Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 288(44): 31458-67, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24047897

ABSTRACT

Both DNA and chromatin need to be duplicated during each cell division cycle. Replication happens in the context of defects in the DNA template and other forms of replication stress that present challenges to both genetic and epigenetic inheritance. The replication machinery is highly regulated by replication stress responses to accomplish this goal. To identify important replication and stress response proteins, we combined isolation of proteins on nascent DNA (iPOND) with quantitative mass spectrometry. We identified 290 proteins enriched on newly replicated DNA at active, stalled, and collapsed replication forks. Approximately 16% of these proteins are known replication or DNA damage response proteins. Genetic analysis indicates that several of the newly identified proteins are needed to facilitate DNA replication, especially under stressed conditions. Our data provide a useful resource for investigators studying DNA replication and the replication stress response and validate the use of iPOND combined with mass spectrometry as a discovery tool.


Subject(s)
DNA Damage , DNA Replication , DNA-Binding Proteins/chemistry , DNA/metabolism , Mass Spectrometry/methods , DNA/biosynthesis , DNA-Binding Proteins/metabolism , Humans
2.
Cold Spring Harb Perspect Biol ; 5(8): a012724, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23813586

ABSTRACT

Genome integrity is challenged by DNA damage from both endogenous and environmental sources. This damage must be repaired to allow both RNA and DNA polymerases to accurately read and duplicate the information in the genome. Multiple repair enzymes scan the DNA for problems, remove the offending damage, and restore the DNA duplex. These repair mechanisms are regulated by DNA damage response kinases including DNA-PKcs, ATM, and ATR that are activated at DNA lesions. These kinases improve the efficiency of DNA repair by phosphorylating repair proteins to modify their activities, by initiating a complex series of changes in the local chromatin structure near the damage site, and by altering the overall cellular environment to make it more conducive to repair. In this review, we focus on these three levels of regulation to illustrate how the DNA damage kinases promote efficient repair to maintain genome integrity and prevent disease.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Chromatin/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA Repair/physiology , DNA-Activated Protein Kinase/metabolism , Genomic Instability/physiology , Models, Genetic , Phosphorylation
3.
PLoS One ; 8(7): e68915, 2013.
Article in English | MEDLINE | ID: mdl-23894374

ABSTRACT

Given the fundamental roles of histone deacetylases (HDACs) in the regulation of DNA repair, replication, transcription and chromatin structure, it is fitting that therapies targeting HDAC activities are now being explored as anti-cancer agents. In fact, two histone deacetylase inhibitors (HDIs), SAHA and Depsipeptide, are FDA approved for single-agent treatment of refractory cutaneous T cell lymphoma (CTCL). An important target of these HDIs, histone deacetylase 3 (HDAC3), regulates processes such as DNA repair, metabolism, and tumorigenesis through the regulation of chromatin structure and gene expression. Here we show that HDAC3 inhibition using a first in class selective inhibitor, RGFP966, resulted in decreased cell growth in CTCL cell lines due to increased apoptosis that was associated with DNA damage and impaired S phase progression. Through isolation of proteins on nascent DNA (iPOND), we found that HDAC3 was associated with chromatin and is present at and around DNA replication forks. DNA fiber labeling analysis showed that inhibition of HDAC3 resulted in a significant reduction in DNA replication fork velocity within the first hour of drug treatment. These results suggest that selective inhibition of HDAC3 could be useful in treatment of CTCL by disrupting DNA replication of the rapidly cycling tumor cells, ultimately leading to cell death.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Replication/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Lymphoma, T-Cell, Cutaneous/genetics , Lymphoma, T-Cell, Cutaneous/pathology , Stress, Physiological/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/drug effects , Chromatin/genetics , DNA Damage , Drug Synergism , Humans , S Phase/drug effects , Stress, Physiological/drug effects
4.
PLoS Genet ; 9(6): e1003518, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23754951

ABSTRACT

Histone acetyltransferase 1 is an evolutionarily conserved type B histone acetyltransferase that is thought to be responsible for the diacetylation of newly synthesized histone H4 on lysines 5 and 12 during chromatin assembly. To understand the function of this enzyme in a complex organism, we have constructed a conditional mouse knockout model of Hat1. Murine Hat1 is essential for viability, as homozygous deletion of Hat1 results in neonatal lethality. The lungs of embryos and pups genetically deficient in Hat1 were much less mature upon histological evaluation. The neonatal lethality is due to severe defects in lung development that result in less aeration and respiratory distress. Many of the Hat1(-/-) neonates also display significant craniofacial defects with abnormalities in the bones of the skull and jaw. Hat1(-/-) mouse embryonic fibroblasts (MEFs) are defective in cell proliferation and are sensitive to DNA damaging agents. In addition, the Hat1(-/-) MEFs display a marked increase in genome instability. Analysis of histone dynamics at sites of replication-coupled chromatin assembly demonstrates that Hat1 is not only responsible for the acetylation of newly synthesized histone H4 but is also required to maintain the acetylation of histone H3 on lysines 9, 18, and 27 during replication-coupled chromatin assembly.


Subject(s)
Embryonic Development/genetics , Genomic Instability , Histone Acetyltransferases/genetics , Histones/genetics , Acetylation , Animals , Cell Proliferation , Cell Survival/genetics , Chromatin Assembly and Disassembly/genetics , DNA Replication/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, Knockout
5.
Nat Protoc ; 7(3): 594-605, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22383038

ABSTRACT

Understanding the processes of DNA replication, chromatin assembly and maturation, and the replication stress response requires the ability to monitor protein dynamics at active and damaged replication forks. Detecting protein accumulation at replication forks or damaged sites has primarily relied on immunofluorescence imaging, which is limited in resolution and antibody sensitivity. Here we describe a procedure to isolate proteins on nascent DNA (iPOND) that permits a high-resolution spatiotemporal analysis of proteins at replication forks or on chromatin following DNA replication in cultured cells. iPOND relies on labeling of nascent DNA with the nucleoside analog 5-ethynyl-2'-deoxyuridine (EdU). Biotin conjugation to EdU-labeled DNA using click chemistry facilitates a single-step streptavidin purification of proteins bound to the nascent DNA. iPOND permits an interrogation of any cellular process linked to DNA synthesis using a 3- to 4-d protocol.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Replication/genetics , DNA/metabolism , Proteins/isolation & purification , Biotin/chemistry , Cells, Cultured , DNA/chemistry , Deoxyuridine/analogs & derivatives , Deoxyuridine/chemistry , Molecular Structure , Proteins/chemistry , Proteins/metabolism
6.
Genes Dev ; 26(2): 151-62, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22279047

ABSTRACT

SMARCAL1 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily A-like1) maintains genome integrity during DNA replication. Here we investigated its mechanism of action. We found that SMARCAL1 travels with elongating replication forks, and its absence leads to MUS81-dependent double-strand break formation. Binding to specific nucleic acid substrates activates SMARCAL1 activity in a reaction that requires its HARP2 (Hep-A-related protein 2) domain. Homology modeling indicates that the HARP domain is similar in structure to the DNA-binding domain of the PUR proteins. Limited proteolysis, small-angle X-ray scattering, and functional assays indicate that the core enzymatic unit consists of the HARP2 and ATPase domains that fold into a stable structure. Surprisingly, SMARCAL1 is capable of binding three-way and four-way Holliday junctions and model replication forks that lack a designed ssDNA region. Furthermore, SMARCAL1 remodels these DNA substrates by promoting branch migration and fork regression. SMARCAL1 mutations that cause Schimke immunoosseous dysplasia or that inactivate the HARP2 domain abrogate these activities. These results suggest that SMARCAL1 continuously surveys replication forks for damage. If damage is present, it remodels the fork to promote repair and restart. Failures in the process lead to activation of an alternative repair mechanism that depends on MUS81-catalyzed cleavage of the damaged fork.


Subject(s)
DNA Helicases/metabolism , DNA Replication/physiology , DNA, Cruciform/metabolism , Genomic Instability/physiology , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Helicases/genetics , DNA Replication/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Genomic Instability/genetics , HEK293 Cells , Humans , Protein Binding , Protein Structure, Tertiary , S Phase
7.
PLoS One ; 6(8): e23053, 2011.
Article in English | MEDLINE | ID: mdl-21857991

ABSTRACT

Homologous recombination (HR) is required for the restart of collapsed DNA replication forks and error-free repair of DNA double-strand breaks (DSB). However, unscheduled or hyperactive HR may lead to genomic instability and promote cancer development. The cellular factors that restrict HR processes in mammalian cells are only beginning to be elucidated. The tumor suppressor p53 has been implicated in the suppression of HR though it has remained unclear why p53, as the guardian of the genome, would impair an error-free repair process. Here, we show for the first time that p53 downregulates foci formation of the RAD51 recombinase in response to replicative stress in H1299 lung cancer cells in a manner that is independent of its role as a transcription factor. We find that this downregulation of HR is not only completely dependent on the binding site of p53 with replication protein A but also the ATR/ATM serine 15 phosphorylation site. Genetic analysis suggests that ATR but not ATM kinase modulates p53's function in HR. The suppression of HR by p53 can be bypassed under experimental conditions that cause DSB either directly or indirectly, in line with p53's role as a guardian of the genome. As a result, transactivation-inactive p53 does not compromise the resistance of H1299 cells to the interstrand crosslinking agent mitomycin C. Altogether, our data support a model in which p53 plays an anti-recombinogenic role in the ATR-dependent mammalian replication checkpoint but does not impair a cell's ability to use HR for the removal of DSB induced by cytotoxic agents.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Repair , Homologous Recombination , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cross-Linking Reagents/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Damage , DNA Replication/drug effects , Flow Cytometry , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mitomycin/pharmacology , Mutation , Protein Serine-Threonine Kinases/genetics , RNA Interference , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Thymidine/pharmacology , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics
8.
Genes Dev ; 25(12): 1320-7, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21685366

ABSTRACT

Successful DNA replication and packaging of newly synthesized DNA into chromatin are essential to maintain genome integrity. Defects in the DNA template challenge genetic and epigenetic inheritance. Unfortunately, tracking DNA damage responses (DDRs), histone deposition, and chromatin maturation at replication forks is difficult in mammalian cells. Here we describe a technology called iPOND (isolation of proteins on nascent DNA) to analyze proteins at active and damaged replication forks at high resolution. Using this methodology, we define the timing of histone deposition and chromatin maturation. Class 1 histone deacetylases are enriched at replisomes and remove predeposition marks on histone H4. Chromatin maturation continues even when decoupled from replisome movement. Furthermore, fork stalling causes changes in the recruitment and phosphorylation of proteins at the damaged fork. Checkpoint kinases catalyze H2AX phosphorylation, which spreads from the stalled fork to include a large chromatin domain even prior to fork collapse and double-strand break formation. Finally, we demonstrate a switch in the DDR at persistently stalled forks that includes MRE11-dependent RAD51 assembly. These data reveal a dynamic recruitment of proteins and post-translational modifications at damaged forks and surrounding chromatin. Furthermore, our studies establish iPOND as a useful methodology to study DNA replication and chromatin maturation.


Subject(s)
DNA Replication/physiology , Chromatin/metabolism , Click Chemistry/methods , DNA Damage , HEK293 Cells , Histones/metabolism , Humans
9.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...