Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 106(3): 356-370, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32109418

RESUMEN

Genetic syndromes frequently present with overlapping clinical features and inconclusive or ambiguous genetic findings which can confound accurate diagnosis and clinical management. An expanding number of genetic syndromes have been shown to have unique genomic DNA methylation patterns (called "episignatures"). Peripheral blood episignatures can be used for diagnostic testing as well as for the interpretation of ambiguous genetic test results. We present here an approach to episignature mapping in 42 genetic syndromes, which has allowed the identification of 34 robust disease-specific episignatures. We examine emerging patterns of overlap, as well as similarities and hierarchical relationships across these episignatures, to highlight their key features as they are related to genetic heterogeneity, dosage effect, unaffected carrier status, and incomplete penetrance. We demonstrate the necessity of multiclass modeling for accurate genetic variant classification and show how disease classification using a single episignature at a time can sometimes lead to classification errors in closely related episignatures. We demonstrate the utility of this tool in resolving ambiguous clinical cases and identification of previously undiagnosed cases through mass screening of a large cohort of subjects with developmental delays and congenital anomalies. This study more than doubles the number of published syndromes with DNA methylation episignatures and, most significantly, opens new avenues for accurate diagnosis and clinical assessment in individuals affected by these disorders.


Asunto(s)
Metilación de ADN , Trastornos del Neurodesarrollo/genética , Fenotipo , Estudios de Cohortes , Heterogeneidad Genética , Humanos , Síndrome
2.
Hum Mol Genet ; 29(19): 3197-3210, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-32916696

RESUMEN

The most distal 2 kb region in the majority of human subtelomeres contains CpG-rich promoters for TERRA, a long non-coding RNA. When the function of the de novo DNA methyltransferase DNMT3B is disrupted, as in ICF1 syndrome, subtelomeres are abnormally hypomethylated, subtelomeric heterochromatin acquires open chromatin characteristics, TERRA is highly expressed, and telomeres shorten rapidly. In this study, we explored whether the regulation of subtelomeric epigenetic characteristics by DNMT3B is conserved between humans and mice. Studying the DNA sequence of the distal 30 kb of the majority of murine q-arm subtelomeres indicated that these regions are relatively CpG-poor and do not contain TERRA promoters similar to those present in humans. Despite the lack of human-like TERRA promoters, we clearly detected TERRA expression originating from at least seven q-arm subtelomeres, and at higher levels in mouse pluripotent stem cells in comparison with mouse embryonic fibroblasts (MEFs). However, these differences in TERRA expression could not be explained by differential methylation of CpG islands present in the TERRA-expressing murine subtelomeres. To determine whether Dnmt3b regulates the expression of TERRA in mice, we characterized subtelomeric methylation and associated telomeric functions in cells derived from ICF1 model mice. Littermate-derived WT and ICF1 MEFs demonstrated no significant differences in subtelomeric DNA methylation, chromatin modifications, TERRA expression levels, telomere sister chromatid exchange or telomere length. We conclude that the epigenetic characteristics of murine subtelomeres differ substantially from their human counterparts and that TERRA transcription in mice is regulated by factors others than Dnmt3b.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Cara/anomalías , Fibroblastos/patología , Enfermedades de Inmunodeficiencia Primaria/patología , Telómero/fisiología , Factores de Transcripción/metabolismo , Animales , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/genética , Proteínas de Unión al ADN/genética , Cara/patología , Fibroblastos/metabolismo , Humanos , Ratones , Enfermedades de Inmunodeficiencia Primaria/genética , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Transcripción Genética , ADN Metiltransferasa 3B
3.
Prog Mol Subcell Biol ; 60: 169-201, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34386876

RESUMEN

Centromeres are chromosomal regions that are essential for the faithful transmission of genetic material through each cell division. They represent the chromosomal platform on which assembles a protein complex, the kinetochore, which mediates attachment to the mitotic spindle. In most organisms, centromeres assemble on large arrays of tandem satellite repeats, although their DNA sequences and organization are highly divergent among species. It has become evident that centromeres are not defined by underlying DNA sequences, but are instead epigenetically defined by the deposition of the centromere-specific histone H3 variant, CENP-A. In addition, and although long regarded as silent chromosomal loci, centromeres are in fact transcriptionally competent in most species, yet at low levels in normal somatic cells, but where the resulting transcripts participate in centromere architecture, identity, and function. In this chapter, we discuss the various roles proposed for centromere transcription and their transcripts, and the potential molecular mechanisms involved. We also discuss pathological cases in which unscheduled transcription of centromeric repeats or aberrant accumulation of their transcripts are pathological signatures of chromosomal instability diseases. In sum, tight regulation of centromeric satellite repeats transcription is critical for healthy development and tissue homeostasis, and thus prevents the emergence of disease states.


Asunto(s)
Centrómero , Cromatina , Centrómero/genética , Proteína A Centromérica/genética , Cinetocoros , Transcripción Genética/genética
4.
Int J Mol Sci ; 22(7)2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-33916664

RESUMEN

DNA methylation (DNAme) profiling is used to establish specific biomarkers to improve the diagnosis of patients with inherited neurodevelopmental disorders and to guide mutation screening. In the specific case of mendelian disorders of the epigenetic machinery, it also provides the basis to infer mechanistic aspects with regard to DNAme determinants and interplay between histone and DNAme that apply to humans. Here, we present comparative methylomes from patients with mutations in the de novo DNA methyltransferases DNMT3A and DNMT3B, in their catalytic domain or their N-terminal parts involved in reading histone methylation, or in histone H3 lysine (K) methylases NSD1 or SETD2 (H3 K36) or KMT2D/MLL2 (H3 K4). We provide disease-specific DNAme signatures and document the distinct consequences of mutations in enzymes with very similar or intertwined functions, including at repeated sequences and imprinted loci. We found that KMT2D and SETD2 germline mutations have little impact on DNAme profiles. In contrast, the overlapping DNAme alterations downstream of NSD1 or DNMT3 mutations underlines functional links, more specifically between NSD1 and DNMT3B at heterochromatin regions or DNMT3A at regulatory elements. Together, these data indicate certain discrepancy with the mechanisms described in animal models or the existence of redundant or complementary functions unforeseen in humans.


Asunto(s)
Metilación de ADN/genética , Enfermedades Genéticas Congénitas/genética , Histonas/genética , Mutación , Enfermedades Raras/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/genética , Enfermedades Genéticas Congénitas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Humanos , Proteínas de Neoplasias/genética , Enfermedades Raras/metabolismo , ADN Metiltransferasa 3B
5.
Hum Mol Genet ; 27(20): 3568-3581, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30010917

RESUMEN

Human telomeres and adjacent subtelomeres are packaged as heterochromatin. Subtelomeric DNA undergoes methylation during development by DNA methyltransferase 3B (DNMT3B), including the CpG-rich promoters of the long non-coding RNA (TERRA) embedded in these regions. The factors that direct DNMT3B methylation to human subtelomeres and maintain this methylation throughout lifetime are yet unknown. The importance of subtelomeric methylation is manifested through the abnormal telomeric phenotype in Immunodeficiency, Centromeric instability and Facial anomalies (ICF) syndrome type 1 patients carrying mutations in DNMT3B. Patient cells demonstrate subtelomeric hypomethylation, accompanied by elevated TERRA transcription, accelerated telomere shortening and premature senescence of fibroblasts. ICF syndrome can arise due to mutations in at least three additional genes, ZBTB24 (ICF2), CDCA7 (ICF3) and HELLS (ICF4). While pericentromeric repeat hypomethylation is evident in all ICF syndrome subtypes, the status of subtelomeric DNA methylation had not been described for patients of subtypes 2-4. Here we explored the telomeric phenotype in cells derived from ICF2-4 patients with the aim to determine whether ZBTB24, CDCA7 and HELLS also play a role in establishing and/or maintaining human subtelomeric methylation. We found normal subtelomeric methylation in ICF2-4 and accordingly low TERRA levels and unperturbed telomere length. Moreover, depleting the ICF2-4-related proteins in normal fibroblasts did not influence subtelomeric methylation. Thus, these gene products are not involved in establishing or maintaining subtelomeric methylation. Our findings indicate that human subtelomeric heterochromatin has specialized methylation regulation and highlight the telomeric phenotype as a characteristic that distinguishes ICF1 from ICF2-4.


Asunto(s)
Anomalías Múltiples/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Helicasas/genética , Metilación de ADN , Mutación , Proteínas Nucleares/genética , Proteínas Represoras/genética , Anomalías Múltiples/metabolismo , Adolescente , Adulto , Línea Celular , Centrómero , Niño , Preescolar , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Helicasas/metabolismo , Cara/anomalías , Fibroblastos , Heterocromatina/metabolismo , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Lactante , Recién Nacido , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Telómero/metabolismo , Adulto Joven , ADN Metiltransferasa 3B
6.
Hum Mol Genet ; 27(14): 2409-2424, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29659838

RESUMEN

Alterations of DNA methylation landscapes and machinery are a hallmark of many human diseases. A prominent case is the ICF syndrome, a rare autosomal recessive immunological/neurological disorder diagnosed by the loss of DNA methylation at (peri)centromeric repeats and its associated chromosomal instability. It is caused by mutations in the de novo DNA methyltransferase DNMT3B in about half of the patients (ICF1). In the remainder, the striking identification of mutations in factors devoid of DNA methyltransferase activity, ZBTB24 (ICF2), CDCA7 (ICF3) or HELLS (ICF4), raised key questions about common or distinguishing DNA methylation alterations downstream of these mutations and hence, about the functional link between the four factors. Here, we established the first comparative methylation profiling in ICF patients with all four genotypes and we provide evidence that, despite unifying hypomethylation of pericentromeric repeats and a few common loci, methylation profiling clearly distinguished ICF1 from ICF2, 3 and 4 patients. Using available genomic and epigenomic annotations to characterize regions prone to loss of DNA methylation downstream of ICF mutations, we found that ZBTB24, CDCA7 and HELLS mutations affect CpG-poor regions with heterochromatin features. Among these, we identified clusters of coding and non-coding genes mostly expressed in a monoallelic manner and implicated in neuronal development, consistent with the clinical spectrum of these patients' subgroups. Hence, beyond providing blood-based biomarkers of dysfunction of ICF factors, our comparative study unveiled new players to consider at certain heterochromatin regions of the human genome.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , ADN Helicasas/genética , Síndromes de Inmunodeficiencia/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Inestabilidad Cromosómica/genética , Metilación de ADN/genética , Femenino , Genoma Humano/genética , Genotipo , Heterocromatina/genética , Humanos , Síndromes de Inmunodeficiencia/fisiopatología , Masculino , Mutación , Neurogénesis/genética , ADN Metiltransferasa 3B
7.
RNA Biol ; 17(12): 1707-1720, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32559119

RESUMEN

The findings that an RNA is not necessarily either coding or non-coding, or that a precursor RNA can produce different types of mature RNAs, whether coding or non-coding, long or short, have challenged the dichotomous view of the RNA world almost 15 years ago. Since then, and despite an increasing number of studies, the diversity of information that can be conveyed by RNAs is rarely searched for, and when it is known, it remains largely overlooked in further functional studies. Here, we provide an update with prominent examples of multiple functions that are carried by the same RNA or are produced by the same precursor RNA, to emphasize their biological relevance in most living organisms. An important consequence is that the overall function of their locus of origin results from the balance between various RNA species with distinct functions and fates. The consideration of the molecular basis of this multiplicity of information is obviously crucial for downstream functional studies when the targeted functional molecule is often not the one that is believed.


Asunto(s)
Regulación de la Expresión Génica , ARN/genética , Animales , Evolución Molecular , Exones , Humanos , Intrones , Sistemas de Lectura Abierta , Empalme del ARN , ARN Circular , ARN no Traducido/genética
9.
Clin Genet ; 95(2): 210-220, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30456829

RESUMEN

Alterations in epigenetic landscapes are hallmarks of many complex human diseases, yet, it is often challenging to assess the underlying mechanisms and causal link with clinical manifestations. In this regard, monogenic diseases that affect actors of the epigenetic machinery are of considerable interest to learn more about the etiology of complex traits. Spectacular breakthroughs in medical genetics are largely the result of advances in genome-wide approaches to identify genomic and epigenomic alterations in patients. These approaches have enabled the identification of an ever-increasing number of hereditary disorders caused by defects in the establishment of epigenetic marks early during development or in the perpetuation of such marks at later stages. We focus our review on particular cases where DNA methylation landscapes are altered at the genome scale, whether it is a direct consequence of mutations in DNA methyltransferases (DNMT) or that it reflects initial alterations of chromatin states or guiding factors caused by mutations in chromatin modifiers or transcription factors. Collectively, increased knowledge of these rare diseases will add to our understanding of the genetic determinants of DNA methylation in humans. Moreover, investigating how perturbations to these determinants affect genome function has far-reaching potential to understand various complex human diseases.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Predisposición Genética a la Enfermedad , Enfermedades Raras/genética , Animales , Cromatina/genética , ADN (Citosina-5-)-Metiltransferasa 1/genética , Regulación de la Expresión Génica , Estudios de Asociación Genética , Marcadores Genéticos , Humanos , Mutación , Enfermedades Raras/diagnóstico , Factores de Transcripción/metabolismo
10.
Nucleic Acids Res ; 45(8): 4768-4781, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28053119

RESUMEN

Introns represent almost half of the human genome, although they are eliminated from transcripts through RNA splicing. Yet, different classes of non-canonical miRNAs have been proposed to originate directly from intron splicing. Here, we considered the alternative splicing of introns as an interesting source of miRNAs, compatible with a developmental switch. We report computational prediction of new Short Intron-Derived ncRNAs (SID), defined as precursors of smaller ncRNAs like miRNAs and snoRNAs produced directly by splicing, and tested their dependence on each key factor in canonical or alternative miRNAs biogenesis (Drosha, DGCR8, DBR1, snRNP70, U2AF65, PRP8, Dicer, Ago2). We found that about half of predicted SID rely on debranching of the excised intron-lariat by the enzyme DBR1, as proposed for mirtrons. However, we identified new classes of SID for which miRNAs biogenesis may rely on intermingling between canonical and alternative pathways. We validated selected SID as putative miRNAs precursors and identified new endogenous miRNAs produced by non-canonical pathways, including one hosted in the first intron of SRA (Steroid Receptor RNA activator). Consistent with increased SRA intron retention during myogenic differentiation, release of SRA intron and its associated mature miRNA decreased in cells from healthy subjects but not from myotonic dystrophy patients with splicing defects.


Asunto(s)
Intrones/genética , MicroARNs/genética , ARN no Traducido/genética , Empalme Alternativo/genética , Biología Computacional , Genoma Humano , Humanos , MicroARNs/biosíntesis , Precursores del ARN/genética
11.
Nucleic Acids Res ; 45(10): 5739-5756, 2017 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-28334849

RESUMEN

Hypomorphic mutations in DNA-methyltransferase DNMT3B cause majority of the rare disorder Immunodeficiency, Centromere instability and Facial anomalies syndrome cases (ICF1). By unspecified mechanisms, mutant-DNMT3B interferes with lymphoid-specific pathways resulting in immune response defects. Interestingly, recent findings report that DNMT3B shapes intragenic CpG-methylation of highly-transcribed genes. However, how the DNMT3B-dependent epigenetic network modulates transcription and whether ICF1-specific mutations impair this process remains unknown. We performed a transcriptomic and epigenomic study in patient-derived B-cell lines to investigate the genome-scale effects of DNMT3B dysfunction. We highlighted that altered intragenic CpG-methylation impairs multiple aspects of transcriptional regulation, like alternative TSS usage, antisense transcription and exon splicing. These defects preferentially associate with changes of intragenic H3K4me3 and at lesser extent of H3K27me3 and H3K36me3. In addition, we highlighted a novel DNMT3B activity in modulating the self-regulatory circuit of sense-antisense pairs and the exon skipping during alternative splicing, through interacting with RNA molecules. Strikingly, altered transcription affects disease relevant genes, as for instance the memory-B cell marker CD27 and PTPRC genes, providing us with biological insights into the ICF1-syndrome pathogenesis. Our genome-scale approach sheds light on the mechanisms still poorly understood of the intragenic function of DNMT3B and DNA methylation in gene expression regulation.


Asunto(s)
Empalme Alternativo , Anorexia/genética , Caquexia/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Anomalías del Ojo/genética , Histonas/genética , Síndromes de Inmunodeficiencia/genética , Mutación , ARN Mensajero/genética , Enfermedades de la Piel/genética , Anorexia/inmunología , Anorexia/patología , Linfocitos B/inmunología , Linfocitos B/patología , Caquexia/inmunología , Caquexia/patología , Línea Celular Transformada , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/inmunología , Metilación de ADN , Epigénesis Genética , Anomalías del Ojo/inmunología , Anomalías del Ojo/patología , Facies , Femenino , Histonas/inmunología , Humanos , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Memoria Inmunológica , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Masculino , Regiones Promotoras Genéticas , ARN Mensajero/inmunología , Enfermedades de la Piel/inmunología , Enfermedades de la Piel/patología , Transcripción Genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , ADN Metiltransferasa 3B
12.
J Clin Immunol ; 36(2): 149-59, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26851945

RESUMEN

PURPOSE: Autosomal recessive deficiencies of DNMT3B or ZBTB24 account for two-thirds of cases of immunodeficiency, centromeric instability and facial dysmorphism (ICF syndrome). This primary immunodeficiency (PID) is characterized mainly by an antibody deficiency, facial abnormalities and centromeric instability. We analyzed the national cohort of patients with ICF syndrome with the aim of providing a more detailed description of the phenotype and management of patients with ICF syndrome. METHODS: Demographic, genetic, immunological, and clinical features were recorded for each patient. RESULTS: In the French cohort, seven of the nine patients carried DNMT3B mutations, six of which had never been described before. One patient had compound heterozygous ZBTB24 mutations. All patients were found to lack CD19(+)CD27(+) memory B cells. This feature is a major diagnostic criterion for both ICF1 and ICF2. Patients suffered both bacterial and viral infections, and three patients developed bronchiectasis. Autoimmune manifestations (hepatitis, nephritis and thyroiditis) not previously reported in ICF1 patients were also detected in two of our ICF1 patients. The mode of treatment and outcome of the French patients are reported, by genetic defect, and compared with those for 68 previously reported ICF patients. Immunoglobulin (Ig) replacement treatment was administered to all nine French patients. One ICF1 patient presented severe autoimmune manifestations and pancytopenia and underwent allogeneic hematopoietic stem cell transplantation (HSCT), but she died from unknown causes 6 years post-transplant. CONCLUSION: Autoimmune signs are uncommon in ICF syndrome, but, when present, they affect patient outcome and require immunosuppressive treatment. The long-term outcome of ICF patients has been improved by the combination of IgG replacement and antibiotic prophylaxis.


Asunto(s)
Predisposición Genética a la Enfermedad , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/etiología , Fenotipo , Autoinmunidad , Niño , Preescolar , Manejo de la Enfermedad , Femenino , Francia/epidemiología , Pruebas Genéticas , Humanos , Síndromes de Inmunodeficiencia/epidemiología , Síndromes de Inmunodeficiencia/terapia , Pruebas Inmunológicas , Lactante , Recién Nacido , Infecciones/etiología , Masculino , Mutación , Evaluación de Resultado en la Atención de Salud , Vigilancia de la Población
13.
Int J Mol Sci ; 16(3): 4429-52, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25710723

RESUMEN

Introns represent almost half of the human genome, yet their vast majority is eliminated from eukaryotic transcripts through RNA splicing. Nevertheless, they feature key elements and functions that deserve further interest. At the level of DNA, introns are genomic segments that can shelter independent transcription units for coding and non-coding RNAs which transcription may interfere with that of the host gene, and regulatory elements that can influence gene expression and splicing itself. From the RNA perspective, some introns can be subjected to alternative splicing. Intron retention appear to provide some plasticity to the nature of the protein produced, its distribution in a given cell type and timing of its translation. Intron retention may also serve as a switch to produce coding or non-coding RNAs from the same transcription unit. Conversely, splicing of introns has been directly implicated in the production of small regulatory RNAs. Hence, splicing of introns also appears to provide plasticity to the type of RNA produced from a genetic locus (coding, non-coding, short or long). We addressed these aspects to add to our understanding of mechanisms that control the fate of introns and could be instrumental in regulating genomic output and hence cell fate.


Asunto(s)
Empalme Alternativo , Genoma/genética , Intrones/genética , ARN Mensajero/genética , Animales , Exones/genética , Regulación de la Expresión Génica , Variación Genética , Humanos , Modelos Genéticos
14.
J Cell Biol ; 223(4)2024 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-38376465

RESUMEN

DNA methylation (DNAme) is a key epigenetic mark that regulates critical biological processes maintaining overall genome stability. Given its pleiotropic function, studies of DNAme dynamics are crucial, but currently available tools to interfere with DNAme have limitations and major cytotoxic side effects. Here, we present cell models that allow inducible and reversible DNAme modulation through DNMT1 depletion. By dynamically assessing whole genome and locus-specific effects of induced passive demethylation through cell divisions, we reveal a cooperative activity between DNMT1 and DNMT3B, but not of DNMT3A, to maintain and control DNAme. We show that gradual loss of DNAme is accompanied by progressive and reversible changes in heterochromatin, compartmentalization, and peripheral localization. DNA methylation loss coincides with a gradual reduction of cell fitness due to G1 arrest, with minor levels of mitotic failure. Altogether, this system allows DNMTs and DNA methylation studies with fine temporal resolution, which may help to reveal the etiologic link between DNAme dysfunction and human disease.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1 , Metilación de ADN , ADN Metiltransferasa 3A , Epigenómica , Humanos , División Celular , Heterocromatina/genética , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN Metiltransferasa 3A/genética , Línea Celular
15.
J Hum Genet ; 58(7): 455-60, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23739126

RESUMEN

Immunodeficiency, centromeric instability and facial anomalies (ICF) syndrome is a rare autosomal recessive disorder that shows DNA hypomethylation at pericentromeric satellite-2 and -3 repeats in chromosomes 1, 9 and 16. ICF syndrome is classified into two groups: type 1 (ICF1) patients have mutations in the DNMT3B gene and about half of type 2 (ICF2) patients have mutations in the ZBTB24 gene. Besides satellite-2 and -3 repeats, α-satellite repeats are also hypomethylated in ICF2. In this study, we report three novel ZBTB24 mutations in ICF2. A Japanese patient was homozygous for a missense mutation (C383Y), and a Cape Verdean patient was compound heterozygous for a nonsense mutation (K263X) and a frame-shift mutation (C327W fsX54). In addition, the second Japanese patient was homozygous for a previously reported nonsense mutation (R320X). The C383Y mutation abolished a C2H2 motif in one of the eight zinc-finger domains, and the other three mutations caused a complete or large loss of the zinc-finger domains. Our immunofluorescence analysis revealed that mouse Zbtb24 proteins possessing a mutation corresponding to either C383Y or R320X are mislocalized from pericentrometic heterochromatin, suggesting the importance of the zinc-finger domains in proper intranuclear localization of this protein. We further revealed that the proper localization of wild-type Zbtb24 protein does not require DNA methylation.


Asunto(s)
Pueblo Asiatico/genética , Población Negra/genética , Cara/anomalías , Síndromes de Inmunodeficiencia/genética , Proteínas Represoras/genética , Adolescente , Adulto , Animales , Línea Celular , Centrómero/metabolismo , Preescolar , Aberraciones Cromosómicas , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo , Cromosomas Humanos Par 16/genética , Clonación Molecular , Metilación de ADN , Femenino , Genómica , Humanos , Síndromes de Inmunodeficiencia/diagnóstico , Masculino , Ratones , Mutación , Células 3T3 NIH , Enfermedades de Inmunodeficiencia Primaria , Proteínas Recombinantes de Fusión/genética , Análisis de Secuencia , Dedos de Zinc/genética
16.
Nucleic Acids Res ; 39(2): 513-25, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20855289

RESUMEN

The steroid receptor RNA activator (SRA) has the unusual property to function as both a non-coding RNA (ncRNA) and a protein SRAP. SRA ncRNA is known to increase the activity of a range of nuclear receptors as well as the master regulator of muscle differentiation MyoD. The contribution of SRA to either a ncRNA or a protein is influenced by alternative splicing of the first intron, the retention of which disrupts the SRAP open reading frame. We reported here that the ratio between non-coding and coding SRA isoforms increased during myogenic differentiation of human satellite cells but not myotonic dystrophy patient satellite cells, in which differentiation capacity is affected. Using constructs that exclusively produce SRA ncRNA or SRAP, we demonstrated that whereas SRA ncRNA was indeed an enhancer of myogenic differentiation and myogenic conversion of non-muscle cells through the co-activation of MyoD activity, SRAP prevented this SRA RNA-dependant co-activation. Interestingly, the SRAP inhibitory effect is mediated through the interaction of SRAP with its RNA counterpart via its RRM-like domain interacting with the functional sub-structure of SRA RNA, STR7. This study thus provides a new model for SRA-mediated regulation of MyoD transcriptional activity in the promotion of normal muscle differentiation, which takes into account the nature of SRA molecules present.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Desarrollo de Músculos/genética , Proteína MioD/metabolismo , ARN no Traducido/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Animales , Sitios de Unión , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Bases de Datos Genéticas , Variación Genética , Humanos , Ratones , Distrofia Miotónica/genética , Unión Proteica , Interferencia de ARN , ARN Largo no Codificante , ARN no Traducido/antagonistas & inhibidores , ARN no Traducido/química , Células Satélite del Músculo Esquelético/citología
17.
Proc Natl Acad Sci U S A ; 107(20): 9281-6, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20439742

RESUMEN

Methylation of cytosine residues within the CpG dinucleotide in mammalian cells is an important mediator of gene expression, genome stability, X-chromosome inactivation, genomic imprinting, chromatin structure, and embryonic development. The majority of CpG sites in mammalian cells is methylated in a nonrandom fashion, raising the question of how DNA methylation is distributed along the genome. Here, we focused on the functions of DNA methyltransferase-3b (Dnmt3b), of which deregulated activity is linked to several human pathologies. We generated Dnmt3b hypomorphic mutant mice with reduced catalytic activity, which first revealed a deregulation of Hox genes expression, consistent with the observed homeotic transformations of the posterior axis. In addition, analysis of deregulated expression programs in Dnmt3b mutant embryos, using DNA microarrays, highlighted illegitimate activation of several germ-line genes in somatic tissues that appeared to be linked directly to their hypomethylation in mutant embryos. We provide evidence that these genes are direct targets of Dnmt3b. Moreover, the recruitment of Dnmt3b to their proximal promoter is dependant on the binding of the E2F6 transcriptional repressor, which emerges as a common hallmark in the promoters of genes found to be up-regulated as a consequence of impaired Dnmt3b activity. Therefore, our results unraveled a coordinated regulation of genes involved in meiosis, through E2F6-dependant methylation and transcriptional silencing in somatic tissues.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Factor de Transcripción E2F6/metabolismo , Silenciador del Gen/fisiología , Meiosis/genética , Proteínas Represoras/metabolismo , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Islas de CpG/genética , Proteínas de Homeodominio/genética , Ratones , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , ADN Metiltransferasa 3B
18.
Noncoding RNA ; 8(5)2022 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-36287115

RESUMEN

Pseudouridylation is one of the most abundant modifications found in RNAs. To identify the Pseudouridylation sites (Psi) in RNAs, several techniques have been developed, but the most common and robust is the CMC (N-cyclohexyl-N'-(2-morpholinoethyl)carbodiimide) treatment, which consists in the addition of an adduct on Psi that inhibits the reverse transcription. Here, we describe a turnkey method and a tool to design the bridging oligo (DBO), which is somewhat difficult to design. Finally, we propose a trouble-shooting guide to help users.

19.
FEBS J ; 289(7): 1858-1875, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34739170

RESUMEN

Cell cycle progression requires control of the abundance of several proteins and RNAs over space and time to properly transit from one phase to the next and to ensure faithful genomic inheritance in daughter cells. The proteasome, the main protein degradation system of the cell, facilitates the establishment of a proteome specific to each phase of the cell cycle. Its activity also strongly influences transcription. Here, we detected the upregulation of repetitive RNAs upon proteasome inhibition in human cancer cells using RNA-seq. The effect of proteasome inhibition on centromeres was remarkable, especially on α-Satellite RNAs. We showed that α-Satellite RNAs fluctuate along the cell cycle and interact with members of the cohesin ring, suggesting that these transcripts may take part in the regulation of mitotic progression. Next, we forced exogenous overexpression and used gapmer oligonucleotide targeting to demonstrate that α-Sat RNAs have regulatory roles in mitosis. Finally, we explored the transcriptional regulation of α-Satellite DNA. Through in silico analyses, we detected the presence of CCAAT transcription factor-binding motifs within α-Satellite centromeric arrays. Using high-resolution three-dimensional immuno-FISH and ChIP-qPCR, we showed an association between the α-Satellite upregulation and the recruitment of the transcription factor NFY-A to the centromere upon MG132-induced proteasome inhibition. Together, our results show that the proteasome controls α-Satellite RNAs associated with the regulation of mitosis.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Satélite de ARN , Centrómero/genética , Centrómero/metabolismo , ADN Satélite/genética , Humanos , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Satélite de ARN/genética , Regulación hacia Arriba
20.
Nucleic Acids Res ; 37(15): 5071-80, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19542185

RESUMEN

Non-coding RNAs are emerging as key players in many fundamental biological processes, including specification of higher-order chromatin structure. We examined the implication of RNA transcribed from mouse centromeric minor satellite repeats in the formation and function of centromere-associated complexes. Here we show that the levels of minor satellite RNA vary during cell-cycle progression, peaking in G2/M phase, concomitant with accumulation of proteins of the chromosomal passenger complex near the centromere. Consistent with this, we describe that murine minor satellite RNA are components of CENP-A-associated centromeric fractions and associate with proteins of the chromosomal passenger complex Aurora B and Survivin at the onset of mitosis. Interactions of endogenous Aurora B with CENP-A and Survivin are sensitive to RNaseA. Likewise, the kinase activity of Aurora B requires an RNA component. More importantly, Aurora B kinase activity can be potentiated by minor satellite RNA. In addition, decreased Aurora B activity after RNA depletion can be specifically rescued by restitution of these transcripts. Together, our data provide new functional evidence for minor satellite transcripts as key partners and regulators of the mitotic kinase Aurora B.


Asunto(s)
Centrómero/química , Cromatina/química , Proteínas Serina-Treonina Quinasas/metabolismo , ARN no Traducido/metabolismo , Animales , Aurora Quinasa B , Aurora Quinasas , Autoantígenos/análisis , Ciclo Celular/genética , Línea Celular , Proteína A Centromérica , Proteínas Cromosómicas no Histona/análisis , ADN Satélite , Proteínas Inhibidoras de la Apoptosis , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/genética , ARN no Traducido/análisis , Proteínas Represoras , Survivin
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA