Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 138
1.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article En | MEDLINE | ID: mdl-34815340

Common fragile sites (CFSs) are difficult-to-replicate genomic regions that form gaps and breaks on metaphase chromosomes under replication stress. They are hotspots for chromosomal instability in cancer. Repetitive sequences located at CFS loci are inefficiently copied by replicative DNA polymerase (Pol) delta. However, translesion synthesis Pol eta has been shown to efficiently polymerize CFS-associated repetitive sequences in vitro and facilitate CFS stability by a mechanism that is not fully understood. Here, by locus-specific, single-molecule replication analysis, we identified a crucial role for Pol eta (encoded by the gene POLH) in the in vivo replication of CFSs, even without exogenous stress. We find that Pol eta deficiency induces replication pausing, increases initiation events, and alters the direction of replication-fork progression at CFS-FRA16D in both lymphoblasts and fibroblasts. Furthermore, certain replication pause sites at CFS-FRA16D were associated with the presence of non-B DNA-forming motifs, implying that non-B DNA structures could increase replication hindrance in the absence of Pol eta. Further, in Pol eta-deficient fibroblasts, there was an increase in fork pausing at fibroblast-specific CFSs. Importantly, while not all pause sites were associated with non-B DNA structures, they were embedded within regions of increased genetic variation in the healthy human population, with mutational spectra consistent with Pol eta activity. From these findings, we propose that Pol eta replicating through CFSs may result in genetic variations found in the human population at these sites.


Chromosome Fragile Sites/genetics , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/physiology , Cell Line , Chromosome Fragility/genetics , Chromosome Fragility/physiology , DNA/genetics , DNA Damage/genetics , DNA Polymerase III/metabolism , DNA Repair/genetics , DNA Repair/physiology , DNA Replication/physiology , Genetic Variation/genetics , Genomic Instability/genetics , Humans , Proliferating Cell Nuclear Antigen/metabolism
2.
Cancer Lett ; 472: 1-7, 2020 03 01.
Article En | MEDLINE | ID: mdl-31830560

Fanconi anemia (FA) is a rare genome instability syndrome characterized by progressive bone marrow failure and predisposition to cancer, especially head and neck squamous cell carcinoma. Surgical resection is the standard of care for solid tumors, as patients with FA do not tolerate genotoxic chemotherapies or radiation, leading to poor prognosis. It is therefore imperative to develop chemoprevention strategies such as the identification of novel biomarkers to detect the formation of the tumor before its emergence and to use them in clinical trials aimed to counteract genome instability of patients with FA in tissues at risk. Micronuclei (MN) are chromosome fragments that are left behind in anaphase and appear in daughter cells as small additional nuclei. In this work, we analyzed MN frequencies in exfoliated buccal cells from 40 patients with FA and 24 controls. We found that MN frequency was significantly increased in the FA cohort indicating that we can detect chromosome fragility in patients with FA in basal conditions and in a tissue that is divided in vivo. Consequently, the MN assay in exfoliated buccal cells of patients with FA could be used in cancer risk studies and clinical trials aimed to identify cancer chemopreventive drugs.


Chromosome Fragility/genetics , Fanconi Anemia/genetics , Micronuclei, Chromosome-Defective , Squamous Cell Carcinoma of Head and Neck/genetics , Adolescent , Adult , Biomarkers, Tumor/genetics , Child , Child, Preschool , DNA Damage/genetics , Epithelium/metabolism , Epithelium/pathology , Fanconi Anemia/drug therapy , Fanconi Anemia/pathology , Female , Genomic Instability/genetics , Humans , Male , Micronucleus Tests/methods , Middle Aged , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Young Adult
3.
PLoS Genet ; 15(5): e1008169, 2019 05.
Article En | MEDLINE | ID: mdl-31100062

The Pol32 protein is one of the universal subunits of DNA polymerase δ (Pol δ), which is responsible for genome replication in eukaryotic cells. Although the role of Pol32 in DNA repair has been well-characterized, its exact function in genome replication remains obscure as studies in single cell systems have not established an essential role for Pol32 in the process. Here we characterize Pol32 in the context of Drosophila melanogaster development. In the rapidly dividing embryonic cells, loss of Pol32 halts genome replication as it specifically disrupts Pol δ localization to the nucleus. This function of Pol32 in facilitating the nuclear import of Pol δ would be similar to that of accessory subunits of DNA polymerases from mammalian Herpes viruses. In post-embryonic cells, loss of Pol32 reveals mitotic fragile sites in the Drosophila genome, a defect more consistent with Pol32's role as a polymerase processivity factor. Interestingly, these fragile sites do not favor repetitive sequences in heterochromatin, with the rDNA locus being a striking exception. Our study uncovers a possibly universal function for DNA polymerase ancillary factors and establishes a powerful system for the study of chromosomal fragile sites in a non-mammalian organism.


Chromosome Fragile Sites/physiology , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , Animals , Chromosome Fragile Sites/genetics , Chromosome Fragility/genetics , Chromosome Fragility/physiology , DNA Repair , DNA Replication/genetics , DNA Replication/physiology , DNA-Directed DNA Polymerase/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Fragile X Mental Retardation Protein/genetics , Mutagenesis , Nuclear Localization Signals/metabolism , Protein Binding
4.
J Invest Dermatol ; 139(3): 522-527, 2019 03.
Article En | MEDLINE | ID: mdl-30393082

The heritable forms of epidermolysis bullosa (EB), a phenotypically heterogeneous group of skin fragility disorders, is currently associated with mutations in as many as 21 distinct genes. EB is primarily a disorder affecting the epithelial layers of skin and mucous membranes, without extracutaneous manifestations, and thus is nonsyndromic. However, recent demonstrations of skin blistering in multisystem disorders with single gene defects highlight the concept of syndromic EB. Here, we review the phenotypic and genotypic features of syndromic forms of EB to delineate the concept of syndromic versus nonsyndromic skin fragility disorders.


Chromosome Fragility/genetics , Epidermolysis Bullosa/genetics , Genetic Predisposition to Disease/epidemiology , Mutation/genetics , Skin Diseases, Vesiculobullous/genetics , Epidermolysis Bullosa/epidemiology , Epidermolysis Bullosa/physiopathology , Female , Genotype , Humans , Male , Phenotype , Skin/pathology , Skin Diseases, Vesiculobullous/epidemiology , Skin Diseases, Vesiculobullous/physiopathology , Syndrome
5.
Bioessays ; 39(5)2017 05.
Article En | MEDLINE | ID: mdl-28345796

Improvements in deep sequencing, together with methods to rapidly deplete essential transcription factors (TFs) and chromatin remodelers, have recently led to a more detailed picture of promoter nucleosome architecture in yeast and its relationship to transcriptional regulation. These studies revealed that ∼40% of all budding yeast protein-coding genes possess a unique promoter structure, where we propose that an unusually unstable nucleosome forms immediately upstream of the transcription start site (TSS). This "fragile" nucleosome (FN) promoter architecture relies on the combined action of the essential RSC (Remodels Structure of Chromatin) nucleosome remodeler and pioneer transcription factors (PTFs). FNs are associated with genes whose expression is high, coupled to cell growth, and characterized by low cell-to-cell variability (noise), suggesting that they may promote these features. Recent studies in metazoans suggest that the presence of dynamic nucleosomes upstream of the TSS at highly expressed genes may be conserved throughout evolution.


Nucleosomes/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Animals , Chromatin Assembly and Disassembly/genetics , Chromosome Fragility/genetics , DNA-Binding Proteins/genetics , Genes, Fungal , Humans , Models, Genetic , Plants/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Schizosaccharomyces/genetics
6.
Adv Exp Med Biol ; 1042: 489-526, 2017.
Article En | MEDLINE | ID: mdl-29357071

Chromosome fragile sites are a fascinating cytogenetic phenomenon now widely implicated in a slew of human diseases ranging from neurological disorders to cancer. Yet, the paths leading to these revelations were far from direct, and the number of fragile sites that have been molecularly cloned with known disease-associated genes remains modest. Moreover, as more fragile sites were being discovered, research interests in some of the earliest discovered fragile sites ebbed away, leaving a number of unsolved mysteries in chromosome biology. In this review we attempt to recount some of the early discoveries of fragile sites and highlight those phenomena that have eluded intense scrutiny but remain extremely relevant in our understanding of the mechanisms of chromosome fragility. We then survey the literature for disease association for a comprehensive list of fragile sites. We also review recent studies addressing the underlying cause of chromosome fragility while highlighting some ongoing debates. We report an observed enrichment for R-loop forming sequences in fragile site-associated genes than genomic average. Finally, we will leave the reader with some lingering questions to provoke discussion and inspire further scientific inquiries.


Chromosome Fragile Sites/physiology , Chromosome Fragility/genetics , Animals , DNA Breaks, Double-Stranded , Genome/genetics , Genomic Instability/genetics , Humans , Neoplasms/genetics , Nervous System Diseases/genetics
7.
Cell Rep ; 16(5): 1300-1314, 2016 08 02.
Article En | MEDLINE | ID: mdl-27487043

The telomeric CTC1/STN1/TEN1 (CST) complex has been implicated in promoting replication recovery under replication stress at genomic regions, yet its precise role is unclear. Here, we report that STN1 is enriched at GC-rich repetitive sequences genome-wide in response to hydroxyurea (HU)-induced replication stress. STN1 deficiency exacerbates the fragility of these sequences under replication stress, resulting in chromosome fragmentation. We find that upon fork stalling, CST proteins form distinct nuclear foci that colocalize with RAD51. Furthermore, replication stress induces physical association of CST with RAD51 in an ATR-dependent manner. Strikingly, CST deficiency diminishes HU-induced RAD51 foci formation and reduces RAD51 recruitment to telomeres and non-telomeric GC-rich fragile sequences. Collectively, our findings establish that CST promotes RAD51 recruitment to GC-rich repetitive sequences in response to replication stress to facilitate replication restart, thereby providing insights into the mechanism underlying genome stability maintenance.


DNA Replication/genetics , GC Rich Sequence/genetics , Rad51 Recombinase/genetics , Repetitive Sequences, Nucleic Acid/genetics , Telomere-Binding Proteins/genetics , Cell Line, Tumor , Chromosome Fragility/genetics , DNA Fragmentation , Genome/genetics , Genomic Instability/genetics , HeLa Cells , Humans , Telomere/genetics , Telomere Homeostasis/genetics
8.
Mutat Res ; 781: 14-21, 2015 Nov.
Article En | MEDLINE | ID: mdl-26379101

Friedreich ataxia (FRDA) is a member of the Repeat Expansion Diseases, a group of genetic conditions resulting from an increase/expansion in the size of a specific tandem array. FRDA results from expansion of a GAA/TTC-tract in the first intron of the frataxin gene (FXN). The disease-associated tandem repeats all form secondary structures that are thought to contribute to the propensity of the repeat to expand. The subset of these diseases that result from a CGG/CCG-repeat expansion, such as Fragile X syndrome, also express a folate-sensitive fragile site coincident with the repeat on the affected chromosome. This chromosome fragility involves the generation of chromosome/chromatid gaps or breaks, or the high frequency loss of one or both copies of the affected gene when cells are grown under folate stress or as we showed previously, in the presence of an inhibitor of the ATM checkpoint kinase. Whether Repeat Expansion Disease loci containing different repeats form similar fragile sites was not known. We show here that the region of chromosome 9 that contains the FXN locus is intrinsically prone to breakage in vivo even in control cells. However, like FXS alleles, FRDA alleles show significantly elevated levels of chromosome abnormalities in the presence of an ATM inhibitor, consistent with the formation of a fragile site.


Chromosome Fragility/genetics , Chromosomes, Human, Pair 9/genetics , Friedreich Ataxia/genetics , Iron-Binding Proteins/genetics , Base Sequence , Cell Line , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Sequence Analysis, DNA , Trinucleotide Repeat Expansion , Frataxin
9.
Curr Opin Genet Dev ; 33: 32-40, 2015 Aug.
Article En | MEDLINE | ID: mdl-26254775

Fanconi anemia (FA) is characterized by bone marrow failure, malformations, and chromosome fragility. We review the recent discovery of FA genes and efforts to develop genetic therapies for FA in the last five years. Because current data exclude FANCM as an FA gene, 15 genes remain bona fide FA genes and three (FANCO, FANCR and FANCS) cause an FA like syndrome. Monoallelic mutations in 6 FA associated genes (FANCD1, FANCJ, FANCM, FANCN, FANCO and FANCS) predispose to breast and ovarian cancer. The products of all these genes are involved in the repair of stalled DNA replication forks by unhooking DNA interstrand cross-links and promoting homologous recombination. The genetic characterization of patients with FA is essential for developing therapies, including hematopoietic stem cell transplantation from a savior sibling donor after embryo selection, gene therapy, or genome editing using genetic recombination or engineered nucleases. Newly acquired knowledge about FA promises to provide therapeutic strategies in the near future.


DNA Helicases/genetics , Fanconi Anemia/genetics , Fanconi Anemia/therapy , Genetic Therapy , Bone Marrow/metabolism , Bone Marrow/pathology , Chromosome Fragility/genetics , Fanconi Anemia/pathology , Hematopoietic Stem Cell Transplantation , Humans , Mutation , RNA Editing/genetics
10.
Cell Cycle ; 14(15): 2501-8, 2015 Aug 03.
Article En | MEDLINE | ID: mdl-26030398

Histone Nuclear Factor P (HINFP) is essential for expression of histone H4 genes. Ablation of Hinfp and consequential depletion of histones alter nucleosome spacing and cause stalled replication and DNA damage that ultimately result in genomic instability. Faithful replication and packaging of newly replicated DNA are required for normal cell cycle control and proliferation. The tumor suppressor protein p53, the guardian of the genome, controls multiple cell cycle checkpoints and its loss leads to cellular transformation. Here we addressed whether the absence of p53 impacts the outcomes/consequences of Hinfp-mediated histone H4 deficiency. We examined mouse embryonic fibroblasts lacking both Hinfp and p53. Our data revealed that the reduced histone H4 expression caused by depletion of Hinfp persists when p53 is also inactivated. Loss of p53 enhanced the abnormalities in nuclear shape and size (i.e. multi-lobed irregularly shaped nuclei) caused by Hinfp depletion and also altered the sub-nuclear organization of Histone Locus Bodies (HLBs). In addition to the polyploid phenotype resulting from deletion of either p53 or Hinfp, inactivation of both p53 and Hinfp increased mitotic defects and generated chromosomal fragility and susceptibility to DNA damage. Thus, our study conclusively establishes that simultaneous loss of both Hinfp and the p53 checkpoint is detrimental to normal cell growth and may predispose to cellular transformation.


Cell Cycle Checkpoints/genetics , DNA Damage/genetics , Histones/biosynthesis , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Line , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Chromosome Fragility/genetics , DNA Replication/genetics , Fibroblasts/cytology , Genomic Instability/genetics , Histones/genetics , Mice , Mice, Knockout
11.
Nat Commun ; 6: 7094, 2015 May 11.
Article En | MEDLINE | ID: mdl-25959793

Recurrent genomic instability in cancer is attributed to positive selection and/or the sensitivity of specific genomic regions to breakage. Among these regions are fragile sites (FSs), genomic regions sensitive to replication stress conditions induced by the DNA polymerase inhibitor aphidicolin. However, the basis for the majority of cancer genomic instability hotspots remains unclear. Aberrant oncogene expression induces replication stress, leading to DNA breaks and genomic instability. Here we map the cytogenetic locations of oncogene-induced FSs and show that in the same cells, each oncogene creates a unique fragility landscape that only partially overlaps with aphidicolin-induced FSs. Oncogene-induced FSs colocalize with cancer breakpoints and large genes, similar to aphidicolin-induced FSs. The observed plasticity in the fragility landscape of the same cell type following oncogene expression highlights an additional level of complexity in the molecular basis for recurrent fragility in cancer.


Chromosome Fragile Sites/physiology , Chromosome Fragility/physiology , Fibroblasts/physiology , Genomic Instability , Oncogenes/physiology , Cell Line, Tumor , Chromosome Fragile Sites/genetics , Chromosome Fragility/genetics , Gene Deletion , Gene Expression Regulation/physiology , Humans , Multigene Family , Oncogenes/genetics , Plasmids
12.
Crit Rev Biochem Mol Biol ; 50(2): 142-67, 2015.
Article En | MEDLINE | ID: mdl-25608779

The expansion of repeated sequences is the cause of over 30 inherited genetic diseases, including Huntington disease, myotonic dystrophy (types 1 and 2), fragile X syndrome, many spinocerebellar ataxias, and some cases of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Repeat expansions are dynamic, and disease inheritance and progression are influenced by the size and the rate of expansion. Thus, an understanding of the various cellular mechanisms that cooperate to control or promote repeat expansions is of interest to human health. In addition, the study of repeat expansion and contraction mechanisms has provided insight into how repair pathways operate in the context of structure-forming DNA, as well as insights into non-canonical roles for repair proteins. Here we review the mechanisms of repeat instability, with a special emphasis on the knowledge gained from the various model systems that have been developed to study this topic. We cover the repair pathways and proteins that operate to maintain genome stability, or in some cases cause instability, and the cross-talk and interactions between them.


DNA Repair/genetics , DNA/genetics , Nucleic Acid Conformation , Trinucleotide Repeat Expansion/genetics , Chromosome Fragility/genetics , DNA/chemistry , DNA Damage , DNA Replication/genetics , Genetic Diseases, Inborn/classification , Genetic Diseases, Inborn/etiology , Genomic Instability , Humans , Recombination, Genetic
13.
Protoplasma ; 252(1): 301-6, 2015 Jan.
Article En | MEDLINE | ID: mdl-25056831

Phleum echinatum Host (2n = 2x = 10) is an annual Mediterranean species which differs from other representatives of the genus Phleum by reduced chromosome number, asymmetric karyotype and unusually high amount of DNA in the genome. Chromosomes of this plant were studied using conventional acetic-orcein staining and fluorescence in situ hybridization (FISH). FISH showed the major 35S ribosomal DNA (rDNA) site at the secondary constriction of satellite chromosome (3) and the minor 35S rDNA site near 5S rDNA cluster in the monobrachial chromosome 5. Telomeric repeats were detected at all chromosome ends within secondary constriction in satellited chromosome 3 and at the centromeric regions of chromosomes 1 and 2. Intrachromosomally located telomeric repeats are probably traces of chromosomal rearrangements that have shaped P.echinatum genome; they were prone to breakage which was manifested in chromosome fragmentation. The most distinct telomeric signals, suggesting massive amplification of interstitial telomeric sequences (ITRs), were observed at the nucleolar organizer region (NOR) of the third chromosome pair. Double FISH confirmed co-localization of telomeric and 35S rDNA repeats in this locus characterized by the biggest fragility in the karyotype. Fragile sites of P.echinatum, composed of amplified telomeric repeats, may bear a resemblance to metazoan rare fragile sites enriched in microsatellite repeats.


Chromosome Banding/methods , Chromosome Fragility/genetics , In Situ Hybridization, Fluorescence/methods , Karyotyping/methods , Phleum/chemistry
14.
BMC Med Genomics ; 7: 24, 2014 May 15.
Article En | MEDLINE | ID: mdl-24885126

BACKGROUND: Fanconi anemia (FA) is a rare inherited genetic syndrome with highly variable clinical manifestations. Fifteen genetic subtypes of FA have been identified. Traditional complementation tests for grouping studies have been used generally in FA patients and in stepwise methods to identify the FA type, which can result in incomplete genetic information from FA patients. METHODS: We diagnosed five pediatric patients with FA based on clinical manifestations, and we performed exome sequencing of peripheral blood specimens from these patients and their family members. The related sequencing data were then analyzed by bioinformatics, and the FANC gene mutations identified by exome sequencing were confirmed by PCR re-sequencing. RESULTS: Homozygous and compound heterozygous mutations of FANC genes were identified in all of the patients. The FA subtypes of the patients included FANCA, FANCM and FANCD2. Interestingly, four FA patients harbored multiple mutations in at least two FA genes, and some of these mutations have not been previously reported. These patients' clinical manifestations were vastly different from each other, as were their treatment responses to androstanazol and prednisone. This finding suggests that heterozygous mutation(s) in FA genes could also have diverse biological and/or pathophysiological effects on FA patients or FA gene carriers. Interestingly, we were not able to identify de novo mutations in the genes implicated in DNA repair pathways when the sequencing data of patients were compared with those of their parents. CONCLUSIONS: Our results indicate that Chinese FA patients and carriers might have higher and more complex mutation rates in FANC genes than have been conventionally recognized. Testing of the fifteen FANC genes in FA patients and their family members should be a regular clinical practice to determine the optimal care for the individual patient, to counsel the family and to obtain a better understanding of FA pathophysiology.


DNA Mutational Analysis/methods , Exome/genetics , Fanconi Anemia/genetics , Mutation/genetics , Bone Marrow/drug effects , Bone Marrow/pathology , Child , Child, Preschool , Chromosome Fragility/drug effects , Chromosome Fragility/genetics , Comet Assay , Fanconi Anemia/diagnosis , Female , Humans , Karyotyping , Male , Mitomycin/pharmacology , Reproducibility of Results , Software , Statistics as Topic
15.
Int J Radiat Biol ; 90(1): 53-9, 2014 Jan.
Article En | MEDLINE | ID: mdl-24164476

PURPOSE: To test the hypothesis that differences in DNA double-strand breaks (DSB) repair fidelity underlies differences in radiosensitivity. MATERIALS AND METHODS: A primary fibroblast culture (C42) derived from a pediatric cancer patient treated with reduced radiation doses consequent to a family history of radiosensitivity reminiscent of chromosomal fragility syndrome, was compared to a normal control (C29). DNA DSB rejoining and repair fidelity were studied by Southern blotting and hybridization to specific fragments: Alu repetitive sequence representing the overall DSB rejoining capacity in the genome and a 3.2 Mbp NotI restriction fragment on chromosome 21 for DSB repair fidelity. RESULTS: Although both assays showed statistically significant difference (p ≤ 0.05) between the two cell strains in residual misrepaired (un-or mis-rejoined) DSB (24 h after 30 or 80 Gy), the residual damage was lower in the Alu enriched genome assay compared to NotI assay (0.01-0.07 and 0.10-0.37, respectively). CONCLUSIONS: These results suggest that, in comparison to classic DSB repair experiment, an assay of measuring DNA DSB repair fidelity can provide better resolution and a more accurate estimate of misrepair of radiation-induced DNA damage, which underlies genomic instability and increased radiosensitivity.


Chromosome Disorders/genetics , Chromosome Fragility/genetics , Chromosome Fragility/radiation effects , DNA Damage/genetics , DNA Mismatch Repair/genetics , DNA Mismatch Repair/radiation effects , Radiation Tolerance/genetics , Base Pair Mismatch/genetics , Base Pair Mismatch/radiation effects , Biological Assay/methods , Child, Preschool , Female , Humans
16.
PLoS Genet ; 9(12): e1003979, 2013.
Article En | MEDLINE | ID: mdl-24339793

Inverted repeats capable of forming hairpin and cruciform structures present a threat to chromosomal integrity. They induce double strand breaks, which lead to gross chromosomal rearrangements, the hallmarks of cancers and hereditary diseases. Secondary structure formation at this motif has been proposed to be the driving force for the instability, albeit the mechanisms leading to the fragility are not well-understood. We carried out a genome-wide screen to uncover the genetic players that govern fragility of homologous and homeologous Alu quasi-palindromes in the yeast Saccharomyces cerevisiae. We found that depletion or lack of components of the DNA replication machinery, proteins involved in Fe-S cluster biogenesis, the replication-pausing checkpoint pathway, the telomere maintenance complex or the Sgs1-Top3-Rmi1 dissolvasome augment fragility at Alu-IRs. Rad51, a component of the homologous recombination pathway, was found to be required for replication arrest and breakage at the repeats specifically in replication-deficient strains. These data demonstrate that Rad51 is required for the formation of breakage-prone secondary structures in situations when replication is compromised while another mechanism operates in DSB formation in replication-proficient strains.


Chromosome Fragility/genetics , DNA Replication/genetics , Homologous Recombination/genetics , Inverted Repeat Sequences/genetics , Alu Elements/genetics , DNA Breaks, Double-Stranded , Genome, Fungal/genetics , Nucleic Acid Conformation , Nucleotide Motifs/genetics , Rad51 Recombinase/genetics , Saccharomyces cerevisiae/genetics
17.
J Cell Sci ; 126(Pt 11): 2502-15, 2013 Jun 01.
Article En | MEDLINE | ID: mdl-23549790

Transcription factor IIH (TFIIH) participates in transcription, nucleotide excision repair and the control of the cell cycle. In the present study, we demonstrate that the Dmp52 subunit of TFIIH in Drosophila physically interacts with the fly p53 homologue, Dp53. The depletion of Dmp52 in the wing disc generates chromosome fragility, increases apoptosis and produces wings with a reduced number of cells; cellular proliferation, however, is not affected. Interestingly, instead of suppressing the apoptotic phenotype, the depletion of Dp53 in Dmp52-depleted wing disc cells increases apoptosis and the number of cells that suffer from chromosome fragility. The apoptosis induced by the depletion of Dmp52 alone is partially dependent on the JNK pathway. In contrast, the enhanced apoptosis caused by the simultaneous depletion of Dp53 and Dmp52 is absolutely JNK-dependent. In this study, we also show that the anti-proliferative drug triptolide, which inhibits the ATPase activity of the XPB subunit of TFIIH, phenocopies the JNK-dependent massive apoptotic phenotype of Dp53-depleted wing disc cells; this observation suggests that the mechanism by which triptolide induces apoptosis in p53-deficient cancer cells involves the activation of the JNK death pathway.


Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Diterpenes/pharmacology , Drosophila Proteins/metabolism , MAP Kinase Kinase 4/metabolism , Phenanthrenes/pharmacology , Transcription Factor TFIIH/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Chromosome Fragility/drug effects , Chromosome Fragility/genetics , Chromosomes, Insect/genetics , Chromosomes, Insect/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Epoxy Compounds/pharmacology , MAP Kinase Kinase 4/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Transcription Factor TFIIH/genetics , Tumor Suppressor Protein p53/genetics
18.
FEBS Lett ; 587(7): 976-82, 2013 Apr 02.
Article En | MEDLINE | ID: mdl-23454638

The genomic architecture of several functional elements in animals and plants, such as microRNAs and tRNA, has been better characterized. As yet, there is very little known about genomic organization and structure of lncRNA in animals and plants. Here, we conducted a genome-wide systematic computational analysis of genomic architecture of lncRNAs, and further provided a more comprehensive comparative view of genomic organization between lncRNAs and several other functional elements in the human genome. Our study not only provides comprehensive knowledge for further studies into the correlations between the genomic architecture of lncRNAs and their important functional roles in diverse cellular processes and in disease, but also will be valuable for understanding the origin and evolution of lncRNAs.


Computational Biology/methods , Genome, Human/genetics , Genomics/methods , RNA, Long Noncoding/genetics , Algorithms , Chromosome Fragility/genetics , Chromosome Mapping , Evolution, Molecular , Humans , Repetitive Sequences, Nucleic Acid/genetics
19.
Otolaryngol Head Neck Surg ; 149(1): 105-11, 2013 Jul.
Article En | MEDLINE | ID: mdl-23535708

OBJECTIVE: To define the prognostic role of multiple epidemiological, clinical, and biological factors for the development of multiple malignancies (MM) in patients with head and neck cancer (HNC). STUDY DESIGN: Historical cohort study. p53 gene status, microsatellite instability (MSI) of the index tumor, and inherited chromosome fragility (CF) were studied. SETTING: Ninety-six consecutive patients affected by primary HNC, between January 1987 and October 1991, who were eligible for curative radiation therapy were followed up. SUBJECTS AND METHODS: p53 gene status, MSI, and CF in 96 curative radiotherapy-treated patients were correlated with the risk for MM. RESULTS: Multiple malignancies occurred in 28.9%. Microsatellite instability (P = 0.05), CF (P < 0.01), and smoking after treatment of the index tumor (P = 0.02) were correlated with an increased risk of MM. CONCLUSION: Genetic susceptibility may play a central role for MM development in patients with HNC.


Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Genetic Predisposition to Disease/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Neoplasms, Second Primary/etiology , Adult , Aged , Carcinoma, Squamous Cell/radiotherapy , Chromosome Fragility/genetics , Cohort Studies , Female , Genes, p53/genetics , Head and Neck Neoplasms/radiotherapy , Humans , Male , Microsatellite Instability , Middle Aged , Neoplasms, Second Primary/pathology , Neoplasms, Second Primary/therapy , Risk Factors
20.
Mol Cell ; 48(2): 254-65, 2012 Oct 26.
Article En | MEDLINE | ID: mdl-22959270

Triplex structure-forming GAA/TTC repeats pose a dual threat to the eukaryotic genome integrity. Their potential to expand can lead to gene inactivation, the cause of Friedreich's ataxia disease in humans. In model systems, long GAA/TTC tracts also act as chromosomal fragile sites that can trigger gross chromosomal rearrangements. The mechanisms that regulate the metabolism of GAA/TTC repeats are poorly understood. We have developed an experimental system in the yeast Saccharomyces cerevisiae that allows us to systematically identify genes crucial for maintaining the repeat stability. Two major groups of mutants defective in DNA replication or transcription initiation are found to be prone to fragility and large-scale expansions. We demonstrate that problems imposed by the repeats during DNA replication in actively dividing cells and during transcription initiation in nondividing cells can culminate in genome instability. We propose that similar mechanisms can mediate detrimental metabolism of GAA/TTC tracts in human cells.


Chromosome Fragility/genetics , Friedreich Ataxia/genetics , Saccharomyces cerevisiae/genetics , Trinucleotide Repeats/genetics , DNA Replication , Genome, Fungal , Genome, Human , Genomic Instability , Humans , Microsatellite Repeats , Mutation , Nucleic Acid Conformation
...