Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4991, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37591859

ABSTRACT

Activation of the KRAS oncogene is a source of replication stress, but how this stress is generated and how it is tolerated by cancer cells remain poorly understood. Here we show that induction of KRASG12V expression in untransformed cells triggers H3K27me3 and HP1-associated chromatin compaction in an RNA transcription dependent manner, resulting in replication fork slowing and cell death. Furthermore, elevated ATR expression is necessary and sufficient for tolerance of KRASG12V-induced replication stress to expand replication stress-tolerant cells (RSTCs). PrimPol is phosphorylated at Ser255, a potential Chk1 substrate site, under KRASG12V-induced replication stress and promotes repriming to maintain fork progression and cell survival in an ATR/Chk1-dependent manner. However, ssDNA gaps are generated at heterochromatin by PrimPol-dependent repriming, leading to genomic instability. These results reveal a role of ATR-PrimPol in enabling precancerous cells to survive KRAS-induced replication stress and expand clonally with accumulation of genomic instability.


Subject(s)
Heterochromatin , Proto-Oncogene Proteins p21(ras) , Humans , Ataxia Telangiectasia Mutated Proteins/genetics , Chromatin , DNA Primase , DNA-Directed DNA Polymerase , Genomic Instability , Heterochromatin/genetics , Multifunctional Enzymes , Proto-Oncogene Proteins p21(ras)/genetics
2.
Cancer Sci ; 114(7): 2709-2721, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37189251

ABSTRACT

DNA replication stress (RS) causes genomic instability and vulnerability in cancer cells. To counteract RS, cells have evolved various mechanisms involving the ATR kinase signaling pathway, which regulates origin firing, cell cycle checkpoints, and fork stabilization to secure the fidelity of replication. However, ATR signaling also alleviates RS to support cell survival by driving RS tolerance, thereby contributing to therapeutic resistance. Cancer cells harboring genetic mutations and other changes that disrupt normal DNA replication increase the risk of DNA damage and the levels of RS, conferring addiction to ATR activity for sustainable replication and susceptibility to therapeutic approaches using ATR inhibitors (ATRis). Therefore, clinical trials are currently being conducted to evaluate the efficacy of ATRis as monotherapies or in combination with other drugs and biomarkers. In this review, we discuss recent advances in the elucidation of the mechanisms by which ATR functions in the RS response and its therapeutic relevance when utilizing ATRis.


Subject(s)
DNA Damage , Neoplasms , Humans , Ataxia Telangiectasia Mutated Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction , Cell Cycle Checkpoints , DNA Replication , Checkpoint Kinase 1/metabolism , Neoplasms/drug therapy , Neoplasms/genetics
3.
Biochim Biophys Acta Mol Cell Res ; 1870(6): 119484, 2023 08.
Article in English | MEDLINE | ID: mdl-37201767

ABSTRACT

Ataxia-telangiectasia mutated and Rad3-related (ATR) kinase is a crucial regulator of the cell cycle checkpoint and activated in response to DNA replication stress by two independent pathways via RPA32-ETAA1 and TopBP1. However, the precise activation mechanism of ATR by the RPA32-ETAA1 pathway remains unclear. Here, we show that p130RB2, a member of the retinoblastoma protein family, participates in the pathway under hydroxyurea-induced DNA replication stress. p130RB2 binds to ETAA1, but not TopBP1, and depletion of p130RB2 inhibits the RPA32-ETAA1 interaction under replication stress. Moreover, p130RB2 depletion reduces ATR activation accompanied by phosphorylation of its targets RPA32, Chk1, and ATR itself. It also causes improper re-progression of S phase with retaining single-stranded DNA after cancelation of the stress, which leads to an increase in the anaphase bridge phenotype and a decrease in cell survival. Importantly, restoration of p130RB2 rescued the disrupted phenotypes of p130RB2 knockdown cells. These results suggest positive involvement of p130RB2 in the RPA32-ETAA1-ATR axis and proper re-progression of the cell cycle to maintain genome integrity.


Subject(s)
DNA Replication , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Phosphorylation , Cell Cycle , Cell Cycle Checkpoints
4.
J Biol Chem ; 297(1): 100882, 2021 07.
Article in English | MEDLINE | ID: mdl-34144037

ABSTRACT

Alteration of RNA splicing is a hallmark of cellular senescence, which is associated with age-related disease and cancer development. However, the roles of splicing factors in cellular senescence are not fully understood. In this study, we identified the splicing factor PRPF19 as a critical regulator of cellular senescence in normal human diploid fibroblasts. PRPF19 was downregulated during replicative senescence, and PRPF19 knockdown prematurely induced senescence-like cell cycle arrest through the p53-p21 pathway. RNA-sequencing analysis revealed that PRPF19 knockdown caused a switch of the MDM4 splicing isoform from stable full-length MDM4-FL to unstable MDM4-S lacking exon 6. We also found that PRPF19 regulates MDM4 splicing by promoting the physical interaction of other splicing factors, PRPF3 and PRPF8, which are key components of the core spliceosome, U4/U6.U5 tri-snRNP. Given that MDM4 is a major negative regulator of p53, our findings imply that PRPF19 downregulation inhibits MDM4-mediated p53 inactivation, resulting in induction of cellular senescence. Thus, PRPF19 plays an important role in the induction of p53-dependent cellular senescence.


Subject(s)
Alternative Splicing , Cell Cycle Proteins/genetics , Cellular Senescence , DNA Repair Enzymes/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/genetics , RNA Splicing Factors/metabolism , Cell Cycle Proteins/metabolism , DNA Repair Enzymes/genetics , HEK293 Cells , Humans , Nuclear Proteins/genetics , Protein Binding , Proto-Oncogene Proteins/metabolism , RNA Splicing Factors/genetics , Spliceosomes/metabolism , Tumor Suppressor Protein p53/metabolism
5.
NAR Cancer ; 2(2): zcaa005, 2020 Jun.
Article in English | MEDLINE | ID: mdl-34316685

ABSTRACT

The SWI/SNF chromatin remodeling complex regulates transcription through the control of chromatin structure and is increasingly thought to play an important role in human cancer. Lung adenocarcinoma (LADC) patients frequently harbor mutations in SMARCA4, a core component of this multisubunit complex. Most of these mutations are loss-of-function mutations, which disrupt critical functions in the regulation of chromatin architecture and can cause DNA replication stress. This study reports that LADC cells deficient in SMARCA4 showed increased DNA replication stress and greater sensitivity to the ATR inhibitor (ATRi) in vitro and in vivo. Mechanistically, loss of SMARCA4 increased heterochromatin formation, resulting in stalled forks, a typical DNA replication stress. In the absence of SMARCA4, severe ATRi-induced single-stranded DNA, which caused replication catastrophe, was generated on nascent DNA near the reversed forks around heterochromatin in an Mre11-dependent manner. Thus, loss of SMARCA4 confers susceptibility to ATRi, both by increasing heterochromatin-associated replication stress and by allowing Mre11 to destabilize reversed forks. These two mechanisms synergistically increase susceptibility of SMARCA4-deficient LADC cells to ATRi. These results provide a preclinical basis for assessing SMARCA4 defects as a biomarker of ATRi efficacy.

6.
Cell ; 175(2): 558-570.e11, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30245011

ABSTRACT

Given that genomic DNA exerts its function by being transcribed, it is critical for the maintenance of homeostasis that DNA damage, such as double-strand breaks (DSBs), within transcriptionally active regions undergoes accurate repair. However, it remains unclear how this is achieved. Here, we describe a mechanism for transcription-associated homologous recombination repair (TA-HRR) in human cells. The process is initiated by R-loops formed upon DSB induction. We identify Rad52, which is recruited to the DSB site in a DNA-RNA-hybrid-dependent manner, as playing pivotal roles in promoting XPG-mediated R-loop processing and initiating subsequent repair by HRR. Importantly, dysfunction of TA-HRR promotes DSB repair via non-homologous end joining, leading to a striking increase in genomic aberrations. Thus, our data suggest that the presence of R-loops around DSBs within transcriptionally active regions promotes accurate repair of DSBs via processing by Rad52 and XPG to protect genomic information in these critical regions from gene alterations.


Subject(s)
DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Nuclear Proteins/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , Recombinational DNA Repair/physiology , Transcription Factors/metabolism , Cell Line , DNA/genetics , DNA Breaks, Double-Stranded , DNA Damage , DNA End-Joining Repair , DNA Repair , DNA-Binding Proteins/physiology , Endonucleases/physiology , Homologous Recombination , Humans , Nuclear Proteins/genetics , Nuclear Proteins/physiology , RNA/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Transcription Factors/physiology
8.
Oncotarget ; 8(8): 12941-12952, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28099935

ABSTRACT

Well-differentiated liposarcoma (WDLPS) and dedifferentiated liposarcoma (DDLPS) are closely related tumors commonly characterized by MDM2/CDK4 gene amplification, and lack clinically effective treatment options when inoperable. To identify novel therapeutic targets, we performed targeted genomic sequencing analysis of 19 WDLPS and 37 DDLPS tumor samples using a panel of 104 cancer-related genes (NCC oncopanel v3) developed specifically for genomic testing to select suitable molecular targeted therapies. The results of this analysis indicated that these sarcomas had very few gene mutations and a high frequency of amplifications of not only MDM2 and CDK4 but also other genes. Potential driver mutations were found in only six (11%) samples; however, gene amplification events (other than MDM2 and CDK4 amplification) were identified in 30 (54%) samples. Receptor tyrosine kinase (RTK) genes in particular were amplified in 18 (32%) samples. In addition, growth of a WDLPS cell line with IGF1R amplification was suppressed by simultaneous inhibition of CDK4 and IGF1R, using palbociclib and NVP-AEW541, respectively. Combination therapy with CDK4 and RTK inhibitors may be an effective therapeutic option for WDLPS/DDLPS patients with RTK gene amplification.


Subject(s)
Liposarcoma/genetics , Receptor Protein-Tyrosine Kinases/genetics , Soft Tissue Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Blotting, Western , Female , Gene Amplification , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Polymerase Chain Reaction
9.
Mol Cell Biol ; 36(3): 394-406, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26572825

ABSTRACT

Histone acetyltransferase binding to ORC-1 (HBO1) is a critically important histone acetyltransferase for forming the prereplicative complex (pre-RC) at the replication origin. Pre-RC formation is completed by loading of the MCM2-7 heterohexameric complex, which functions as a helicase in DNA replication. HBO1 recruited to the replication origin by CDT1 acetylates histone H4 to relax the chromatin conformation and facilitates loading of the MCM complex onto replication origins. However, the acetylation status and mechanism of regulation of histone H3 at replication origins remain elusive. HBO1 positively regulates cell proliferation under normal cell growth conditions. Whether HBO1 regulates proliferation in response to DNA damage is poorly understood. In this study, we demonstrated that HBO1 was degraded after DNA damage to suppress cell proliferation. Ser50 and Ser53 of HBO1 were phosphorylated in an ATM/ATR DNA damage sensor-dependent manner after UV treatment. ATM/ATR-dependently phosphorylated HBO1 preferentially interacted with DDB2 and was ubiquitylated by CRL4(DDB2). Replacement of endogenous HBO1 in Ser50/53Ala mutants maintained acetylation of histone H3K14 and impaired cell cycle regulation in response to UV irradiation. Our findings demonstrate that HBO1 is one of the targets in the DNA damage checkpoint. These results show that ubiquitin-dependent control of the HBO1 protein contributes to cell survival during UV irradiation.


Subject(s)
Cell Proliferation/radiation effects , DNA Damage/radiation effects , DNA-Binding Proteins/metabolism , Histone Acetyltransferases/metabolism , Phosphorylation/radiation effects , Ubiquitin-Protein Ligases/metabolism , Acetylation/radiation effects , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Histones/metabolism , Humans , Point Mutation , Protein Interaction Maps , Protein Stability/radiation effects , Proteolysis , Ubiquitin/metabolism , Ubiquitination/radiation effects , Ultraviolet Rays
10.
Cancer Res ; 74(14): 3880-9, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24876101

ABSTRACT

CBP-93872 was previously identified as a G2 checkpoint inhibitor using a cell-based high-throughput screening system. However, its molecular actions as well as cellular targets are largely unknown. Here, we uncovered the molecular mechanisms underlying abrogation of the G2 checkpoint by CBP-93872. CBP-93872 specifically abrogates the DNA double-stranded break (DSB)-induced G2 checkpoint through inhibiting maintenance but not initiation of G2 arrest because of specific inhibition of DSB-dependent ATR activation. Hence, ATR-dependent phosphorylation of Nbs1 and replication protein A 2 upon DSB was strongly suppressed in the presence of CBP-93872. CBP-93872 did not seem to inhibit DNA-end resection, but did inhibit Nbs1-dependent and ssDNA-induced ATR activation in vitro in a dose-dependent manner. Taken together, our results suggest that CBP-93872 is an inhibitor of maintenance of the DSB-specific G2 checkpoint and thus might be a strong candidate as the basis for a drug that specifically sensitizes p53-mutated cancer cells to DSB-inducing DNA damage therapy.


Subject(s)
Aniline Compounds/pharmacology , Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Nuclear Proteins/metabolism , Propanolamines/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Mutation , Nuclear Proteins/genetics , Phosphorylation , Protein Binding
11.
Cancer Sci ; 105(7): 870-4, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24724610

ABSTRACT

Nuclear factor-κB (NF-κB) is a key regulator of cancer progression and the inflammatory effects of disease. To identify inhibitors of DNA binding to NF-κB, we developed a new homogeneous method for detection of sequence-specific DNA-binding proteins. This method, which we refer to as DSE-FRET, is based on two phenomena: protein-dependent blocking of spontaneous DNA strand exchange (DSE) between partially double-stranded DNA probes, and fluorescence resonance energy transfer (FRET). If a probe labeled with a fluorophore and quencher is mixed with a non-labeled probe in the absence of a target protein, strand exchange occurs between the probes and results in fluorescence elevation. In contrast, blocking of strand exchange by a target protein results in lower fluorescence intensity. Recombinant human NF-κB (p50) suppressed the fluorescence elevation of a specific probe in a concentration-dependent manner, but had no effect on a non-specific probe. Competitors bearing a NF-κB binding site restored fluorescence, and the degree of restoration was inversely correlated with the number of nucleotide substitutions within the NF-κB binding site of the competitor. Evaluation of two NF-κB inhibitors, Evans Blue and dehydroxymethylepoxyquinomicin ([-]-DHMEQ), was carried out using p50 and p52 (another form of NF-κB), and IC50 values were obtained. The DSE-FRET technique also detected the differential effect of (-)-DHMEQ on p50 and p52 inhibition. These data indicate that DSE-FRET can be used for high throughput screening of anticancer drugs targeted to DNA-binding proteins.


Subject(s)
DNA-Binding Proteins/analysis , Drug Screening Assays, Antitumor/methods , Fluorescence Resonance Energy Transfer/methods , Benzamides/pharmacology , Binding Sites , Cyclohexanones/pharmacology , DNA Probes , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Evans Blue/pharmacology , High-Throughput Screening Assays/methods , Humans , Inhibitory Concentration 50 , NF-kappa B/genetics , NF-kappa B/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Carcinogenesis ; 34(11): 2486-97, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23825154

ABSTRACT

Inhibitors of poly(ADP-ribose) polymerase (PARP) are promising anticancer drugs, particularly for the treatment of tumors deficient in the DNA damage response (DDR). However, it is challenging to design effective therapeutic strategies for use of these compounds against cancers without DDR deficiencies. In this context, combination therapies in which PARP inhibitors are used alongside DDR inhibitors have elicited a great deal of interest. Curcumin, a component of turmeric (Curcuma longa), has been tested in clinical studies for its chemosensitizing potential; however, the mechanisms of chemosensitization by curcumin have not been fully elucidated. This study demonstrates that curcumin suppresses three major DDR pathways: non-homologous end joining (NHEJ), homologous recombination (HR) and the DNA damage checkpoint. Curcumin suppresses the histone acetylation at DNA double-strand break (DSB) sites by inhibiting histone acetyltransferase activity, thereby reducing recruitment of the key NHEJ factor KU70/KU80 to DSB sites. Curcumin also suppresses HR by reducing expression of the BRCA1 gene, which regulates HR, by impairing histone acetylation at the BRCA1 promoter. Curcumin also inhibits ataxia telangiectasia and Rad3-related protein (ATR) kinase (IC50 in vitro = 493 nM), resulting in impaired activation of ATR-CHK1 signaling, which is necessary for HR and the DNA damage checkpoint pathway. Thus, curcumin suppresses three DDR pathways by inhibiting histone acetyltransferases and ATR. Concordantly, curcumin sensitizes cancer cells to PARP inhibitors by enhancing apoptosis and mitotic catastrophe via inhibition of both the DNA damage checkpoint and DSB repair. Our results indicate that curcumin is a promising sensitizer for PARP inhibitor-based therapy.


Subject(s)
Curcumin/pharmacology , DNA Damage/drug effects , Homologous Recombination/drug effects , Neoplasms/pathology , Poly(ADP-ribose) Polymerase Inhibitors , Signal Transduction/drug effects , Acetylation/drug effects , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/metabolism , Blotting, Western , Cell Cycle Checkpoints , Cell Proliferation/drug effects , Checkpoint Kinase 1 , Cobalt Radioisotopes , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Enzyme Inhibitors/pharmacology , Gamma Rays , Histone Acetyltransferases/metabolism , Histones/metabolism , Homologous Recombination/radiation effects , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Poly (ADP-Ribose) Polymerase-1 , Promoter Regions, Genetic , Protein Kinases/metabolism , Sialoglycoproteins/antagonists & inhibitors , Sialoglycoproteins/metabolism , Signal Transduction/radiation effects , Tumor Cells, Cultured , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/metabolism
13.
Cell Rep ; 3(5): 1651-62, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23684611

ABSTRACT

The ATM- and Rad3-related (ATR) kinase is a master regulator of the DNA damage response, yet how ATR is activated toward different substrates is still poorly understood. Here, we show that ATR phosphorylates Chk1 and RPA32 through distinct mechanisms at replication-associated DNA double-stranded breaks (DSBs). In contrast to the rapid phosphorylation of Chk1, RPA32 is progressively phosphorylated by ATR at Ser33 during DSB resection prior to the phosphorylation of Ser4/Ser8 by DNA-PKcs. Surprisingly, despite its reliance on ATR and TopBP1, substantial RPA32 Ser33 phosphorylation occurs in a Rad17-independent but Nbs1-dependent manner in vivo and in vitro. Importantly, the role of Nbs1 in RPA32 phosphorylation can be separated from ATM activation and DSB resection, and it is dependent upon the interaction of Nbs1 with RPA. An Nbs1 mutant that is unable to bind RPA fails to support proper recovery of collapsed replication forks, suggesting that the Nbs1-mediated mode of ATR activation is important for the repair of replication-associated DSBs.


Subject(s)
Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Breaks, Double-Stranded , DNA Replication , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Phosphorylation , Protein Binding , Protein Kinases , RNA Interference , RNA, Small Interfering/metabolism , Replication Protein A/metabolism
14.
Proc Natl Acad Sci U S A ; 109(8): 2760-5, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-21930940

ABSTRACT

Homeobox 9 (HOXB9), a nontransforming transcription factor overexpressed in breast cancer, alters tumor cell fate and promotes tumor progression and metastasis. Here we show that HOXB9 confers resistance to ionizing radiation by promoting DNA damage response. In nonirradiated cells, HOXB9 induces spontaneous DNA damage, phosphorylated histone 2AX and p53 binding protein 1 foci, and increases baseline ataxia telangiectasia mutated (ATM) phosphorylation. Upon ionizing radiation, ATM is hyperactivated in HOXB9-expressing cells during the early stages of the double-stranded DNA break (DSB) response, accelerating accumulation of phosphorylated histone 2AX, mediator of DNA-damage checkpoint 1, and p53 binding protein 1, at DSBs and enhances DSB repair. The effect of HOXB9 on the response to ionizing radiation requires the baseline ATM activity before irradiation and epithelial-to-mesenchymal transition induced by TGF-ß, a HOXB9 transcriptional target. Our results reveal the impact of a HOXB9-TGF-ß-ATM axis on checkpoint activation and DNA repair, suggesting that TGF-ß may be a key factor that links tumor microenvironment, tumor cell fate, DNA damage response, and radioresistance in a subset of HOXB9-overexpressing breast tumors.


Subject(s)
DNA Damage , Epithelial-Mesenchymal Transition , Homeodomain Proteins/metabolism , Radiation Tolerance , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/radiation effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , Enzyme Activation/radiation effects , Epithelial-Mesenchymal Transition/radiation effects , Female , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Radiation Tolerance/radiation effects , Radiation, Ionizing , Signal Transduction/radiation effects , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1
15.
Methods Mol Biol ; 782: 181-91, 2011.
Article in English | MEDLINE | ID: mdl-21870292

ABSTRACT

The Ataxia telangiectasia-mutated (ATM) and the ATM-Rad3-related (ATR) kinases are master regulators of the DNA damage-signaling pathways that respond to a wide variety of DNA damage. In this chapter, we describe an in vitro biochemical assay to study the activation of ATM and ATR by double-stranded DNA breaks (DSBs) (Shiotani and Zou, 2009, Mol Cell 33, 547-58). In this assay, DNA fragments with different structural features are used to activate ATM and ATR in human cell extracts, and the activation of ATM and ATR is monitored by the phosphorylation of specific ATM and ATR substrates. Importantly, in this assay both ATM and ATR are activated in a DNA structure-regulated manner, providing a useful tool to characterize the DNA structural determinants for their activation. The four primary steps of this assay are as follows: (1) preparation of nuclear extracts from cultured human cells; (2) generation of various DNA fragments using DNA oligonucleotides or plasmids; (3) incubation of DNA fragments in extracts; (4) analysis of the phosphorylation of specific ATM or ATR substrates.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Enzyme Assays/methods , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics
16.
Mol Cell ; 43(2): 192-202, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21777809

ABSTRACT

The ataxia telangiectasia-mutated and Rad3-related (ATR) kinase is a master checkpoint regulator safeguarding the genome. Upon DNA damage, the ATR-ATRIP complex is recruited to sites of DNA damage by RPA-coated single-stranded DNA and activated by an elusive process. Here, we show that ATR is transformed into a hyperphosphorylated state after DNA damage, and that a single autophosphorylation event at Thr 1989 is crucial for ATR activation. Phosphorylation of Thr 1989 relies on RPA, ATRIP, and ATR kinase activity, but unexpectedly not on the ATR stimulator TopBP1. Recruitment of ATR-ATRIP to RPA-ssDNA leads to congregation of ATR-ATRIP complexes and promotes Thr 1989 phosphorylation in trans. Phosphorylated Thr 1989 is directly recognized by TopBP1 via the BRCT domains 7 and 8, enabling TopBP1 to engage ATR-ATRIP, to stimulate the ATR kinase, and to facilitate ATR substrate recognition. Thus, ATR autophosphorylation on RPA-ssDNA is a molecular switch to launch robust checkpoint response.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , DNA Damage , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Switch , Genes, cdc , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Replication Protein A/genetics , Replication Protein A/metabolism , Threonine/genetics
17.
Mol Cell ; 39(2): 259-68, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20670894

ABSTRACT

Cells from Fanconi anemia (FA) patients are extremely sensitive to DNA interstrand crosslinking (ICL) agents, but the molecular basis of the hypersensitivity remains to be explored. FANCM (FA complementation group M), and its binding partner, FAAP24, anchor the multisubunit FA core complex to chromatin after DNA damage and may contribute to ICL-specific cellular response. Here we show that the FANCM/FAAP24 complex is specifically required for the recruitment of replication protein A (RPA) to ICL-stalled replication forks. ICL-induced RPA foci formation requires the DNA-binding activity of FAAP24 but not the DNA translocase activity of FANCM. Furthermore, FANCM/FAAP24-dependent RPA foci formation is required for efficient ATR-mediated checkpoint activation in response to ICL. Therefore, we propose that FANCM/FAAP24 plays a role in ICL-induced checkpoint activation through regulating RPA recruiment at ICL-stalled replication forks.


Subject(s)
Chromatin/metabolism , DNA Damage , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/genetics , Cross-Linking Reagents/pharmacology , DNA Helicases/genetics , DNA Replication/drug effects , DNA-Binding Proteins/genetics , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group Proteins , HeLa Cells , Humans , Multiprotein Complexes/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Replication Protein A/genetics , Replication Protein A/metabolism
18.
Mol Cell ; 33(5): 547-58, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19285939

ABSTRACT

ATM and ATR are two master checkpoint kinases activated by double-stranded DNA breaks (DSBs). ATM is critical for the initial response and the subsequent ATR activation. Here we show that ATR activation is coupled with loss of ATM activation, an unexpected ATM-to-ATR switch during the biphasic DSB response. ATM is activated by DSBs with blunt ends or short single-stranded overhangs (SSOs). Surprisingly, the activation of ATM in the presence of SSOs, like that of ATR, relies on single- and double-stranded DNA junctions. In a length-dependent manner, SSOs attenuate ATM activation and potentiate ATR activation through a swap of DNA-damage sensors. Progressive resection of DSBs directly promotes the ATM-to-ATR switch in vitro. In cells, the ATM-to-ATR switch is driven by both ATM and the nucleases participating in DSB resection. Thus, single-stranded DNA orchestrates ATM and ATR to function in an orderly and reciprocal manner in two distinct phases of DSB response.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Acid Anhydride Hydrolases , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Checkpoint Kinase 1 , Checkpoint Kinase 2 , DNA Repair/drug effects , DNA Repair/radiation effects , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/antagonists & inhibitors , Endodeoxyribonucleases , Enzyme Activation , Exodeoxyribonucleases/metabolism , HeLa Cells , Humans , MRE11 Homologue Protein , Nuclear Proteins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Time Factors , Transfection , Tumor Suppressor Proteins/antagonists & inhibitors
20.
Genes Dev ; 22(9): 1147-52, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451105

ABSTRACT

Chk1 is a kinase crucial for genomic integrity and an effector of ATR (ATM and Rad3-related) in DNA damage response. Here, we show that Chk1 regulates the DNA damage-induced ubiquitination of proliferating cell nuclear antigen (PCNA), which facilitates the continuous replication of damaged DNA. Surprisingly, this Chk1 function requires the DNA replication protein Claspin but not ATR. Claspin, which is stabilized by Chk1, regulates the binding of the ubiquitin ligase Rad18 to chromatin. Timeless, a Claspin-associating protein, is also required for efficient PCNA ubiquitination. Thus, Chk1 and the Claspin-Timeless module of replication forks not only participate in ATR signaling, but also protect stressed forks independently of ATR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Replication/genetics , HeLa Cells , Humans , Hydroxyurea/pharmacology , Morpholines/pharmacology , Plasmids/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyrones/pharmacology , RNA, Small Interfering/genetics , Transfection , Ubiquitination/drug effects , Ubiquitination/radiation effects , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...