Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Cell ; 60(1): 163-76, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26344098

ABSTRACT

Human Timeless helps stabilize replication forks during normal DNA replication and plays a critical role in activation of the S phase checkpoint and proper establishment of sister chromatid cohesion. However, it remains elusive whether Timeless is involved in the repair of damaged DNA. Here, we identify that Timeless physically interacts with PARP-1 independent of poly(ADP-ribosyl)ation. We present high-resolution crystal structures of Timeless PAB (PARP-1-binding domain) in free form and in complex with PARP-1 catalytic domain. Interestingly, Timeless PAB domain specifically recognizes PARP-1, but not PARP-2 or PARP-3. Timeless-PARP-1 interaction does not interfere with PARP-1 enzymatic activity. We demonstrate that rapid and transient accumulation of Timeless at laser-induced DNA damage sites requires PARP-1, but not poly(ADP-ribosyl)ation and that Timeless is co-trapped with PARP-1 at DNA lesions upon PARP inhibition. Furthermore, we show that Timeless and PARP-1 interaction is required for efficient homologous recombination repair.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Recombinational DNA Repair , Binding Sites , Crystallography, X-Ray , DNA Breaks, Double-Stranded , HeLa Cells , Homologous Recombination , Humans , Models, Molecular , Poly (ADP-Ribose) Polymerase-1 , Protein Multimerization , Substrate Specificity
2.
Nucleic Acids Res ; 48(21): 12234-12251, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33211885

ABSTRACT

Altered oncogene expression in cancer cells causes loss of redox homeostasis resulting in oxidative DNA damage, e.g. 8-oxoguanine (8-oxoG), repaired by base excision repair (BER). PARP1 coordinates BER and relies on the upstream 8-oxoguanine-DNA glycosylase (OGG1) to recognise and excise 8-oxoG. Here we hypothesize that OGG1 may represent an attractive target to exploit reactive oxygen species (ROS) elevation in cancer. Although OGG1 depletion is well tolerated in non-transformed cells, we report here that OGG1 depletion obstructs A3 T-cell lymphoblastic acute leukemia growth in vitro and in vivo, validating OGG1 as a potential anti-cancer target. In line with this hypothesis, we show that OGG1 inhibitors (OGG1i) target a wide range of cancer cells, with a favourable therapeutic index compared to non-transformed cells. Mechanistically, OGG1i and shRNA depletion cause S-phase DNA damage, replication stress and proliferation arrest or cell death, representing a novel mechanistic approach to target cancer. This study adds OGG1 to the list of BER factors, e.g. PARP1, as potential targets for cancer treatment.


Subject(s)
Colonic Neoplasms/drug therapy , DNA Glycosylases/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Poly (ADP-Ribose) Polymerase-1/immunology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , DNA Damage , DNA Glycosylases/antagonists & inhibitors , DNA Glycosylases/metabolism , DNA Repair/drug effects , DNA Replication/drug effects , DNA, Neoplasm/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , Guanine/metabolism , HCT116 Cells , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Oxidative Stress , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction , Survival Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Nucleic Acids Res ; 47(14): 7418-7429, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31127309

ABSTRACT

Affinity maturation of the humoral immune response depends on somatic hypermutation (SHM) of immunoglobulin (Ig) genes, which is initiated by targeted lesion introduction by activation-induced deaminase (AID), followed by error-prone DNA repair. Stringent regulation of this process is essential to prevent genetic instability, but no negative feedback control has been identified to date. Here we show that poly(ADP-ribose) polymerase-1 (PARP-1) is a key factor restricting AID activity during somatic hypermutation. Poly(ADP-ribose) (PAR) chains formed at DNA breaks trigger AID-PAR association, thus preventing excessive DNA damage induction at sites of AID action. Accordingly, AID activity and somatic hypermutation at the Ig variable region is decreased by PARP-1 activity. In addition, PARP-1 regulates DNA lesion processing by affecting strand biased A:T mutagenesis. Our study establishes a novel function of the ancestral genome maintenance factor PARP-1 as a critical local feedback regulator of both AID activity and DNA repair during Ig gene diversification.


Subject(s)
Cytidine Deaminase/genetics , Genes, Immunoglobulin/genetics , Immunoglobulin Variable Region/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Cells, Cultured , Cytidine Deaminase/metabolism , DNA Damage , DNA Repair , Humans , Mice , Mutation , Poly (ADP-Ribose) Polymerase-1/metabolism
4.
Int J Mol Sci ; 22(9)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925271

ABSTRACT

DNA damage caused by reactive oxygen species may result in genetic mutations or cell death. Base excision repair (BER) is the major pathway that repairs DNA oxidative damage in order to maintain genomic integrity. In mammals, eleven DNA glycosylases have been reported to initiate BER, where each recognizes a few related DNA substrate lesions with some degree of overlapping specificity. 7,8-dihydro-8-oxoguanine (8-oxoG), one of the most abundant DNA oxidative lesions, is recognized and excised mainly by 8-oxoguanine DNA glycosylase 1 (OGG1). Further oxidation of 8-oxoG generates hydantoin lesions, which are recognized by NEIL glycosylases. Here, we demonstrate that NEIL1, and to a lesser extent NEIL2, can potentially function as backup BER enzymes for OGG1 upon pharmacological inhibition or depletion of OGG1. NEIL1 recruitment kinetics and chromatin binding after DNA damage induction increase in cells treated with OGG1 inhibitor TH5487 in a dose-dependent manner, whereas NEIL2 accumulation at DNA damage sites is prolonged following OGG1 inhibition. Furthermore, depletion of OGG1 results in increased retention of NEIL1 and NEIL2 at damaged chromatin. Importantly, oxidatively stressed NEIL1- or NEIL2-depleted cells show excessive genomic 8-oxoG lesions accumulation upon OGG1 inhibition, suggesting a prospective compensatory role for NEIL1 and NEIL2. Our study thus exemplifies possible backup mechanisms within the base excision repair pathway.


Subject(s)
DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Benzimidazoles/pharmacology , Cell Line , DNA/metabolism , DNA Damage/genetics , DNA Glycosylases/drug effects , DNA Repair/drug effects , DNA Repair/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Guanine/analogs & derivatives , Humans , Kinetics , Mutation , Oxidative Stress , Piperidines/pharmacology , Prospective Studies , Reactive Oxygen Species/metabolism
6.
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
7.
Mol Oncol ; 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38533616

ABSTRACT

The one-carbon metabolism enzyme bifunctional methylenetetrahydrofolate dehydrogenase/cyclohydrolase 2 (MTHFD2) is among the most overexpressed proteins across tumors and is widely recognized as a promising anticancer target. While MTHFD2 is mainly described as a mitochondrial protein, a new nuclear function is emerging. Here, we observe that nuclear MTHFD2 protein levels and association with chromatin increase following ionizing radiation (IR) in an ataxia telangiectasia mutated (ATM)- and DNA-dependent protein kinase (DNA-PK)-dependent manner. Furthermore, repair of IR-induced DNA double-strand breaks (DSBs) is delayed upon MTHFD2 knockdown, suggesting a role for MTHFD2 in DSB repair. In support of this, we observe impaired recruitment of replication protein A (RPA), reduced resection, decreased IR-induced DNA repair protein RAD51 homolog 1 (RAD51) levels and impaired homologous recombination (HR) activity in MTHFD2-depleted cells following IR. In conclusion, we identify a key role for MTHFD2 in HR repair and describe an interdependency between MTHFD2 and HR proficiency that could potentially be exploited for cancer therapy.

8.
Nucleic Acids Res ; 39(12): 5045-56, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21398629

ABSTRACT

Post-translational poly(ADP-ribosyl)ation has diverse essential functions in the cellular response to DNA damage as it contributes to avid DNA damage detection and assembly of the cellular repair machinery but extensive modification eventually also induces cell death. While there are 17 human poly(ADP-ribose) polymerase (PARP) genes, there is only one poly(ADP-ribose) glycohydrolase (PARG) gene encoding several PARG isoforms located in different subcellular compartments. To investigate the recruitment of PARG isoforms to DNA repair sites we locally introduced DNA damage by laser microirradiation. All PARG isoforms were recruited to DNA damage sites except for a mitochondrial localized PARG fragment. Using PARP knock out cells and PARP inhibitors, we showed that PARG recruitment was only partially dependent on PARP-1 and PAR synthesis, indicating a second, PAR-independent recruitment mechanism. We found that PARG interacts with PCNA, mapped a PCNA binding site and showed that binding to PCNA contributes to PARG recruitment to DNA damage sites. This dual recruitment mode of the only nuclear PARG via the versatile loading platform PCNA and by a PAR dependent mechanism likely contributes to the dynamic regulation of this posttranslational modification and ensures the tight control of the switch between efficient DNA repair and cell death.


Subject(s)
DNA Damage , Glycoside Hydrolases/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Animals , Biocatalysis , Cells, Cultured , Glycoside Hydrolases/analysis , Glycoside Hydrolases/chemistry , Humans , Lasers , Mice , Protein Isoforms/metabolism , Protein Structure, Tertiary
9.
Nucleic Acids Res ; 39(19): 8445-56, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21745822

ABSTRACT

Active chromatin remodelling is integral to the DNA damage response in eukaryotes, as damage sensors, signalling molecules and repair enzymes gain access to lesions. A variety of nucleosome remodelling complexes is known to promote different stages of DNA repair. The nucleosome sliding factors CHRAC/ACF of Drosophila are involved in chromatin organization during development. Involvement of corresponding hACF1-containing mammalian nucleosome sliding factors in replication, transcription and very recently also non-homologous end-joining of DNA breaks have been suggested. We now found that hACF1-containing factors are more generally involved in the DNA damage response. hACF1 depletion increases apoptosis, sensitivity to radiation and compromises the G2/M arrest that is activated in response to UV- and X-rays. In the absence of hACF1, γH2AX and CHK2ph signals are diminished. hACF1 and its ATPase partner SNF2H rapidly accumulate at sites of laser-induced DNA damage. hACF1 is also required for a tight checkpoint that is induced upon replication fork collapse. ACF1-depleted cells that are challenged with aphidicolin enter mitosis despite persistence of lesions and accumulate breaks in metaphase chromosomes. hACF1-containing remodellers emerge as global facilitators of the cellular response to a variety of different types of DNA damage.


Subject(s)
DNA Damage , DNA Repair , G2 Phase Cell Cycle Checkpoints , M Phase Cell Cycle Checkpoints , Transcription Factors/physiology , Aphidicolin/toxicity , Apoptosis , Cell Line , Chromosomal Proteins, Non-Histone , Chromosome Fragility , Humans , RNA Interference , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
10.
Oncogene ; 42(16): 1282-1293, 2023 04.
Article in English | MEDLINE | ID: mdl-36871087

ABSTRACT

The NUDIX hydrolase NUDT22 converts UDP-glucose into glucose-1-phosphate and the pyrimidine nucleotide uridine monophosphate but a biological significance for this biochemical reaction has not yet been established. Glucose-1-phosphate is an important metabolite for energy and biomass production through glycolysis and nucleotides required for DNA replication are produced through energetically expensive de novo or energy-efficient salvage pathways. Here, we describe p53-regulated pyrimidine salvage through NUDT22-dependent hydrolysis of UDP-glucose to maintain cancer cell growth and to prevent replication stress. NUDT22 expression is consistently elevated in cancer tissues and high NUDT22 expression correlates with worse survival outcomes in patients indicating an increased dependency of cancer cells to NUDT22. Furthermore, we show that NUDT22 transcription is induced after inhibition of glycolysis, MYC-mediated oncogenic stress, and DNA damage directly through p53. NUDT22-deficient cancer cells suffer from growth retardation, S-phase delay, and slower DNA replication fork speed. Uridine supplementation rescues replication fork progression and alleviates replication stress and DNA damage. Conversely, NUDT22 deficiency sensitizes cells to de novo pyrimidine synthesis inhibition in vitro and reduces cancer growth in vivo. In conclusion, NUDT22 maintains pyrimidine supply in cancer cells and depletion of NUDT22 leads to genome instability. Targeting NUDT22 therefore has high potential for therapeutic applications in cancer therapy.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Humans , Glucose , Neoplasms/drug therapy , Neoplasms/genetics , Pyrimidines/pharmacology , Uridine/metabolism , Uridine Diphosphate
11.
Nat Metab ; 5(4): 642-659, 2023 04.
Article in English | MEDLINE | ID: mdl-37012496

ABSTRACT

Cancer cells fuel their increased need for nucleotide supply by upregulating one-carbon (1C) metabolism, including the enzymes methylenetetrahydrofolate dehydrogenase-cyclohydrolase 1 and 2 (MTHFD1 and MTHFD2). TH9619 is a potent inhibitor of dehydrogenase and cyclohydrolase activities in both MTHFD1 and MTHFD2, and selectively kills cancer cells. Here, we reveal that, in cells, TH9619 targets nuclear MTHFD2 but does not inhibit mitochondrial MTHFD2. Hence, overflow of formate from mitochondria continues in the presence of TH9619. TH9619 inhibits the activity of MTHFD1 occurring downstream of mitochondrial formate release, leading to the accumulation of 10-formyl-tetrahydrofolate, which we term a 'folate trap'. This results in thymidylate depletion and death of MTHFD2-expressing cancer cells. This previously uncharacterized folate trapping mechanism is exacerbated by physiological hypoxanthine levels that block the de novo purine synthesis pathway, and additionally prevent 10-formyl-tetrahydrofolate consumption for purine synthesis. The folate trapping mechanism described here for TH9619 differs from other MTHFD1/2 inhibitors and antifolates. Thus, our findings uncover an approach to attack cancer and reveal a regulatory mechanism in 1C metabolism.


Subject(s)
Methylenetetrahydrofolate Dehydrogenase (NADP) , Neoplasms , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Folic Acid/metabolism , Formates , Purines , Tetrahydrofolates
13.
Nat Commun ; 13(1): 1015, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35197472

ABSTRACT

Evidence that long non-coding RNAs (lncRNAs) participate in DNA repair is accumulating, however, whether they can control DNA repair pathway choice is unknown. Here we show that the small Cajal body-specific RNA 2 (scaRNA2) can promote HR by inhibiting DNA-dependent protein kinase (DNA-PK) and, thereby, NHEJ. By binding to the catalytic subunit of DNA-PK (DNA-PKcs), scaRNA2 weakens its interaction with the Ku70/80 subunits, as well as with the LINP1 lncRNA, thereby preventing catalytic activation of the enzyme. Inhibition of DNA-PK by scaRNA2 stimulates DNA end resection by the MRN/CtIP complex, activation of ATM at DNA lesions and subsequent repair by HR. ScaRNA2 is regulated in turn by WRAP53ß, which binds this RNA, sequestering it away from DNA-PKcs and allowing NHEJ to proceed. These findings reveal that RNA-dependent control of DNA-PK catalytic activity is involved in regulating whether the cell utilizes NHEJ or HR.


Subject(s)
Protein Kinases , RNA , DNA/genetics , DNA/metabolism , DNA End-Joining Repair , DNA Repair , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Protein Kinases/metabolism
14.
Science ; 376(6600): 1471-1476, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35737787

ABSTRACT

Oxidative DNA damage is recognized by 8-oxoguanine (8-oxoG) DNA glycosylase 1 (OGG1), which excises 8-oxoG, leaving a substrate for apurinic endonuclease 1 (APE1) and initiating repair. Here, we describe a small molecule (TH10785) that interacts with the phenylalanine-319 and glycine-42 amino acids of OGG1, increases the enzyme activity 10-fold, and generates a previously undescribed ß,δ-lyase enzymatic function. TH10785 controls the catalytic activity mediated by a nitrogen base within its molecular structure. In cells, TH10785 increases OGG1 recruitment to and repair of oxidative DNA damage. This alters the repair process, which no longer requires APE1 but instead is dependent on polynucleotide kinase phosphatase (PNKP1) activity. The increased repair of oxidative DNA lesions with a small molecule may have therapeutic applications in various diseases and aging.


Subject(s)
DNA Damage , DNA Glycosylases , DNA Repair , Oxidative Stress , Biocatalysis/drug effects , DNA Damage/drug effects , DNA Glycosylases/chemistry , DNA Glycosylases/drug effects , DNA Repair/drug effects , Enzyme Activation , Glycine/chemistry , Humans , Ligands , Oxidative Stress/genetics , Phenylalanine/chemistry , Substrate Specificity
15.
Mol Cell Proteomics ; 7(11): 2279-87, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18622019

ABSTRACT

Genetic high throughput screens have yielded large sets of potential protein-protein interactions now to be verified and further investigated. Here we present a simple assay to directly visualize protein-protein interactions in single living cells. Using a modified lac repressor system, we tethered a fluorescent bait at a chromosomal lac operator array and assayed for co-localization of fluorescent prey fusion proteins. With this fluorescent two-hybrid assay we successfully investigated the interaction of proteins from different subcellular compartments including nucleus, cytoplasm, and mitochondria. In combination with an S phase marker we also studied the cell cycle dependence of protein-protein interactions. These results indicate that the fluorescent two-hybrid assay is a powerful tool to investigate protein-protein interactions within their cellular environment and to monitor the response to external stimuli in real time.


Subject(s)
Protein Interaction Mapping/methods , Two-Hybrid System Techniques , Animals , Base Sequence , Cell Cycle , Cell Line , Cricetinae , DNA Primers/genetics , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Lac Operon , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Proteomics/methods , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transfection
16.
Biomolecules ; 10(11)2020 10 26.
Article in English | MEDLINE | ID: mdl-33114607

ABSTRACT

8-oxoguanine DNA glycosylase (OGG1) is the main DNA glycosylase responsible for the excision of 7,8-dihydro-8-oxoguanine (8-oxoG) from duplex DNA to initiate base excision repair. This glycosylase activity is relevant in many pathological conditions including cancer, inflammation, and neurodegenerative diseases. To have a better understanding of the role of OGG1, we previously reported TH5487, a potent active site inhibitor of OGG1. Here, we further investigate the consequences of inhibiting OGG1 with TH5487. TH5487 treatment induces accumulation of genomic 8-oxoG lesions. Furthermore, it impairs the chromatin binding of OGG1 and results in lower recruitment of OGG1 to regions of DNA damage. Inhibiting OGG1 with TH5487 interferes with OGG1's incision activity, resulting in fewer DNA double-strand breaks in cells exposed to oxidative stress. This study validates TH5487 as a potent OGG1 inhibitor that prevents the repair of 8-oxoG and alters OGG1-chromatin dynamics and OGG1's recruitment kinetics.


Subject(s)
Benzimidazoles/pharmacology , Chromatin/drug effects , DNA Glycosylases/antagonists & inhibitors , Piperidines/pharmacology , Chromatin/metabolism , DNA Glycosylases/metabolism , Humans , Tumor Cells, Cultured
17.
Cancer Res ; 80(17): 3530-3541, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32312836

ABSTRACT

Reactive oxygen species (ROS) oxidize nucleotide triphosphate pools (e.g., 8-oxodGTP), which may kill cells if incorporated into DNA. Whether cancers avoid poisoning from oxidized nucleotides by preventing incorporation via the oxidized purine diphosphatase MTH1 remains under debate. Also, little is known about DNA polymerases incorporating oxidized nucleotides in cells or how oxidized nucleotides in DNA become toxic. Here we show that replacement of one of the main DNA replicases in human cells, DNA polymerase delta (Pol δ), with an error-prone variant allows increased 8-oxodG accumulation into DNA following treatment with TH588, a dual MTH1 inhibitor and microtubule targeting agent. The resulting elevated genomic 8-oxodG correlated with increased cytotoxicity of TH588. Interestingly, no substantial perturbation of replication fork progression was observed, but rather mitotic progression was impaired and mitotic DNA synthesis triggered. Reducing mitotic arrest by reversin treatment prevented accumulation of genomic 8-oxodG and reduced cytotoxicity of TH588, in line with the notion that mitotic arrest is required for ROS buildup and oxidation of the nucleotide pool. Furthermore, delayed mitosis and increased mitotic cell death was observed following TH588 treatment in cells expressing the error-prone but not wild-type Pol δ variant, which is not observed following treatments with antimitotic agents. Collectively, these results link accumulation of genomic oxidized nucleotides with disturbed mitotic progression. SIGNIFICANCE: These findings uncover a novel link between accumulation of genomic 8-oxodG and perturbed mitotic progression in cancer cells, which can be exploited therapeutically using MTH1 inhibitors.See related commentary by Alnajjar and Sweasy, p. 3459.


Subject(s)
8-Hydroxy-2'-Deoxyguanosine , Phosphoric Monoester Hydrolases , DNA Repair Enzymes/genetics , Genomics , Humans , Mitosis/genetics , Phosphoric Monoester Hydrolases/genetics , Pyrimidines/pharmacology
18.
Nucleic Acids Res ; 35(22): 7665-75, 2007.
Article in English | MEDLINE | ID: mdl-17982172

ABSTRACT

Genome integrity is constantly threatened by DNA lesions arising from numerous exogenous and endogenous sources. Survival depends on immediate recognition of these lesions and rapid recruitment of repair factors. Using laser microirradiation and live cell microscopy we found that the DNA-damage dependent poly(ADP-ribose) polymerases (PARP) PARP-1 and PARP-2 are recruited to DNA damage sites, however, with different kinetics and roles. With specific PARP inhibitors and mutations, we could show that the initial recruitment of PARP-1 is mediated by the DNA-binding domain. PARP-1 activation and localized poly(ADP-ribose) synthesis then generates binding sites for a second wave of PARP-1 recruitment and for the rapid accumulation of the loading platform XRCC1 at repair sites. Further PARP-1 poly(ADP-ribosyl)ation eventually initiates the release of PARP-1. We conclude that feedback regulated recruitment of PARP-1 and concomitant local poly(ADP-ribosyl)ation at DNA lesions amplifies a signal for rapid recruitment of repair factors enabling efficient restoration of genome integrity.


Subject(s)
DNA Damage , Poly(ADP-ribose) Polymerases/metabolism , Animals , Cell Nucleolus/enzymology , Cells, Cultured , DNA Repair , DNA-Binding Proteins/metabolism , Feedback, Physiological , Gene Deletion , HeLa Cells , Humans , Kinetics , Mice , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Protein Structure, Tertiary , X-ray Repair Cross Complementing Protein 1
19.
Nucleus ; 10(1): 231-253, 2019 12.
Article in English | MEDLINE | ID: mdl-31744372

ABSTRACT

Constitutive heterochromatin is considered as a functionally inert genome compartment, important for its architecture and stability. How such stable structure is maintained is not well understood. Here, we apply four different visualization schemes to label it and investigate its dynamics during DNA replication and repair. We show that replisomes assemble over the heterochromatin in a temporally ordered manner. Furthermore, heterochromatin undergoes transient decompaction locally at the active sites of DNA synthesis. Using selective laser microirradiation conditions that lead to damage repaired via processive DNA synthesis, we measured similarly local decompaction of heterochromatin. In both cases, we could not observe large-scale movement of heterochromatin to the domain surface. Instead, the processive DNA synthesis machinery assembled at the replication/repair sites. Altogether, our data are compatible with a progression of DNA replication/repair along the chromatin in a dynamic mode with localized and transient decompaction that does not globally remodels the whole heterochromatin compartment.


Subject(s)
DNA Repair , DNA Replication , DNA/biosynthesis , Heterochromatin/metabolism , Animals , Cells, Cultured , DNA/chemistry , HeLa Cells , Heterochromatin/chemistry , Humans , Mice
20.
J Cell Biochem ; 104(5): 1562-9, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18384127

ABSTRACT

Mammalian cells are constantly threatened by multiple types of DNA lesions arising from various sources like irradiation, environmental agents, replication errors or by-products of the normal cellular metabolism. If not readily detected and repaired these lesions can lead to cell death or to the transformation of cells giving rise to life-threatening diseases like cancer. Multiple specialized repair pathways have evolved to preserve the genetic integrity of a cell. The increasing number of DNA damage sensors, checkpoint regulators, and repair factors identified in the numerous interconnected repair pathways raises the question of how DNA repair is coordinated. In the last decade, various methods have been developed that allow the induction of DNA lesions and subsequent real-time analysis of repair factor assembly at DNA repair sites in living cells. This combination of biophysical and molecular cell biology methods has yielded interesting new insights into the order and kinetics of protein recruitment and identified regulatory sequences and selective loading platforms for the efficient restoration of the genetic and epigenetic integrity of mammalian cells.


Subject(s)
DNA Damage , Nuclear Proteins/metabolism , Animals , Cell Survival , DNA Repair , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL