Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Genome Res ; 33(2): 169-183, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36828588

RESUMEN

Bi-allelic hypomorphic mutations in DNMT3B disrupt DNA methyltransferase activity and lead to immunodeficiency, centromeric instability, facial anomalies syndrome, type 1 (ICF1). Although several ICF1 phenotypes have been linked to abnormally hypomethylated repetitive regions, the unique genomic regions responsible for the remaining disease phenotypes remain largely uncharacterized. Here we explored two ICF1 patient-derived induced pluripotent stem cells (iPSCs) and their CRISPR-Cas9-corrected clones to determine whether DNMT3B correction can globally overcome DNA methylation defects and related changes in the epigenome. Hypomethylated regions throughout the genome are highly comparable between ICF1 iPSCs carrying different DNMT3B variants, and significantly overlap with those in ICF1 patient peripheral blood and lymphoblastoid cell lines. These regions include large CpG island domains, as well as promoters and enhancers of several lineage-specific genes, in particular immune-related, suggesting that they are premarked during early development. CRISPR-corrected ICF1 iPSCs reveal that the majority of phenotype-related hypomethylated regions reacquire normal DNA methylation levels following editing. However, at the most severely hypomethylated regions in ICF1 iPSCs, which also display the highest increases in H3K4me3 levels and/or abnormal CTCF binding, the epigenetic memory persists, and hypomethylation remains uncorrected. Overall, we demonstrate that restoring the catalytic activity of DNMT3B can reverse the majority of the aberrant ICF1 epigenome. However, a small fraction of the genome is resilient to this rescue, highlighting the challenge of reverting disease states that are due to genome-wide epigenetic perturbations. Uncovering the basis for the persistent epigenetic memory will promote the development of strategies to overcome this obstacle.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes Inducidas/metabolismo , Epigenoma , Memoria Epigenética , Histonas/metabolismo , Metilación de ADN , ADN (Citosina-5-)-Metiltransferasas/genética
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.
J Hum Genet ; 66(11): 1101-1112, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33980986

RESUMEN

RBL2/p130, a member of the retinoblastoma family of proteins, is a key regulator of cell division and propagates irreversible senescence. RBL2/p130 is also involved in neuronal differentiation and survival, and eliminating Rbl2 in certain mouse strains leads to embryonic lethality accompanied by an abnormal central nervous system (CNS) phenotype. Conflicting reports exist regarding a role of RBL2/p130 in transcriptional regulation of DNA methyltransferases (DNMTs), as well as the control of telomere length. Here we describe the phenotype of three patients carrying bi-allelic RBL2-truncating variants. All presented with infantile hypotonia, severe developmental delay and microcephaly. Malignancies were not reported in carriers or patients. Previous studies carried out on mice and human cultured cells, associated RBL2 loss to DNA methylation and telomere length dysregulation. Here, we investigated whether patient cells lacking RBL2 display related abnormalities. The study of primary patient fibroblasts did not detect abnormalities in expression of DNMTs. Furthermore, methylation levels of whole genome DNA, and specifically of pericentromeric repeats and subtelomeric regions, were unperturbed. RBL2-null fibroblasts show no evidence for abnormal elongation by telomeric recombination. Finally, gradual telomere shortening, and normal onset of senescence were observed following continuous culturing of RBL2-mutated fibroblasts. Thus, this study resolves uncertainties regarding a potential non-redundant role for RBL2 in DNA methylation and telomere length regulation, and indicates that loss of function variants in RBL2 cause a severe autosomal recessive neurodevelopmental disorder in humans.


Asunto(s)
Disfunción Cognitiva/genética , Metilación de ADN/genética , Proteína p130 Similar a la del Retinoblastoma/genética , Acortamiento del Telómero/genética , Adolescente , Adulto , Alelos , Animales , Niño , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/fisiopatología , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/fisiopatología , Femenino , Fibroblastos/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Masculino , Metiltransferasas/genética , Ratones , Microcefalia/complicaciones , Microcefalia/genética , Microcefalia/fisiopatología , Actividad Motora/fisiología , Hipotonía Muscular/complicaciones , Hipotonía Muscular/genética , Hipotonía Muscular/fisiopatología , Telómero/genética , Secuenciación del Exoma
4.
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
5.
Hum Mol Genet ; 26(21): 4244-4256, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28973513

RESUMEN

Mutations in the de novo DNA methyltransferase DNMT3B lead to Immunodeficiency, Centromeric Instability and Facial anomalies (ICF) syndrome, type I. This syndrome is characterized, among other hypomethylated genomic loci, by severe subtelomeric hypomethylation that is associated with abnormally short telomere length. While it was demonstrated that the mean telomere length is significantly shorter in ICF type I cells, it is unknown whether all telomeres are equally vulnerable to shortening. To study this question we determined by combined telomere-FISH and spectral karyotyping the relative length of each individual telomere in lymphoblastoid cell lines (LCLs) generated from multiple ICF syndrome patients and control individuals. Here we confirm the short telomere lengths, and demonstrate that telomere length variance in the ICF patient group is much larger than in the control group, suggesting that not all telomeres shorten in a uniform manner. We identified a subgroup of telomeres whose relatively short lengths can distinguish with a high degree of certainty between a control and an ICF metaphase, proposing that in ICF syndrome cells, certain individual telomeres are consistently at greater risk to shorten than others. The majority of these telomeres display high sequence identity at the distal 2 kb of their subtelomeres, suggesting that the attenuation in DNMT3B methylation capacity affects individual telomeres to different degrees based, at least in part, on the adjacent subtelomeric sequence composition.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Telómero/genética , Anomalías Múltiples/genética , Línea Celular , Centrómero/genética , Centrómero/fisiología , Aberraciones Cromosómicas , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Cara/anomalías , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Masculino , Mutación , Linaje , Enfermedades de Inmunodeficiencia Primaria , Telómero/fisiología , Acortamiento del Telómero/genética , ADN Metiltransferasa 3B
6.
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
7.
J Dermatolog Treat ; 35(1): 2363318, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39103159

RESUMEN

Background: Hidradenitis suppurativa (HS) is a chronic inflammatory skin disease. Most patients with moderate-to-severe disease require long-term antibiotic treatment, or biologic treatments to control their disease. Despite these interventions, relapses are common. This study evaluated the effectiveness of piperacillin/tazobactam treatment in patients with Hurley stage II and III HS who experienced disease flares and did not respond to conventional antibiotic and biologic treatment. Methods: Patients with HS hospitalized at the Department of Dermatology, Sheba Medical Center between August 2021 and January 2023 were retrospectively analyzed. Results: A cohort of ten such patients were treated with piperacillin/tazobactam for 6-21 days. Eight (80%) and two (20%) patients respectively demonstrated 2- and 1-grade improvements, from their baseline HS-Physician Global Assessment score. During the follow-up period, nine patients were monitored. HS Clinical Response (HiSCR) was achieved in six (66.7%) and five (55.6%) patients at the 3- and 6-month follow-up visits, respectively. Conclusions: In conclusion, Piperacillin/tazobactam emerges as a promising therapeutic option for disease flare-up in patients with Hurley stage II and III HS who do not respond to conventional treatment. Thus, piperacillin/tazobactam should be considered as crisis therapy for this patient subset.


Asunto(s)
Antibacterianos , Hidradenitis Supurativa , Combinación Piperacilina y Tazobactam , Índice de Severidad de la Enfermedad , Humanos , Hidradenitis Supurativa/tratamiento farmacológico , Combinación Piperacilina y Tazobactam/uso terapéutico , Femenino , Masculino , Estudios Retrospectivos , Adulto , Antibacterianos/uso terapéutico , Persona de Mediana Edad , Resultado del Tratamiento , Brote de los Síntomas , Adulto Joven
8.
Front Oncol ; 12: 1004014, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36300096

RESUMEN

Among the main metabolic pathways implicated in cancer cell proliferation are those of cholesterol and fatty acid synthesis, both of which are tightly regulated by sterol regulatory element-binding proteins (SREBPs). SREBPs are activated through specific cleavage by membrane-bound transcription factor protease 1 (MBTPS1), a serine protease that cleaves additional substrates (ATF6, BDNF, CREBs and somatostatin), some of which are also implicated in cell proliferation. The goal of this study was to determine whether MBTPS1 may serve as a master regulator in proliferation of colorectal cancer (CRC). Tumors from CRC patients showed variable levels of MBTPS1 mRNA, which were in positive correlation with the levels of SREBPs and ATF6, and in reverse correlation with BDNF levels. Chemical inhibition of MBTPS1 activity in two CRC-derived cell lines resulted in a marked decrease in the levels of SREBPs, but not of its other substrates and a marked decrease in cell proliferation, which suggested that MBTPS1 activity is critical for proliferation of these cells. In accordance, CRISPR/Cas9 targeted knockout (KO) of the MBTPS1 gene resulted in the survival of only a single clone that presented a phenotype of severely attenuated proliferation and marked downregulation of several energy metabolism pathways. We further showed that survival of the MBTPS1 KO clone was dependent upon significant upregulation of the type-1 interferon pathway, the inhibition of which halted proliferation entirely. Finally, rescue of the MBTPS1 KO cells, resulted in partial restoration of MBTPS1 levels, which was in accordance with partial recovery in proliferation and in SREBP levels. These finding suggest that MBTPS1 plays a critical role in regulating colon cancer proliferation primarily through SREBP-associated lipid metabolism, and as such may serve as a possible therapeutic target in CRC.

9.
Life (Basel) ; 11(4)2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33810393

RESUMEN

Most human cancers circumvent senescence by activating a telomere length maintenance mechanism, most commonly involving telomerase activation. A minority of cancers utilize the recombination-based alternative lengthening of telomeres (ALT) pathway. The exact requirements for unleashing normally repressed recombination at telomeres are yet unclear. Epigenetic modifications at telomeric regions were suggested to be pivotal for activating ALT; however, conflicting data exist regarding their exact nature and necessity. To uncover common ALT-positive epigenetic characteristics, we performed a comprehensive analysis of subtelomeric DNA methylation, histone modifications, and TERRA expression in several ALT-positive and ALT-negative cell lines. We found that subtelomeric DNA methylation does not differentiate between the ALT-positive and ALT-negative groups, and most of the analyzed subtelomeres within each group do not share common DNA methylation patterns. Additionally, similar TERRA levels were measured in the ALT-positive and ALT-negative groups, and TERRA levels varied significantly among the members of the ALT-positive group. Subtelomeric H3K4 and H3K9 trimethylation also differed significantly between samples in the ALT-positive group. Our findings do not support a common route by which epigenetic modifications activate telomeric recombination in ALT-positive cells, and thus, different therapeutic approaches will be necessary to overcome ALT-dependent cellular immortalization.

10.
Curr Opin Genet Dev ; 60: 9-16, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32109830

RESUMEN

Subtelomeres are the regions at chromosome ends, immediately adjacent to the terminal telomeric repeats. The majority of human subtelomeres are CpG-rich in their distal two kilobases, and are methylated during early embryonic development by the de novo DNA methyltransferase DNMT3B. The biological relevance of subtelomeric DNA methylation is highlighted by the presence of promoters for the long non-coding TERRA transcripts in these CpG-rich regions. Indeed, deviant subtelomeric methylation has been linked with abnormal telomeric phenotypes, as most strikingly found in ICF syndrome. Here we review recent studies that explore new aspects of subtelomeric methylation regulation and demonstrate the significance of maintaining proper DNA methylation at the extreme distal human subtelomeric regions.


Asunto(s)
Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Neoplasias/patología , Homeostasis del Telómero , Telómero , Humanos , Regiones Promotoras Genéticas
11.
Elife ; 82019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31738163

RESUMEN

DNA methyltransferase 3B (DNMT3B) is the major DNMT that methylates mammalian genomes during early development. Mutations in human DNMT3B disrupt genome-wide DNA methylation patterns and result in ICF syndrome type 1 (ICF1). To study whether normal DNA methylation patterns may be restored in ICF1 cells, we corrected DNMT3B mutations in induced pluripotent stem cells from ICF1 patients. Focusing on repetitive regions, we show that in contrast to pericentromeric repeats, which reacquire normal methylation, the majority of subtelomeres acquire only partial DNA methylation and, accordingly, the ICF1 telomeric phenotype persists. Subtelomeres resistant to de novo methylation were characterized by abnormally high H3K4 trimethylation (H3K4me3), and short-term reduction of H3K4me3 by pharmacological intervention partially restored subtelomeric DNA methylation. These findings demonstrate that the abnormal epigenetic landscape established in ICF1 cells restricts the recruitment of DNMT3B, and suggest that rescue of epigenetic diseases with genome-wide disruptions will demand further manipulation beyond mutation correction.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , Cara/anomalías , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedades de Inmunodeficiencia Primaria/genética , Epigénesis Genética/genética , Cara/patología , Genoma/genética , Histonas/genética , Humanos , Mutación , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Enfermedades de Inmunodeficiencia Primaria/patología , Regiones Promotoras Genéticas/genética , Telómero/genética , ADN Metiltransferasa 3B
12.
FEBS J ; 285(14): 2552-2566, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29637701

RESUMEN

R-loops (RLs) are three-stranded nucleic acid structures that contain a DNA:RNA hybrid and a displaced DNA strand. Genomic regions with GC skew and a G-rich transcript are particularly prone to form RLs. RLs play important physiological roles in cells; however, when present at abnormally high levels, they may threaten genome stability. The perfect GC skew of telomeric repeats and the discovery of telomeric repeat-containing RNA (TERRA), a long noncoding transcript that consists of the G-rich telomeric sequence, make telomeric sequences the perfect candidates for generating RLs. Indeed, in the past 5 years, telomere R-loops (TRLs) have been demonstrated in Saccharomyces cerevisiae, Trypanosoma brucei, and human cells. The presence of TRLs in normal human cells that transcribe low levels of TERRA, suggests a physiological role for these nucleic structures in telomere maintenance. Abnormally enhanced TERRA transcription, as found in several human pathological conditions, leads to high TRL levels and various cellular outcomes, depending on the recombinogenic capabilities of the cells. Study of TRLs in various organisms highlights the necessity for tight regulation of these structures, which can switch from beneficial to detrimental under different conditions. Here, we review the current state of knowledge on TRLs, describe several means by which TRLs are regulated, and discuss how findings from yeast are relevant to human pathological scenarios in which TRLs are deregulated.


Asunto(s)
ADN/química , Cara/anomalías , Genoma , Síndromes de Inmunodeficiencia/genética , Hibridación de Ácido Nucleico , ARN Largo no Codificante/química , Telómero/química , Composición de Base , ADN/genética , ADN/metabolismo , Cara/patología , Regulación de la Expresión Génica , Inestabilidad Genómica , Humanos , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Conformación de Ácido Nucleico , Enfermedades de Inmunodeficiencia Primaria , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Telómero/metabolismo , Homeostasis del Telómero , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo
13.
Nat Commun ; 8: 14015, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28117327

RESUMEN

DNA:RNA hybrids, nucleic acid structures with diverse physiological functions, can disrupt genome integrity when dysregulated. Human telomeres were shown to form hybrids with the lncRNA TERRA, yet the formation and distribution of these hybrids among telomeres, their regulation and their cellular effects remain elusive. Here we predict and confirm in several human cell types that DNA:RNA hybrids form at many subtelomeric and telomeric regions. We demonstrate that ICF syndrome cells, which exhibit short telomeres and elevated TERRA levels, are enriched for hybrids at telomeric regions throughout the cell cycle. Telomeric hybrids are associated with high levels of DNA damage at chromosome ends in ICF cells, which are significantly reduced with overexpression of RNase H1. Our findings suggest that abnormally high TERRA levels in ICF syndrome lead to accumulation of telomeric hybrids that, in turn, can result in telomeric dysfunction.


Asunto(s)
Daño del ADN/genética , ADN/metabolismo , Cara/anomalías , Síndromes de Inmunodeficiencia/genética , ARN Largo no Codificante/metabolismo , Telómero/genética , Línea Celular , Inestabilidad Cromosómica/genética , ADN/genética , Humanos , Síndromes de Inmunodeficiencia/sangre , Linfocitos , Cultivo Primario de Células , Enfermedades de Inmunodeficiencia Primaria , ARN Largo no Codificante/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Ribonucleasa H/metabolismo , Acortamiento del Telómero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA