic conditions resulting from expansion of a trinucleotide repeat in a region of the affected gene that is transcribed but not translated. In the case of the FXDs, pathology results from expansion of CGG•CCG-repeat tract in the 5' UTR of the FMR1 gene, while pathology in FRDA results from expansion of a GAA•TTC-repeat in intron 1 of the FXN gene. Expansion occurs during gametogenesis or early embryogenesis by a mechanism that is not well understood. Associated Expansion then produces disease pathology in various ways that are not completely understood either. In the case of the FXDs, alleles with 55-200 repeats express higher than normal levels of a transcript that is thought to be toxic, while alleles with >200 repeats are silenced. In addition, alleles with >200 repeats are associated with a cytogenetic abnormality known as a fragile site, which is apparent as a constriction or gap in the chromatin that is seen when cells are grown in presence of inhibitors of thymidylate synthase. FRDA alleles show a deficit of the FXN transcript. This review will address the role of repeat-mediated chromatin changes in these aspects of FXD and FRDA disease pathology. This article is part of a Special Issue entitled: Chromatin in time and space.


Asunto(s)
Cromatina/metabolismo , Síndrome del Cromosoma X Frágil/genética , Ataxia de Friedreich/genética , Mutación , Animales , Cromatina/genética , Fragilidad Cromosómica , Expansión de las Repeticiones de ADN , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Ataxia de Friedreich/metabolismo , Silenciador del Gen , Heterocigoto , Humanos , Secuencias Repetidas en Tándem
13.
J Biol Chem ; 286(6): 4209-15, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21127046

RESUMEN

Expansion of a GAA · TTC repeat in the first intron of the frataxin (FXN) gene causes an mRNA deficit that results in Friedreich ataxia (FRDA). The region flanking the repeat on FRDA alleles is associated with more extensive DNA methylation than is seen on normal alleles and histone modifications typical of repressed genes. However, whether these changes are responsible for the mRNA deficit is controversial. Using chromatin immunoprecipitation and cell lines from affected and unaffected individuals, we show that certain marks of active chromatin are also reduced in the promoter region of the FXN gene in patient cells. Thus, the promoter chromatin may be less permissive for transcription initiation than it is on normal alleles. Furthermore, we show that the initiating form of RNA polymerase II and histone H3 trimethylated on lysine 4, a chromatin mark tightly linked to transcription initiation, are both present at lower levels on FRDA alleles. In addition, a mark of transcription elongation, trimethylated H3K36, shows a reduced rate of accumulation downstream of the repeat. Our data thus suggest that repeat expansion reduces both transcription initiation and elongation in FRDA cells. Our findings may have implications for understanding the mechanism responsible for FRDA as well as for therapeutic approaches to reverse the transcription deficit.


Asunto(s)
Expansión de las Repeticiones de ADN , Ataxia de Friedreich/metabolismo , Proteínas de Unión a Hierro/biosíntesis , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , Transcripción Genética , Alelos , Células Cultivadas , Inmunoprecipitación de Cromatina/métodos , Metilación de ADN/genética , Femenino , Ataxia de Friedreich/genética , Histonas/genética , Histonas/metabolismo , Humanos , Intrones , Proteínas de Unión a Hierro/genética , Masculino , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , Frataxina
14.
Hum Mol Genet ; 19(23): 4634-42, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20843831

RESUMEN

Fragile X syndrome (FXS) is the most common heritable cause of intellectual disability and the most common known cause of autism. Most cases of FXS result from the expansion of a CGG·CCG repeat in the 5' UTR of the FMR1 gene that leads to gene silencing. It has previously been shown that silenced alleles are associated with histone H3 dimethylated at lysine 9 (H3K9Me2) and H3 trimethylated at lysine 27 (H3K27Me3), modified histones typical of developmentally repressed genes. We show here that these alleles are also associated with elevated levels of histone H3 trimethylated at lysine 9 (H3K9Me3) and histone H4 trimethylated at lysine 20 (H4K20Me3). All four of these modified histones are present on exon 1 of silenced alleles at levels comparable to that seen on pericentric heterochromatin. The two groups of histone modifications show a different distribution on fragile X alleles: H3K9Me2 and H3K27Me3 have a broad distribution, whereas H3K9Me3 and H4K20Me3 have a more focal distribution with the highest level of these marks being present in the vicinity of the repeat. This suggests that the trigger for gene silencing may be local to the repeat itself and perhaps involves a mechanism similar to that involved in the formation of pericentric heterochromatin.


Asunto(s)
Metilación de ADN , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Silenciador del Gen , Histonas , Repeticiones de Trinucleótidos , Alelos , Northern Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Cromosomas Humanos X , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/química , Histonas/genética , Histonas/metabolismo , Humanos , Metilación , Reacción en Cadena de la Polimerasa , Procesamiento Proteico-Postraduccional , ARN/genética , ARN/metabolismo , Interferencia de ARN
15.
Nucleic Acids Res ; 37(13): 4385-92, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19465392

RESUMEN

FRAXA is one of a number of fragile sites in human chromosomes that are induced by agents like fluorodeoxyuridine (FdU) that affect intracellular thymidylate levels. FRAXA coincides with a >200 CGG*CCG repeat tract in the 5' UTR of the FMR1 gene, and alleles prone to fragility are associated with Fragile X (FX) syndrome, one of the leading genetic causes of intellectual disability. Using siRNA depletion, we show that ATR is involved in protecting the genome against FdU-induced chromosome fragility. We also show that FdU increases the number of gamma-H2AX foci seen in both normal and patient cells and increases the frequency with which the FMR1 gene colocalizes with these foci in patient cells. In the presence of FdU and KU55933, an ATM inhibitor, the incidence of chromosome fragility is reduced, suggesting that ATM contributes to FdU-induced chromosome fragility. Since both ATR and ATM are involved in preventing aphidicolin-sensitive fragile sites, our data suggest that the lesions responsible for aphidicolin-induced and FdU-induced fragile sites differ. FRAXA also displays a second form of chromosome fragility in absence of FdU, which our data suggest is normally prevented by an ATM-dependent process.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Sitios Frágiles del Cromosoma , Fragilidad Cromosómica , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Síndrome del Cromosoma X Frágil/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Afidicolina/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular , Rotura Cromosómica , Reparación del ADN , Proteínas de Unión al ADN/antagonistas & inhibidores , Floxuridina/farmacología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/enzimología , Técnicas de Silenciamiento del Gen , Histonas/análisis , Humanos , Masculino , Morfolinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Pironas/farmacología , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Proteínas Supresoras de Tumor/antagonistas & inhibidores
16.
PLoS Genet ; 4(3): e1000017, 2008 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-18369442

RESUMEN

Expansion of the CGG.CCG-repeat tract in the 5' UTR of the FMR1 gene to >200 repeats leads to heterochromatinization of the promoter and gene silencing. This results in Fragile X syndrome (FXS), the most common heritable form of mental retardation. The mechanism of gene silencing is unknown. We report here that a Class III histone deacetylase, SIRT1, plays an important role in this silencing process and show that the inhibition of this enzyme produces significant gene reactivation. This contrasts with the much smaller effect of inhibitors like trichostatin A (TSA) that inhibit Class I, II and IV histone deacetylases. Reactivation of silenced FMR1 alleles was accompanied by an increase in histone H3 lysine 9 acetylation as well as an increase in the amount of histone H4 that is acetylated at lysine 16 (H4K16) by the histone acetyltransferase, hMOF. DNA methylation, on the other hand, is unaffected. We also demonstrate that deacetylation of H4K16 is a key downstream consequence of DNA methylation. However, since DNA methylation inhibitors require DNA replication in order to be effective, SIRT1 inhibitors may be more useful for FMR1 gene reactivation in post-mitotic cells like neurons where the effect of the gene silencing is most obvious.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Síndrome del Cromosoma X Frágil/enzimología , Síndrome del Cromosoma X Frágil/genética , Silenciador del Gen/efectos de los fármacos , Sirtuinas/antagonistas & inhibidores , Alelos , Azacitidina/análogos & derivados , Azacitidina/farmacología , Secuencia de Bases , Línea Celular , Metilación de ADN , Cartilla de ADN/genética , Decitabina , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Modelos Biológicos , Mutación , Naftalenos/farmacología , Niacinamida/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Pironas/farmacología , Sirtuina 1 , Expansión de Repetición de Trinucleótido
17.
Front Genet ; 12: 708860, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34567068

RESUMEN

The human genome has many chromosomal regions that are fragile, demonstrating chromatin breaks, gaps, or constrictions on exposure to replication stress. Common fragile sites (CFSs) are found widely distributed in the population, with the largest subset of these sites being induced by aphidicolin (APH). Other fragile sites are only found in a subset of the population. One group of these so-called rare fragile sites (RFSs) is induced by folate stress. APH-inducible CFSs are generally located in large transcriptionally active genes that are A + T rich and often enriched for tracts of AT-dinucleotide repeats. In contrast, all the folate-sensitive sites mapped to date consist of transcriptionally silenced CGG microsatellites. Thus, all the folate-sensitive fragile sites may have a very similar molecular basis that differs in key ways from that of the APH CFSs. The folate-sensitive FSs include FRAXA that is associated with Fragile X syndrome (FXS), the most common heritable form of intellectual disability. Both CFSs and RFSs can cause chromosomal abnormalities. Recent work suggests that both APH-inducible fragile sites and FRAXA undergo Mitotic DNA synthesis (MiDAS) when exposed to APH or folate stress, respectively. Interestingly, blocking MiDAS in both cases prevents chromosome fragility but increases the risk of chromosome mis-segregation. MiDAS of both APH-inducible and FRAXA involves conservative DNA replication and POLD3, an accessory subunit of the replicative polymerase Pol δ that is essential for break-induced replication (BIR). Thus, MiDAS is thought to proceed via some form of BIR-like process. This review will discuss the recent work that highlights the similarities and differences between these two groups of fragile sites and the growing evidence for the presence of many more novel fragile sites in the human genome.

18.
J Huntingtons Dis ; 10(1): 149-163, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33579860

RESUMEN

Huntington's disease (HD) is one of a large group of human disorders that are caused by expanded DNA repeats. These repeat expansion disorders can have repeat units of different size and sequence that can be located in any part of the gene and, while the pathological consequences of the expansion can differ widely, there is evidence to suggest that the underlying mutational mechanism may be similar. In the case of HD, the expanded repeat unit is a CAG trinucleotide located in exon 1 of the huntingtin (HTT) gene, resulting in an expanded polyglutamine tract in the huntingtin protein. Expansion results in neuronal cell death, particularly in the striatum. Emerging evidence suggests that somatic CAG expansion, specifically expansion occurring in the brain during the lifetime of an individual, contributes to an earlier disease onset and increased severity. In this review we will discuss mouse models of two non-CAG repeat expansion diseases, specifically the Fragile X-related disorders (FXDs) and Friedreich ataxia (FRDA). We will compare and contrast these models with mouse and patient-derived cell models of various other repeat expansion disorders and the relevance of these findings for somatic expansion in HD. We will also describe additional genetic factors and pathways that modify somatic expansion in the FXD mouse model for which no comparable data yet exists in HD mice or humans. These additional factors expand the potential druggable space for diseases like HD where somatic expansion is a significant contributor to disease impact.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , Síndrome del Cromosoma X Frágil/genética , Ataxia de Friedreich/genética , Genes Modificadores/genética , Inestabilidad Genómica/genética , Enfermedad de Huntington/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Humanos , Ratones
19.
Genes (Basel) ; 11(4)2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32230785

RESUMEN

In fragile X syndrome (FXS), expansion of a CGG repeat tract in the 5'-untranslated region of the FMR1 gene to >200 repeats causes transcriptional silencing by inducing heterochromatin formation. Understanding the mechanism of FMR1 silencing is important as gene reactivation is a potential treatment approach for FXS. To date, only the DNA demethylating drug 5-azadeoxycytidine (AZA) has proved effective at gene reactivation; however, this drug is toxic. The repressive H3K9 methylation mark is enriched on the FMR1 gene in FXS patient cells and is thus a potential druggable target. However, its contribution to the silencing process is unclear. Here, we studied the effect of small molecule inhibitors of H3K9 methylation on FMR1 expression in FXS patient cells. Chaetocin showed a small effect on FMR1 gene reactivation and a synergistic effect on FMR1 mRNA levels when used in combination with AZA. Additionally, chaetocin, BIX01294 and 3-Deazaneplanocin A (DZNep) were able to significantly delay the re-silencing of AZA-reactivated FMR1 alleles. These data are consistent with the idea that H3K9 methylation precedes DNA methylation and that removal of DNA methylation is necessary to see the optimal effect of histone methyl-transferase (HMT) inhibitors on FMR1 gene expression. Nonetheless, our data also show that drugs targeting repressive H3K9 methylation marks are able to produce sustained reactivation of the FMR1 gene after a single dose of AZA.


Asunto(s)
Metilación de ADN , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/antagonistas & inhibidores , Síndrome del Cromosoma X Frágil/genética , Silenciador del Gen , Preparaciones Farmacéuticas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Alelos , Células Cultivadas , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Síndrome del Cromosoma X Frágil/patología , Humanos , Repeticiones de Trinucleótidos
20.
Nucleic Acids Res ; 35(10): 3383-90, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17478498

RESUMEN

Friedreich ataxia (FRDA), the most common hereditary ataxia, is caused by mutations in the frataxin (FXN) gene. The vast majority of FRDA mutations involve expansion of a GAA*TTC-repeat tract in intron 1, which leads to an FXN mRNA deficit. Bisulfite mapping demonstrates that the region adjacent to the repeat was methylated in both unaffected and affected individuals. However, methylation was more extensive in patients. Additionally, three residues were almost completely methylation-free in unaffected individuals but almost always methylated in those with FRDA. One of these residues is located within an E-box whose deletion caused a significant drop in promoter activity in reporter assays. Elevated levels of histone H3 dimethylated on lysine 9 were seen in FRDA cells consistent with a more repressive chromatin organization. Such chromatin is known to reduce transcription elongation. This may be one way in which the expanded repeats contribute to the frataxin deficit in FRDA. Our data also suggest that repeat-mediated chromatin changes may also affect transcription initiation by blocking binding of factors that increase frataxin promoter activity. Our results also raise the possibility that the repeat-mediated increases in DNA methylation in the FXN gene in FRDA patients are secondary to the chromatin changes.


Asunto(s)
Expansión de las Repeticiones de ADN , Epigénesis Genética , Ataxia de Friedreich/genética , Intrones , Proteínas de Unión a Hierro/genética , Secuencia de Aminoácidos , Línea Celular , Cromatina/química , Metilación de ADN , Elementos E-Box , Histonas/metabolismo , Humanos , Modelos Genéticos , Datos de Secuencia Molecular , Células Musculares/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Frataxina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA