Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Genes Dev ; 38(1-2): 70-94, 2024 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-38316520

RESUMEN

Since genome instability can drive cancer initiation and progression, cells have evolved highly effective and ubiquitous DNA damage response (DDR) programs. However, some cells (for example, in skin) are normally exposed to high levels of DNA-damaging agents. Whether such high-risk cells possess lineage-specific mechanisms that tailor DNA repair to the tissue remains largely unknown. Using melanoma as a model, we show here that the microphthalmia-associated transcription factor MITF, a lineage addition oncogene that coordinates many aspects of melanocyte and melanoma biology, plays a nontranscriptional role in shaping the DDR. On exposure to DNA-damaging agents, MITF is phosphorylated at S325, and its interactome is dramatically remodeled; most transcription cofactors dissociate, and instead MITF interacts with the MRE11-RAD50-NBS1 (MRN) complex. Consequently, cells with high MITF levels accumulate stalled replication forks and display defects in homologous recombination-mediated repair associated with impaired MRN recruitment to DNA damage. In agreement with this, high MITF levels are associated with increased single-nucleotide and copy number variant burdens in melanoma. Significantly, the SUMOylation-defective MITF-E318K melanoma predisposition mutation recapitulates the effects of DNA-PKcs-phosphorylated MITF. Our data suggest that a nontranscriptional function of a lineage-restricted transcription factor contributes to a tissue-specialized modulation of the DDR that can impact cancer initiation.


Asunto(s)
Melanoma , Humanos , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Daño del ADN , Inestabilidad Genómica/genética , ADN
2.
Nucleic Acids Res ; 51(7): 3205-3222, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-36951111

RESUMEN

Chromosomal instability (CIN) drives cell-to-cell heterogeneity, and the development of genetic diseases, including cancer. Impaired homologous recombination (HR) has been implicated as a major driver of CIN, however, the underlying mechanism remains unclear. Using a fission yeast model system, we establish a common role for HR genes in suppressing DNA double-strand break (DSB)-induced CIN. Further, we show that an unrepaired single-ended DSB arising from failed HR repair or telomere loss is a potent driver of widespread CIN. Inherited chromosomes carrying a single-ended DSB are subject to cycles of DNA replication and extensive end-processing across successive cell divisions. These cycles are enabled by Cullin 3-mediated Chk1 loss and checkpoint adaptation. Subsequent propagation of unstable chromosomes carrying a single-ended DSB continues until transgenerational end-resection leads to fold-back inversion of single-stranded centromeric repeats and to stable chromosomal rearrangements, typically isochromosomes, or to chromosomal loss. These findings reveal a mechanism by which HR genes suppress CIN and how DNA breaks that persist through mitotic divisions propagate cell-to-cell heterogeneity in the resultant progeny.


Asunto(s)
Schizosaccharomyces , Humanos , Inestabilidad Cromosómica , Roturas del ADN de Doble Cadena , Reparación del ADN , Recombinación Homóloga , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo
3.
Mol Cell Proteomics ; 21(11): 100410, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36089194

RESUMEN

Little is known about the pathways regulating MHC antigen presentation and the identity of treatment-specific T cell antigens induced by ionizing radiation. For this reason, we investigated the radiation-specific changes in the colorectal tumor cell proteome. We found an increase in DDX58 and ZBP1 protein expression, two nucleic acid sensing molecules likely involved in induction of the dominant interferon response signature observed after genotoxic insult. We further observed treatment-induced changes in key regulators and effector proteins of the antigen processing and presentation machinery. Differential regulation of MHC allele expression was further driving the presentation of a significantly broader MHC-associated peptidome postirradiation, defining a radiation-specific peptide repertoire. Interestingly, treatment-induced peptides originated predominantly from proteins involved in catecholamine synthesis and metabolic pathways. A nuanced relationship between protein expression and antigen presentation was observed where radiation-induced changes in proteins do not correlate with increased presentation of associated peptides. Finally, we detected an increase in the presentation of a tumor-specific neoantigen derived from Mtch1. This study provides new insights into how radiation enhances antigen processing and presentation that could be suitable for the development of combinatorial therapies. Data are available via ProteomeXchange with identifier PXD032003.


Asunto(s)
Presentación de Antígeno , Proteoma , Proteoma/metabolismo , Péptidos/metabolismo , Proteómica , Radiación Ionizante
4.
PLoS Genet ; 17(7): e1009526, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34228709

RESUMEN

Somatic and germline mutations in the proofreading domain of the replicative DNA polymerase ε (POLE-exonuclease domain mutations, POLE-EDMs) are frequently found in colorectal and endometrial cancers and, occasionally, in other tumours. POLE-associated cancers typically display hypermutation, and a unique mutational signature, with a predominance of C > A transversions in the context TCT and C > T transitions in the context TCG. To understand better the contribution of hypermutagenesis to tumour development, we have modelled the most recurrent POLE-EDM (POLE-P286R) in Schizosaccharomyces pombe. Whole-genome sequencing analysis revealed that the corresponding pol2-P287R allele also has a strong mutator effect in vivo, with a high frequency of base substitutions and relatively few indel mutations. The mutations are equally distributed across different genomic regions, but in the immediate vicinity there is an asymmetry in AT frequency. The most abundant base-pair changes are TCT > TAT transversions and, in contrast to human mutations, TCG > TTG transitions are not elevated, likely due to the absence of cytosine methylation in fission yeast. The pol2-P287R variant has an increased sensitivity to elevated dNTP levels and DNA damaging agents, and shows reduced viability on depletion of the Pfh1 helicase. In addition, S phase is aberrant and RPA foci are elevated, suggestive of ssDNA or DNA damage, and the pol2-P287R mutation is synthetically lethal with rad3 inactivation, indicative of checkpoint activation. Significantly, deletion of genes encoding some translesion synthesis polymerases, most notably Pol κ, partially suppresses pol2-P287R hypermutation, indicating that polymerase switching contributes to this phenotype.


Asunto(s)
ADN Polimerasa II/genética , Replicación del ADN , Mutación , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/genética , Quinasa de Punto de Control 2/genética , ADN Helicasas/genética , ADN Polimerasa II/metabolismo , Genoma Fúngico , Humanos , Neoplasias/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Fase S/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo
5.
BMC Cancer ; 23(1): 721, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37528416

RESUMEN

SETD2-dependent H3 Lysine-36 trimethylation (H3K36me3) has been recently linked to the deposition of de-novo DNA methylation. SETD2 is frequently mutated in cancer, however, the functional impact of SETD2 loss and depletion on DNA methylation across cancer types and tumorigenesis is currently unknown. Here, we perform a pan-cancer analysis and show that both SETD2 mutation and reduced expression are associated with DNA methylation dysregulation across 21 out of the 24 cancer types tested. In renal cancer, these DNA methylation changes are associated with altered gene expression of oncogenes, tumour suppressors, and genes involved in neoplasm invasiveness, including TP53, FOXO1, and CDK4. This suggests a new role for SETD2 loss in tumorigenesis and cancer aggressiveness through DNA methylation dysregulation. Moreover, using a robust machine learning methodology, we develop and validate a 3-CpG methylation signature which is sufficient to predict SETD2 mutation status with high accuracy and correlates with patient prognosis.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Metilación de ADN , Histonas/metabolismo , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Carcinogénesis/genética , Transformación Celular Neoplásica/genética
6.
Nucleic Acids Res ; 48(3): 1271-1284, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31828313

RESUMEN

The healing of broken chromosomes by de novo telomere addition, while a normal developmental process in some organisms, has the potential to cause extensive loss of heterozygosity, genetic disease, or cell death. However, it is unclear how de novo telomere addition (dnTA) is regulated at DNA double-strand breaks (DSBs). Here, using a non-essential minichromosome in fission yeast, we identify roles for the HR factors Rqh1 helicase, in concert with Rad55, in suppressing dnTA at or near a DSB. We find the frequency of dnTA in rqh1Δ rad55Δ cells is reduced following loss of Exo1, Swi5 or Rad51. Strikingly, in the absence of the distal homologous chromosome arm dnTA is further increased, with nearly half of the breaks being healed in rqh1Δ rad55Δ or rqh1Δ exo1Δ cells. These findings provide new insights into the genetic context of highly efficient dnTA within HR intermediates, and how such events are normally suppressed to maintain genome stability.


Asunto(s)
ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Reparación del ADN por Recombinación/genética , Proteínas de Schizosaccharomyces pombe/genética , Telómero/genética , Cromosomas Fúngicos/genética , Roturas del ADN de Doble Cadena , Exodesoxirribonucleasas/genética , Regulación Fúngica de la Expresión Génica/genética , Genoma Fúngico/genética , Inestabilidad Genómica/genética , Pérdida de Heterocigocidad/genética , Recombinasa Rad51/genética , Schizosaccharomyces/genética
7.
J Cell Sci ; 132(6)2019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30674555

RESUMEN

Replication stress is a common feature of cancer cells, and thus a potentially important therapeutic target. Here, we show that cyclin-dependent kinase (CDK)-induced replication stress, resulting from Wee1 inactivation, is synthetic lethal with mutations disrupting dNTP homeostasis in fission yeast. Wee1 inactivation leads to increased dNTP demand and replication stress through CDK-induced firing of dormant replication origins. Subsequent dNTP depletion leads to inefficient DNA replication, DNA damage and to genome instability. Cells respond to this replication stress by increasing dNTP supply through histone methyltransferase Set2-dependent MBF-induced expression of Cdc22, the catalytic subunit of ribonucleotide reductase (RNR). Disrupting dNTP synthesis following Wee1 inactivation, through abrogating Set2-dependent H3K36 tri-methylation or DNA integrity checkpoint inactivation results in critically low dNTP levels, replication collapse and cell death, which can be rescued by increasing dNTP levels. These findings support a 'dNTP supply and demand' model in which maintaining dNTP homeostasis is essential to prevent replication catastrophe in response to CDK-induced replication stress.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Nucleótidos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Puntos de Control del Ciclo Celular , Daño del ADN , Replicación del ADN , Código de Histonas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Homeostasis , Metilación , Schizosaccharomyces/metabolismo , Mutaciones Letales Sintéticas , Factores de Transcripción/metabolismo
8.
Nucleic Acids Res ; 46(15): 7731-7746, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-29986057

RESUMEN

The nuclear pore complex (NPC) machinery is emerging as an important determinant in the maintenance of genome integrity and sensitivity to DNA double-strand break (DSB)-inducing agents, such as ionising radiation (IR). In this study, using a high-throughput siRNA screen, we identified the central channel NPC protein Nup54, and concomitantly its molecular partners Nup62 and Nup58, as novel factors implicated in radiosensitivity. Nup54 depletion caused an increase in cell death by mitotic catastrophe after IR, and specifically enhanced both the duration of the G2 arrest and the radiosensitivity of cells that contained replicated DNA at the time of IR exposure. Nup54-depleted cells also exhibited increased formation of chromosome aberrations arisen from replicated DNA. Interestingly, we found that Nup54 is epistatic with the homologous recombination (HR) factor Rad51. Moreover, using specific DNA damage repair reporters, we observed a decreased HR repair activity upon Nup54 knockdown. In agreement with a role in HR repair, we also demonstrated a decreased formation of HR-linked DNA synthesis foci and sister chromatid exchanges after IR in cells depleted of Nup54. Our study reveals a novel role for Nup54 in the response to IR and the maintenance of HR-mediated genome integrity.


Asunto(s)
Replicación del ADN , ADN/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Reparación del ADN por Recombinación , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , ADN/genética , Roturas del ADN de Doble Cadena/efectos de la radiación , Células HeLa , Humanos , Células MCF-7 , Poro Nuclear/genética , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Interferencia de ARN , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Radiación Ionizante , Intercambio de Cromátides Hermanas/efectos de la radiación
9.
Proc Natl Acad Sci U S A ; 114(29): 7671-7676, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28673974

RESUMEN

The partner and localiser of BRCA2 (PALB2) plays important roles in the maintenance of genome integrity and protection against cancer. Although PALB2 is commonly described as a repair factor recruited to sites of DNA breaks, recent studies provide evidence that PALB2 also associates with unperturbed chromatin. Here, we investigated the previously poorly described role of chromatin-associated PALB2 in undamaged cells. We found that PALB2 associates with active genes through its major binding partner, MRG15, which recognizes histone H3 trimethylated at lysine 36 (H3K36me3) by the SETD2 methyltransferase. Missense mutations that ablate PALB2 binding to MRG15 confer elevated sensitivity to the topoisomerase inhibitor camptothecin (CPT) and increased levels of aberrant metaphase chromosomes and DNA stress in gene bodies, which were suppressed by preventing DNA replication. Remarkably, the level of PALB2 at genic regions was frequently decreased, rather than increased, upon CPT treatment. We propose that the steady-state presence of PALB2 at active genes, mediated through the SETD2/H3K36me3/MRG15 axis, ensures an immediate response to DNA stress and therefore effective protection of these regions during DNA replication. This study provides a conceptual advance in demonstrating that the constitutive chromatin association of repair factors plays a key role in the maintenance of genome stability and furthers our understanding of why PALB2 defects lead to human genome instability syndromes.


Asunto(s)
Cromatina/ultraestructura , Daño del ADN , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Factores de Transcripción/metabolismo , Proteína BRCA2/genética , Línea Celular Tumoral , Cromosomas/ultraestructura , Reparación del ADN , Replicación del ADN , Genoma Humano , Células HEK293 , Células HeLa , Humanos , Concentración 50 Inhibidora , Mutación , Unión Proteica , Proteómica , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo
10.
Nucleic Acids Res ; 44(12): 5743-57, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27131361

RESUMEN

DNA double-strand breaks (DSBs) are toxic lesions, which if improperly repaired can result in cell death or genomic instability. DSB repair is usually facilitated by the classical non-homologous end joining (C-NHEJ), or homologous recombination (HR) pathways. However, a mutagenic alternative NHEJ pathway, microhomology-mediated end joining (MMEJ), can also be deployed. While MMEJ is suppressed by C-NHEJ, the relationship between HR and MMEJ is less clear. Here, we describe a role for HR genes in suppressing MMEJ in human cells. By monitoring DSB mis-repair using a sensitive HPRT assay, we found that depletion of HR proteins, including BRCA2, BRCA1 or RPA, resulted in a distinct mutational signature associated with significant increases in break-induced mutation frequencies, deletion lengths and the annealing of short regions of microhomology (2-6 bp) across the break-site. This signature was dependent on CtIP, MRE11, POLQ and PARP, and thus indicative of MMEJ. In contrast to CtIP or MRE11, depletion of BRCA1 resulted in increased partial resection and MMEJ, thus revealing a functional distinction between these early acting HR factors. Together these findings indicate that HR factors suppress mutagenic MMEJ following DSB resection.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , ADN/metabolismo , Reparación del ADN por Recombinación , Proteína de Replicación A/genética , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/metabolismo , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/metabolismo , Secuencia de Bases , Bioensayo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Endodesoxirribonucleasas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Proteína Homóloga de MRE11 , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteína de Replicación A/antagonistas & inhibidores , Proteína de Replicación A/metabolismo , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , ADN Polimerasa theta
11.
Nucleic Acids Res ; 44(4): 1703-17, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26682798

RESUMEN

The formation of RNA-DNA hybrids, referred to as R-loops, can promote genome instability and cancer development. Yet the mechanisms by which R-loops compromise genome instability are poorly understood. Here, we establish roles for the evolutionarily conserved Nrl1 protein in pre-mRNA splicing regulation, R-loop suppression and in maintaining genome stability. nrl1Δ mutants exhibit endogenous DNA damage, are sensitive to exogenous DNA damage, and have defects in homologous recombination (HR) repair. Concomitantly, nrl1Δ cells display significant changes in gene expression, similar to those induced by DNA damage in wild-type cells. Further, we find that nrl1Δ cells accumulate high levels of R-loops, which co-localize with HR repair factors and require Rad51 and Rad52 for their formation. Together, our findings support a model in which R-loop accumulation and subsequent DNA damage sequesters HR factors, thereby compromising HR repair at endogenously or exogenously induced DNA damage sites, leading to genome instability.


Asunto(s)
Empalme Alternativo/genética , Inestabilidad Genómica/genética , Recombinación Homóloga/genética , Precursores del ARN/genética , Proteínas de Schizosaccharomyces pombe/genética , ADN/química , ADN/genética , Reparación del ADN/genética , ARN/química , ARN/genética , Recombinasa Rad51/genética , Proteína Recombinante y Reparadora de ADN Rad52/genética , Schizosaccharomyces/genética , Empalmosomas/genética , Empalmosomas/metabolismo
12.
Genes Dev ; 24(23): 2705-16, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21123655

RESUMEN

Nucleotide synthesis is a universal response to DNA damage, but how this response facilitates DNA repair and cell survival is unclear. Here we establish a role for DNA damage-induced nucleotide synthesis in homologous recombination (HR) repair in fission yeast. Using a genetic screen, we found the Ddb1-Cul4(Cdt)² ubiquitin ligase complex and ribonucleotide reductase (RNR) to be required for HR repair of a DNA double-strand break (DSB). The Ddb1-Cul4(Cdt)² ubiquitin ligase complex is required for degradation of Spd1, an inhibitor of RNR in fission yeast. Accordingly, deleting spd1(+) suppressed the DNA damage sensitivity and the reduced HR efficiency associated with loss of ddb1(+) or cdt2(+). Furthermore, we demonstrate a role for nucleotide synthesis in postsynaptic gap filling of resected ssDNA ends during HR repair. Finally, we define a role for Rad3 (ATR) in nucleotide synthesis and HR through increasing Cdt2 nuclear levels in response to DNA damage. Our findings support a model in which break-induced Rad3 and Ddb1-Cul4(Cdt)² ubiquitin ligase-dependent Spd1 degradation and RNR activation promotes postsynaptic ssDNA gap filling during HR repair.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Quinasas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Quinasa de Punto de Control 2 , Roturas del ADN de Doble Cadena , Reparación del ADN , Eliminación de Gen , Nucleótidos/metabolismo , Recombinación Genética , Ribonucleótido Reductasas/metabolismo
13.
Hum Mol Genet ; 24(24): 7097-110, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26423459

RESUMEN

Understanding the mechanisms of chromosomal double-strand break repair (DSBR) provides insight into genome instability, oncogenesis and genome engineering, including disease gene correction. Research into DSBR exploits rare-cutting endonucleases to cleave exogenous reporter constructs integrated into the genome. Multiple reporter constructs have been developed to detect various DSBR pathways. Here, using a single endogenous reporter gene, the X-chromosomal disease gene encoding hypoxanthine phosphoribosyltransferase (HPRT), we monitor the relative utilization of three DSBR pathways following cleavage by I-SceI or CRISPR/Cas9 nucleases. For I-SceI, our estimated frequencies of accurate or mutagenic non-homologous end-joining and gene correction by homologous recombination are 4.1, 1.5 and 0.16%, respectively. Unexpectedly, I-SceI and Cas9 induced markedly different DSBR profiles. Also, using an I-SceI-sensitive HPRT minigene, we show that gene correction is more efficient when using long double-stranded DNA than single- or double-stranded oligonucleotides. Finally, using both endogenous HPRT and exogenous reporters, we validate novel cell cycle phase-specific I-SceI derivatives for investigating cell cycle variations in DSBR. The results obtained using these novel approaches provide new insights into template design for gene correction and the relationships between multiple DSBR pathways at a single endogenous disease gene.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Endonucleasas/metabolismo , Hipoxantina Fosforribosiltransferasa/genética , Animales , Proteínas Bacterianas/metabolismo , Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Ciclo Celular , Línea Celular Tumoral , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Genes Reporteros , Células HeLa , Humanos , Ratones , Mutagénesis , Proteínas de Saccharomyces cerevisiae/metabolismo
14.
Nucleic Acids Res ; 42(9): 5644-56, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24623809

RESUMEN

DNA double-strand breaks (DSBs) can cause chromosomal rearrangements and extensive loss of heterozygosity (LOH), hallmarks of cancer cells. Yet, how such events are normally suppressed is unclear. Here we identify roles for the DNA damage checkpoint pathway in facilitating homologous recombination (HR) repair and suppressing extensive LOH and chromosomal rearrangements in response to a DSB. Accordingly, deletion of Rad3(ATR), Rad26ATRIP, Crb2(53BP1) or Cdc25 overexpression leads to reduced HR and increased break-induced chromosome loss and rearrangements. We find the DNA damage checkpoint pathway facilitates HR, in part, by promoting break-induced Cdt2-dependent nucleotide synthesis. We also identify additional roles for Rad17, the 9-1-1 complex and Chk1 activation in facilitating break-induced extensive resection and chromosome loss, thereby suppressing extensive LOH. Loss of Rad17 or the 9-1-1 complex results in a striking increase in break-induced isochromosome formation and very low levels of chromosome loss, suggesting the 9-1-1 complex acts as a nuclease processivity factor to facilitate extensive resection. Further, our data suggest redundant roles for Rad3ATR and Exo1 in facilitating extensive resection. We propose that the DNA damage checkpoint pathway coordinates resection and nucleotide synthesis, thereby promoting efficient HR repair and genome stability.


Asunto(s)
Roturas del ADN de Doble Cadena , División del ADN , Inestabilidad Genómica , Reparación del ADN por Recombinación , Schizosaccharomyces/genética , Puntos de Control del Ciclo Celular , Quinasa de Punto de Control 2/metabolismo , Cromosomas Fúngicos/genética , Hibridación Genómica Comparativa , Exodesoxirribonucleasas/metabolismo , Genoma Fúngico , Pérdida de Heterocigocidad , Nucleótidos/biosíntesis , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo
15.
Int J Radiat Oncol Biol Phys ; 119(5): 1517-1529, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38447610

RESUMEN

PURPOSE: This study investigated how isoform switching affects the cellular response to ionizing radiation (IR), an understudied area despite its relevance to radiation therapy in cancer treatment. We aimed to identify changes in transcript isoform expression post-IR exposure and the proteins mediating these changes, with a focus on their potential to modulate radiosensitivity. METHODS AND MATERIALS: Using RNA sequencing, we analyzed the B-cell lines derived from 10 healthy individuals at 3 timepoints, applying the mixture of isoforms algorithm to quantify alternative splicing. We examined RNA binding protein motifs within the sequences of IR-responsive isoforms and validated the serine/arginine-rich splicing factor 1 (SRSF1) as a predominant mediator through RNA immunoprecipitation. We further investigated the effects of SRSF1 on radiosensitivity by RNA interference and by analyzing publicly available data on patients with cancer. RESULTS: We identified ∼1900 radiation-responsive alternatively spliced isoforms. Many isoforms were differentially expressed without changes in their overall gene expression. Over a third of these transcripts underwent exon skipping, while others used proximal last exons. These IR-responsive isoforms tended to be shorter transcripts missing vital domains for preventing apoptosis and promoting cell division but retaining those necessary for DNA repair. Our combined computational, genetic, and molecular analyses identified the proto-oncogene SRSF1 as a mediator of these radiation-induced isoform-switching events that promote apoptosis. After exposure to DNA double-strand break-inducing agents, SRSF1 expression decreased. A reduction in SRSF1 increased radiosensitivity in vitro and among patients with cancer. CONCLUSIONS: We establish a pivotal role for isoform switching in the cellular response to IR and propose SRSF1 as a promising biomarker for assessing radiation therapy effectiveness.


Asunto(s)
Empalme Alternativo , Isoformas de Proteínas , Proto-Oncogenes Mas , Tolerancia a Radiación , Radiación Ionizante , Factores de Empalme Serina-Arginina , Factores de Empalme Serina-Arginina/metabolismo , Factores de Empalme Serina-Arginina/genética , Humanos , Tolerancia a Radiación/genética , Exones
16.
Semin Cell Dev Biol ; 22(8): 886-97, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22027614

RESUMEN

Chromosomal rearrangements, which can lead to oncogene activation and tumour suppressor loss, are a hallmark of cancer cells. Such outcomes can result from both the repair and misrepair of DNA ends, which arise from a variety of lesions including DNA double strand breaks (DSBs), collapsed replication forks and dysfunctional telomeres. Here we review the mechanisms by which non-homologous end joining (NHEJ) and homologous recombination (HR) repair pathways can both promote chromosomal rearrangements and also suppress them in response to such lesions, in accordance with their increasingly recognised tumour suppressor function. Further, we consider how chromosomal rearrangements, together with a modular approach towards understanding their etiology, may be exploited for cancer therapy.


Asunto(s)
Aberraciones Cromosómicas , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Neoplasias/genética , Humanos
17.
Trends Genet ; 26(5): 209-13, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20382440

RESUMEN

Centromeres are essential for chromosome segregation during both mitosis and meiosis. There are no obvious or conserved DNA sequence motif determinants for centromere function, but the complex centromeres found in the majority of eukaryotes studied to date consist of repetitive DNA sequences. A striking feature of these repeats is that they maintain a high level of inter-repeat sequence identity within the centromere. This observation is suggestive of a recombination mechanism that operates at centromeres. Here we postulate that inter-repeat homologous recombination plays an intrinsic role in centromere function by forming covalently closed DNA loops. Moreover, the model provides an explanation of why both inverted and direct repeats are maintained and how they contribute to centromere function.


Asunto(s)
Centrómero/fisiología , Recombinación Genética , Animales , Humanos , Meiosis , Levaduras/citología , Levaduras/genética , Levaduras/fisiología
18.
EMBO J ; 28(21): 3400-12, 2009 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-19798055

RESUMEN

Loss of heterozygosity (LOH), a causal event in cancer and human genetic diseases, frequently encompasses multiple genetic loci and whole chromosome arms. However, the mechanisms by which such extensive LOH arises, and how it is suppressed in normal cells is poorly understood. We have developed a genetic system to investigate the mechanisms of DNA double-strand break (DSB)-induced extensive LOH, and its suppression, using a non-essential minichromosome, Ch(16), in fission yeast. We find extensive LOH to arise from a new break-induced mechanism of isochromosome formation. Our data support a model in which Rqh1 and Exo1-dependent end processing from an unrepaired DSB leads to removal of the broken chromosome arm and to break-induced replication of the intact arm from the centromere, a considerable distance from the initial lesion. This process also promotes genome-wide copy number variation. A genetic screen revealed Rhp51, Rhp55, Rhp57 and the MRN complex to suppress both isochromosome formation and chromosome loss, in accordance with these events resulting from extensive end processing associated with failed homologous recombination repair.


Asunto(s)
Cromosomas Fúngicos/metabolismo , Roturas del ADN de Doble Cadena , Conversión Génica , Pérdida de Heterocigocidad , Schizosaccharomyces/genética , Adenosina Trifosfatasas/metabolismo , Centrómero/genética , Cromosomas Fúngicos/genética , Proteínas de Unión al ADN/metabolismo , Recombinasa Rad51/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo
19.
PLoS One ; 18(1): e0271016, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36626373

RESUMEN

We constructed a panel of S. pombe strains expressing DNA polymerase ε variants associated with cancer, specifically POLES297F, POLEV411L, POLEL424V, POLES459F, and used these to compare mutation rates determined by canavanine resistance with other selective methods. Canavanine-resistance mutation rates are broadly similar to those seen with reversion of the ade-485 mutation to adenine prototrophy, but lower than 5-fluoroorotic acid (FOA)-resistance rates (inactivation of ura4+ or ura5+ genes). Inactivation of several genes has been associated with canavanine resistance in S. pombe but surprisingly whole genome sequencing showed that 8/8 spontaneous canavanine-resistant mutants have an R175C mutation in the any1/arn1 gene. This gene encodes an α-arrestin-like protein involved in mediating Pub1 ubiquitylation of target proteins, and the phenotypic resistance to canavanine by this single mutation is similar to that shown by the original "can1-1" strain, which also has the any1R175C mutation. Some of the spontaneous mutants have additional mutations in arginine transporters, suggesting that this may marginally increase resistance to canavanine. The any1R175C strain showed internalisation of the Cat1 arginine transporter as previously reported, explaining the canavanine-resistance phenotype.


Asunto(s)
Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Canavanina/farmacología , Canavanina/metabolismo , Tasa de Mutación , Proteínas de Schizosaccharomyces pombe/metabolismo , Mutación , Arginina/metabolismo , Arrestinas/metabolismo
20.
bioRxiv ; 2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37131595

RESUMEN

Since genome instability can drive cancer initiation and progression, cells have evolved highly effective and ubiquitous DNA Damage Response (DDR) programs. However, some cells, in skin for example, are normally exposed to high levels of DNA damaging agents. Whether such high-risk cells possess lineage-specific mechanisms that tailor DNA repair to the tissue remains largely unknown. Here we show, using melanoma as a model, that the microphthalmia-associated transcription factor MITF, a lineage addition oncogene that coordinates many aspects of melanocyte and melanoma biology, plays a non-transcriptional role in shaping the DDR. On exposure to DNA damaging agents, MITF is phosphorylated by ATM/DNA-PKcs, and unexpectedly its interactome is dramatically remodelled; most transcription (co)factors dissociate, and instead MITF interacts with the MRE11-RAD50-NBS1 (MRN) complex. Consequently, cells with high MITF levels accumulate stalled replication forks, and display defects in homologous recombination-mediated repair associated with impaired MRN recruitment to DNA damage. In agreement, high MITF levels are associated with increased SNV burden in melanoma. Significantly, the SUMOylation-defective MITF-E318K melanoma predisposition mutation recapitulates the effects of ATM/DNA-PKcs-phosphorylated MITF. Our data suggest that a non-transcriptional function of a lineage-restricted transcription factor contributes to a tissue-specialised modulation of the DDR that can impact cancer initiation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA