Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 52(3): 1136-1155, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38038252

ABSTRACT

Maintaining chromatin integrity at the repetitive non-coding DNA sequences underlying centromeres is crucial to prevent replicative stress, DNA breaks and genomic instability. The concerted action of transcriptional repressors, chromatin remodelling complexes and epigenetic factors controls transcription and chromatin structure in these regions. The histone chaperone complex ATRX/DAXX is involved in the establishment and maintenance of centromeric chromatin through the deposition of the histone variant H3.3. ATRX and DAXX have also evolved mutually-independent functions in transcription and chromatin dynamics. Here, using paediatric glioma and pancreatic neuroendocrine tumor cell lines, we identify a novel ATRX-independent function for DAXX in promoting genome stability by preventing transcription-associated R-loop accumulation and DNA double-strand break formation at centromeres. This function of DAXX required its interaction with histone H3.3 but was independent of H3.3 deposition and did not reflect a role in the repression of centromeric transcription. DAXX depletion mobilized BRCA1 at centromeres, in line with BRCA1 role in counteracting centromeric R-loop accumulation. Our results provide novel insights into the mechanisms protecting the human genome from chromosomal instability, as well as potential perspectives in the treatment of cancers with DAXX alterations.


Subject(s)
Centromere , DNA Breaks, Double-Stranded , Molecular Chaperones , Nuclear Proteins , R-Loop Structures , X-linked Nuclear Protein , Child , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Centromere/metabolism , Chromatin , Co-Repressor Proteins/metabolism , DNA , Histones/genetics , Histones/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
2.
Cell Death Dis ; 12(10): 896, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599155

ABSTRACT

Centromeres are defined by chromatin containing the histone H3 variant CENP-A assembled onto repetitive α-satellite sequences, which are actively transcribed throughout the cell cycle. Centromeres play an essential role in chromosome inheritance and genome stability through coordinating kinetochores assembly during mitosis. Structural and functional alterations of the centromeres cause aneuploidy and chromosome aberrations which can induce cell death. In human cells, the tumor suppressor BRCA1 associates with centromeric chromatin in the absence of exogenous damage. While we previously reported that BRCA1 contributes to proper centromere homeostasis, the mechanism underlying its centromeric function and recruitment was not fully understood. Here, we show that BRCA1 association with centromeric chromatin depends on the presence of R-loops, which are non-canonical three-stranded structures harboring a DNA:RNA hybrid and are frequently formed during transcription. Subsequently, BRCA1 counteracts the accumulation of R-loops at centromeric α-satellite repeats. Strikingly, BRCA1-deficient cells show impaired localization of CENP-A, higher transcription of centromeric RNA, increased breakage at centromeres and formation of acentric micronuclei, all these features being R-loop-dependent. Finally, BRCA1 depletion reveals a Rad52-dependent hyper-recombination process between centromeric satellite repeats, associated with centromere instability and missegregation. Altogether, our findings provide molecular insights into the key function of BRCA1 in maintaining centromere stability and identity.


Subject(s)
BRCA1 Protein/metabolism , Centromere/metabolism , R-Loop Structures , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA, Satellite/genetics , Humans , Models, Biological , Rad52 DNA Repair and Recombination Protein/metabolism , Recombination, Genetic/genetics
3.
Mol Cancer Ther ; 16(10): 2166-2177, 2017 10.
Article in English | MEDLINE | ID: mdl-28611105

ABSTRACT

Poisons of topoisomerase II (TOP2) kill cancer cells by preventing religation of intermediate DNA breaks during the enzymatic process and thus by accumulating enzyme-drug-DNA complexes called TOP2 cleavage-complex (TOP2cc). F14512 is a highly cytotoxic polyamine-vectorized TOP2 inhibitor derived from etoposide and currently in clinical trials. It was shown in vitro that F14512 has acquired DNA-binding properties and that the stability of TOP2cc was strongly increased. Paradoxically, at equitoxic concentrations in cells, F14512 induced less DNA breaks than etoposide. Here, we directly compared etoposide and F14512 for their rates of TOP2cc production and resolution in human cells. We report that targeting of TOP2α and not TOP2ß impacts cell killing by F14512, contrary to etoposide that kills cells through targeting both isoforms. Then, we show that despite being more cytotoxic, F14512 is less efficient than etoposide at producing TOP2α cleavage-complex (TOP2αcc) in cells. Finally, we report that compared with TOP2αcc mediated by etoposide, those generated by F14512 persist longer in the genome, are not dependent on TDP2 for cleaning break ends from TOP2α, are channeled to a larger extent to resection-based repair processes relying on CtIP and BRCA1 and promote RAD51 recruitment to damaged chromatin. In addition to the addressing of F14512 to the polyamine transport system, the properties uncovered here would be particularly valuable for a therapeutic usage of this new anticancer compound. More generally, the concept of increasing drug cytotoxicity by switching the repair mode of the induced DNA lesions via addition of a DNA-binding moiety deserves further developments. Mol Cancer Ther; 16(10); 2166-77. ©2017 AACR.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA Topoisomerases, Type II/genetics , Neoplasms/drug therapy , Topoisomerase II Inhibitors/administration & dosage , Apoptosis/drug effects , BRCA1 Protein/genetics , Chromatin/genetics , Genetic Vectors/drug effects , Humans , Neoplasms/genetics , Neoplasms/pathology , Podophyllotoxin/administration & dosage , Podophyllotoxin/analogs & derivatives , Polyamines/administration & dosage , Rad51 Recombinase/genetics
4.
Oncotarget ; 7(3): 2269-83, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26745677

ABSTRACT

Accurate repair of DNA double-strand breaks (DSB) caused during DNA replication and by exogenous stresses is critical for the maintenance of genomic integrity. There is growing evidence that the Polo-like kinase 1 (Plk1) that plays a number of pivotal roles in cell proliferation can directly participate in regulation of DSB repair. In this study, we show that Plk1 regulates BRCA1, a key mediator protein required to efficiently repair DSB through homologous recombination (HR). Following induction of DSB, BRCA1 concentrates in distinctive large nuclear foci at damage sites where multiple DNA repair factors accumulate. First, we found that inhibition of Plk1 shortly before DNA damage sensitizes cells to ionizing radiation and reduces DSB repair by HR. Second, we provide evidence that BRCA1 foci formation induced by DSB is reduced when Plk1 is inhibited or depleted. Third, we identified BRCA1 as a novel Plk1 substrate and determined that Ser1164 is the major phosphorylation site for Plk1 in vitro. In cells, mutation of Plk1 sites on BRCA1 significantly delays BRCA1 foci formation following DSB, recapitulating the phenotype observed upon Plk1 inhibition. Our data then assign a key function to Plk1 in BRCA1 foci formation at DSB, emphasizing Plk1 importance in the HR repair of human cells.


Subject(s)
BRCA1 Protein/metabolism , Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA Replication/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation , DNA/genetics , DNA-Binding Proteins/genetics , HeLa Cells , Homologous Recombination/genetics , Humans , MCF-7 Cells , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Radiation, Ionizing , Polo-Like Kinase 1
5.
FASEB J ; 28(12): 5250-61, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25205741

ABSTRACT

In contrast to its well-known role in the DNA damage response during interphase, the function of BRCA1 in the maintenance of chromosomal stability during mitosis remains to be defined. In this study, we uncover a novel role of BRCA1 in preserving centromere integrity in mitotic human cells. Using immunofluorescence and chromatin immunoprecipitation approaches, we report BRCA1 association with centromeric chromatin during mitosis. BRCA1 depletion impairs centromeric cohesion, leading to an increase in interkinetochore distance and in unpaired sister-chromatids frequency during prometaphase. Moreover, BRCA1 loss partially decreased accumulation of the Aurora B kinase at the centromere. We found that proper recruitment of the DNMT3b DNA methyltransferase to satellite sequences is BRCA1-dependent during mitosis, suggesting that DNA hypomethylation contributes to Aurora B mislocalization. BRCA1-deficient cells exhibited decreased ability to correct improper Aurora B-dependent chromosome-spindle attachments and to align chromosomes at metaphase. Finally, we show that BRCA1 disruption promotes merotelic kinetochore attachments that represent a major mechanism of aneuploidy in human cells. In summary, we report here a novel function of BRCA1 in maintaining chromosomal stability through its contribution to the mitotic centromere integrity necessary for faithful segregation of sister-chromatids during cell division.


Subject(s)
Centromere , Chromosomal Instability , Genes, BRCA1 , Base Sequence , Cell Line, Tumor , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Humans , Microscopy, Fluorescence , RNA, Small Interfering , DNA Methyltransferase 3B
6.
Mol Biol Cell ; 22(7): 999-1013, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21289092

ABSTRACT

Although the anti-apoptotic activity of Bcl-2 has been extensively studied, its mode of action remains incompletely understood. Deciphering the network of Bcl-2 interacting factors is necessary to better understand the key function of Bcl-2 in apoptosis initiation. To identify novel Bcl-2 mitochondrial partners, we have combined a Bcl-2 immunocapture with a mass spectrometry analysis using highly pure mitochondrial fractions isolated from human cancer cells. We identified at high confidence 127 potential Bcl-2-interacting proteins. Gene ontology mining reveals enrichment for mitochondrial proteins, endoplasmic reticulum-associated proteins, and cytoskeleton-associated proteins. Importantly, we report the identification of galectin-7 (Gal7), a member of a family of ß-galactoside-binding lectins that was already known to exhibit a pro-apoptotic function, as a new mitochondrial Bcl-2 interacting partner. Our data further show that endogenous Bcl-2 coimmunoprecipitates with Gal7 and that recombinant Gal7 directly interacts with recombinant Bcl-2. A fraction of Gal7 is constitutively localized at mitochondria in a Bcl-2-dependent manner and sensitizes the mitochondria to the apoptotic signal. In addition, we show that the Bcl-2/Gal7 interaction is abolished following genotoxic stress. Taken together, our findings suggest that the binding of Gal7 to Bcl-2 may constitute a new target for enhancing the intrinsic apoptosis pathway.


Subject(s)
Apoptosis/physiology , Galectins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Cell Line , Galectins/genetics , Humans , Mitochondria/genetics , Mitochondria/radiation effects , Mitochondrial Proteins/genetics , Molecular Sequence Data , Protein Binding , Protein Interaction Maps , Proto-Oncogene Proteins c-bcl-2/genetics , Ultraviolet Rays
7.
Cell Biol Int ; 33(5): 572-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19269343

ABSTRACT

We previously demonstrated that phospho-Thr56 Bcl-2 colocalizes with Ki-67 and nucleolin in nuclear structures in prophase cells and is detected on mitotic chromosomes in later mitotic phases. To gain insight into the fine localization of Bcl-2 on mitotic chromosomes, we further investigated Bcl-2 localization by immunostaining of Bcl-2 with known components of metaphase chromosomes and electron microscopic immunocytochemistry. Immunofluorescence analysis on HeLa mitotic cells together with chromatin immunoprecipitation assays showed that Bcl-2 is associated with the condensed chromatin. Co-immunostaining experiments performed on mitotic chromosome spreads demonstrated that Bcl-2 is not localized on the longitudinal axis of chromatids with the condensin complex, but partially colocalizes with histone H3 on some regions of the mitotic chromosome. Finally, most of the Bcl-2 staining overlaps with Ki-67 staining at the chromosome periphery. Bcl-2 localization at the periphery and over the mitotic chromosome was confirmed by immunoelectron microscopy on mitotic cells. Our results indicate that Bcl-2 is an integral component of the mitotic chromosome.


Subject(s)
Chromosomes, Human/metabolism , Mitosis/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Human/ultrastructure , HeLa Cells , Histones/metabolism , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics
8.
Biochim Biophys Acta ; 1783(12): 2223-33, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18804494

ABSTRACT

Inherited mutations of the breast cancer susceptibility gene 1 (BRCA1) confer an increased risk for breast, ovarian and prostate cancer. BRCA1 has been involved in regulation of cell cycle progression, DNA damage signaling and repair, maintenance of genome integrity, ubiquitination and regulation of transcription. Aside from its essential functions in the DNA damage response BRCA1 has been also involved in the cellular response to microtubule damage. Emerging evidence indicates that BRCA1 regulates the duplication and the function of centrosomes, participates in mitotic spindle assembly and is required in the spindle checkpoint. Given BRCA1 distinct functions in microtubule-dependent pathways, we hypothesized that BRCA1 might be regulated following microtubule damage. In the present study, we report the novel finding that BRCA1 is phosphorylated by the checkpoint kinase Chk2 on the previously identified site Ser988 following anti-mitotic treatment in human cancer cells. Ser988-phosphorylated BRCA1 accumulates at centrosomes in response to microtubule damage but Ser988 is not essential for BRCA1 localization at the microtubule-organizing centers. We further demonstrate that the Ser988 phosphorylation is important for the inhibiting microtubule nucleation activity of BRCA1 and for BRCA1 function in cell survival following microtubule damage. These findings reveal a striking outcome of BRCA1 phosphorylation by Chk2 on its role in microtubule-dependent pathways and suggest a fine cross-talk between DNA damage and spindle damage responses.


Subject(s)
BRCA1 Protein/metabolism , DNA Damage , Microtubules/metabolism , Protein Serine-Threonine Kinases/physiology , Spindle Apparatus/metabolism , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/genetics , Blotting, Western , Cell Survival , Centrosome/metabolism , Checkpoint Kinase 2 , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Microtubules/pathology , Mitosis/drug effects , Mitosis/physiology , Mitotic Index , Mutation , Nocodazole/pharmacology , Paclitaxel/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Serine/chemistry , Serine/genetics , Serine/metabolism , Spindle Apparatus/drug effects , Tubulin Modulators/pharmacology , Tumor Cells, Cultured
9.
Exp Cell Res ; 313(17): 3800-8, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17826766

ABSTRACT

OPA1, an intra-mitochondrial dynamin GTPase, is a key actor of outer and inner mitochondrial membrane dynamic. OPA1 amino-terminal cleavage by PARL and m-AAA proteases was recently proposed to participate to the mitochondrial network dynamic in a DeltaPsi(m)-dependent way, and to apoptosis. Here, by an in vitro approach combining the use of purified mitochondrial fractions and mitochondrial targeting drugs, we intended to identify the central stimulus responsible for OPA1 cleavage. We confirm that apoptosis induction and PTPore opening, as well as DeltaPsi(m) dissipation induce OPA1 cleavage. Nevertheless, our experiments evidenced that decreased mitochondrial ATP levels, either generated by apoptosis induction, DeltaPsi(m) dissipation or inhibition of ATP synthase, is the common and crucial stimulus that controls OPA1 processing. In addition, we report that ectopic iron addition activates OPA1 cleavage, whereas zinc inhibits this process. These results suggest that the ATP-dependent OPA1 processing plays a central role in correlating the energetic metabolism to mitochondrial dynamic and might be involved in the pathophysiology of diseases associated to excess of iron or depletion of zinc and ATP.


Subject(s)
GTP Phosphohydrolases/metabolism , Iron/metabolism , Mitochondria/enzymology , Zinc/metabolism , Adenosine Triphosphate/analysis , Adenosine Triphosphate/deficiency , Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Cations, Divalent/analysis , Cations, Divalent/metabolism , HeLa Cells , Humans , Iron/analysis , Jurkat Cells , Mitochondria/chemistry , Reactive Oxygen Species/metabolism , Zinc/analysis
10.
Mutagenesis ; 20(2): 111-4, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15755800

ABSTRACT

Zeocin is a member of the bleomycin/phleomycin family of antibiotics, known to bind and cleave DNA. We established human SK-OV-3 cells that stably express the Zeocin resistance gene (Sh ble) using an ecdysone-inducible mammalian expression system. Surprisingly, our results demonstrated that Zeocin, added in the culture medium to maintain the expression of the ecdysone receptor, was responsible for the formation of DNA strand breaks in the recombinant cells. This suggests that the Zeocin is not completely detoxified and is still able to cleave DNA, despite the stable expression of the Sh ble gene in the recombinant clones. Our study indicates that one needs to be very cautious in the interpretation of data involving stable cell lines selected with Zeocin.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , DNA Damage , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Gene Expression , Humans , Receptors, Steroid/genetics
11.
Biol Cell ; 96(7): 545-52, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15380621

ABSTRACT

DNA interstrand cross-links (ICL)-inducing agents such as cisplatin, mitomycin C (MMC) and nitrogen mustards are widely used as potent antitumor drugs. Although ICL repair mechanism is not yet well characterized in mammalian cells, this pathway is thought to involve a sequential action of nucleotide excision repair (NER) and homologous recombination (HR). The importance of unraveling ICL repair pathways is highlighted by the hypersensitivity to ICL-inducing agents in cells of patients with the genetic disease Fanconi anemia (FA) and in cells mutated in the Breast Cancer susceptibility genes BRCA1 and BRCA2. To better characterize the involvement of HR in the sensitivity to ICL-inducing agents, we examined spontaneous and ICL-induced HR in rodent FA-like V-H4 cells. In this report, we show that MMC-hypersensitive V-H4 cells exhibit an increased spontaneous homology-directed repair (HDR) activity compared to the resistant V79 parental cells. Elevated HDR activity results mainly in increased conservative Rad51-dependent recombination, without affecting non-conservative single-strand annealing process (SSA). We also show that HDR activity is enhanced following MMC treatment in parental cells, but not in rodent FA-like V-H4 cells. Moreover, our data indicate that Rad51 foci formation is significantly delayed in these FA-like cells in response to crosslinking agent. These findings provide evidence for an impairment of HR control in V-H4 cells and emphasize the involvement of the FA pathway in HR-mediated repair.


Subject(s)
DNA Repair , Fanconi Anemia/genetics , Recombination, Genetic , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Line , Cricetinae , DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Mutation , Rad51 Recombinase
12.
Oncogene ; 21(33): 5188-92, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12140769

ABSTRACT

The BRCA2 tumor suppressor has been implicated in the maintenance of genomic integrity through a function in cellular responses to DNA damage. The BRCA2 protein directly associates with Rad51, that is essential for repair of double-strand breaks (DSBs) by homologous recombination (HR). In this report, we study the BRCA2-defective Chinese hamster cell mutant V-C8 for its ability to perform homology-directed repair (HDR) between repeated sequences. V-C8 cells were recently shown to be defective in Rad51 foci formation in response to DNA damage. Strikingly, we find that these BRCA2 mutant cells exhibit a strong stimulation of HDR activity compared to the V79 parental cells, which harbor a wild-type BRCA2. Furthermore, molecular characterization of the HDR products shows that loss of BRCA2 in V-C8 cells leads to significant reduction in Rad51-dependent gene conversion but strong enhancement of Rad51-independent single-strand annealing (SSA) events frequency. These data imply that, when HDR by conservative gene conversion is impaired, DSBs usually repaired by this pathway are instead resolved by other non-conservative HDR subpathways. Therefore, high chromosomal instability in BRCA2-deficient cells presumably results from enhancement of error-prone repair mechanisms, such as SSA.


Subject(s)
BRCA2 Protein/deficiency , BRCA2 Protein/metabolism , Recombination, Genetic/genetics , Animals , BRCA2 Protein/genetics , CHO Cells , Cloning, Molecular , Cricetinae , DNA Damage/genetics , DNA Repair/genetics , DNA-Binding Proteins/metabolism , Drug Resistance/genetics , Gene Conversion/genetics , Gentamicins/pharmacology , Mutation/genetics , Rad51 Recombinase
13.
J Biol Chem ; 277(7): 4764-9, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11713249

ABSTRACT

DNA cross-linking agents such as mitomycin C (MMC) and cisplatin are used as chemotherapeutic agents in cancer treatment. However, the molecular mechanism underlying their antitumor activity is not entirely clear. Critical steps in cytotoxicity toward cross-linking agents can involve DNA repair efficiency, inhibition of replication, cell-cycle checkpoints, regulation, and induction of apoptosis. The complexity of the mechanisms of the mammalian cell defense against cross-linking agents is reflected by the existence of many complementation groups identified in rodent cells that are specifically sensitive to MMC. We recently showed that increased induction of apoptosis contributes to the MMC sensitivity of the group represented by the V-H4 hamster mutant cell line. In this study, through the analyses of a substractive library, we discovered that sensitive V-H4 cells display a 40-fold increase of steady-state expression of metallothionein II (MT-II) mRNA compared with resistant parental V79 cells. Down-regulation of MT-II by antisense oligonucleotides partially restores MMC resistance in V-H4 cells, indicating that MT-II overexpression is directly involved in MMC hypersensitivity of these cells. MTs have been reported to regulate the activation of NF-kappaB, one of the key proteins that modulates the apoptotic response. Here we found that NF-kappaB activation by MMC is impaired in V-H4 cells and is partially restored following down-regulation of MT-II by antisense oligonucleotides. All these data suggest that the overexpression of MT-II in V-H4 cells impairs NF-kappaB activation by MMC, resulting in decreased cell survival and enhanced induction of apoptosis.


Subject(s)
Apoptosis , Cross-Linking Reagents/pharmacology , DNA/metabolism , Metallothionein/biosynthesis , Mitomycin/pharmacology , NF-kappa B/metabolism , Animals , Antineoplastic Agents/pharmacology , Blotting, Northern , Cadmium Chloride/pharmacology , Cell Line , Cisplatin/pharmacology , Cricetinae , DNA, Complementary/metabolism , Down-Regulation , Gene Library , Mutation , Nucleic Acid Hybridization , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...