Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
EMBO J ; 41(12): e110632, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35578785

ABSTRACT

Topoisomerase II (TOP2) unlinks chromosomes during vertebrate DNA replication. TOP2 "poisons" are widely used chemotherapeutics that stabilize TOP2 complexes on DNA, leading to cytotoxic DNA breaks. However, it is unclear how these drugs affect DNA replication, which is a major target of TOP2 poisons. Using Xenopus egg extracts, we show that the TOP2 poisons etoposide and doxorubicin both inhibit DNA replication through different mechanisms. Etoposide induces TOP2-dependent DNA breaks and TOP2-dependent fork stalling by trapping TOP2 behind replication forks. In contrast, doxorubicin does not lead to appreciable break formation and instead intercalates into parental DNA to stall replication forks independently of TOP2. In human cells, etoposide stalls forks in a TOP2-dependent manner, while doxorubicin stalls forks independently of TOP2. However, both drugs exhibit TOP2-dependent cytotoxicity. Thus, etoposide and doxorubicin inhibit DNA replication through distinct mechanisms despite shared genetic requirements for cytotoxicity.


Subject(s)
DNA Topoisomerases, Type II , Poisons , Animals , DNA , DNA Replication , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Doxorubicin/pharmacology , Etoposide/pharmacology , Humans , Vertebrates/genetics , Vertebrates/metabolism
2.
Sci Adv ; 8(13): eabm0314, 2022 04.
Article in English | MEDLINE | ID: mdl-35353580

ABSTRACT

Replication-coupled DNA repair and damage tolerance mechanisms overcome replication stress challenges and complete DNA synthesis. These pathways include fork reversal, translesion synthesis, and repriming by specialized polymerases such as PRIMPOL. Here, we investigated how these pathways are used and regulated in response to varying replication stresses. Blocking lagging-strand priming using a POLα inhibitor slows both leading- and lagging-strand synthesis due in part to RAD51-, HLTF-, and ZRANB3-mediated, but SMARCAL1-independent, fork reversal. ATR is activated, but CHK1 signaling is dampened compared to stalling both the leading and lagging strands with hydroxyurea. Increasing CHK1 activation by overexpressing CLASPIN in POLα-inhibited cells promotes replication elongation through PRIMPOL-dependent repriming. CHK1 phosphorylates PRIMPOL to promote repriming irrespective of the type of replication stress, and this phosphorylation is important for cellular resistance to DNA damage. However, PRIMPOL activation comes at the expense of single-strand gap formation, and constitutive PRIMPOL activity results in reduced cell fitness.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase , DNA Damage , DNA Repair , DNA-Directed DNA Polymerase/genetics , Phosphorylation
3.
Cell Rep ; 31(9): 107705, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492421

ABSTRACT

5-Hydroxymethylcytosine (5hmC) binding, ES-cell-specific (HMCES) crosslinks to apurinic or apyrimidinic (AP, abasic) sites in single-strand DNA (ssDNA). To determine whether HMCES responds to the ssDNA abasic site in cells, we exploited the activity of apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3A (APOBEC3A). APOBEC3A preferentially deaminates cytosines to uracils in ssDNA, which are then converted to abasic sites by uracil DNA glycosylase. We find that HMCES-deficient cells are hypersensitive to nuclear APOBEC3A localization. HMCES relocalizes to chromatin in response to nuclear APOBEC3A and protects abasic sites from processing into double-strand breaks (DSBs). Abasic sites induced by APOBEC3A slow both leading and lagging strand synthesis, and HMCES prevents further slowing of the replication fork by translesion synthesis (TLS) polymerases zeta (Polζ) and kappa (Polκ). Thus, our study provides direct evidence that HMCES responds to ssDNA abasic sites in cells to prevent DNA cleavage and balance the engagement of TLS polymerases.


Subject(s)
Cytidine Deaminase/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , Cytidine Deaminase/genetics , DNA Replication , DNA, Single-Stranded/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Deamination , Endonucleases/antagonists & inhibitors , Endonucleases/genetics , Endonucleases/metabolism , Humans , Multifunctional Enzymes/antagonists & inhibitors , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Uracil/metabolism , Uracil-DNA Glycosidase/metabolism
4.
Cell ; 176(1-2): 144-153.e13, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30554877

ABSTRACT

Abasic sites are one of the most common DNA lesions. All known abasic site repair mechanisms operate only when the damage is in double-stranded DNA. Here, we report the discovery of 5-hydroxymethylcytosine (5hmC) binding, ESC-specific (HMCES) as a sensor of abasic sites in single-stranded DNA. HMCES acts at replication forks, binds PCNA and single-stranded DNA, and generates a DNA-protein crosslink to shield abasic sites from error-prone processing. This unusual HMCES DNA-protein crosslink intermediate is resolved by proteasome-mediated degradation. Acting as a suicide enzyme, HMCES prevents translesion DNA synthesis and the action of endonucleases that would otherwise generate mutations and double-strand breaks. HMCES is evolutionarily conserved in all domains of life, and its biochemical properties are shared with its E. coli ortholog. Thus, HMCES is an ancient DNA lesion recognition protein that preserves genome integrity by promoting error-free repair of abasic sites in single-stranded DNA.


Subject(s)
5-Methylcytosine/analogs & derivatives , DNA Repair/physiology , DNA, Single-Stranded/physiology , 5-Methylcytosine/metabolism , Apurinic Acid/metabolism , DNA/metabolism , DNA Damage/physiology , DNA Replication/physiology , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases , Escherichia coli/metabolism , Polynucleotides/metabolism , Proliferating Cell Nuclear Antigen/metabolism
5.
Mol Cell ; 67(3): 374-386.e5, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28735897

ABSTRACT

RAD51 promotes homology-directed repair (HDR), replication fork reversal, and stalled fork protection. Defects in these functions cause genomic instability and tumorigenesis but also generate hypersensitivity to cancer therapeutics. Here we describe the identification of RADX as an RPA-like, single-strand DNA binding protein. RADX is recruited to replication forks, where it prevents fork collapse by regulating RAD51. When RADX is inactivated, excessive RAD51 activity slows replication elongation and causes double-strand breaks. In cancer cells lacking BRCA2, RADX deletion restores fork protection without restoring HDR. Furthermore, RADX inactivation confers chemotherapy and PARP inhibitor resistance to cancer cells with reduced BRCA2/RAD51 pathway function. By antagonizing RAD51 at forks, RADX allows cells to maintain a high capacity for HDR while ensuring that replication functions of RAD51 are properly regulated. Thus, RADX is essential to achieve the proper balance of RAD51 activity to maintain genome stability.


Subject(s)
DNA, Neoplasm/biosynthesis , Drug Resistance, Neoplasm , Genomic Instability , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rad51 Recombinase/metabolism , Replication Origin , A549 Cells , Animals , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , CRISPR-Cas Systems , DNA Breaks, Double-Stranded , DNA Repair , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Models, Molecular , Mutation , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , RNA Interference , Rad51 Recombinase/genetics , Transfection
6.
Proc Natl Acad Sci U S A ; 112(48): 14864-9, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26578802

ABSTRACT

The SMARCAL1 (SWI/SNF related, matrix-associated, actin-dependent, regulator of chromatin, subfamily A-like 1) DNA translocase is one of several related enzymes, including ZRANB3 (zinc finger, RAN-binding domain containing 3) and HLTF (helicase-like transcription factor), that are recruited to stalled replication forks to promote repair and restart replication. These enzymes can perform similar biochemical reactions such as fork reversal; however, genetic studies indicate they must have unique cellular activities. Here, we present data showing that SMARCAL1 has an important function at telomeres, which present an endogenous source of replication stress. SMARCAL1-deficient cells accumulate telomere-associated DNA damage and have greatly elevated levels of extrachromosomal telomere DNA (C-circles). Although these telomere phenotypes are often found in tumor cells using the alternative lengthening of telomeres (ALT) pathway for telomere elongation, SMARCAL1 deficiency does not yield other ALT phenotypes such as elevated telomere recombination. The activity of SMARCAL1 at telomeres can be separated from its genome-maintenance activity in bulk chromosomal replication because it does not require interaction with replication protein A. Finally, this telomere-maintenance function is not shared by ZRANB3 or HLTF. Our results provide the first identification, to our knowledge, of an endogenous source of replication stress that requires SMARCAL1 for resolution and define differences between members of this class of replication fork-repair enzymes.


Subject(s)
Chromosomes, Human/metabolism , DNA Helicases/metabolism , DNA Replication/physiology , Telomere Homeostasis/physiology , Telomere/metabolism , Animals , Chromosomes, Human/genetics , DNA Damage/physiology , DNA Helicases/genetics , HeLa Cells , Humans , Mice , Recombination, Genetic/physiology , Telomere/genetics
7.
Mol Cell Biol ; 35(17): 2979-90, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26100022

ABSTRACT

Accurate replication of DNA is imperative for the maintenance of genomic integrity. We identified Enhancer of Rudimentary Homolog (ERH) using a whole-genome RNA interference (RNAi) screen to discover novel proteins that function in the replication stress response. Here we report that ERH is important for DNA replication and recovery from replication stress. ATR pathway activity is diminished in ERH-deficient cells. The reduction in ATR signaling corresponds to a decrease in the expression of multiple ATR pathway genes, including ATR itself. ERH interacts with multiple RNA processing complexes, including splicing regulators. Furthermore, splicing of ATR transcripts is deficient in ERH-depleted cells. Transcriptome-wide analysis indicates that ERH depletion affects the levels of ∼1,500 transcripts, with DNA replication and repair genes being highly enriched among those with reduced expression. Splicing defects were evident in ∼750 protein-coding genes, which again were enriched for DNA metabolism genes. Thus, ERH regulation of RNA processing is needed to ensure faithful DNA replication and repair.


Subject(s)
Cell Cycle Proteins/genetics , DNA Repair/genetics , DNA Replication/genetics , RNA Splicing/genetics , Stress, Physiological/genetics , Transcription Factors/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Base Sequence , Cell Line , DNA Damage/genetics , Gene Expression Profiling , HEK293 Cells , Humans , RNA Interference , RNA, Small Interfering , Regulatory Sequences, Nucleic Acid/genetics , Sequence Analysis, RNA , Signal Transduction/genetics
8.
PLoS One ; 9(6): e99397, 2014.
Article in English | MEDLINE | ID: mdl-24901225

ABSTRACT

Subcellular localization, protein interactions, and post-translational modifications regulate the DNA damage response kinases ATR, ATM, and DNA-PK. During an analysis of putative ATR phosphorylation sites, we found that a single mutation at S1333 creates a hyperactive kinase. In vitro and in cells, mutation of S1333 to alanine (S1333A-ATR) causes elevated levels of kinase activity with and without the addition of the protein activator TOPBP1. S1333 mutations to glycine, arginine, or lysine also create a hyperactive kinase, while mutation to aspartic acid decreases ATR activity. S1333A-ATR maintains the G2 checkpoint and promotes completion of DNA replication after transient exposure to replication stress but the less active kinase, S1333D-ATR, has modest defects in both of these functions. While we find no evidence that S1333 is phosphorylated in cultured cells, our data indicate that small changes in the HEAT repeats can have large effects on kinase activity. These mutants may serve as useful tools for future studies of the ATR pathway.


Subject(s)
Serine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins/metabolism , Checkpoint Kinase 1 , DNA Replication/drug effects , DNA Replication/radiation effects , DNA-Binding Proteins/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/radiation effects , HCT116 Cells , HEK293 Cells , Humans , Hydroxyurea/pharmacology , Molecular Sequence Data , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , Protein Binding , Protein Kinases/metabolism , Protein Structure, Secondary , Radiation, Ionizing , Signal Transduction/drug effects , Signal Transduction/radiation effects , Ultraviolet Rays
9.
Nucleic Acids Res ; 42(2): 918-25, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24150942

ABSTRACT

SMARCAL1 promotes the repair and restart of damaged replication forks. Either overexpression or silencing SMARCAL1 causes the accumulation of replication-associated DNA damage. SMARCAL1 is heavily phosphorylated. Here we identify multiple phosphorylation sites, including S889, which is phosphorylated even in undamaged cells. S889 is highly conserved through evolution and it regulates SMARCAL1 activity. Specifically, S889 phosphorylation increases the DNA-stimulated ATPase activity of SMARCAL1 and increases its ability to catalyze replication fork regression. A phosphomimetic S889 mutant is also hyperactive when expressed in cells, while a non-phosphorylatable mutant is less active. S889 lies within a C-terminal region of the SMARCAL1 protein. Deletion of the C-terminal region also creates a hyperactive SMARCAL1 protein suggesting that S889 phosphorylation relieves an auto-inhibitory function of this SMARCAL1 domain. Thus, S889 phosphorylation is one mechanism by which SMARCAL1 activity is regulated to ensure the proper level of fork remodeling needed to maintain genome integrity during DNA synthesis.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/metabolism , Amino Acid Sequence , Cell Line , DNA Helicases/genetics , HEK293 Cells , Humans , Molecular Sequence Data , Mutation , Phosphorylation , Protein Structure, Tertiary
10.
PLoS One ; 8(5): e63149, 2013.
Article in English | MEDLINE | ID: mdl-23671665

ABSTRACT

SMARCAL1 is an ATPase in the SNF2 family that functions at damaged replication forks to promote their stability and restart. It acts by translocating on DNA to catalyze DNA strand annealing, branch migration, and fork regression. Many SNF2 enzymes work as motor subunits of large protein complexes. To determine if SMARCAL1 is also a member of a protein complex and to further understand how it functions in the replication stress response, we used a proteomics approach to identify interacting proteins. In addition to the previously characterized interaction with replication protein A (RPA), we found that SMARCAL1 forms complexes with several additional proteins including DNA-PKcs and the WRN helicase. SMARCAL1 and WRN co-localize at stalled replication forks independently of one another. The SMARCAL1 interaction with WRN is indirect and is mediated by RPA acting as a scaffold. SMARCAL1 and WRN act independently to prevent MUS81 cleavage of the stalled fork. Biochemical experiments indicate that both catalyze fork regression with SMARCAL1 acting more efficiently and independently of WRN. These data suggest that RPA brings a complex of SMARCAL1 and WRN to stalled forks, but that they may act in different pathways to promote fork repair and restart.


Subject(s)
DNA Helicases/metabolism , DNA Replication , Exodeoxyribonucleases/metabolism , RecQ Helicases/metabolism , Replication Protein A/metabolism , Cell Line, Tumor , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Helicases/genetics , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Exodeoxyribonucleases/genetics , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Mass Spectrometry , Nucleic Acid Conformation , Protein Binding , Proteomics/methods , RecQ Helicases/genetics , Replication Protein A/genetics , S Phase/genetics , Werner Syndrome Helicase
11.
J Biol Chem ; 286(43): 37320-7, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21908846

ABSTRACT

ATR (ataxia telangiectasia-mutated and Rad3-related) contains 16 conserved candidate autophosphorylation sites that match its preferred S/TQ consensus. To determine whether any is functionally important, we mutated the 16 candidate residues to alanine in a single cDNA to create a 16A-ATR mutant. The 16A-ATR mutant maintains kinase and G(2) checkpoint activities. However, it fails to rescue the essential function of ATR in maintaining cell viability and fails to promote replication recovery from a transient exposure to replication stress. Further analysis identified T1566A/T1578A/T1589A (3A-ATR) as critical mutations causing this separation of function activity. Secondary structure predictions indicate that these residues occur in a region between ATR HEAT repeats 31R and 32R that aligns with regions of ATM and DNA-PK containing regulatory autophosphorylation sites. Although this region is important for ATR function, the 3A-ATR residues do not appear to be sites of autophosphorylation. Nevertheless, our analysis identifies an important regulatory region of ATR that is shared among the PI3K-related protein kinase family. Furthermore, our data indicate that the essential function of ATR for cell viability is linked to its function in promoting proper replication in the context of replication stress and is independent of G(2) checkpoint activity.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , G2 Phase/physiology , Mutation, Missense , Protein Serine-Threonine Kinases/metabolism , S Phase/physiology , Tumor Suppressor Proteins/metabolism , Amino Acid Substitution , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line , Cell Survival/physiology , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/genetics , Humans , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Structure, Secondary , Tumor Suppressor Proteins/genetics
12.
J Biol Chem ; 286(33): 28707-28714, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21705319

ABSTRACT

The DNA damage response kinases ataxia telangiectasia-mutated (ATM), DNA-dependent protein kinase (DNA-PK), and ataxia telangiectasia-mutated and Rad3-related (ATR) signal through multiple pathways to promote genome maintenance. These related kinases share similar methods of regulation, including recruitment to specific nucleic acid structures and association with protein activators. ATM and DNA-PK also are regulated via phosphorylation, which provides a convenient biomarker for their activity. Whether phosphorylation regulates ATR is unknown. Here we identify ATR Thr-1989 as a DNA damage-regulated phosphorylation site. Selective inhibition of ATR prevents Thr-1989 phosphorylation, and phosphorylation requires ATR activation. Cells engineered to express only a non-phosphorylatable T1989A mutant exhibit a modest ATR functional defect. Our results suggest that, like ATM and DNA-PK, phosphorylation regulates ATR, and phospho-peptide specific antibodies to Thr-1989 provide a proximal marker of ATR activation.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Threonine/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , DNA Damage/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation/physiology , Humans , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Threonine/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
EMBO Rep ; 11(11): 876-82, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20930849

ABSTRACT

Cyclin-dependent kinase 9 (CDK9) is a well-characterized subunit of the positive transcription elongation factor b complex in which it regulates transcription elongation in cooperation with cyclin T. However, CDK9 also forms a complex with cyclin K, the function of which is less clear. Using a synthetic lethal RNA interference screen in human cells, we identified CDK9 as a component of the replication stress response. Loss of CDK9 activity causes an increase in spontaneous levels of DNA damage signalling in replicating cells and a decreased ability to recover from a transient replication arrest. This activity is restricted to CDK9-cyclin K complexes and is independent of CDK9-cyclin T complex. CDK9 accumulates on chromatin in response to replication stress and limits the amount of single-stranded DNA in cells under stress. Furthermore, we show that CDK9 and cyclin K interact with ataxia telangiectasia and Rad3-related protein and other checkpoint signalling proteins. These results reveal an unexpectedly direct role for CDK9-cyclin K in checkpoint pathways that maintain genome integrity in response to replication stress.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Cyclins/metabolism , DNA Replication , Stress, Physiological , Cell Cycle/drug effects , Cell Line, Tumor , Chromatin/metabolism , DNA/biosynthesis , DNA Replication/drug effects , Gene Silencing/drug effects , Humans , Hydroxyurea/pharmacology , Protein Binding/drug effects , Replication Protein A/metabolism , Stress, Physiological/drug effects
14.
Proc Natl Acad Sci U S A ; 106(46): 19304-9, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19889979

ABSTRACT

The DNA damage response (DDR) has a critical role in maintaining genome integrity and serves as a barrier to tumorigenesis by promoting cell-cycle arrest, DNA repair, and apoptosis. The DDR is activated not only by genotoxic agents that induce DNA damage, but also during aberrant cell-division cycles caused by activated oncogenes and inactivated tumor suppressors. Here we use RNAi and cDNA overexpression screens in human cells to identify genes that, when deregulated, lead to activation of the DDR. The RNAi screen identified 73 genes that, when silenced in at least two cell types, cause DDR activation. Silencing several of these genes also caused an increased frequency of micronuclei, a marker of genetically unstable cells. The cDNA screen identified 97 genes that when overexpressed induce DDR activation in the absence of any exogenous genotoxic agent, with an overrepresentation of genes linked to cancer. Secondary RNAi screens identified CDK2-interacting protein (CINP) as a cell-cycle checkpoint protein. CINP interacts with ATR-interacting protein and regulates ATR-dependent signaling, resistance to replication stress, and G2 checkpoint integrity.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Genome, Human , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/genetics , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Genomics , HeLa Cells , Humans , Two-Hybrid System Techniques
15.
Genes Dev ; 22(11): 1478-89, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519640

ABSTRACT

The ATR (ATM and Rad3-related) kinase and its regulatory partner ATRIP (ATR-interacting protein) coordinate checkpoint responses to DNA damage and replication stress. TopBP1 functions as a general activator of ATR. However, the mechanism by which TopBP1 activates ATR is unknown. Here, we show that ATRIP contains a TopBP1-interacting region that is necessary for the association of TopBP1 and ATR, for TopBP1-mediated activation of ATR, and for cells to survive and recover DNA synthesis following replication stress. We demonstrate that this region is functionally conserved in the Saccharomyces cerevisiae ATRIP ortholog Ddc2, suggesting a conserved mechanism of regulation. In addition, we identify a domain of ATR that is critical for its activation by TopBP1. Mutations of the ATR PRD (PIKK [phosphoinositide 3-kinase related kinase] Regulatory Domain) do not affect the basal kinase activity of ATR but prevent its activation. Cellular complementation experiments demonstrate that TopBP1-mediated ATR activation is required for checkpoint signaling and cellular viability. The PRDs of ATM and mTOR (mammalian target of rapamycin) were shown previously to regulate the activities of these kinases, and our data indicate that the DNA-PKcs (DNA-dependent protein kinase catalytic subunit) PRD is important for DNA-PKcs regulation. Therefore, divergent amino acid sequences within the PRD and a unique protein partner allow each of these PIK kinases to respond to distinct cellular events.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Amino Acid Motifs , Binding Sites , Cell Line , Cell Survival , Enzyme Activation , Humans , Protein Binding , Regulatory Sequences, Nucleic Acid , Saccharomyces cerevisiae , Signal Transduction , Transfection
16.
Cancer Res ; 67(14): 6685-90, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17638878

ABSTRACT

The ATR-ATRIP kinase complex regulates cellular responses to DNA damage and replication stress. Mass spectrometry was used to identify phosphorylation sites on ATR and ATRIP to understand how the kinase complex is regulated by post-translational modifications. Two novel phosphorylation sites on ATRIP were identified, S224 and S239. Phosphopeptide-specific antibodies to S224 indicate that it is phosphorylated in a cell cycle-dependent manner. S224 matches a consensus site for cyclin-dependent kinase (CDK) phosphorylation and is phosphorylated by CDK2-cyclin A in vitro. S224 phosphorylation in cells is sensitive to CDK2 inhibitors. Mutation of S224 to alanine causes a defect in the ATR-ATRIP-dependent maintenance of the G(2)-M checkpoint to ionizing and UV radiation. Thus, ATRIP is a CDK2 substrate, and CDK2-dependent phosphorylation of S224 regulates the ability of ATR-ATRIP to promote cell cycle arrest in response to DNA damage.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cyclin-Dependent Kinase 2/physiology , Exodeoxyribonucleases/biosynthesis , Gene Expression Regulation, Neoplastic , Phosphoproteins/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle , Cyclin-Dependent Kinase 2/metabolism , DNA Damage , DNA-Binding Proteins , HeLa Cells , Humans , Molecular Sequence Data , Phosphopeptides/chemistry , Phosphorylation , Protein Processing, Post-Translational , Sequence Homology, Amino Acid
17.
Exp Cell Res ; 313(8): 1667-74, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17376433

ABSTRACT

The ATM and ATR kinases function at the apex of checkpoint signaling pathways. These kinases share significant sequence similarity, phosphorylate many of the same substrates, and have overlapping roles in initiating cell cycle checkpoints. However, they sense DNA damage through distinct mechanisms. ATR primarily senses single stranded DNA (ssDNA) through its interaction with ATRIP, and ATM senses double strand breaks through its interaction with Nbs1. We determined that the N-terminus of ATR contains a domain that binds ATRIP. Attaching this domain to ATM allowed the fusion protein (ATM*) to bind ATRIP and associate with RPA-coated ssDNA. ATM* also gained the ability to localize efficiently to stalled replication forks as well as double strand breaks. Despite having normal kinase activity when tested in vitro and being phosphorylated on S1981 in vivo, ATM* is defective in checkpoint signaling and does not complement cellular deficiencies in either ATM or ATR. These data indicate that the N-terminus of ATR is sufficient to bind ATRIP and to promote localization to sites of replication stress.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line , DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , Exodeoxyribonucleases/metabolism , Humans , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Replication Origin , Replication Protein A/metabolism , Tumor Suppressor Proteins/genetics
18.
J Biol Chem ; 280(24): 22788-92, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15863514

ABSTRACT

Polycythemia vera (PV) is a human clonal hematological disorder. The molecular etiology of the disease has not been identified. PV hematopoietic progenitor cells exhibit hypersensitivity to growth factors and cytokines, suggesting possible abnormalities in protein-tyrosine kinases and phosphatases. By sequencing the entire coding regions of cDNAs of candidate enzymes, we identified a G:C--> T:A point mutation of the JAK2 tyrosine kinase in 20 of 24 PV blood samples but none in 12 normal samples. The mutation has varying degrees of heterozygosity and is apparently acquired. It changes conserved Val(617) to Phe in the pseudokinase domain of JAK2 that is known to have an inhibitory role. The mutant JAK2 has enhanced kinase activity, and when overexpressed together with the erythropoietin receptor in cells, it caused hyperactivation of erythropoietin-induced cell signaling. This gain-of-function mutation of JAK may explain the hypersensitivity of PV progenitor cells to growth factors and cytokines. Our study thus defines a molecular defect of PV.


Subject(s)
Mutation , Polycythemia Vera/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cytokines/metabolism , DNA Mutational Analysis , DNA, Complementary/metabolism , Erythropoietin/metabolism , HeLa Cells , Heterozygote , Humans , Immunoblotting , Janus Kinase 2 , Molecular Sequence Data , Phenylalanine/chemistry , Point Mutation , Polymerase Chain Reaction , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Signal Transduction , Transfection , Tyrosine/chemistry , Valine/chemistry
19.
Proc Natl Acad Sci U S A ; 101(8): 2311-6, 2004 Feb 24.
Article in English | MEDLINE | ID: mdl-14983006

ABSTRACT

Mutations of the polycystic kidney and hepatic disease 1 (PKHD1) gene have been shown to cause autosomal recessive polycystic kidney disease (ARPKD), but the cellular functions of the gene product (PKHD1) remain uncharacterized. To illuminate its properties, the spatial and temporal expression patterns of PKHD1 were determined in mouse, rat, and human tissues by using polyclonal Abs and mAbs recognizing various specific regions of the gene product. During embryogenesis, PKHD1 is widely expressed in epithelial derivatives, including neural tubules, gut, pulmonary bronchi, and hepatic cells. In the kidneys of the pck rats, the rat model of which is genetically homologous to human ARPKD, the level of PKHD1 was significantly reduced but not completely absent. In cultured renal cells, the PKHD1 gene product colocalized with polycystin-2, the gene product of autosomal dominant polycystic disease type 2, at the basal bodies of primary cilia. Immunoreactive PKHD1 localized predominantly at the apical domain of polarized epithelial cells, suggesting it may be involved in the tubulogenesis and/or maintenance of duct-lumen architecture. Reduced PKHD1 levels in pck rat kidneys and its colocalization with polycystins may underlie the pathogenic basis for cystogenesis in polycystic kidney diseases.


Subject(s)
Kidney/enzymology , Receptors, Cell Surface/genetics , Adult , Animals , Base Sequence , Cell Line , Cell Line, Tumor , Cloning, Molecular , DNA Primers , Fetus , Humans , Immunohistochemistry , Mice , Polycystic Kidney Diseases/enzymology , Polycystic Kidney Diseases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription, Genetic
20.
J Biol Chem ; 278(44): 42893-8, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12913007

ABSTRACT

SHP-2 is an Src homology 2 (SH2) domain-containing tyrosine phosphatase with crucial functions in cell signaling and major pathological implications. It stays inactive in the cytosol and is activated by binding through its SH2 domains to tyrosine-phosphorylated receptors on the cell surface. One such cell surface protein is PZR, which contains two tyrosine-based inhibition motifs responsible for binding of SHP-2. We have generated a glutathione S-transferase fusion protein carrying the tandem tyrosine-based inhibition motifs of PZR, and the protein was tyrosine-phosphorylated by co-expressing c-Src in Escherichia coli cells. The purified phosphoprotein displays a strong binding to SHP-2 and causes its activation in vitro. However, when introduced into NIH 3T3 cells by using a protein delivery reagent, it effectively inhibited the activation of ERK1/2 induced by growth factors and serum but not by phorbol ester, in reminiscence of the effects caused by expression of dominant negative SHP-2 mutants and deletion of functional SHP-2. The data suggest that the exogenously introduced PZR protein specifically binds SHP-2, blocks its translocation, and renders it functionally incompetent. This is further supported by the fact that the phosphorylated PZR protein had no inhibitory effects on fibroblasts derived from mice expressing only a mutant SHP-2 protein lacking most of the N-terminal SH2 domain. This study thus provides a novel and highly specific method to interrupt the function of SHP-2 in cells.


Subject(s)
Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/physiology , Amino Acid Motifs , Animals , Blotting, Western , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Escherichia coli/metabolism , Glutathione Transferase/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Mutation , NIH 3T3 Cells , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Recombinant Fusion Proteins/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Time Factors , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...