Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 176(3): 505-519.e22, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30612738

ABSTRACT

Genomic instability can be a hallmark of both human genetic disease and cancer. We identify a deleterious UBQLN4 mutation in families with an autosomal recessive syndrome reminiscent of genome instability disorders. UBQLN4 deficiency leads to increased sensitivity to genotoxic stress and delayed DNA double-strand break (DSB) repair. The proteasomal shuttle factor UBQLN4 is phosphorylated by ATM and interacts with ubiquitylated MRE11 to mediate early steps of homologous recombination-mediated DSB repair (HRR). Loss of UBQLN4 leads to chromatin retention of MRE11, promoting non-physiological HRR activity in vitro and in vivo. Conversely, UBQLN4 overexpression represses HRR and favors non-homologous end joining. Moreover, we find UBQLN4 overexpressed in aggressive tumors. In line with an HRR defect in these tumors, UBQLN4 overexpression is associated with PARP1 inhibitor sensitivity. UBQLN4 therefore curtails HRR activity through removal of MRE11 from damaged chromatin and thus offers a therapeutic window for PARP1 inhibitor treatment in UBQLN4-overexpressing tumors.


Subject(s)
Carrier Proteins/genetics , Nuclear Proteins/genetics , Carrier Proteins/metabolism , Chromatin/metabolism , DNA , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Female , Genomic Instability , Germ-Line Mutation , Homologous Recombination , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Male , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/metabolism , Primary Cell Culture , Recombinational DNA Repair
2.
Nat Rev Mol Cell Biol ; 21(10): 569, 2020 10.
Article in English | MEDLINE | ID: mdl-32807976
3.
Blood ; 144(11): 1193-1205, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-38917355

ABSTRACT

ABSTRACT: Ataxia-telangiectasia (A-T) is an autosomal-recessive disorder caused by pathogenic variants (PVs) of the ATM gene, predisposing children to hematological malignancies. We investigated their characteristics and outcomes to generate data-based treatment recommendations. In this multinational, observational study we report 202 patients aged ≤25 years with A-T and hematological malignancies from 25 countries. Ninety-one patients (45%) presented with mature B-cell lymphomas, 82 (41%) with acute lymphoblastic leukemia/lymphoma, 21 (10%) with Hodgkin lymphoma and 8 (4%) with other hematological malignancies. Four-year overall survival and event-free survival (EFS) were 50.8% (95% confidence interval [CI], 43.6-59.1) and 47.9% (95% CI 40.8-56.2), respectively. Cure rates have not significantly improved over the last four decades (P = .76). The major cause of treatment failure was treatment-related mortality (TRM) with a four-year cumulative incidence of 25.9% (95% CI, 19.5-32.4). Germ line ATM PVs were categorized as null or hypomorphic and patients with available genetic data (n = 110) were classified as having absent (n = 81) or residual (n = 29) ATM kinase activity. Four-year EFS was 39.4% (95% CI, 29-53.3) vs 78.7% (95% CI, 63.7-97.2), (P < .001), and TRM rates were 37.6% (95% CI, 26.4-48.7) vs 4.0% (95% CI, 0-11.8), (P = .017), for those with absent and residual ATM kinase activity, respectively. Absence of ATM kinase activity was independently associated with decreased EFS (HR = 0.362, 95% CI, 0.16-0.82; P = .009) and increased TRM (hazard ratio [HR] = 14.11, 95% CI, 1.36-146.31; P = .029). Patients with A-T and leukemia/lymphoma may benefit from deescalated therapy for patients with absent ATM kinase activity and near-standard therapy regimens for those with residual kinase activity.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Ataxia Telangiectasia , Germ-Line Mutation , Hematologic Neoplasms , Humans , Ataxia Telangiectasia Mutated Proteins/genetics , Child , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/complications , Ataxia Telangiectasia/mortality , Male , Female , Adolescent , Hematologic Neoplasms/genetics , Hematologic Neoplasms/mortality , Child, Preschool , Infant , Young Adult , Adult
4.
Mol Cell ; 69(5): 866-878.e7, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29499138

ABSTRACT

Double-strand breaks (DSBs) are critical DNA lesions that robustly activate the elaborate DNA damage response (DDR) network. We identified a critical player in DDR fine-tuning: the E3/E4 ubiquitin ligase UBE4A. UBE4A's recruitment to sites of DNA damage is dependent on primary E3 ligases in the DDR and promotes enhancement and sustainment of K48- and K63-linked ubiquitin chains at these sites. This step is required for timely recruitment of the RAP80 and BRCA1 proteins and proper organization of RAP80- and BRCA1-associated protein complexes at DSB sites. This pathway is essential for optimal end resection at DSBs, and its abrogation leads to upregulation of the highly mutagenic alternative end-joining repair at the expense of error-free homologous recombination repair. Our data uncover a critical regulatory level in the DSB response and underscore the importance of fine-tuning the complex DDR network for accurate and balanced execution of DSB repair.


Subject(s)
BRCA1 Protein/metabolism , Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , Nuclear Proteins/metabolism , Recombinational DNA Repair/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , BRCA1 Protein/genetics , Carrier Proteins/genetics , DNA-Binding Proteins , HeLa Cells , Histone Chaperones , Humans , Nuclear Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitins/genetics , Ubiquitins/metabolism
5.
EMBO J ; 40(2): e104400, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33215756

ABSTRACT

The DNA damage response (DDR) is a complex signaling network that relies on cascades of protein phosphorylation, which are initiated by three protein kinases of the family of PI3-kinase-related protein kinases (PIKKs): ATM, ATR, and DNA-PK. ATM is missing or inactivated in the genome instability syndrome, ataxia-telangiectasia (A-T). The relative shares of these PIKKs in the response to genotoxic stress and the functional relationships among them are central questions in the genome stability field. We conducted a comprehensive phosphoproteomic analysis in human wild-type and A-T cells treated with the double-strand break-inducing chemical, neocarzinostatin, and validated the results with the targeted proteomic technique, selected reaction monitoring. We also matched our results with 34 published screens for DDR factors, creating a valuable resource for identifying strong candidates for novel DDR players. We uncovered fine-tuned dynamics between the PIKKs following genotoxic stress, such as DNA-PK-dependent attenuation of ATM. In A-T cells, partial compensation for ATM absence was provided by ATR and DNA-PK, with distinct roles and kinetics. The results highlight intricate relationships between these PIKKs in the DDR.


Subject(s)
DNA Damage/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Phosphatidylinositol 3-Kinases/genetics , Proteomics/methods , Signal Transduction/genetics
6.
Nat Rev Mol Cell Biol ; 14(4): 197-210, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23486281

ABSTRACT

The protein kinase ataxia-telangiectasia mutated (ATM) is best known for its role as an apical activator of the DNA damage response in the face of DNA double-strand breaks (DSBs). Following induction of DSBs, ATM mobilizes one of the most extensive signalling networks that responds to specific stimuli and modifies directly or indirectly a broad range of targets. Although most ATM research has focused on this function, evidence suggests that ATM-mediated phosphorylation has a role in the response to other types of genotoxic stress. Moreover, it has become apparent that ATM is active in other cell signalling pathways involved in maintaining cellular homeostasis.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , DNA Damage , DNA Repair , Homeostasis/genetics , Humans , Models, Genetic , Phosphorylation , Signal Transduction/genetics
7.
Nat Rev Mol Cell Biol ; 14(4): 197-210, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23847781

ABSTRACT

The protein kinase ataxia-telangiectasia mutated (ATM) is best known for its role as an apical activator of the DNA damage response in the face of DNA double-strand breaks (DSBs). Following induction of DSBs, ATM mobilizes one of the most extensive signalling networks that responds to specific stimuli and modifies directly or indirectly a broad range of targets. Although most ATM research has focused on this function, evidence suggests that ATM-mediated phosphorylation has a role in the response to other types of genotoxic stress. Moreover, it has become apparent that ATM is active in other cell signalling pathways involved in maintaining cellular homeostasis.


Subject(s)
Cell Cycle Proteins/physiology , DNA Damage , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Nucleus/enzymology , Homeostasis , Humans , Neurons/enzymology , Oxidation-Reduction , Protein Interaction Maps , Protein Processing, Post-Translational , Protein Stability , Retinal Vessels/enzymology , Signal Transduction , Tumor Suppressor Protein p53/metabolism
8.
Cell ; 140(3): 308-10, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20144755

ABSTRACT

The tumor suppressor protein p53, a crucial player in the DNA damage response, is regulated in many ways, most notably through ubiquitination. In this issue, Yuan et al. (2010) identify the deubiquitinating protease USP10 as a new regulator of p53 in the DNA damage response and tumor development.


Subject(s)
DNA Repair , Ubiquitin Thiolesterase/metabolism , Gene Expression Regulation, Neoplastic , Humans , Tumor Suppressor Protein p53/metabolism , Ubiquitination
9.
Nucleic Acids Res ; 46(2): 730-747, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29253183

ABSTRACT

The DNA damage response (DDR) is an extensive signaling network that is robustly mobilized by DNA double-strand breaks (DSBs). The primary transducer of the DSB response is the protein kinase, ataxia-telangiectasia, mutated (ATM). Here, we establish nuclear poly(A)-binding protein 1 (PABPN1) as a novel target of ATM and a crucial player in the DSB response. PABPN1 usually functions in regulation of RNA processing and stability. We establish that PABPN1 is recruited to the DDR as a critical regulator of DSB repair. A portion of PABPN1 relocalizes to DSB sites and is phosphorylated on Ser95 in an ATM-dependent manner. PABPN1 depletion sensitizes cells to DSB-inducing agents and prolongs the DSB-induced G2/M cell-cycle arrest, and DSB repair is hampered by PABPN1 depletion or elimination of its phosphorylation site. PABPN1 is required for optimal DSB repair via both nonhomologous end-joining (NHEJ) and homologous recombination repair (HRR), and specifically is essential for efficient DNA-end resection, an initial, key step in HRR. Using mass spectrometry analysis, we capture DNA damage-induced interactions of phospho-PABPN1, including well-established DDR players as well as other RNA metabolizing proteins. Our results uncover a novel ATM-dependent axis in the rapidly growing interface between RNA metabolism and the DDR.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Nuclear Proteins/metabolism , Poly(A)-Binding Protein I/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line, Tumor , DNA/genetics , DNA/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , HeLa Cells , Humans , Nuclear Proteins/genetics , Phosphorylation , Poly(A)-Binding Protein I/genetics , Protein Binding , Protein Interaction Maps , RNA Interference
11.
Mol Cell ; 41(5): 529-42, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21362549

ABSTRACT

The cellular response to DNA double-strand breaks (DSBs) is mobilized by the protein kinase ATM, which phosphorylates key players in the DNA damage response (DDR) network. A major question is how ATM controls DSB repair. Optimal repair requires chromatin relaxation at damaged sites. Chromatin reorganization is coupled to dynamic alterations in histone posttranslational modifications. Here, we show that in human cells, DSBs induce monoubiquitylation of histone H2B, a modification that is associated in undamaged cells with transcription elongation. We find that this process relies on recruitment to DSB sites and ATM-dependent phosphorylation of the responsible E3 ubiquitin ligase: the RNF20-RNF40 heterodimer. H2B monoubiquitylation is required for timely recruitment of players in the two major DSB repair pathways-nonhomologous end-joining and homologous recombination repair-and optimal repair via both pathways. Our data and previous data suggest a two-stage model for chromatin decondensation that facilitates DSB repair.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Histones/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin/chemistry , Ataxia Telangiectasia Mutated Proteins , Chromatin/chemistry , Chromatin/metabolism , Comet Assay/methods , HeLa Cells , Histones/chemistry , Humans , Kinetics , Phosphorylation , Protein Processing, Post-Translational , RNA Interference , Recombination, Genetic , Ubiquitin-Protein Ligases/metabolism
12.
Semin Cancer Biol ; 37-38: 26-35, 2016 06.
Article in English | MEDLINE | ID: mdl-26773346

ABSTRACT

The genome is constantly attacked by a variety of genotoxic insults. The causal role for DNA damage in aging and cancer is exemplified by genetic defects in DNA repair that underlie a broad spectrum of acute and chronic human disorders that are characterized by developmental abnormalities, premature aging, and cancer predisposition. The disease symptoms are typically tissue-specific with uncertain genotype-phenotype correlation. The cellular DNA damage response (DDR) has been extensively investigated ever since yeast geneticists discovered DNA damage checkpoint mechanisms, several decades ago. In recent years, it has become apparent that not only cell-autonomous but also systemic DNA damage responses determine the outcome of genome instability in organisms. Understanding the mechanisms of non-cell-autonomous DNA damage responses will provide important new insights into the role of genome instability in human aging and a host of diseases including cancer and might better explain the complex phenotypes caused by genome instability.


Subject(s)
Aging/genetics , DNA Damage , DNA Repair , Genomic Instability , Neoplasms/genetics , Animals , Cellular Senescence/genetics , DNA Breaks, Double-Stranded , Genetic Association Studies , Humans , Immunity, Innate/genetics , Mutation
13.
Nucleic Acids Res ; 43(9): 4517-30, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25855810

ABSTRACT

The DNA damage response is vigorously activated by DNA double-strand breaks (DSBs). The chief mobilizer of the DSB response is the ATM protein kinase. We discovered that the COP9 signalosome (CSN) is a crucial player in the DSB response and an ATM target. CSN is a protein complex that regulates the activity of cullin ring ubiquitin ligase (CRL) complexes by removing the ubiquitin-like protein, NEDD8, from their cullin scaffold. We find that the CSN is physically recruited to DSB sites in a neddylation-dependent manner, and is required for timely repair of DSBs, affecting the balance between the two major DSB repair pathways-nonhomologous end-joining and homologous recombination repair (HRR). The CSN is essential for the processivity of deep end-resection-the initial step in HRR. Cullin 4a (CUL4A) is recruited to DSB sites in a CSN- and neddylation-dependent manner, suggesting that CSN partners with CRL4 in this pathway. Furthermore, we found that ATM-mediated phosphorylation of CSN subunit 3 on S410 is critical for proper DSB repair, and that loss of this phosphorylation site alone is sufficient to cause a DDR deficiency phenotype in the mouse. This novel branch of the DSB response thus significantly affects genome stability.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Multiprotein Complexes/metabolism , Peptide Hydrolases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , COP9 Signalosome Complex , Cell Line , Cells, Cultured , Cullin Proteins/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Protein Kinases/metabolism
14.
Mol Cell ; 31(2): 167-77, 2008 Jul 25.
Article in English | MEDLINE | ID: mdl-18657500

ABSTRACT

Ataxia Telangiectasia Mutated (ATM) signaling is essential for the repair of a subset of DNA double-strand breaks (DSBs); however, its precise role is unclear. Here, we show that < or =25% of DSBs require ATM signaling for repair, and this percentage correlates with increased chromatin but not damage complexity. Importantly, we demonstrate that heterochromatic DSBs are generally repaired more slowly than euchromatic DSBs, and ATM signaling is specifically required for DSB repair within heterochromatin. Significantly, knockdown of the transcriptional repressor KAP-1, an ATM substrate, or the heterochromatin-building factors HP1 or HDAC1/2 alleviates the requirement for ATM in DSB repair. We propose that ATM signaling temporarily perturbs heterochromatin via KAP-1, which is critical for DSB repair/processing within otherwise compacted/inflexible chromatin. In support of this, ATM signaling alters KAP-1 affinity for chromatin enriched for heterochromatic factors. These data suggest that the importance of ATM signaling for DSB repair increases as the heterochromatic component of a genome expands.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Heterochromatin/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , Deoxyribonucleases/metabolism , Embryo, Mammalian/cytology , Fibroblasts/enzymology , Fibroblasts/radiation effects , Heterochromatin/radiation effects , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Mice , NIH 3T3 Cells , Radiation, Ionizing , Repressor Proteins/metabolism , Signal Transduction/radiation effects , Tripartite Motif-Containing Protein 28
15.
EMBO J ; 28(23): 3667-80, 2009 Dec 02.
Article in English | MEDLINE | ID: mdl-19851285

ABSTRACT

Human tyrosyl-DNA phosphodiesterase (TDP1) hydrolyzes the phosphodiester bond at a DNA 3' end linked to a tyrosyl moiety. This type of linkage is found at stalled topoisomerase I (Top1)-DNA covalent complexes, and TDP1 has been implicated in the repair of such complexes. Here we show that Top1-associated DNA double-stranded breaks (DSBs) induce the phosphorylation of TDP1 at S81. This phosphorylation is mediated by the protein kinases: ataxia-telangiectasia-mutated (ATM) and DNA-dependent protein kinase (DNA-PK). Phosphorylated TDP1 forms nuclear foci that co-localize with those of phosphorylated histone H2AX (gammaH2AX). Both Top1-induced replication- and transcription-mediated DNA damages induce TDP1 phosphorylation. Furthermore, we show that S81 phosphorylation stabilizes TDP1, induces the formation of XRCC1 (X-ray cross-complementing group 1)-TDP1 complexes and enhances the mobilization of TDP1 to DNA damage sites. Finally, we provide evidence that TDP1-S81 phosphorylation promotes cell survival and DNA repair in response to CPT-induced DSBs. Together; our findings provide a new mechanism for TDP1 post-translational regulation by ATM and DNA-PK.


Subject(s)
Cell Cycle Proteins/chemistry , DNA Repair , DNA-Activated Protein Kinase/chemistry , DNA-Binding Proteins/chemistry , Phosphoric Diester Hydrolases/metabolism , Protein Serine-Threonine Kinases/chemistry , Tumor Suppressor Proteins/chemistry , Ataxia Telangiectasia/enzymology , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Carnitine O-Palmitoyltransferase/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cell Survival/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Humans , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/physiology , Phosphorylation/genetics , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Serine/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology , X-ray Repair Cross Complementing Protein 1
16.
Nat Cell Biol ; 8(8): 870-6, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16862143

ABSTRACT

The cellular DNA-damage response is a signaling network that is vigorously activated by cytotoxic DNA lesions, such as double-strand breaks (DSBs). The DSB response is mobilized by the nuclear protein kinase ATM, which modulates this process by phosphorylating key players in these pathways. A long-standing question in this field is whether DSB formation affects chromatin condensation. Here, we show that DSB formation is followed by ATM-dependent chromatin relaxation. ATM's effector in this pathway is the protein KRAB-associated protein (KAP-1, also known as TIF1beta, KRIP-1 or TRIM28), previously known as a corepressor of gene transcription. In response to DSB induction, KAP-1 is phosphorylated in an ATM-dependent manner on Ser 824. KAP-1 is phosphorylated exclusively at the damage sites, from which phosphorylated KAP-1 spreads rapidly throughout the chromatin. Ablation of the phosphorylation site of KAP-1 leads to loss of DSB-induced chromatin decondensation and renders the cells hypersensitive to DSB-inducing agents. Knocking down KAP-1, or mimicking a constitutive phosphorylation of this protein, leads to constitutive chromatin relaxation. These results suggest that chromatin relaxation is a fundamental pathway in the DNA-damage response and identify its primary mediators.


Subject(s)
Cell Cycle Proteins/physiology , Chromatin/metabolism , DNA Damage , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Repressor Proteins/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins/physiology , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Microscopy, Fluorescence , Mutation/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tripartite Motif-Containing Protein 28 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Zinostatin/pharmacology
17.
EMBO Rep ; 12(7): 713-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21637298

ABSTRACT

The cellular response to double-strand breaks (DSBs) in DNA is a complex signalling network, mobilized by the nuclear protein kinase ataxia-telangiectasia mutated (ATM), which phosphorylates many factors in the various branches of this network. A main question is how ATM regulates DSB repair. Here, we identify the DNA repair enzyme polynucleotide kinase/phosphatase (PNKP) as an ATM target. PNKP phosphorylates 5'-OH and dephosphorylates 3'-phosphate DNA ends that are formed at DSB termini caused by DNA-damaging agents, thereby regenerating legitimate ends for further processing. We establish that the ATM phosphorylation targets on human PNKP-Ser 114 and Ser 126-are crucial for cellular survival following DSB induction and for effective DSB repair, being essential for damage-induced enhancement of the activity of PNKP and its proper accumulation at the sites of DNA damage. These findings show a direct functional link between ATM and the DSB-repair machinery.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair Enzymes/metabolism , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cytotoxins/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Repair Enzymes/genetics , HEK293 Cells , Humans , Mice , Phosphorylation/drug effects , Zinostatin/pharmacology
18.
Nat Rev Cancer ; 3(3): 155-68, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12612651

ABSTRACT

Maintenance of genome stability is essential for avoiding the passage to neoplasia. The DNA-damage response--a cornerstone of genome stability--occurs by a swift transduction of the DNA-damage signal to many cellular pathways. A prime example is the cellular response to DNA double-strand breaks, which activate the ATM protein kinase that, in turn, modulates numerous signalling pathways. ATM mutations lead to the cancer-predisposing genetic disorder ataxia-telangiectasia (A-T). Understanding ATM's mode of action provides new insights into the association between defective responses to DNA damage and cancer, and brings us closer to resolving the issue of cancer predisposition in some A-T carriers.


Subject(s)
Ataxia Telangiectasia/genetics , Cell Transformation, Neoplastic/genetics , DNA Repair/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Ataxia Telangiectasia/enzymology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Chromosome Breakage , DNA Damage , DNA Repair/genetics , DNA-Binding Proteins , Enzyme Activation , Genes, cdc , Genetic Predisposition to Disease , Humans , Mice , Mice, Knockout , Models, Genetic , Multigene Family , Neoplastic Syndromes, Hereditary/enzymology , Neoplastic Syndromes, Hereditary/genetics , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Tumor Suppressor Proteins
19.
Nucleic Acids Res ; 39(Database issue): D793-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21097778

ABSTRACT

The rapid accumulation of knowledge on biological signaling pathways and their regulatory mechanisms has highlighted the need for specific repositories that can store, organize and allow retrieval of pathway information in a way that will be useful for the research community. SPIKE (Signaling Pathways Integrated Knowledge Engine; http://www.cs.tau.ac.il/&~spike/) is a database for achieving this goal, containing highly curated interactions for particular human pathways, along with literature-referenced information on the nature of each interaction. To make database population and pathway comprehension straightforward, a simple yet informative data model is used, and pathways are laid out as maps that reflect the curator’s understanding and make the utilization of the pathways easy. The database currently focuses primarily on pathways describing DNA damage response, cell cycle, programmed cell death and hearing related pathways. Pathways are regularly updated, and additional pathways are gradually added. The complete database and the individual maps are freely exportable in several formats. The database is accompanied by a stand-alone software tool for analysis and dynamic visualization of pathways.


Subject(s)
Databases, Factual , Intercellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Apoptosis , Cell Cycle , DNA Damage , Humans
20.
Proc Natl Acad Sci U S A ; 107(5): 2207-12, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20133864

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality worldwide and is considered to be the outcome of chronic liver inflammation. Currently, the main treatment for HCC is surgical resection. However, survival rates are suboptimal partially because of tumor recurrence in the remaining liver. Our aim was to understand the molecular mechanisms linking liver regeneration under chronic inflammation to hepatic tumorigenesis. Mdr2-KO mice, a model of inflammation-associated cancer, underwent partial hepatectomy (PHx), which led to enhanced hepatocarcinogenesis. Moreover, liver regeneration in these mice was severely attenuated. We demonstrate the activation of the DNA damage-response machinery and increased genomic instability during early liver inflammatory stages resulting in hepatocyte apoptosis, cell-cycle arrest, and senescence and suggest their involvement in tumor growth acceleration subsequent to PHx. We propose that under the regenerative proliferative stress induced by liver resection, the genomic unstable hepatocytes generated during chronic inflammation escape senescence and apoptosis and reenter the cell cycle, triggering the enhanced tumorigenesis. Thus, we clarify the immediate and long-term contributions of the DNA damage response to HCC development and recurrence.


Subject(s)
Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/physiopathology , Liver Regeneration/physiology , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , DNA Breaks, Double-Stranded , Gene Expression , Genomic Instability , Hepatectomy , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/physiopathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Liver Regeneration/genetics , Mice , Mice, Knockout , Models, Biological , ATP-Binding Cassette Sub-Family B Member 4
SELECTION OF CITATIONS
SEARCH DETAIL