Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nucleic Acids Res ; 49(13): 7507-7524, 2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34181717

RESUMEN

Impaired replication progression leads to de novo copy number variant (CNV) formation at common fragile sites (CFSs). We previously showed that these hotspots for genome instability reside in late-replicating domains associated with large transcribed genes and provided indirect evidence that transcription is a factor in their instability. Here, we compared aphidicolin (APH)-induced CNV and CFS frequency between wild-type and isogenic cells in which FHIT gene transcription was ablated by promoter deletion. Two promoter-deletion cell lines showed reduced or absent CNV formation and CFS expression at FHIT despite continued instability at the NLGN1 control locus. APH treatment led to critical replication delays that remained unresolved in G2/M in the body of many, but not all, large transcribed genes, an effect that was reversed at FHIT by the promoter deletion. Altering RNase H1 expression did not change CNV induction frequency and DRIP-seq showed a paucity of R-loop formation in the central regions of large genes, suggesting that R-loops are not the primary mediator of the transcription effect. These results demonstrate that large gene transcription is a determining factor in replication stress-induced genomic instability and support models that CNV hotspots mainly result from the transcription-dependent passage of unreplicated DNA into mitosis.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Variaciones en el Número de Copia de ADN , Replicación del ADN , Proteínas de Neoplasias/genética , Transcripción Genética , Ácido Anhídrido Hidrolasas/biosíntesis , Animales , Afidicolina/farmacología , Línea Celular , Sitios Frágiles del Cromosoma , Sitios Genéticos , Humanos , Ratones , Mutación , Proteínas de Neoplasias/biosíntesis , Regiones Promotoras Genéticas , Estructuras R-Loop , Ribonucleasa H/metabolismo , Estrés Fisiológico
2.
Hum Genet ; 137(11-12): 921-939, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30450527

RESUMEN

Juvenile segmental progeroid syndromes are rare, heterogeneous disorders characterized by signs of premature aging affecting more than one tissue or organ starting in childhood. Hutchinson-Gilford progeria syndrome (HGPS), caused by a recurrent de novo synonymous LMNA mutation resulting in aberrant splicing and generation of a mutant product called progerin, is a prototypical example of such disorders. Here, we performed a joint collaborative study using massively parallel sequencing and targeted Sanger sequencing, aimed at delineating the underlying genetic cause of 14 previously undiagnosed, clinically heterogeneous, non-LMNA-associated juvenile progeroid patients. The molecular diagnosis was achieved in 11 of 14 cases (~ 79%). Furthermore, we firmly establish biallelic mutations in POLR3A as the genetic cause of a recognizable, neonatal, Wiedemann-Rautenstrauch-like progeroid syndrome. Thus, we suggest that POLR3A mutations are causal for a portion of under-diagnosed early-onset segmental progeroid syndromes. We additionally expand the clinical spectrum associated with PYCR1 mutations by showing that they can somewhat resemble HGPS in the first year of life. Moreover, our results lead to clinical reclassification in one single case. Our data emphasize the complex genetic and clinical heterogeneity underlying progeroid disorders.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Progeria/genética , Pirrolina Carboxilato Reductasas/genética , ARN Polimerasa III/genética , Adolescente , Empalme Alternativo/genética , Niño , Femenino , Retardo del Crecimiento Fetal/diagnóstico , Retardo del Crecimiento Fetal/patología , Predisposición Genética a la Enfermedad , Humanos , Lactante , Lamina Tipo A/genética , Masculino , Mutación , Fenotipo , Progeria/diagnóstico , Progeria/patología , Progeria/fisiopatología , delta-1-Pirrolina-5-Carboxilato Reductasa
3.
Genome Res ; 25(2): 189-200, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25373142

RESUMEN

Copy number variants (CNVs) resulting from genomic deletions and duplications and common fragile sites (CFSs) seen as breaks on metaphase chromosomes are distinct forms of structural chromosome instability precipitated by replication inhibition. Although they share a common induction mechanism, it is not known how CNVs and CFSs are related or why some genomic loci are much more prone to their occurrence. Here we compare large sets of de novo CNVs and CFSs in several experimental cell systems to each other and to overlapping genomic features. We first show that CNV hotpots and CFSs occurred at the same human loci within a given cultured cell line. Bru-seq nascent RNA sequencing further demonstrated that although genomic regions with low CNV frequencies were enriched in transcribed genes, the CNV hotpots that matched CFSs specifically corresponded to the largest active transcription units in both human and mouse cells. Consistently, active transcription units >1 Mb were robust cell-type-specific predictors of induced CNV hotspots and CFS loci. Unlike most transcribed genes, these very large transcription units replicated late and organized deletion and duplication CNVs into their transcribed and flanking regions, respectively, supporting a role for transcription in replication-dependent lesion formation. These results indicate that active large transcription units drive extreme locus- and cell-type-specific genomic instability under replication stress, resulting in both CNVs and CFSs as different manifestations of perturbed replication dynamics.


Asunto(s)
Sitios Frágiles del Cromosoma , Variaciones en el Número de Copia de ADN , Replicación del ADN , Transcripción Genética , Animales , Línea Celular , Rotura Cromosómica , Mapeo Cromosómico , Sitios Genéticos , Inestabilidad Genómica , Humanos , Hibridación Fluorescente in Situ , Ratones , Especificidad de Órganos , Reproducibilidad de los Resultados
4.
J Med Genet ; 54(3): 212-216, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27920058

RESUMEN

BACKGROUND: Hutchinson-Gilford progeria syndrome (HGPS) is a fatal sporadic autosomal dominant premature ageing disease caused by single base mutations that optimise a cryptic splice site within exon 11 of the LMNA gene. The resultant disease-causing protein, progerin, acts as a dominant negative. Disease severity relies partly on progerin levels. METHODS AND RESULTS: We report a novel form of somatic mosaicism, where a child possessed two cell populations with different HGPS disease-producing mutations of the same nucleotide-one producing severe HGPS and one mild HGPS. The proband possessed an intermediate phenotype. The mosaicism was initially discovered when Sanger sequencing showed a c.1968+2T>A mutation in blood DNA and a c.1968+2T>C in DNA from cultured fibroblasts. Deep sequencing of DNA from the proband's blood revealed 4.7% c.1968+2T>C mutation, and 41.3% c.1968+2T>A mutation. CONCLUSIONS: We hypothesise that the germline mutation was c.1968+2T>A, but a rescue event occurred during early development, where the somatic mutation from A to C at 1968+2 provided a selective advantage. This type of mosaicism where a partial phenotypic rescue event results from a second but milder disease-causing mutation in the same nucleotide has not been previously characterised for any disease.


Asunto(s)
Núcleo Celular/genética , Lamina Tipo A/genética , Progeria/genética , Adolescente , Núcleo Celular/patología , Células Cultivadas , Niño , Preescolar , Exones/genética , Femenino , Fibroblastos/patología , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Mosaicismo , Progeria/patología
5.
Hum Mol Genet ; 22(24): 4901-13, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23863462

RESUMEN

SNM1B/Apollo is a DNA nuclease that has important functions in telomere maintenance and repair of DNA interstrand crosslinks (ICLs) within the Fanconi anemia (FA) pathway. SNM1B is required for efficient localization of key repair proteins, such as the FA protein, FANCD2, to sites of ICL damage and functions epistatically to FANCD2 in cellular survival to ICLs and homology-directed repair. The FA pathway is also activated in response to replication fork stalling. Here, we sought to determine the importance of SNM1B in cellular responses to stalled forks in the absence of a blocking lesion, such as ICLs. We found that depletion of SNM1B results in hypersensitivity to aphidicolin, a DNA polymerase inhibitor that causes replication stress. We observed that the SNM1B nuclease is required for efficient localization of the DNA repair proteins, FANCD2 and BRCA1, to subnuclear foci upon aphidicolin treatment, thereby indicating SNM1B facilitates direct repair of stalled forks. Consistent with a role for SNM1B subsequent to recognition of the lesion, we found that SNM1B is dispensable for upstream events, including activation of ATR-dependent signaling and localization of RPA, γH2AX and the MRE11/RAD50/NBS1 complex to aphidicolin-induced foci. We determined that a major consequence of SNM1B depletion is a marked increase in spontaneous and aphidicolin-induced chromosomal gaps and breaks, including breakage at common fragile sites. Thus, this study provides evidence that SNM1B functions in resolving replication stress and preventing accumulation of genomic damage.


Asunto(s)
Sitios Frágiles del Cromosoma , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN , Inestabilidad Genómica , Proteínas Nucleares/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Afidicolina/farmacología , Proteína BRCA1/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cromatina/metabolismo , Daño del ADN , Reparación del ADN , Enzimas Reparadoras del ADN/química , Enzimas Reparadoras del ADN/genética , Exodesoxirribonucleasas , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Expresión Génica , Histonas/metabolismo , Humanos , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteína de Replicación A/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitinación
6.
PLoS Genet ; 8(9): e1002981, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23028374

RESUMEN

Spontaneous copy number variant (CNV) mutations are an important factor in genomic structural variation, genomic disorders, and cancer. A major class of CNVs, termed nonrecurrent CNVs, is thought to arise by nonhomologous DNA repair mechanisms due to the presence of short microhomologies, blunt ends, or short insertions at junctions of normal and de novo pathogenic CNVs, features recapitulated in experimental systems in which CNVs are induced by exogenous replication stress. To test whether the canonical nonhomologous end joining (NHEJ) pathway of double-strand break (DSB) repair is involved in the formation of this class of CNVs, chromosome integrity was monitored in NHEJ-deficient Xrcc4(-/-) mouse embryonic stem (ES) cells following treatment with low doses of aphidicolin, a DNA replicative polymerase inhibitor. Mouse ES cells exhibited replication stress-induced CNV formation in the same manner as human fibroblasts, including the existence of syntenic hotspot regions, such as in the Auts2 and Wwox loci. The frequency and location of spontaneous and aphidicolin-induced CNV formation were not altered by loss of Xrcc4, as would be expected if canonical NHEJ were the predominant pathway of CNV formation. Moreover, de novo CNV junctions displayed a typical pattern of microhomology and blunt end use that did not change in the absence of Xrcc4. A number of complex CNVs were detected in both wild-type and Xrcc4(-/-) cells, including an example of a catastrophic, chromothripsis event. These results establish that nonrecurrent CNVs can be, and frequently are, formed by mechanisms other than Xrcc4-dependent NHEJ.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Reparación del ADN por Unión de Extremidades/genética , Proteínas de Unión al ADN/genética , Células Madre Embrionarias , Animales , Afidicolina/farmacología , Proteínas del Citoesqueleto , Roturas del ADN de Doble Cadena , Replicación del ADN/efectos de los fármacos , Humanos , Ratones , Proteínas Nucleares/genética , Oxidorreductasas/genética , Factores de Transcripción , Oxidorreductasa que Contiene Dominios WW
7.
Proc Natl Acad Sci U S A ; 108(42): 17360-5, 2011 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-21987784

RESUMEN

Copy number variants (CNVs) are widely distributed throughout the human genome, where they contribute to genetic variation and phenotypic diversity. Spontaneous CNVs are also a major cause of genetic and developmental disorders and arise frequently in cancer cells. As with all mutation classes, genetic and environmental factors almost certainly increase the risk for new and deleterious CNVs. However, despite the importance of CNVs, there is limited understanding of these precipitating risk factors and the mechanisms responsible for a large percentage of CNVs. Here we report that low doses of hydroxyurea, an inhibitor of ribonucleotide reductase and an important drug in the treatment of sickle cell disease and other diseases induces a high frequency of de novo CNVs in cultured human cells that resemble pathogenic and aphidicolin-induced CNVs in size and breakpoint structure. These CNVs are distributed throughout the genome, with some hotspots of de novo CNV formation. Sequencing revealed that CNV breakpoint junctions are characterized by short microhomologies, blunt ends, and short insertions. These data provide direct experimental support for models of replication-error origins of CNVs and suggest that any agent or condition that leads to replication stress has the potential to induce deleterious CNVs. In addition, they point to a need for further study of the genomic consequences of the therapeutic use of hydroxyurea.


Asunto(s)
Variaciones en el Número de Copia de ADN/efectos de los fármacos , Hidroxiurea/farmacología , Antidrepanocíticos/farmacología , Afidicolina/farmacología , Secuencia de Bases , Células Cultivadas , Cromosomas Humanos Par 3/efectos de los fármacos , Cromosomas Humanos Par 3/genética , Roturas del ADN/efectos de los fármacos , Variaciones en el Número de Copia de ADN/genética , Replicación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Hidroxiurea/administración & dosificación , Hidroxiurea/efectos adversos , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Ribonucleótido Reductasas/antagonistas & inhibidores
8.
Am J Hum Genet ; 84(3): 339-50, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19232554

RESUMEN

Copy number variants (CNVs) are an important component of genomic variation in humans and other mammals. Similar de novo deletions and duplications, or copy number changes (CNCs), are now known to be a major cause of genetic and developmental disorders and to arise somatically in many cancers. A major mechanism leading to both CNVs and disease-associated CNCs is meiotic unequal crossing over, or nonallelic homologous recombination (NAHR), mediated by flanking repeated sequences or segmental duplications. Others appear to involve nonhomologous end joining (NHEJ) or aberrant replication suggesting a mitotic cell origin. Here we show that aphidicolin-induced replication stress in normal human cells leads to a high frequency of CNCs of tens to thousands of kilobases across the human genome that closely resemble CNVs and disease-associated CNCs. Most deletion and duplication breakpoint junctions were characterized by short (<6 bp) microhomologies, consistent with the hypothesis that these rearrangements were formed by NHEJ or a replication-coupled process, such as template switching. This is a previously unrecognized consequence of replication stress and suggests that replication fork stalling and subsequent error-prone repair are important mechanisms in the formation of CNVs and pathogenic CNCs in humans.


Asunto(s)
Replicación del ADN/genética , Dosificación de Gen , Variación Genética , Genoma Humano , Afidicolina/farmacología , Células Cultivadas , Replicación del ADN/efectos de los fármacos , Replicación del ADN/fisiología , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Estudio de Asociación del Genoma Completo , Humanos , Recombinación Genética
9.
Proc Natl Acad Sci U S A ; 105(1): 246-51, 2008 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-18162546

RESUMEN

Common fragile sites (CFSs) are loci that preferentially exhibit metaphase chromosome gaps and breaks after partial inhibition of DNA synthesis. The fragile site FRA3B, which lies within the FHIT tumor-suppressor gene, is a site of frequent heterozygous and homozygous deletions in many cancer cells and precancerous lesions. The great majority of FHIT and other CFS-associated gene rearrangements in tumors are submicroscopic, intralocus deletions of hundreds of kilobases that often result in inactivation of associated genes. Although CFS instability leads to chromosome gaps and breaks and translocations, there has been no direct evidence showing that CFS instability or replication stress can generate large submicroscopic deletions of the type seen in cancer cells. Here, we have produced FHIT/FRA3B deletions closely resembling those in tumors by exposing human-mouse chromosome 3 somatic hybrid cells to aphidicolin-mediated replication stress. Clonal cell populations were analyzed for deletions by using PCR, array comparative genomic hybridization (aCGH), and FISH. Thirteen percent to 23% of clones exhibited submicroscopic FHIT deletions spanning approximately 200-600 kb within FRA3B. Chromosomes with FRA3B deletions exhibited significantly decreased fragility of this locus, with a 2- to 12-fold reduction in metaphase gaps and breaks compared with controls. Sequence analysis showed no regions of homology at breakpoints and suggests involvement of NHEJ in generating the deletions. Our results demonstrate that replication stress induces a remarkably high frequency of tumor-like microdeletions that reduce fragility at a CFS in cultured cells and suggests that similar conditions during tumor formation lead to intralocus deletion and inactivation of genes at CFSs and perhaps elsewhere in the genome.


Asunto(s)
Ácido Anhídrido Hidrolasas/biosíntesis , Ácido Anhídrido Hidrolasas/genética , Replicación del ADN , Eliminación de Gen , Regulación de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neoplasias/genética , Animales , Cromosomas/ultraestructura , Genoma Humano , Humanos , Células Híbridas , Hibridación Fluorescente in Situ , Ratones , Modelos Genéticos , Neoplasias/metabolismo , Hibridación de Ácido Nucleico , Reacción en Cadena de la Polimerasa
10.
G3 (Bethesda) ; 10(6): 1997-2005, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32253194

RESUMEN

Mouse sex chromosomes are enriched for co-amplified gene families, present in tens to hundreds of copies. Co-amplification of Slx/Slxl1 on the X chromosome and Sly on the Y chromosome are involved in dose-dependent meiotic drive, however the role of other co-amplified genes remains poorly understood. Here we demonstrate that the co-amplified gene family on the X chromosome, Srsx, along with two additional partial gene annotations, is actually part of a larger transcription unit, which we name LaidxLaidx is harbored in a 229 kb amplicon that represents the ancestral state as compared to a 525 kb Y-amplicon containing the rearranged LaidyLaidx contains a 25,011 nucleotide open reading frame, predominantly expressed in round spermatids, predicted to encode an 871 kD protein. Laidx has orthologous copies with the rat and also the 825-MY diverged parasitic Chinese liver fluke, Clonorchis sinensis, the likely result of a horizontal gene transfer of rodent Laidx to an ancestor of the liver fluke. To assess the male reproductive functions of Laidx, we generated mice carrying a multi-megabase deletion of the Laidx-ampliconic region. Laidx-deficient male mice do not show detectable reproductive defects in fertility, fecundity, testis histology, and offspring sex ratio. We speculate that Laidx and Laidy represent a now inactive X vs. Y chromosome conflict that occurred in an ancestor of present day mice.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Animales , Genómica , Masculino , Ratones , Ratas , Espermátides , Cromosoma X/genética , Cromosoma Y/genética
11.
Mamm Genome ; 20(6): 375-85, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19504344

RESUMEN

The ATR checkpoint pathway responds to DNA damage during the S/G2 phases of the cell cycle and is activated early in tumorigenesis. Investigation of ATR's role in development and tumorigenesis is complicated by the lethality of homozygous knockout mice and the limited effects of heterozygous deficiency. To overcome this limitation, we sought to create mice with a hypomorphic Atr mutation based on the ATR mutation in the human disease Seckel syndrome-1 (SCKL1). Homozygous SCKL1 mice were generated by targeted knock-in of the A --> G SCKL1 mutation. Western blot and RT-PCR analysis established that homozygotes have no reduction in Atr protein or increase in missplicing as is seen in humans. Thus, the A --> G substitution alone is not sufficient to reproduce in mice the effects that are seen in humans. However, homozygous SCKL1 mice that retain the neo cassette used for targeting have an estimated 66-82% reduction in total Atr protein levels due to missplicing into the neo cassette. Under conditions of APH-induced replication stress, primary fibroblasts from homozygous mice displayed an increase in overall chromosome damage and an increase in gaps and breaks at specific common fragile sites. In addition, mutant cells display a significant delay in checkpoint induction and an increase in DNA damage as assayed by Chk1 phosphorylation and gamma-H2ax levels, respectively. These mice provide a novel model system for studies of Atr deficiency and replication stress.


Asunto(s)
Proteínas de Ciclo Celular/genética , Ciclo Celular , Daño del ADN , Enanismo/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Modelos Animales de Enfermedad , Enanismo/metabolismo , Enanismo/fisiopatología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Mutación , Proteínas Serina-Treonina Quinasas/metabolismo
12.
Genes Chromosomes Cancer ; 47(10): 860-72, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18615677

RESUMEN

Common fragile sites (CFSs) are loci that are especially prone to forming gaps and breaks on metaphase chromosomes under conditions of replication stress. Although much has been learned about the cellular responses to gaps and breaks at CFSs, less is known about what makes these sites inherently unstable. CFS sequences are highly conserved in mammalian evolution and contain a number of sequence motifs that are hypothesized to contribute to their instability. To examine the role of CFS sequences in chromosome breakage, we stably transfected two BACs containing FRA3B sequences and two nonCFS control BACs containing similar sequence content into HCT116 cells and isolated cell clones with BACs integrated at ectopic sites. Integrated BACs were present at just a few to several hundred contiguous copies. Cell clones containing integrated FRA3B BACs showed a significant, three to sevenfold increase in aphidicolin-induced gaps and breaks at the integration site as compared to control BACs. Furthermore, many FRA3B integration sites displayed additional chromosome rearrangements associated with CFS instability. Clones were examined for replication timing and it was found that the integrated FRA3B sequences were not dependent on late replication for their fragility. This is the first direct evidence in human cells that introduction of CFS sequences into ectopic nonfragile loci is sufficient to recapitulate the instability found at CFSs. These data support the hypothesis that sequences at CFSs are inherently unstable, and are a major factor in the formation of replication stress induced gaps and breaks at CFSs.


Asunto(s)
Ácido Anhídrido Hidrolasas/fisiología , Sitios Frágiles del Cromosoma , Fragilidad Cromosómica/genética , Proteínas de Neoplasias/fisiología , Mapeo Cromosómico , Cromosomas Artificiales Bacterianos , Cromosomas Humanos/genética , Replicación del ADN , Biblioteca Genómica , Células HCT116 , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Reacción en Cadena de la Polimerasa , Transfección
13.
Environ Mol Mutagen ; 59(8): 698-714, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30218578

RESUMEN

Copy number variants (CNVs) are important in genome variation and genetic disease, with new mutations arising frequently in the germline and somatic cells. Replication stress caused by aphidicolin and hydroxyurea (HU) is a potent inducer of de novo CNVs in cultured mammalian cells. HU is used extensively for long-term management of sickle cell disease. Here, we examined the effects of HU treatment on germline CNVs in vivo in male mice to explore whether replication stress can act as a CNV mutagen in germline mitotic divisions as in cultured cells and whether this would support a concern for increased CNV mutations in offspring of men treated with HU. Several trials of HU administration were performed by oral gavage and subcutaneous pump, with CNVs characterized in C57BL/6 x C3H/HeJ hybrid mouse offspring by microarray and mate-pair sequencing. HU had a short half-life of ~14 min and a narrow dose window over which studies could be performed while maintaining fertility. Tissue histopathology and reticulocyte micronucleus assays verified that doses had a substantial tissue and genetic toxicity. CNVs were readily detected in offspring that originated in both paternal and maternal mouse strains, as de novo and inherited events. However, HU did not increase CNV formation above baseline levels. These results reveal a high rate of CNV mutagenesis in the mouse germline but do not support the hypothesis that HU would increase CNV formation during mammalian spermatogenesis, perhaps due to highly toxic effects on sperm development or experimental variables related to HU pharmacology in mice. Environ. Mol. Mutagen. 59:698-714, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Replicación del ADN/genética , Células Germinativas/efectos de los fármacos , Hidroxiurea/toxicidad , Espermatogénesis/efectos de los fármacos , Espermatozoides/crecimiento & desarrollo , Animales , Variaciones en el Número de Copia de ADN/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Replicación del ADN/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL
14.
DNA Repair (Amst) ; 5(9-10): 1126-35, 2006 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-16807141

RESUMEN

Common fragile sites are large chromosomal regions that preferentially exhibit gaps or breaks after DNA synthesis is partially perturbed. Fragile site instability in cultured cells is well documented and includes gaps and breaks on metaphase chromosomes, translocation and deletions breakpoints, and sister chromosome exchanges. In recent years, much has been learned about the genomic structure at fragile sites and the cellular mechanisms that monitor their stability. The study of fragile sites has merged with that of cell cycle checkpoints and DNA repair, with multiple proteins from these pathways implicated in fragile site stability, including ATR, BRCA1, CHK1, and RAD51. Since their discovery, fragile sites have been implicated in constitutional and cancer chromosome rearrangements in vivo and recent studies suggest that common fragile sites may serve as markers of chromosome damage caused by replication stress during early tumorigenesis. Here we review the relationship of fragile sites to chromosome rearrangements, particularly in tumor cells, and discuss the mechanisms that may be involved.


Asunto(s)
Sitios Frágiles del Cromosoma , Neoplasias/genética , Translocación Genética , Fragilidad Cromosómica , Cromosomas Humanos , Inestabilidad Genómica , Humanos , Modelos Genéticos , Células Tumorales Cultivadas
15.
Mol Cell Biol ; 24(15): 6701-9, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15254237

RESUMEN

Common fragile sites are loci that form chromosome gaps or breaks when DNA synthesis is partially inhibited. Fragile sites are prone to deletions, translocations, and other rearrangements that can cause the inactivation of associated tumor suppressor genes in cancer cells. It was previously shown that ATR is critical to fragile-site stability and that ATR-deficient cells have greatly elevated fragile-site expression (A. M. Casper, P. Nghiem, M. F. Arlt, and T. W. Glover, Cell 111:779-789, 2002). Here we demonstrate that mouse and human cells deficient for BRCA1, due to mutation or knockdown by RNA interference, also have elevated fragile-site expression. We further show that BRCA1 functions in the induction of the G(2)/M checkpoint after aphidicolin-induced replication stalling and that this checkpoint function is involved in fragile-site stability. These data indicate that BRCA1 is important in fragile-site stability and that fragile sites are recognized by the G(2)/M checkpoint pathway, in which BRCA1 plays a key role. Furthermore, they suggest that mutations in BRCA1 or interacting proteins could lead to rearrangements at fragile sites in cancer cells.


Asunto(s)
Proteína BRCA1/fisiología , Sitios Frágiles del Cromosoma , Fragilidad Cromosómica , Genes BRCA1 , Animales , Sitios de Unión , Línea Celular Tumoral , Daño del ADN , Citometría de Flujo , Fase G2 , Humanos , Ratones , Microscopía Fluorescente , Mitosis , Mutación , Neoplasias/genética , Pruebas de Precipitina , Interferencia de ARN , Transfección
16.
Nat Rev Cancer ; 17(8): 489-501, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28740117

RESUMEN

Ever since initial suggestions that instability at common fragile sites (CFSs) could be responsible for chromosome rearrangements in cancers, CFSs and associated genes have been the subject of numerous studies, leading to questions and controversies about their role and importance in cancer. It is now clear that CFSs are not frequently involved in translocations or other cancer-associated recurrent gross chromosome rearrangements. However, recent studies have provided new insights into the mechanisms of CFS instability, their effect on genome instability, and their role in generating focal copy number alterations that affect the genomic landscape of many cancers.


Asunto(s)
Inestabilidad Cromosómica , Sitios Frágiles del Cromosoma , Variaciones en el Número de Copia de ADN , Neoplasias/genética , Oncogenes/genética , Anafase , Animales , Rotura Cromosómica , Roturas del ADN de Doble Cadena , Replicación del ADN , Reordenamiento Génico , Humanos , Metafase
17.
Environ Mol Mutagen ; 55(2): 103-13, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24327335

RESUMEN

Despite their importance to human genetic variation and disease, little is known about the molecular mechanisms and environmental risk factors that impact copy number variant (CNV) formation. While it is clear that replication stress can lead to de novo CNVs, for example, following treatment of cultured mammalian cells with aphidicolin (APH) and hydroxyurea (HU), the effect of different types of mutagens on CNV induction is unknown. Here we report that ionizing radiation (IR) in the range of 1.5-3.0 Gy effectively induces de novo CNV mutations in cultured normal human fibroblasts. These IR-induced CNVs are found throughout the genome, with the same hotspot regions seen after APH- and HU-induced replication stress. IR produces duplications at a higher frequency relative to deletions than do APH and HU. At most hotspots, these duplications are physically shifted from the regions typically deleted after APH or HU, suggesting different pathways involved in their formation. CNV breakpoint junctions from irradiated samples are characterized by microhomology, blunt ends, and insertions like those seen in spontaneous and APH/HU-induced CNVs and most nonrecurrent CNVs in vivo. The similarity to APH/HU-induced CNVs suggests that low-dose IR induces CNVs through a replication-dependent mechanism, as opposed to replication-independent repair of DSBs. Consistent with this mechanism, a lower yield of CNVs was observed when cells were held for 48 hr before replating after irradiation. These results predict that any environmental DNA damaging agent that impairs replication is capable of creating CNVs.


Asunto(s)
Variaciones en el Número de Copia de ADN/efectos de la radiación , Puntos de Control del Ciclo Celular , Línea Celular , Puntos de Rotura del Cromosoma , Replicación del ADN , Fibroblastos/citología , Fibroblastos/efectos de la radiación , Eliminación de Gen , Duplicación de Gen , Genoma Humano , Humanos , Polimorfismo de Nucleótido Simple
18.
Cancer Genet ; 207(4): 133-40, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24813807

RESUMEN

It has emerged that palindrome-mediated genomic instability generates DNA-based rearrangements. The presence of palindromic AT-rich repeats (PATRRs) at the translocation breakpoints suggested a palindrome-mediated mechanism in the generation of several recurrent constitutional rearrangements: the t(11;22), t(17;22), and t(8;22). To date, all reported PATRR-mediated translocations include the PATRR on chromosome 22 (PATRR22) as a translocation partner. Here, the constitutional rearrangement, t(3;8)(p14.2;q24.1), segregating with renal cell carcinoma in two families, is examined. The chromosome 8 breakpoint lies in PATRR8 in the first intron of the RNF139 (TRC8) gene, whereas the chromosome 3 breakpoint is located in an AT-rich palindromic sequence in intron 3 of the FHIT gene (PATRR3). Thus, the t(3;8) is the first PATRR-mediated, recurrent, constitutional translocation that does not involve PATRR22. Furthermore, we detect de novo translocations similar to the t(11;22) and t(8;22), involving PATRR3 in normal sperm. The breakpoint on chromosome 3 is in proximity to FRA3B, the most common fragile site in the human genome and a site of frequent deletions in tumor cells. However, the lack of involvement of PATRR3 sequence in numerous FRA3B-related deletions suggests that there are several different DNA sequence-based etiologies responsible for chromosome 3p14.2 genomic rearrangements.


Asunto(s)
Carcinoma de Células Renales/genética , Cromosomas Humanos Par 3/genética , Cromosomas Humanos Par 8/genética , Neoplasias Renales/genética , Translocación Genética , Secuencia Rica en At/genética , Ácido Anhídrido Hidrolasas/genética , Animales , Secuencia de Bases , Carcinoma de Células Renales/patología , Línea Celular , Puntos de Rotura del Cromosoma , Humanos , Secuencias Invertidas Repetidas/genética , Neoplasias Renales/patología , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Receptores de Superficie Celular/genética , Análisis de Secuencia de ADN
19.
Curr Opin Genet Dev ; 22(3): 204-10, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22365495

RESUMEN

Copy number variants (CNVs) are widely distributed throughout the human genome, where they contribute to genetic variation and phenotypic diversity. De novo CNVs are also a major cause of numerous genetic and developmental disorders. However, unlike many other types of mutations, little is known about the genetic and environmental risk factors for new and deleterious CNVs. DNA replication errors have been implicated in the generation of a major class of CNVs, the nonrecurrent CNVs. We have found that agents that perturb normal replication and create conditions of replication stress, including hydroxyurea and aphidicolin, are potent inducers of nonrecurrent CNVs in cultured human cells. These findings have broad implications for identifying CNV risk factors and for hydroxyurea-related therapies in humans.


Asunto(s)
Variaciones en el Número de Copia de ADN , Replicación del ADN , Genoma Humano , Estrés Fisiológico , Afidicolina/efectos adversos , Células Cultivadas , Puntos de Rotura del Cromosoma , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/genética , Ambiente , Genes cdc , Variación Genética , Humanos , Hidroxiurea/efectos adversos , Factores de Riesgo
20.
Genetics ; 187(3): 675-83, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21212237

RESUMEN

Copy-number variants (CNVs) are a major source of genetic variation in human health and disease. Previous studies have implicated replication stress as a causative factor in CNV formation. However, existing data are technically limited in the quality of comparisons that can be made between human CNVs and experimentally induced variants. Here, we used two high-resolution strategies-single nucleotide polymorphism (SNP) arrays and mate-pair sequencing-to compare CNVs that occur constitutionally to those that arise following aphidicolin-induced DNA replication stress in the same human cells. Although the optimized methods provided complementary information, sequencing was more sensitive to small variants and provided superior structural descriptions. The majority of constitutional and all aphidicolin-induced CNVs appear to be formed via homology-independent mechanisms, while aphidicolin-induced CNVs were of a larger median size than constitutional events even when mate-pair data were considered. Aphidicolin thus appears to stimulate formation of CNVs that closely resemble human pathogenic CNVs and the subset of larger nonhomologous constitutional CNVs.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Replicación del ADN , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia de ADN/métodos , Afidicolina/farmacología , Línea Celular , Variaciones en el Número de Copia de ADN/efectos de los fármacos , Daño del ADN/genética , Genoma Humano , Humanos , Polimorfismo de Nucleótido Simple/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA