Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
2.
Nucleic Acids Res ; 48(19): 10953-10972, 2020 11 04.
Article En | MEDLINE | ID: mdl-33045735

Mechanistic studies in DNA repair have focused on roles of multi-protein DNA complexes, so how long non-coding RNAs (lncRNAs) regulate DNA repair is less well understood. Yet, lncRNA LINP1 is over-expressed in multiple cancers and confers resistance to ionizing radiation and chemotherapeutic drugs. Here, we unveil structural and mechanistic insights into LINP1's ability to facilitate non-homologous end joining (NHEJ). We characterized LINP1 structure and flexibility and analyzed interactions with the NHEJ factor Ku70/Ku80 (Ku) and Ku complexes that direct NHEJ. LINP1 self-assembles into phase-separated condensates via RNA-RNA interactions that reorganize to form filamentous Ku-containing aggregates. Structured motifs in LINP1 bind Ku, promoting Ku multimerization and stabilization of the initial synaptic event for NHEJ. Significantly, LINP1 acts as an effective proxy for PAXX. Collective results reveal how lncRNA effectively replaces a DNA repair protein for efficient NHEJ with implications for development of resistance to cancer therapy.


DNA Breaks, Double-Stranded , DNA End-Joining Repair , Ku Autoantigen/metabolism , RNA, Long Noncoding/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Protein Binding , Protein Multimerization
3.
Br J Cancer ; 122(12): 1872, 2020 Jun.
Article En | MEDLINE | ID: mdl-32303715

This Article was originally published under Nature Research's License to Publish, but has now been made available under a CC BY 4.0 license. This has now been corrected in both the PDF and HTML versions of the Article.

4.
Mol Cell Biol ; 40(13)2020 06 15.
Article En | MEDLINE | ID: mdl-32284347

The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) has well-established roles in DNA double-strand break repair, and recently, nonrepair functions have also been reported. To better understand its cellular functions, we deleted DNA-PKcs from HeLa and A549 cells using CRISPR/Cas9. The resulting cells were radiation sensitive, had reduced expression of ataxia-telangiectasia mutated (ATM), and exhibited multiple mitotic defects. Mechanistically, nocodazole-induced upregulation of cyclin B1, anillin, and securin was decreased in DNA-PKcs-deficient cells, as were phosphorylation of Aurora A on threonine 288, phosphorylation of Polo-like kinase 1 (PLK1) on threonine 210, and phosphorylation of targeting protein for Xenopus Klp2 (TPX2) on serine 121. Moreover, reduced nocodazole-induced expression of anillin, securin, and cyclin B1 and phosphorylation of PLK1, Aurora A, and TPX2 were rescued by inhibition of the anaphase-promoting complex/cyclosome (APC/C) by proTAME, which prevents binding of the APC/C-activating proteins Cdc20 and Cdh1 to the APC/C. Altogether, our studies suggest that loss of DNA-PKcs prevents inactivation of the APC/C in nocodazole-treated cells.


Anaphase-Promoting Complex-Cyclosome/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Contractile Proteins/genetics , DNA-Activated Protein Kinase/genetics , Enzyme Inhibitors/pharmacology , Nocodazole/pharmacology , A549 Cells , Anaphase-Promoting Complex-Cyclosome/metabolism , Animals , Aurora Kinase A/metabolism , CRISPR-Cas Systems , Carbamates/pharmacology , Cell Cycle Proteins/metabolism , Contractile Proteins/metabolism , DNA-Activated Protein Kinase/metabolism , Diamines/pharmacology , Down-Regulation/drug effects , HeLa Cells , Humans , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Up-Regulation/drug effects , Xenopus , Polo-Like Kinase 1
5.
Br J Cancer ; 121(7): 600-610, 2019 10.
Article En | MEDLINE | ID: mdl-31481733

BACKGROUND: Up to 40% of lung adenocarcinoma have been reported to lack ataxia-telangiectasia mutated (ATM) protein expression. We asked whether ATM-deficient lung cancer cell lines are sensitive to poly-ADP ribose polymerase (PARP) inhibitors and determined the mechanism of action of olaparib in ATM-deficient A549 cells. METHODS: We analysed drug sensitivity data for olaparib and talazoparib in lung adenocarcinoma cell lines from the Genomics of Drug Sensitivity in Cancer (GDSC) project. We deleted ATM from A549 lung adenocarcinoma cells using CRISPR/Cas9 and determined the effects of olaparib and the ATM/Rad3-related (ATR) inhibitor VE-821 on cell viability. RESULTS: IC50 values for both olaparib and talazoparib positively correlated with ATM mRNA levels and gene amplification status in lung adenocarcinoma cell lines. ATM mutation was associated with a significant decrease in the IC50 for olaparib while a similar trend was observed for talazoparib. A549 cells with deletion of ATM were sensitive to ionising radiation and olaparib. Olaparib induced phosphorylation of DNA damage markers and reversible G2 arrest in ATM-deficient cells, while the combination of olaparib and VE-821 induced cell death. CONCLUSIONS: Patients with tumours characterised by ATM-deficiency may benefit from treatment with a PARP inhibitor in combination with an ATR inhibitor.


Adenocarcinoma/drug therapy , Ataxia Telangiectasia Mutated Proteins/deficiency , Lung Neoplasms/drug therapy , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Pyrazines/pharmacology , Sulfones/pharmacology , Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Deletion , Histones/metabolism , Humans , Lung Neoplasms/metabolism , Mutation , Nitroso Compounds/pharmacology , Phosphorylation , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/metabolism
6.
Nucleic Acids Res ; 45(10): 6238-6251, 2017 Jun 02.
Article En | MEDLINE | ID: mdl-28453785

Non-homologous end joining (NHEJ) repairs DNA double strand breaks in non-cycling eukaryotic cells. NHEJ relies on polynucleotide kinase/phosphatase (PNKP), which generates 5΄-phosphate/3΄-hydroxyl DNA termini that are critical for ligation by the NHEJ DNA ligase, LigIV. PNKP and LigIV require the NHEJ scaffolding protein, XRCC4. The PNKP FHA domain binds to the CK2-phosphorylated XRCC4 C-terminal tail, while LigIV uses its tandem BRCT repeats to bind the XRCC4 coiled-coil. Yet, the assembled PNKP-XRCC4-LigIV complex remains uncharacterized. Here, we report purification and characterization of a recombinant PNKP-XRCC4-LigIV complex. We show that the stable binding of PNKP in this complex requires XRCC4 phosphorylation and that only one PNKP protomer binds per XRCC4 dimer. Small angle X-ray scattering (SAXS) reveals a flexible multi-state complex that suggests that both the PNKP FHA and catalytic domains contact the XRCC4 coiled-coil and LigIV BRCT repeats. Hydrogen-deuterium exchange indicates protection of a surface on the PNKP phosphatase domain that may contact XRCC4-LigIV. A mutation on this surface (E326K) causes the hereditary neuro-developmental disorder, MCSZ. This mutation impairs PNKP recruitment to damaged DNA in human cells and provides a possible disease mechanism. Together, this work unveils multipoint contacts between PNKP and XRCC4-LigIV that regulate PNKP recruitment and activity within NHEJ.


DNA End-Joining Repair/physiology , DNA Ligase ATP/physiology , DNA Repair Enzymes/physiology , DNA-Binding Proteins/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Catalytic Domain , DNA Damage , DNA Ligase ATP/chemistry , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/genetics , DNA-Binding Proteins/chemistry , Deuterium/metabolism , Developmental Disabilities/genetics , Humans , Mass Spectrometry , Microcephaly/genetics , Models, Molecular , Multiprotein Complexes , Mutation, Missense , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Point Mutation , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Scattering, Small Angle , Seizures/genetics , Syndrome , X-Ray Diffraction
7.
Mol Cell Biol ; 35(15): 2699-713, 2015 Aug.
Article En | MEDLINE | ID: mdl-25986610

Scaffold attachment factor A (SAF-A), also called heterogenous nuclear ribonuclear protein U (hnRNP-U), is phosphorylated on serine 59 by the DNA-dependent protein kinase (DNA-PK) in response to DNA damage. Since SAF-A, DNA-PK catalytic subunit (DNA-PKcs), and protein phosphatase 6 (PP6), which interacts with DNA-PKcs, have all been shown to have roles in mitosis, we asked whether DNA-PKcs phosphorylates SAF-A in mitosis. We show that SAF-A is phosphorylated on serine 59 in mitosis, that phosphorylation requires polo-like kinase 1 (PLK1) rather than DNA-PKcs, that SAF-A interacts with PLK1 in nocodazole-treated cells, and that serine 59 is dephosphorylated by protein phosphatase 2A (PP2A) in mitosis. Moreover, cells expressing SAF-A in which serine 59 is mutated to alanine have multiple characteristics of aberrant mitoses, including misaligned chromosomes, lagging chromosomes, polylobed nuclei, and delayed passage through mitosis. Our findings identify serine 59 of SAF-A as a new target of both PLK1 and PP2A in mitosis and reveal that both phosphorylation and dephosphorylation of SAF-A serine 59 by PLK1 and PP2A, respectively, are required for accurate and timely exit from mitosis.


Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Mitosis/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , CDC2 Protein Kinase , Cell Line, Tumor , Cyclin B1/metabolism , DNA Damage/genetics , HeLa Cells , Humans , Nocodazole/pharmacology , Paclitaxel/pharmacology , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering , Securin/metabolism , Tubulin Modulators/pharmacology , Polo-Like Kinase 1
8.
Cell Cycle ; 13(13): 2129-37, 2014.
Article En | MEDLINE | ID: mdl-24841718

Small-molecule inhibitors of poly (ADP-ribose) polymerase (PARP) have shown considerable promise in the treatment of homologous recombination (HR)-defective tumors, such as BRCA1- and BRCA2-deficient breast and ovarian cancers. We previously reported that mantle cell lymphoma cells with deficiency in ataxia telangiectasia mutated (ATM) are sensitive to PARP-1 inhibitors in vitro and in vivo. Here, we report that PARP inhibitors can potentially target ATM deficiency arising in a solid malignancy. We show that ATM protein expression varies between gastric cancer cell lines, with NUGC4 having significantly reduced protein levels. Significant correlation was found between ATM protein expression and sensitivity to the PARP inhibitor olaparib, with NUGC4 being the most sensitive. Moreover, reducing ATM kinase activity using a small-molecule inhibitor (KU55933) or shRNA-mediated depletion of ATM protein enhanced olaparib sensitivity in gastric cancer cell lines with depletion or inactivation of p53. Our results demonstrate that ATM is a potential predictive biomarker for PARP-1 inhibitor activity in gastric cancer harboring disruption of p53, and that combined inhibition of ATM and PARP-1 is a rational strategy for expanding the utility of PARP-1 inhibitors to gastric cancer with p53 disruption.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/genetics , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , DNA Damage , Humans , Morpholines/pharmacology , Pyrones/pharmacology
9.
Biosci Rep ; 34(3)2014 Jun 25.
Article En | MEDLINE | ID: mdl-24844881

The protein kinase activity of the DNA-PKcs (DNA-dependent protein kinase catalytic subunit) and its autophosphorylation are critical for DBS (DNA double-strand break) repair via NHEJ (non-homologous end-joining). Recent studies have shown that depletion or inactivation of DNA-PKcs kinase activity also results in mitotic defects. DNA-PKcs is autophosphorylated on Ser2056, Thr2647 and Thr2609 in mitosis and phosphorylated DNA-PKcs localize to centrosomes, mitotic spindles and the midbody. DNA-PKcs also interacts with PP6 (protein phosphatase 6), and PP6 has been shown to dephosphorylate Aurora A kinase in mitosis. Here we report that DNA-PKcs is phosphorylated on Ser3205 and Thr3950 in mitosis. Phosphorylation of Thr3950 is DNA-PK-dependent, whereas phosphorylation of Ser3205 requires PLK1 (polo-like kinase 1). Moreover, PLK1 phosphorylates DNA-PKcs on Ser3205 in vitro and interacts with DNA-PKcs in mitosis. In addition, PP6 dephosphorylates DNA-PKcs at Ser3205 in mitosis and after IR (ionizing radiation). DNA-PKcs also phosphorylates Chk2 on Thr68 in mitosis and both phosphorylation of Chk2 and autophosphorylation of DNA-PKcs in mitosis occur in the apparent absence of Ku and DNA damage. Our findings provide mechanistic insight into the roles of DNA-PKcs and PP6 in mitosis and suggest that DNA-PKcs' role in mitosis may be mechanistically distinct from its well-established role in NHEJ.


Cell Cycle Proteins/metabolism , DNA-Activated Protein Kinase/metabolism , Mitosis/physiology , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins/genetics , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA-Activated Protein Kinase/genetics , HeLa Cells , Humans , Nuclear Proteins/genetics , Phosphoprotein Phosphatases/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Polo-Like Kinase 1
10.
EMBO Mol Med ; 4(6): 515-27, 2012 Jun.
Article En | MEDLINE | ID: mdl-22416035

Poly-ADP ribose polymerase (PARP) inhibitors have shown promise in the treatment of human malignancies characterized by deficiencies in the DNA damage repair proteins BRCA1 and BRCA2 and preclinical studies have demonstrated the potential effectiveness of PARP inhibitors in targeting ataxia-telangiectasia mutated (ATM)-deficient tumours. Here, we show that mantle cell lymphoma (MCL) cells deficient in both ATM and p53 are more sensitive to the PARP inhibitor olaparib than cells lacking ATM function alone. In ATM-deficient MCL cells, olaparib induced DNA-PK-dependent phosphorylation and stabilization of p53 as well as expression of p53-responsive cell cycle checkpoint regulators, and inhibition of DNA-PK reduced the toxicity of olaparib in ATM-deficient MCL cells. Thus, both DNA-PK and p53 regulate the response of ATM-deficient MCL cells to olaparib. In addition, small molecule inhibition of both ATM and PARP was cytotoxic in normal human fibroblasts with disruption of p53, implying that the combination of ATM and PARP inhibitors may have utility in targeting p53-deficient malignancies.


Antineoplastic Agents/pharmacology , Cell Cycle Proteins/deficiency , DNA-Binding Proteins/deficiency , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Protein Serine-Threonine Kinases/deficiency , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Proteins/deficiency , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Humans , Lymphoma, Mantle-Cell/pathology , Mice , Mutation
11.
Nucleic Acids Res ; 39(21): 9224-37, 2011 Nov.
Article En | MEDLINE | ID: mdl-21824916

Human polynucleotide kinase/phosphatase (PNKP) is a dual specificity 5'-DNA kinase/3'-DNA phosphatase, with roles in base excision repair, DNA single-strand break repair and non-homologous end joining (NHEJ); yet precisely how PNKP functions in the repair of DNA double strand breaks (DSBs) remains unclear. We demonstrate that PNKP is phosphorylated by the DNA-dependent protein kinase (DNA-PK) and ataxia-telangiectasia mutated (ATM) in vitro. The major phosphorylation site for both kinases was serine 114, with serine 126 being a minor site. Ionizing radiation (IR)-induced phosphorylation of cellular PNKP on S114 was ATM dependent, whereas phosphorylation of PNKP on S126 required both ATM and DNA-PK. Inactivation of DNA-PK and/or ATM led to reduced PNKP at DNA damage sites in vivo. Cells expressing PNKP with alanine or aspartic acid at serines 114 and 126 were modestly radiosensitive and IR enhanced the association of PNKP with XRCC4 and DNA ligase IV; however, this interaction was not affected by mutation of PNKP phosphorylation sites. Purified PNKP protein with mutation of serines 114 and 126 had decreased DNA kinase and DNA phosphatase activities and reduced affinity for DNA in vitro. Together, our results reveal that IR-induced phosphorylation of PNKP by ATM and DNA-PK regulates PNKP function at DSBs.


Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair Enzymes/metabolism , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , HeLa Cells , Humans , Mutation , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Radiation Tolerance , Radiation, Ionizing , Serine/metabolism
12.
Mol Cell Biol ; 30(6): 1368-81, 2010 Mar.
Article En | MEDLINE | ID: mdl-20065038

The catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs) plays a major role in the repair of DNA double-strand breaks (DSBs) by nonhomologous end joining (NHEJ). We have previously shown that DNA-PKcs is autophosphorylated in response to ionizing radiation (IR) and that dephosphorylation by a protein phosphatase 2A (PP2A)-like protein phosphatase (PP2A, PP4, or PP6) regulates the protein kinase activity of DNA-PKcs. Here we report that DNA-PKcs interacts with the catalytic subunits of PP6 (PP6c) and PP2A (PP2Ac), as well as with the PP6 regulatory subunits PP6R1, PP6R2, and PP6R3. Consistent with a role in the DNA damage response, silencing of PP6c by small interfering RNA (siRNA) induced sensitivity to IR and delayed release from the G(2)/M checkpoint. Furthermore, siRNA silencing of either PP6c or PP6R1 led to sustained phosphorylation of histone H2AX on serine 139 (gamma-H2AX) after IR. In contrast, silencing of PP6c did not affect the autophosphorylation of DNA-PKcs on serine 2056 or that of the ataxia-telangiectasia mutated (ATM) protein on serine 1981. We propose that a novel function of DNA-PKcs is to recruit PP6 to sites of DNA damage and that PP6 contributes to the dephosphorylation of gamma-H2AX, the dissolution of IR-induced foci, and release from the G(2)/M checkpoint in vivo.


Catalytic Domain , DNA-Activated Protein Kinase/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Extracts , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Checkpoint Kinase 2 , Chromosomal Proteins, Non-Histone/metabolism , Comet Assay , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , G2 Phase/radiation effects , Gene Silencing/radiation effects , HeLa Cells , Humans , Mitosis/radiation effects , Models, Biological , Phosphorylation/radiation effects , Protein Binding/radiation effects , Protein Serine-Threonine Kinases/metabolism , Radiation, Ionizing , Tumor Suppressor Proteins/metabolism
13.
J Biol Chem ; 285(2): 1414-23, 2010 Jan 08.
Article En | MEDLINE | ID: mdl-19893054

DNA double strand break (DSB) repair by non-homologous end joining (NHEJ) is initiated by DSB detection by Ku70/80 (Ku) and DNA-dependent protein kinase catalytic subunit (DNA-PKcs) recruitment, which promotes pathway progression through poorly defined mechanisms. Here, Ku and DNA-PKcs solution structures alone and in complex with DNA, defined by x-ray scattering, reveal major structural reorganizations that choreograph NHEJ initiation. The Ku80 C-terminal region forms a flexible arm that extends from the DNA-binding core to recruit and retain DNA-PKcs at DSBs. Furthermore, Ku- and DNA-promoted assembly of a DNA-PKcs dimer facilitates trans-autophosphorylation at the DSB. The resulting site-specific autophosphorylation induces a large conformational change that opens DNA-PKcs and promotes its release from DNA ends. These results show how protein and DNA interactions initiate large Ku and DNA-PKcs rearrangements to control DNA-PK biological functions as a macromolecular machine orchestrating assembly and disassembly of the initial NHEJ complex on DNA.


Antigens, Nuclear/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , DNA/metabolism , Antigens, Nuclear/chemistry , Antigens, Nuclear/genetics , DNA/chemistry , DNA/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Humans , Ku Autoantigen , Protein Binding/physiology , Protein Structure, Tertiary/physiology
14.
DNA Repair (Amst) ; 7(10): 1680-92, 2008 Oct 01.
Article En | MEDLINE | ID: mdl-18644470

Nonhomologous end joining (NHEJ) is the major pathway for the repair of DNA double strand breaks (DSBs) in human cells. NHEJ requires the catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs), Ku70, Ku80, XRCC4, DNA ligase IV and Artemis, as well as DNA polymerases mu and lambda and polynucleotide kinase. Recent studies have identified an additional participant, XLF, for XRCC4-like factor (also called Cernunnos), which interacts with the XRCC4-DNA ligase IV complex and stimulates its activity in vitro, however, its precise role in the DNA damage response is not fully understood. Since the protein kinase activity of DNA-PKcs is required for NHEJ, we asked whether XLF might be a physiological target of DNA-PK. Here, we have identified two major in vitro DNA-PK phosphorylation sites in the C-terminal region of XLF, serines 245 and 251. We show that these represent the major phosphorylation sites in XLF in vivo and that serine 245 is phosphorylated in vivo by DNA-PK, while serine 251 is phosphorylated by Ataxia-Telangiectasia Mutated (ATM). However, phosphorylation of XLF did not have a significant effect on the ability of XLF to interact with DNA in vitro or its recruitment to laser-induced DSBs in vivo. Similarly, XLF in which the identified in vivo phosphorylation sites were mutated to alanine was able to complement the DSB repair defect as well as radiation sensitivity in XLF-deficient 2BN cells. We conclude that phosphorylation of XLF at these sites does not play a major role in the repair of IR-induced DSBs in vivo.


Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair Enzymes/metabolism , DNA Repair , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Survival/radiation effects , DNA/metabolism , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/isolation & purification , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , HeLa Cells , Humans , Molecular Sequence Data , Mutant Proteins/metabolism , Phosphorylation/radiation effects , Phosphoserine/metabolism , Protein Binding/radiation effects , Protein Structure, Tertiary , Protein Transport/radiation effects , Radiation, Ionizing , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
15.
Mol Cell Biol ; 27(5): 1581-91, 2007 Mar.
Article En | MEDLINE | ID: mdl-17158925

The protein kinase activity of the DNA-dependent protein kinase (DNA-PK) is required for the repair of DNA double-strand breaks (DSBs) via the process of nonhomologous end joining (NHEJ). However, to date, the only target shown to be functionally relevant for the enzymatic role of DNA-PK in NHEJ is the large catalytic subunit DNA-PKcs itself. In vitro, autophosphorylation of DNA-PKcs induces kinase inactivation and dissociation of DNA-PKcs from the DNA end-binding component Ku70/Ku80. Phosphorylation within the two previously identified clusters of phosphorylation sites does not mediate inactivation of the assembled complex and only partially regulates kinase disassembly, suggesting that additional autophosphorylation sites may be important for DNA-PK function. Here, we show that DNA-PKcs contains a highly conserved amino acid (threonine 3950) in a region similar to the activation loop or t-loop found in the protein kinase domain of members of the typical eukaryotic protein kinase family. We demonstrate that threonine 3950 is an in vitro autophosphorylation site and that this residue, as well as other previously identified sites in the ABCDE cluster, is phosphorylated in vivo in irradiated cells. Moreover, we show that mutation of threonine 3950 to the phosphomimic aspartic acid abrogates V(D)J recombination and leads to radiation sensitivity. Together, these data suggest that threonine 3950 is a functionally important, DNA damage-inducible phosphorylation site and that phosphorylation of this site regulates the activity of DNA-PKcs.


DNA-Activated Protein Kinase/metabolism , Protein Kinases/chemistry , Threonine/metabolism , Amino Acid Motifs , Amino Acid Sequence , Catalytic Domain , Cell Line , Cell Line, Tumor , Conserved Sequence , DNA-Activated Protein Kinase/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Sequence Data , Okadaic Acid/pharmacology , Phosphorylation , Protein Structure, Secondary , Protein Structure, Tertiary , Radiation, Ionizing , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
16.
EMBO J ; 25(16): 3880-9, 2006 Aug 23.
Article En | MEDLINE | ID: mdl-16874298

The Artemis nuclease is defective in radiosensitive severe combined immunodeficiency patients and is required for the repair of a subset of ionising radiation induced DNA double-strand breaks (DSBs) in an ATM and DNA-PK dependent process. Here, we show that Artemis phosphorylation by ATM and DNA-PK in vitro is primarily attributable to S503, S516 and S645 and demonstrate ATM dependent phosphorylation at serine 645 in vivo. However, analysis of multisite phosphorylation mutants of Artemis demonstrates that Artemis phosphorylation is dispensable for endonuclease activity in vitro and for DSB repair and V(D)J recombination in vivo. Importantly, DNA-dependent protein kinase catalytic subunit (DNA-PKcs) autophosphorylation at the T2609-T2647 cluster, in the presence of Ku and target DNA, is required for Artemis-mediated endonuclease activity. Moreover, autophosphorylated DNA-PKcs stably associates with Ku-bound DNA with large single-stranded overhangs until overhang cleavage by Artemis. We propose that autophosphorylation triggers conformational changes in DNA-PK that enhance Artemis cleavage at single-strand to double-strand DNA junctions. These findings demonstrate that DNA-PK autophosphorylation regulates Artemis access to DNA ends, providing insight into the mechanism of Artemis mediated DNA end processing.


DNA Repair , DNA-Activated Protein Kinase/chemistry , Nuclear Proteins/chemistry , Catalytic Domain , DNA/chemistry , DNA Helicases/chemistry , DNA, Single-Stranded/chemistry , DNA-Binding Proteins , Endonucleases/chemistry , Endonucleases/genetics , Humans , Ku Autoantigen , Nuclear Proteins/genetics , Phosphorylation , Protein Conformation , Recombination, Genetic , Serine/chemistry
17.
Cancer Res ; 65(5): 1670-7, 2005 Mar 01.
Article En | MEDLINE | ID: mdl-15753361

Previous reports have suggested a connection between reduced levels of the catalytic subunit of DNA-dependent protein kinases (DNA-PKcs), a component of the nonhomologous DNA double-strand breaks end-joining system, and a reduction in ATM. We studied this possible connection in other DNA-PKcs-deficient cell types, and following knockdown of DNA-PKcs with small interfering RNA, Chinese hamster ovary V3 cells, lacking DNA-PKcs, had reduced levels of ATM and hSMG-1, but both were restored after transfection with PRKDC. Atm levels were also reduced in murine scid cells. Reduction of ATM in a human glioma cell line lacking DNA-PKcs was accompanied by defective signaling through downstream substrates, post-irradiation. A large reduction of DNA-PKcs was achieved in normal human fibroblasts after transfection with two DNA-PKcs small interfering RNA sequences. This was accompanied by a reduction in ATM. These data were confirmed using immunocytochemical detection of the proteins. Within hours after transfection, a decline in PRKDC mRNA was seen, followed by a more gradual decline in DNA-PKcs protein beginning 1 day after transfection. No change in ATM mRNA was observed for 2 days post-transfection. Only after the DNA-PKcs reduction occurred was a reduction in ATM mRNA observed, beginning 2 days post-transfection. The amount of ATM began to decline, starting about 3 days post-treatment, then it declined to levels comparable to DNA-PKcs. Both proteins returned to normal levels at later times. These data illustrate a potentially important cross-regulation between the nonhomologous end-joining system for rejoining of DNA double-strand breaks and the ATM-dependent damage response network of pathways, both of which operate to maintain the integrity of the genome.


Cell Cycle Proteins/metabolism , DNA-Binding Proteins , DNA-Binding Proteins/metabolism , DNA/genetics , Gene Expression Regulation , Protein Serine-Threonine Kinases , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Antigens, Surface , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia Mutated Proteins , CHO Cells , Catalytic Domain , Cell Cycle Proteins/genetics , Cricetinae , DNA/metabolism , DNA/radiation effects , DNA Damage/genetics , DNA Repair/genetics , DNA-Activated Protein Kinase , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Down-Regulation , Fibroblasts/metabolism , Fibroblasts/pathology , Glioma/metabolism , Glioma/pathology , Humans , Immunoenzyme Techniques , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Proteins , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Subunits , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Transfection , Tumor Suppressor Proteins/genetics
18.
EMBO J ; 23(22): 4451-61, 2004 Nov 10.
Article En | MEDLINE | ID: mdl-15510216

Ionizing radiation induces autophosphorylation of the ataxia-telangiectasia mutated (ATM) protein kinase on serine 1981; however, the precise mechanisms that regulate ATM activation are not fully understood. Here, we show that the protein phosphatase inhibitor okadaic acid (OA) induces autophosphorylation of ATM on serine 1981 in unirradiated cells at concentrations that inhibit protein phosphatase 2A-like activity in vitro. OA did not induce gamma-H2AX foci, suggesting that it induces ATM autophosphorylation by inactivation of a protein phosphatase rather than by inducing DNA double-strand breaks. In support of this, we show that ATM interacts with the scaffolding (A) subunit of protein phosphatase 2A (PP2A), that the scaffolding and catalytic (C) subunits of PP2A interact with ATM in undamaged cells and that immunoprecipitates of ATM from undamaged cells contain PP2A-like protein phosphatase activity. Moreover, we show that IR induces phosphorylation-dependent dissociation of PP2A from ATM and loss of the associated protein phosphatase activity. We propose that PP2A plays an important role in the regulation of ATM autophosphorylation and activity in vivo.


Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Phosphoprotein Phosphatases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia , Ataxia Telangiectasia Mutated Proteins , Catalytic Domain , Cell Cycle Proteins/chemistry , Cell Line , Comet Assay , DNA-Binding Proteins/chemistry , Enzyme Inhibitors/pharmacology , Fibroblasts/radiation effects , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Gene Deletion , Genes, Dominant , Green Fluorescent Proteins/metabolism , Humans , Hydrazines , Microscopy, Confocal , Models, Biological , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/drug effects , Phosphoprotein Phosphatases/genetics , Phosphorylation , Precipitin Tests , Protein Phosphatase 2 , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Radiation, Ionizing , Serine/chemistry , Signal Transduction , Tumor Suppressor Proteins/chemistry , Two-Hybrid System Techniques
19.
DNA Repair (Amst) ; 3(3): 235-44, 2004 Mar 04.
Article En | MEDLINE | ID: mdl-15177039

The ataxia-telangiectasia mutated (ATM) protein kinase is activated in response to ionizing radiation (IR) and activates downstream DNA-damage signaling pathways. Although the role of ATM in the cellular response to ionizing radiation has been well characterized, its role in response to other DNA-damaging agents is less well defined. We previously showed that genistein, a naturally occurring isoflavonoid, induced increased ATM protein kinase activity, ATM-dependent phosphorylation of p53 on serine 15 and activation of the DNA-binding properties of p53. Here, we show that genistein also induces phosphorylation of p53 at serines 6, 9, 20, 46, and 392, and that genistein-induced accumulation and phosphorylation of p53 is reduced in two ATM-deficient human cell lines. Also, we show that genistein induces phosphorylation of ATM on serine 1981 and phosphorylation of histone H2AX on serine 139. The related bioflavonoids, daidzein and biochanin A, did not induce either phosphorylation of p53 or ATM at these sites. Like genistein, quercetin induced phosphorylation of ATM on serine 1981, and ATM-dependent phosphorylation of histone H2AX on serine 139; however, p53 accumulation and phosphorylation on serines 6, 9, 15, 20, 46, and 392 occurred in ATM-deficient cells, indicating that ATM is not required for quercetin-induced phosphorylation of p53. Our data suggest that genistein and quercetin induce different DNA-damage induced signaling pathways that, in the case of genistein, are highly ATM-dependent but, in the case of quercetin, may be ATM-dependent only for some downstream targets.


DNA Damage/drug effects , Genistein/pharmacology , Quercetin/pharmacology , Signal Transduction/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Enzyme Activation/drug effects , Genistein/chemistry , Histones/metabolism , Humans , Isoflavones/chemistry , Isoflavones/pharmacology , Phosphorylation/drug effects , Precipitin Tests , Protein Serine-Threonine Kinases/metabolism , Quercetin/chemistry , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
20.
DNA Repair (Amst) ; 2(11): 1239-52, 2003 Nov 21.
Article En | MEDLINE | ID: mdl-14599745

Nonhomologous end joining (NHEJ) is a major pathway for the repair of DNA double-strand breaks (DSBs) in higher eukaryotes. Several proteins, including the DNA-dependent protein kinase (DNA-PK), XRCC4 and DNA ligase IV, are required for nonhomologous end joining both in vitro and in vivo. Since XRCC4 is recruited to the DNA double-strand break with DNA-PK, and because the protein kinase activity of DNA-PK is required for its in vivo function, we reasoned that XRCC4 could be a potential physiological substrate of DNA-PK. Here, we have used mass spectrometry to map the DNA-PK phosphorylation sites in XRCC4. Two major phosphorylation sites (serines 260 and 318), as well as several minor sites were identified. All of the identified sites lie within the carboxy-terminal 100 amino acids of XRCC4. Substitution of each of these sites to alanine (in combination) reduced the ability of DNA-PK to phosphorylate XRCC4 in vitro by at least two orders of magnitude. However, XRCC4-deficient cells that were complemented with XRCC4 lacking DNA-PK phosphorylation sites were analogous to wild type XRCC4 with respect to survival after ionizing radiation and ability to repair DSBs introduced during V(D)J recombination.


Cell Survival/radiation effects , DNA-Binding Proteins/chemistry , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , VDJ Recombinases/metabolism , Alanine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , DNA Damage , DNA-Activated Protein Kinase , Dose-Response Relationship, Radiation , HeLa Cells , Humans , Nuclear Proteins , Phosphorylation , Radiation, Ionizing
...