Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Nucleic Acids Res ; 49(20): 11728-11745, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34718776

ABSTRACT

Canonical non-homologous end-joining (cNHEJ) is the prominent mammalian DNA double-strand breaks (DSBs) repair pathway operative throughout the cell cycle. Phosphorylation of Ku70 at ser27-ser33 (pKu70) is induced by DNA DSBs and has been shown to regulate cNHEJ activity, but the underlying mechanism remained unknown. Here, we established that following DNA damage induction, Ku70 moves from nucleoli to the sites of damage, and once linked to DNA, it is phosphorylated. Notably, the novel emanating functions of pKu70 are evidenced through the recruitment of RNA Pol II and concomitant formation of phospho-53BP1 foci. Phosphorylation is also a prerequisite for the dynamic release of Ku70 from the repair complex through neddylation-dependent ubiquitylation. Although the non-phosphorylable ala-Ku70 form does not compromise the formation of the NHEJ core complex per se, cells expressing this form displayed constitutive and stress-inducible chromosomal instability. Consistently, upon targeted induction of DSBs by the I-SceI meganuclease into an intrachromosomal reporter substrate, cells expressing pKu70, rather than ala-Ku70, are protected against the joining of distal DNA ends. Collectively, our results underpin the essential role of pKu70 in the orchestration of DNA repair execution in living cells and substantiated the way it paves the maintenance of genome stability.


Subject(s)
DNA End-Joining Repair , Ku Autoantigen/metabolism , Cell Line , Cell Line, Tumor , DNA Damage , Humans , Phosphorylation , Protein Binding , RNA Polymerase II/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
2.
Mol Cell ; 48(5): 785-98, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23102699

ABSTRACT

Poly-(ADP-ribose) glycohydrolase (PARG) is a catabolic enzyme that cleaves ADP-ribose polymers synthesized by poly-(ADP-ribose) polymerases. Here, transcriptome profiling and differentiation assay revealed a requirement of PARG for retinoic acid receptor (RAR)-mediated transcription. Mechanistically, PARG accumulates early at promoters of RAR-responsive genes upon retinoic acid treatment to promote the formation of an appropriate chromatin environment suitable for transcription. Silencing of PARG or knockout of its enzymatic activity maintains the H3K9me2 mark at the promoter of the RAR-dependent genes, leading to the absence of preinitiation complex formation. In the absence of PARG, we found that the H3K9 demethylase KDM4D/JMJD2D became PARsylated. Mutation of two glutamic acids located in the Jumonji N domain of KDM4D inhibited PARsylation. PARG becomes dispensable for ligand-dependent transcription when either a PARP inhibitor or a non-PARsylable KDM4D/JMJD2D mutant is used. Our results define PARG as a coactivator regulating chromatin remodeling during RA-dependent gene expression.


Subject(s)
Glycoside Hydrolases/metabolism , Receptors, Retinoic Acid/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Chromatin Assembly and Disassembly , Enzyme Inhibitors/pharmacology , Gene Expression Profiling/methods , Gene Expression Regulation , Glutamic Acid , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/genetics , HeLa Cells , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Methylation , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , RNA Interference , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/metabolism , Retinoic Acid Receptor alpha , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Tretinoin/pharmacology
3.
Nucleic Acids Res ; 46(5): 2417-2431, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29361132

ABSTRACT

Poly(ADP-ribose) polymerases (PARPs) act as DNA break sensors and catalyze the synthesis of polymers of ADP-ribose (PAR) covalently attached to acceptor proteins at DNA damage sites. It has been demonstrated that both mammalian PARP1 and PARP2 PARylate double-strand break termini in DNA oligonucleotide duplexes in vitro. Here, we show that mammalian PARP2 and PARP3 can PARylate and mono(ADP-ribosyl)ate (MARylate), respectively, 5'- and 3'-terminal phosphate residues at double- and single-strand break termini of a DNA molecule containing multiple strand breaks. PARP3-catalyzed DNA MARylation can be considered a new type of reversible post-replicative DNA modification. According to DNA substrate specificity of PARP3 and PARP2, we propose a putative mechanistic model of PARP-catalyzed strand break-oriented ADP-ribosylation of DNA termini. Notably, PARP-mediated DNA ADP-ribosylation can be more effective than PARPs' auto-ADP-ribosylation depending on the DNA substrates and reaction conditions used. Finally, we show an effective PARP3- or PARP2-catalyzed ADP-ribosylation of high-molecular-weight (∼3-kb) DNA molecules, PARP-mediated DNA PARylation in cell-free extracts and a persisting signal of anti-PAR antibodies in a serially purified genomic DNA from bleomycin-treated poly(ADP-ribose) glycohydrolase-depleted HeLa cells. These results suggest that certain types of complex DNA breaks can be effectively ADP-ribosylated by PARPs in cellular response to DNA damage.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks , DNA/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Adenosine Diphosphate Ribose/metabolism , DNA/chemistry , DNA Adducts/metabolism , DNA Breaks, Double-Stranded , HeLa Cells , Humans , Phosphates/metabolism , Substrate Specificity
4.
J Cell Sci ; 129(16): 3167-77, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27383768

ABSTRACT

Cytidine deaminase (CDA) deficiency induces an excess of cellular dCTP, which reduces basal PARP-1 activity, thereby compromising complete DNA replication, leading to ultrafine anaphase bridge (UFB) formation. CDA dysfunction has pathological implications, notably in cancer and in Bloom syndrome. It remains unknown how reduced levels of PARP-1 activity and pyrimidine pool imbalance lead to the accumulation of unreplicated DNA during mitosis. We report that a decrease in PARP-1 activity in CDA-deficient cells impairs DNA-damage-induced Chk1 activation, and, thus, the downstream checkpoints. Chemical inhibition of the ATR-Chk1 pathway leads to UFB accumulation, and we found that this pathway was compromised in CDA-deficient cells. Our data demonstrate that ATR-Chk1 acts downstream from PARP-1, preventing the accumulation of unreplicated DNA in mitosis, and, thus, UFB formation. Finally, delaying entry into mitosis is sufficient to prevent UFB formation in both CDA-deficient and CDA-proficient cells, suggesting that both physiological and pathological UFBs are derived from unreplicated DNA. Our findings demonstrate an unsuspected requirement for a balanced nucleotide pool for optimal Chk1 activation both in unchallenged cells and in response to genotoxic stress.


Subject(s)
Anaphase , Checkpoint Kinase 1/metabolism , Pyrimidines/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Checkpoints , Cytidine Deaminase/metabolism , DNA Damage , DNA Replication , Enzyme Activation , HeLa Cells , Humans , Models, Biological , Poly(ADP-ribose) Polymerases/metabolism , S Phase
5.
Biochem Biophys Res Commun ; 498(4): 715-722, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29550484

ABSTRACT

Manganese Superoxide dismutase 2 (SOD2) plays a crucial role in antioxidant defense but there are no data suggesting its role in genetic instability in CML. We evaluated the effects of SOD2 silencing in human UT7 cell line expressing either non-mutated or T315I-mutated BCR-ABL. Array-CGH experiments detected in BCR-ABL-expressing cells silenced for SOD2 a major genetic instability within several chromosomal loci, especially in regions carrying the glypican family (duplicated) and ß-defensin genes (deleted). In a large cohort of patients with chronic myeloid leukemia (CML), a significant decrease of SOD2 mRNA was observed. This reduction appeared inversely correlated with leukocytosis and Sokal score, high-risk patients showing lower SOD2 levels. The analysis of anti-oxidant gene expression analysis revealed a specific down-regulation of the expression of PRDX2 in UT7-BCR-ABL and UT7-T315I cells silenced for SOD2 expression. Gene set enrichment analysis performed between the two SOD2-dependent classes of CML patients revealed a significant enrichment of Reactive Oxygen Species (ROS) Pathway. Our data provide the first evidence for a link between SOD2 expression and genetic instability in CML. Consequently, SOD2 mRNA levels should be analyzed in prospective studies as patients with low SOD2 expression could be more prone to develop a mutator phenotype under TKI therapies.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Superoxide Dismutase/genetics , Cell Line, Tumor , Cohort Studies , Gene Silencing , Humans , Mutation , Peroxiredoxins/genetics , Point Mutation
6.
Mol Cell ; 38(1): 54-66, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20385089

ABSTRACT

Upon gene activation, we found that RNA polymerase II transcription machinery assembles sequentially with the nucleotide excision repair (NER) factors at the promoter. This recruitment occurs in absence of exogenous genotoxic attack, is sensitive to transcription inhibitors, and depends on the XPC protein. The presence of these repair proteins at the promoter of activated genes is necessary in order to achieve optimal DNA demethylation and histone posttranslational modifications (H3K4/H3K9 methylation, H3K9/14 acetylation) and thus efficient RNA synthesis. Deficiencies in some NER factors impede the recruitment of others and affect nuclear receptor transactivation. Our data suggest that there is a functional difference between the presence of the NER factors at the promoters (which requires XPC) and the NER factors at the distal regions of the gene (which requires CSB). While the latter may be a repair function, the former is a function with respect to transcription unveiled in the current study.


Subject(s)
Chromatin/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Mutagens/metabolism , Promoter Regions, Genetic , Transcription, Genetic , Chromatin Assembly and Disassembly , DNA Damage , DNA Methylation , DNA-Binding Proteins/genetics , HeLa Cells , Histones/metabolism , Humans , Models, Molecular , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism
7.
Nucleic Acids Res ; 44(12): 5717-31, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27095204

ABSTRACT

Ultraviolet-induced 6-4 photoproducts (6-4PP) and cyclobutane pyrimidine dimers (CPD) can be tolerated by translesion DNA polymerases (TLS Pols) at stalled replication forks or by gap-filling. Here, we investigated the involvement of Polη, Rev1 and Rev3L (Polζ catalytic subunit) in the specific bypass of 6-4PP and CPD in repair-deficient XP-C human cells. We combined DNA fiber assay and novel methodologies for detection and quantification of single-stranded DNA (ssDNA) gaps on ongoing replication forks and postreplication repair (PRR) tracts in the human genome. We demonstrated that Rev3L, but not Rev1, is required for postreplicative gap-filling, while Polη and Rev1 are responsible for TLS at stalled replication forks. Moreover, specific photolyases were employed to show that in XP-C cells, CPD arrest replication forks, while 6-4PP are responsible for the generation of ssDNA gaps and PRR tracts. On the other hand, in the absence of Polη or Rev1, both types of lesion block replication forks progression. Altogether, the data directly show that, in the human genome, Polη and Rev1 bypass CPD and 6-4PP at replication forks, while only 6-4PP are also tolerated by a Polζ-dependent gap-filling mechanism, independent of S phase.


Subject(s)
DNA Repair , DNA Replication , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Nuclear Proteins/genetics , Nucleotidyltransferases/genetics , Adenoviridae/genetics , Adenoviridae/metabolism , Cell Line, Transformed , DNA Damage , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Deoxyribodipyrimidine Photo-Lyase , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Genome, Human , Humans , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Pyrimidine Dimers/metabolism , S Phase/genetics , Transduction, Genetic , Ultraviolet Rays
8.
Proc Natl Acad Sci U S A ; 112(22): E2910-9, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-26038566

ABSTRACT

UV-sensitive syndrome (UV(S)S) and Cockayne syndrome (CS) are human disorders caused by CSA or CSB gene mutations; both conditions cause defective transcription-coupled repair and photosensitivity. Patients with CS also display neurological and developmental abnormalities and dramatic premature aging, and their cells are hypersensitive to oxidative stress. We report CSA/CSB-dependent depletion of the mitochondrial DNA polymerase-γ catalytic subunit (POLG1), due to HTRA3 serine protease accumulation in CS, but not in UV(s)S or control fibroblasts. Inhibition of serine proteases restored physiological POLG1 levels in either CS fibroblasts and in CSB-silenced cells. Moreover, patient-derived CS cells displayed greater nitroso-redox imbalance than UV(S)S cells. Scavengers of reactive oxygen species and peroxynitrite normalized HTRA3 and POLG1 levels in CS cells, and notably, increased mitochondrial oxidative phosphorylation, which was altered in CS cells. These data reveal critical deregulation of proteases potentially linked to progeroid phenotypes in CS, and our results suggest rescue strategies as a therapeutic option.


Subject(s)
Cockayne Syndrome/drug therapy , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Mitochondrial Diseases/drug therapy , Progeria/pathology , Serine Proteinase Inhibitors/pharmacology , Blotting, Western , Cells, Cultured , Cockayne Syndrome/pathology , DNA Polymerase gamma , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/metabolism , Fluorescent Antibody Technique , HeLa Cells , Humans , Mitochondrial Diseases/pathology , Peroxynitrous Acid/metabolism , Poly-ADP-Ribose Binding Proteins , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Serine Proteinase Inhibitors/metabolism
9.
PLoS Genet ; 11(7): e1005384, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26181065

ABSTRACT

Genome stability is jeopardized by imbalances of the dNTP pool; such imbalances affect the rate of fork progression. For example, cytidine deaminase (CDA) deficiency leads to an excess of dCTP, slowing the replication fork. We describe here a novel mechanism by which pyrimidine pool disequilibrium compromises the completion of replication and chromosome segregation: the intracellular accumulation of dCTP inhibits PARP-1 activity. CDA deficiency results in incomplete DNA replication when cells enter mitosis, leading to the formation of ultrafine anaphase bridges between sister-chromatids at "difficult-to-replicate" sites such as centromeres and fragile sites. Using molecular combing, electron microscopy and a sensitive assay involving cell imaging to quantify steady-state PAR levels, we found that DNA replication was unsuccessful due to the partial inhibition of basal PARP-1 activity, rather than slower fork speed. The stimulation of PARP-1 activity in CDA-deficient cells restores replication and, thus, chromosome segregation. Moreover, increasing intracellular dCTP levels generates under-replication-induced sister-chromatid bridges as efficiently as PARP-1 knockdown. These results have direct implications for Bloom syndrome (BS), a rare genetic disease combining susceptibility to cancer and genomic instability. BS results from mutation of the BLM gene, encoding BLM, a RecQ 3'-5' DNA helicase, a deficiency of which leads to CDA downregulation. BS cells thus have a CDA defect, resulting in a high frequency of ultrafine anaphase bridges due entirely to dCTP-dependent PARP-1 inhibition and independent of BLM status. Our study describes previously unknown pathological consequences of the distortion of dNTP pools and reveals an unexpected role for PARP-1 in preventing DNA under-replication and chromosome segregation defects.


Subject(s)
Bloom Syndrome/genetics , Cytidine Deaminase/genetics , Poly(ADP-ribose) Polymerases/genetics , Pyrimidines/metabolism , Bloom Syndrome/pathology , Cell Line , Centromere/genetics , Chromosome Fragile Sites/genetics , Chromosome Segregation/genetics , Cytidine Deaminase/deficiency , DNA Replication/genetics , Genomic Instability , Humans , Mitosis/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/biosynthesis , RecQ Helicases/genetics , Sister Chromatid Exchange/genetics
10.
Hum Mol Genet ; 23(16): 4232-48, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24694934

ABSTRACT

The MRE11/RAD50/NBN (MRN) complex plays a key role in detecting DNA double-strand breaks, recruiting and activating ataxia-telangiectasia mutated and in processing the breaks. Members of this complex also act as adaptor molecules for downstream signaling to the cell cycle and other cellular processes. Somewhat more controversial are the results to support a role for MRN in the ataxia-telangiectasia and Rad3-related (ATR) activation and signaling. We provide evidence that RAD50 is required for ATR activation in mammalian cells in response to DNA replication stress. It is in turn phosphorylated at a specific site (S635) by ATR, which is required for ATR signaling through Chk1 and other downstream substrates. We find that RAD50 phosphorylation is essential for DNA replication restart by promoting loading of cohesin at these sites. We also demonstrate that replication stress-induced RAD50 phosphorylation is functionally significant for cell survival and cell cycle checkpoint activation. These results highlight the importance of the adaptor role for a member of the MRN complex in all aspects of the response to DNA replication stress.


Subject(s)
Ataxia Telangiectasia/pathology , DNA Breaks, Double-Stranded , DNA Repair Enzymes/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Signal Transduction , Acid Anhydride Hydrolases , Cell Line , Checkpoint Kinase 1 , Fibroblasts , Humans , Phosphorylation , Protein Kinases/metabolism , Serine/metabolism , Stress, Physiological
11.
Mol Carcinog ; 55(3): 245-54, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25641732

ABSTRACT

Along with CD44, CD24 is a key marker of breast cancer stem cells (CSCs), frequently defined by CD24(-)/CD44(+) labeling. Among all phenotypes classically attributed to breast CD24(-)/CD44(+) cancer cells, radiation resistance has been extensively described and seen as being implicated in radiotherapy failure. Our previous data indicated that CD24(-) cells constitute a radiation-resistant subpopulation transitory selected by high doses of ionizing radiation. However, little is known about the biological role of CD24 in breast cancers, and no function has been assigned to CD24 in radiation response. Here, CD24 expression was induced in CD24(-) cells or knocked-down in CD24(+) cells. We show that forced extinction of CD24 expression is associated with decreased proliferation rate, lower levels of reactive oxygen species (ROS) and decreased genomic instability. On the opposite when CD24 is artificially expressed in CD24(-) cells, proliferation rates in vitro and in vivo, ROS levels and genomic instability are enhanced. Moreover, we observe that loss of CD24 expression leads to radiation resistance, by preventing radiation-induced cell death and promoting generation of progeny in relation to lower G2/M blockade and a smaller proportion of polyploid cells. Finally, control of ROS levels appears to be the key event in the CD24-mediated radiation response. For the first time, CD24 is proposed as a direct actor in radiation response of breast cancer cells, independently of CD44 expression. These findings could have interesting applications in evaluating the intrinsic radiation response of primary tumors.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Breast/radiation effects , CD24 Antigen/genetics , Oxidative Stress , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD24 Antigen/analysis , Cell Line, Tumor , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Hyaluronan Receptors/analysis , Hyaluronan Receptors/genetics , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects
12.
Cell Mol Life Sci ; 72(8): 1585-97, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25336152

ABSTRACT

Poly(ADP-ribose) polymerase-2 (PARP-2) activity contributes to a cells' poly(ADP-ribosyl)ating potential and like PARP-1, has been implicated in several DNA repair pathways including base excision repair and DNA single strand break repair. Here the consequences of its stable depletion in HeLa, U20S, and AS3WT2 cells were examined. All three PARP-2 depleted models showed increased sensitivity to the cell killing effects on ionizing radiation as reported in PARP-2 depleted mouse embryonic fibroblasts providing further evidence for a role in DNA strand break repair. The PARP-2 depleted HeLa cells also showed both higher constitutive and DNA damage-induced levels of polymers of ADP-ribose (PAR) associated with unchanged PARP-1 protein levels, but higher PARP activity and a concomitant lower PARG protein levels and activity. These changes were accompanied by a reduced maximal recruitment of PARP-1, XRCC1, PCNA, and PARG to DNA damage sites. This PAR-associated phenotype could be reversed in HeLa cells on re-expression of PARP-2 and was not seen in U20S and AS3WT2 cells. These results highlight the complexity of the relationship between different members of the PARP family on PAR metabolism and suggest that cell model dependent phenotypes associated with the absence of PARP-2 exist within a common background of radiation sensitivity.


Subject(s)
Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , Cell Line , Cell Survival/radiation effects , DNA Damage/radiation effects , DNA Repair , DNA-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , HeLa Cells , Humans , Mice , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Proliferating Cell Nuclear Antigen/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Radiation, Ionizing , X-ray Repair Cross Complementing Protein 1
13.
Nucleic Acids Res ; 42(7): 4435-49, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24493735

ABSTRACT

Poly(ADP-ribose) polymerases (PARP) attach poly(ADP-ribose) (PAR) chains to various proteins including themselves and chromatin. Topoisomerase I (Top1) regulates DNA supercoiling and is the target of camptothecin and indenoisoquinoline anticancer drugs, as it forms Top1 cleavage complexes (Top1cc) that are trapped by the drugs. Endogenous and carcinogenic DNA lesions can also trap Top1cc. Tyrosyl-DNA phosphodiesterase 1 (TDP1), a key repair enzyme for trapped Top1cc, hydrolyzes the phosphodiester bond between the DNA 3'-end and the Top1 tyrosyl moiety. Alternative repair pathways for Top1cc involve endonuclease cleavage. However, it is unknown what determines the choice between TDP1 and the endonuclease repair pathways. Here we show that PARP1 plays a critical role in this process. By generating TDP1 and PARP1 double-knockout lymphoma chicken DT40 cells, we demonstrate that TDP1 and PARP1 are epistatic for the repair of Top1cc. The N-terminal domain of TDP1 directly binds the C-terminal domain of PARP1, and TDP1 is PARylated by PARP1. PARylation stabilizes TDP1 together with SUMOylation of TDP1. TDP1 PARylation enhances its recruitment to DNA damage sites without interfering with TDP1 catalytic activity. TDP1-PARP1 complexes, in turn recruit X-ray repair cross-complementing protein 1 (XRCC1). This work identifies PARP1 as a key component driving the repair of trapped Top1cc by TDP1.


Subject(s)
DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Phosphoric Diester Hydrolases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , Epistasis, Genetic , Humans , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Protein Interaction Domains and Motifs , Sumoylation , X-ray Repair Cross Complementing Protein 1
14.
Gastroenterology ; 146(2): 401-11.e1, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24512910

ABSTRACT

BACKGROUND & AIMS: Patients with colorectal tumors with microsatellite instability (MSI) have better prognoses than patients with tumors without MSI, but have a poor response to 5-fluorouracil­based chemotherapy. A dominant-negative form of heat shock protein (HSP)110 (HSP110DE9) expressed by cancer cells with MSI, via exon skipping caused by somatic deletions in the T(17) intron repeat, sensitizes the cells to 5-fluorouracil and oxaliplatin.We investigated whether HSP110 T(17) could be used to identify patients with colorectal cancer who would benefit from adjuvant chemotherapy with 5-fluorouracil and oxaliplatin. METHODS: We characterized the interaction between HSP110 and HSP110DE9 using surface plasmon resonance. By using polymerase chain reaction and fragment analysis, we examined how the size of somatic allelic deletions in HSP110 T(17) affected the HSP110 protein expressed by tumor cells. We screened 329 consecutive patients with stage II­III colorectal tumors with MSI who underwent surgical resection at tertiary medical centers for HSP110 T(17). RESULTS: HSP110 and HSP110DE9 interacted in a1:1 ratio. Tumor cells with large deletions in T(17) had increased ratios of HSP110DE9:HSP110, owing to the loss of expression of full-length HSP110. Deletions in HSP110 T(17) were mostly biallelic in primary tumor samples with MSI. Patients with stage II­III cancer who received chemotherapy and had large HSP110 T(17) deletions (≥5 bp; 18 of 77 patients, 23.4%) had longer times of relapse-free survival than patients with small or no deletions (≤4 bp; 59 of 77 patients, 76.6%) in multivariate analysis (hazard ratio, 0.16; 95% confidence interval, 0.012­0.8; P = .03). We found a significant interaction between chemotherapy and T17 deletion (P =.009). CONCLUSIONS: About 25% of patients with stages II­III colorectal tumors with MSI have an excellent response to chemotherapy, due to large, biallelic deletions in the T(17) intron repeat of HSP110 in tumor DNA.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Base Sequence , Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , HSP110 Heat-Shock Proteins/genetics , Microsatellite Instability , Sequence Deletion , Aged , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Line, Tumor , Chemotherapy, Adjuvant , Colectomy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/surgery , Female , Fluorouracil/administration & dosage , Follow-Up Studies , HSP110 Heat-Shock Proteins/chemistry , HSP110 Heat-Shock Proteins/metabolism , Humans , Introns , Leucovorin/administration & dosage , Male , Models, Molecular , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Retrospective Studies , Surface Plasmon Resonance , Survival Analysis , Treatment Outcome
15.
Proc Natl Acad Sci U S A ; 108(7): 2783-8, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21270334

ABSTRACT

The ADP ribosyl transferase [poly(ADP-ribose) polymerase] ARTD3(PARP3) is a newly characterized member of the ARTD(PARP) family that catalyzes the reaction of ADP ribosylation, a key posttranslational modification of proteins involved in different signaling pathways from DNA damage to energy metabolism and organismal memory. This enzyme shares high structural similarities with the DNA repair enzymes PARP1 and PARP2 and accordingly has been found to catalyse poly(ADP ribose) synthesis. However, relatively little is known about its in vivo cellular properties. By combining biochemical studies with the generation and characterization of loss-of-function human and mouse models, we describe PARP3 as a newcomer in genome integrity and mitotic progression. We report a particular role of PARP3 in cellular response to double-strand breaks, most likely in concert with PARP1. We identify PARP3 as a critical player in the stabilization of the mitotic spindle and in telomere integrity notably by associating and regulating the mitotic components NuMA and tankyrase 1. Both functions open stimulating prospects for specifically targeting PARP3 in cancer therapy.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , Genomic Instability/genetics , Mitosis/genetics , Poly(ADP-ribose) Polymerases/metabolism , Spindle Apparatus/physiology , Adenosine Diphosphate/metabolism , Animals , Antigens, Nuclear/metabolism , Blotting, Western , Cell Line, Tumor , Colony-Forming Units Assay , Comet Assay , DNA Primers/genetics , Fluorescent Antibody Technique, Indirect , Genomic Instability/physiology , Humans , Immunoprecipitation , In Situ Hybridization, Fluorescence , Mass Spectrometry , Mice , Mice, Knockout , Microscopy, Video , Mitosis/physiology , Nuclear Matrix-Associated Proteins/metabolism , Poly(ADP-ribose) Polymerases/deficiency , Tankyrases/metabolism
16.
Photochem Photobiol ; 100(1): 4-18, 2024.
Article in English | MEDLINE | ID: mdl-37926965

ABSTRACT

Xeroderma pigmentosum (XP) variant cells are deficient in the translesion synthesis (TLS) DNA polymerase Polη (eta). This protein contributes to DNA damage tolerance, bypassing unrepaired UV photoproducts and allowing S-phase progression with minimal delay. In the absence of Polη, backup polymerases perform TLS of UV lesions. However, which polymerase plays this role in human cells remains an open question. Here, we investigated the potential role of Polι (iota) in bypassing ultraviolet (UV) induced photoproducts in the absence of Polη, using NER-deficient (XP-C) cells knocked down for Polι and/or Polη genes. Our results indicate that cells lacking either Polι or Polη have increased sensitivity to UVC radiation. The lack of both TLS polymerases led to increased cell death and defects in proliferation and migration. Loss of both polymerases induces a significant replication fork arrest and G1/S-phase blockage, compared to the lack of Polη alone. In conclusion, we propose that Polι acts as a bona fide backup for Polη in the TLS of UV-photoproducts.


Subject(s)
DNA Polymerase iota , Xeroderma Pigmentosum , Humans , DNA Damage , Translesion DNA Synthesis , DNA Replication , Xeroderma Pigmentosum/genetics , Ultraviolet Rays , DNA Repair
17.
J Cell Sci ; 124(Pt 11): 1943-51, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21576354

ABSTRACT

DNA-dependent protein kinase (DNA-PK) is involved in DNA double-strand break (DSB) signalling and repair. We report that DNA-PK is activated by mild hypoxia conditions (0.1-1% O2) as shown by (1) its autophosphorylation on Ser2056, and (2) its mobilisation from a soluble nucleoplasmic compartment to a less extractable nuclear fraction. The recruitment of DNA-PK was not followed by activation and recruitment of the XRCC4-DNA-ligase-IV complex, suggesting that DSBs are not responsible for activation of DNA-PK. To unravel the mechanism of DNA-PK activation, we show that exposure of cells to trichostatin A, a histone deacetylase inhibitor, leads to DNA-PK autophosphorylation and relocalisation to DNA. Histone acetylation (mainly H3K14) is increased in hypoxic cells and treatment with anacardic acid, an inhibitor of histone acetyl transferase, prevented both histone modifications and DNA-PK activation in hypoxic conditions. Importantly, in using either silenced DNA-PK cells or cells exposed to a specific DNA-PK inhibitor (NU7026), we demonstrated that hypoxic DNA-PK activation positively regulates the key transcription factor HIF-1 and one subsequent target gene, GLUT1. Our results show that hypoxia initiates chromatin modification and consequently DNA-PK activation, which positively regulate cellular oxygen-sensing and oxygen-signalling pathways.


Subject(s)
Chromatin/metabolism , DNA-Activated Protein Kinase/metabolism , Stress, Physiological , Acetylation , Adaptation, Physiological , Aminoglycosides/pharmacology , Antigens, Nuclear/metabolism , Cell Hypoxia , Cell Line , Chromones/pharmacology , DNA Damage , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Enediynes/pharmacology , Enzyme Activation , Glucose Transporter Type 1/metabolism , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ku Autoantigen , Morpholines/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction
18.
Cell Mol Life Sci ; 69(4): 629-40, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21773671

ABSTRACT

Functional telomeres are protected from non-homologous end-joining (NHEJ) and homologous recombination (HR) DNA repair pathways. Replication is a critical period for telomeres because of the requirement for reconstitution of functional protected telomere conformations, a process that involves DNA repair proteins. Using knockdown of DNA-PKcs and Rad51 expression in three different cell lines, we demonstrate the respective involvement of NHEJ and HR in the formation of telomere aberrations induced by the G-quadruplex ligand 360A during or after replication. HR contributed to specific chromatid-type aberrations (telomere losses and doublets) affecting the lagging strand telomeres, whereas DNA-PKcs-dependent NHEJ was responsible for sister telomere fusions as a direct consequence of G-quadruplex formation and/or stabilization induced by 360A on parental telomere G strands. NHEJ and HR activation at telomeres altered mitotic progression in treated cells. In particular, NHEJ-mediated sister telomere fusions were associated with altered metaphase-anaphase transition and anaphase bridges and resulted in cell death during mitosis or early G1. Collectively, these data elucidate specific molecular and cellular mechanisms triggered by telomere targeting by the G-quadruplex ligand 360A, leading to cancer cell death.


Subject(s)
Apoptosis , DNA-Activated Protein Kinase/metabolism , G-Quadruplexes , Mitosis/genetics , Nuclear Proteins/metabolism , Pyridines/pharmacology , Quinolines/pharmacology , Rad51 Recombinase/metabolism , Telomere , Anaphase , Cell Line , DNA End-Joining Repair , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/genetics , Homologous Recombination , Humans , Ligands , Metaphase , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/genetics , Telomere/metabolism , Telomere/pathology
19.
Cell Mol Life Sci ; 69(17): 2933-49, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22460582

ABSTRACT

DNA ligase I (LigI) plays a central role in the joining of strand interruptions during replication and repair. In our current study, we provide evidence that DNA ligase III (LigIII) and XRCC1, which form a complex that functions in single-strand break repair, are required for the proliferation of mammalian LigI-depleted cells. We show from our data that in cells with either dysfunctional LigI activity or depleted of this enzyme, both LigIII and XRCC1 are retained on the chromatin and accumulate at replication foci. We also demonstrate that the LigI and LigIII proteins cooperate to inhibit sister chromatid exchanges but that only LigI prevents telomere sister fusions. Taken together, these results suggest that in cells with dysfunctional LigI, LigIII contributes to the ligation of replication intermediates but not to the prevention of telomeric instability.


Subject(s)
DNA Ligases/physiology , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Fibroblasts/cytology , Mitosis/physiology , Telomere/chemistry , Animals , Blotting, Western , Cell Survival , Cells, Cultured , Chromatin/genetics , Colony-Forming Units Assay , DNA Damage/genetics , DNA Ligase ATP , DNA Ligases/antagonists & inhibitors , DNA Repair , DNA Replication , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Embryo, Mammalian/enzymology , Fibroblasts/enzymology , Fluorescent Antibody Technique , Genetic Complementation Test , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Knockout , Poly-ADP-Ribose Binding Proteins , RNA, Small Interfering/genetics , Sister Chromatid Exchange/genetics , Telomere/genetics , X-ray Repair Cross Complementing Protein 1 , Xenopus Proteins
20.
Cell Mol Life Sci ; 69(6): 951-62, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21922195

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) has been identified as a determinant of sensitivity to poly(ADP-ribose) polymerase (PARP) inhibitors. Here, the consequences of its depletion on cell survival, PARP activity, the recruitment of base excision repair (BER) proteins to DNA damage sites, and overall DNA single-strand break (SSB) repair were investigated using isogenic HeLa stably depleted (KD) and Control cell lines. Synthetic lethality achieved by disrupting PARP activity in Cdk5-deficient cells was confirmed, and the Cdk5(KD) cells were also found to be sensitive to the killing effects of ionizing radiation (IR) but not methyl methanesulfonate or neocarzinostatin. The recruitment profiles of GFP-PARP-1 and XRCC1-YFP to sites of micro-irradiated Cdk5(KD) cells were slower and reached lower maximum values, while the profile of GFP-PCNA recruitment was faster and attained higher maximum values compared to Control cells. Higher basal, IR, and hydrogen peroxide-induced polymer levels were observed in Cdk5(KD) compared to Control cells. Recruitment of GFP-PARP-1 in which serines 782, 785, and 786, potential Cdk5 phosphorylation targets, were mutated to alanines in micro-irradiated Control cells was also reduced. We hypothesize that Cdk5-dependent PARP-1 phosphorylation on one or more of these serines results in an attenuation of its ribosylating activity facilitating persistence at DNA damage sites. Despite these deficiencies, Cdk5(KD) cells are able to effectively repair SSBs probably via the long patch BER pathway, suggesting that the enhanced radiation sensitivity of Cdk5(KD) cells is due to a role of Cdk5 in other pathways or the altered polymer levels.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Poly(ADP-ribose) Polymerases/metabolism , Radiation Tolerance , Base Sequence , DNA Damage , DNA Repair , HeLa Cells , Humans , Molecular Sequence Data , Poly(ADP-ribose) Polymerase Inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL