Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Cell ; 72(6): 925-941.e4, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30576655

ABSTRACT

BRCA1-deficient tumor cells have defects in homologous-recombination repair and replication fork stability, resulting in PARP inhibitor sensitivity. Here, we demonstrate that a deubiquitinase, USP1, is upregulated in tumors with mutations in BRCA1. Knockdown or inhibition of USP1 resulted in replication fork destabilization and decreased viability of BRCA1-deficient cells, revealing a synthetic lethal relationship. USP1 binds to and is stimulated by fork DNA. A truncated form of USP1, lacking its DNA-binding region, was not stimulated by DNA and failed to localize and protect replication forks. Persistence of monoubiquitinated PCNA at the replication fork was the mechanism of cell death in the absence of USP1. Taken together, USP1 exhibits DNA-mediated activation at the replication fork, protects the fork, and promotes survival in BRCA1-deficient cells. Inhibition of USP1 may be a useful treatment for a subset of PARP-inhibitor-resistant BRCA1-deficient tumors with acquired replication fork stabilization.


Subject(s)
BRCA1 Protein/deficiency , Breast Neoplasms/enzymology , DNA Replication , DNA, Neoplasm/biosynthesis , Ubiquitin-Specific Proteases/metabolism , Uterine Cervical Neoplasms/enzymology , Animals , BRCA1 Protein/genetics , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Cell Survival , DNA, Neoplasm/genetics , Drug Resistance , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Mice, Nude , Mutation , Nucleic Acid Denaturation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Nucleic Acids Res ; 46(8): 3891-3905, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29529298

ABSTRACT

CHD1 is a conserved chromatin remodeling enzyme required for development and linked to prostate cancer in adults, yet its role in human cells is poorly understood. Here, we show that targeted disruption of the CHD1 gene in human cells leads to a defect in early double-strand break (DSB) repair via homologous recombination (HR), resulting in hypersensitivity to ionizing radiation as well as PARP and PTEN inhibition. CHD1 knockout cells show reduced H2AX phosphorylation (γH2AX) and foci formation as well as impairments in CtIP recruitment to the damaged sites. Chromatin immunoprecipitation following a single DSB shows that the reduced levels of γH2AX accumulation at DSBs in CHD1-KO cells are due to both a global reduction in H2AX incorporation and poor retention of H2AX at the DSBs. We also identified a unique N-terminal region of CHD1 that inhibits the DNA binding, ATPase, and chromatin assembly and remodeling activities of CHD1. CHD1 lacking the N terminus was more active in rescuing the defects in γH2AX formation and CtIP recruitment in CHD1-KO cells than full-length CHD1, suggesting the N terminus is a negative regulator in cells. Our data point to a role for CHD1 in the DSB repair process and identify a novel regulatory region of the protein.


Subject(s)
DNA Damage , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Binding Sites , Carrier Proteins/metabolism , Cell Line , Chromatin Assembly and Disassembly , DNA Breaks, Double-Stranded , DNA Helicases/chemistry , DNA Helicases/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Endodeoxyribonucleases , Gene Knockout Techniques , Histones/metabolism , Homologous Recombination , Humans , Nuclear Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
3.
Cell Rep ; 20(9): 2044-2056, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28854357

ABSTRACT

Oxidative damage to telomere DNA compromises telomere integrity. We recently reported that the DNA glycosylase NEIL3 preferentially repairs oxidative lesions in telomere sequences in vitro. Here, we show that loss of NEIL3 causes anaphase DNA bridging because of telomere dysfunction. NEIL3 expression increases during S phase and reaches maximal levels in late S/G2. NEIL3 co-localizes with TRF2 and associates with telomeres during S phase, and this association increases upon oxidative stress. Mechanistic studies reveal that NEIL3 binds to single-stranded DNA via its intrinsically disordered C terminus in a telomere-sequence-independent manner. Moreover, NEIL3 is recruited to telomeres through its interaction with TRF1, and this interaction enhances the enzymatic activity of purified NEIL3. Finally, we show that NEIL3 interacts with AP Endonuclease 1 (APE1) and the long-patch base excision repair proteins PCNA and FEN1. Taken together, we propose that NEIL3 protects genome stability through targeted repair of oxidative damage in telomeres during S/G2 phase.


Subject(s)
Chromosome Segregation , DNA Damage , DNA Repair , Mitosis , N-Glycosyl Hydrolases/metabolism , S Phase , Telomere/pathology , CD4-Positive T-Lymphocytes/metabolism , Cell Cycle Checkpoints , Cell Nucleus/metabolism , DNA/metabolism , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Humans , Microtubules/metabolism , N-Glycosyl Hydrolases/chemistry , Oxidative Stress , Protein Binding , Protein Domains , Spindle Apparatus/metabolism
4.
Sci Transl Med ; 9(389)2017 05 10.
Article in English | MEDLINE | ID: mdl-28490664

ABSTRACT

Multiple myeloma (MM) is a frequently incurable hematological cancer in which overactivity of MYC plays a central role, notably through up-regulation of ribosome biogenesis and translation. To better understand the oncogenic program driven by MYC and investigate its potential as a therapeutic target, we screened a chemically diverse small-molecule library for anti-MM activity. The most potent hits identified were rocaglate scaffold inhibitors of translation initiation. Expression profiling of MM cells revealed reversion of the oncogenic MYC-driven transcriptional program by CMLD010509, the most promising rocaglate. Proteome-wide reversion correlated with selective depletion of short-lived proteins that are key to MM growth and survival, most notably MYC, MDM2, CCND1, MAF, and MCL-1. The efficacy of CMLD010509 in mouse models of MM confirmed the therapeutic relevance of these findings in vivo and supports the feasibility of targeting the oncogenic MYC-driven translation program in MM with rocaglates.


Subject(s)
Multiple Myeloma/genetics , Multiple Myeloma/therapy , Animals , Cell Line, Tumor , Cyclin D1/genetics , Humans , Mice , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-myc/genetics , Xenograft Model Antitumor Assays
5.
J Biol Chem ; 292(28): 11927-11936, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28533432

ABSTRACT

Proper chromatin regulation is central to genome function and maintenance. The group III chromodomain-helicase-DNA-binding (CHD) family of ATP-dependent chromatin remodeling enzymes, comprising CHD6, CHD7, CHD8, and CHD9, has well-documented roles in transcription regulation, impacting both organism development and disease etiology. These four enzymes are similar in their constituent domains, but they fill surprisingly non-redundant roles in the cell, with deficiencies in individual enzymes leading to dissimilar disease states such as CHARGE syndrome or autism spectrum disorders. The mechanisms explaining their divergent, non-overlapping functions are unclear. In this study, we performed an in-depth biochemical analysis of purified CHD6, CHD7, and CHD8 and discovered distinct differences in chromatin remodeling specificities and activities among them. We report that CHD6 and CHD7 both bind with high affinity to short linker DNA, whereas CHD8 requires longer DNA for binding. As a result, CHD8 slides nucleosomes into positions with more flanking linker DNA than CHD7. Moreover, we found that, although CHD7 and CHD8 slide nucleosomes, CHD6 disrupts nucleosomes in a distinct non-sliding manner. The different activities of these enzymes likely lead to differences in chromatin structure and, thereby, transcriptional control, at the enhancer and promoter loci where these enzymes bind. Overall, our work provides a mechanistic basis for both the non-redundant roles and the diverse mutant disease states of these enzymes in vivo.


Subject(s)
Adenosine Triphosphate/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Nerve Tissue Proteins/metabolism , Nucleosomes/enzymology , Transcription Factors/metabolism , Animals , Biological Transport , DNA/chemistry , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Helicases/isolation & purification , DNA, Recombinant/chemistry , DNA, Recombinant/metabolism , DNA, Viral/chemistry , DNA, Viral/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , HeLa Cells , Humans , Hydrolysis , Kinetics , Molecular Weight , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Nucleosomes/metabolism , Phylogeny , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/isolation & purification
6.
Nucleic Acids Res ; 45(8): 4687-4695, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28334870

ABSTRACT

We investigate the mechanistic nature of the Snf2 family protein HARP, mutations of which are responsible for Schimke immuno-osseous dysplasia. Using a single-molecule magnetic tweezers assay, we construct RPA-stabilized DNA bubbles within torsionally constrained DNA to investigate the annealing action of HARP on a physiologically relevant substrate. We find that HARP closes RPA-stabilized bubbles in a slow reaction, taking on the order of tens of minutes for ∼600 bp of DNA to be re-annealed. The data indicate that DNA re-anneals through the removal of RPA, which is observed as clear steps in the bubble-closing traces. The dependence of the closing rate on both ionic strength and HARP concentration indicates that removal of RPA occurs via an association-dissociation mechanism where HARP does not remain associated with the DNA. The enzyme exhibits classical Michaelis-Menten kinetics and acts cooperatively with a Hill coefficient of 3 ± 1. Our work also allows the determination of some important features of RPA-bubble structures at low supercoiling, including the existence of multiple bubbles and that RPA molecules are mis-registered on the two strands.


Subject(s)
DNA Helicases/chemistry , DNA, Superhelical/chemistry , Replication Protein A/chemistry , Bacteriophage lambda/genetics , Bacteriophage lambda/metabolism , Biomechanical Phenomena , DNA Helicases/genetics , DNA Helicases/metabolism , DNA, Superhelical/genetics , DNA, Superhelical/metabolism , Humans , Kinetics , Magnetic Fields , Optical Tweezers , Osmolar Concentration , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Protein A/metabolism
7.
Nature ; 518(7538): 258-62, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25642963

ABSTRACT

Large-scale genomic studies have shown that half of epithelial ovarian cancers (EOCs) have alterations in genes regulating homologous recombination (HR) repair. Loss of HR accounts for the genomic instability of EOCs and for their cellular hyper-dependence on alternative poly-ADP ribose polymerase (PARP)-mediated DNA repair mechanisms. Previous studies have implicated the DNA polymerase θ (Polθ also known as POLQ, encoded by POLQ) in a pathway required for the repair of DNA double-strand breaks, referred to as the error-prone microhomology-mediated end-joining (MMEJ) pathway. Whether Polθ interacts with canonical DNA repair pathways to prevent genomic instability remains unknown. Here we report an inverse correlation between HR activity and Polθ expression in EOCs. Knockdown of Polθ in HR-proficient cells upregulates HR activity and RAD51 nucleofilament assembly, while knockdown of Polθ in HR-deficient EOCs enhances cell death. Consistent with these results, genetic inactivation of an HR gene (Fancd2) and Polq in mice results in embryonic lethality. Moreover, Polθ contains RAD51 binding motifs and it blocks RAD51-mediated recombination. Our results reveal a synthetic lethal relationship between the HR pathway and Polθ-mediated repair in EOCs, and identify Polθ as a novel druggable target for cancer therapy.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Directed DNA Polymerase/metabolism , Homologous Recombination , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Amino Acid Motifs , Animals , Carcinoma, Ovarian Epithelial , Cell Cycle , Cell Death , Cell Line, Tumor , DNA End-Joining Repair/genetics , DNA Replication , DNA-Directed DNA Polymerase/deficiency , Embryo Loss , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Genomic Instability , Homologous Recombination/genetics , Humans , Mice , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Protein Binding , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism , Recombinational DNA Repair/genetics , DNA Polymerase theta
8.
J Biol Chem ; 290(1): 25-34, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25384982

ABSTRACT

Chromodomain helicase DNA-binding protein 2 (CHD2) is an ATPase and a member of the SNF2-like family of helicase-related enzymes. Although deletions of CHD2 have been linked to developmental defects in mice and epileptic disorders in humans, little is known about its biochemical and cellular activities. In this study, we investigate the ATP-dependent activity of CHD2 and show that CHD2 catalyzes the assembly of chromatin into periodic arrays. We also show that the N-terminal region of CHD2, which contains tandem chromodomains, serves an auto-inhibitory role in both the DNA-binding and ATPase activities of CHD2. While loss of the N-terminal region leads to enhanced chromatin-stimulated ATPase activity, the N-terminal region is required for ATP-dependent chromatin remodeling by CHD2. In contrast, the C-terminal region, which contains a putative DNA-binding domain, selectively senses double-stranded DNA of at least 40 base pairs in length and enhances the ATPase and chromatin remodeling activities of CHD2. Our study shows that the accessory domains of CHD2 play central roles in both regulating the ATPase domain and conferring selectivity to chromatin substrates.


Subject(s)
Adenosine Triphosphatases/genetics , Chromatin Assembly and Disassembly/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Histones/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Animals , Baculoviridae/genetics , Binding Sites , Chromatin/chemistry , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Histones/metabolism , Humans , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sf9 Cells , Spodoptera
9.
PLoS One ; 9(9): e108066, 2014.
Article in English | MEDLINE | ID: mdl-25247294

ABSTRACT

Loss of the chromatin remodeling ATPase CHD5 has been linked to the progression of neuroblastoma tumors, yet the underlying mechanisms behind the tumor suppressor role of CHD5 are unknown. In this study, we purified the human CHD5 complex and found that CHD5 is a component of the full NuRD transcriptional repressor complex, which also contains methyl-CpG binding proteins and histone deacetylases. The CHD5/NuRD complex appears mutually exclusive with the related CHD4/NuRD complex as overexpression of CHD5 results in loss of the CHD4 protein in cells. Following a search for genes that are regulated by CHD5 in neuroblastoma cells, we found that CHD5 binds to and represses the G2/M checkpoint gene WEE1. Reintroduction of CHD5 into neuroblastoma cells represses WEE1 expression, demonstrating that CHD5 can function as a repressor in cells. A catalytically inactive mutant version of CHD5 is able to associate with a NuRD cofactor but fails to repress transcription. Our study shows that CHD5 is a NuRD-associated transcriptional repressor and identifies WEE1 as one of the CHD5-regulated genes that may link CHD5 to tumor suppression.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroblastoma/metabolism , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Alcohol Oxidoreductases , Cadherins/metabolism , Catalytic Domain , Cell Cycle Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Molecular Sequence Data , Mutation , Neuroblastoma/genetics , Nuclear Proteins/genetics , Protein-Tyrosine Kinases/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic
10.
J Biol Chem ; 289(30): 20717-26, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24923445

ABSTRACT

Although mutations or deletions of chromodomain helicase DNA-binding protein 5 (CHD5) have been linked to cancer and implicate CHD5 in tumor suppression, the ATP-dependent activity of CHD5 is currently unknown. In this study, we discovered that CHD5 is a chromatin remodeling factor with a unique enzymatic activity. CHD5 can expose nucleosomal DNA at one or two discrete positions in the nucleosome. The exposure of the nucleosomal DNA by CHD5 is dependent on ATP hydrolysis, but continued ATP hydrolysis is not required to maintain the nucleosomes in their remodeled state. The activity of CHD5 is distinct from other related chromatin remodeling ATPases, such as ACF and BRG1, and does not lead to complete disruption or destabilization of the nucleosome. Rather, CHD5 likely initiates remodeling in a manner similar to that of other remodeling factors but does not significantly reposition the nucleosome. While the related factor CHD4 shows strong ATPase activity, it does not unwrap nucleosomes as efficiently as CHD5. Our findings add to the growing evidence that chromatin remodeling ATPases have diverse roles in modulating chromatin structure.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA Helicases/metabolism , DNA/metabolism , Nerve Tissue Proteins/metabolism , Nucleosomes/metabolism , Tumor Suppressor Proteins/metabolism , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Cadherins/chemistry , Cadherins/genetics , Cadherins/metabolism , DNA/chemistry , DNA/genetics , DNA Helicases/chemistry , DNA Helicases/genetics , Drosophila melanogaster , Humans , Hydrolysis , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nucleosomes/chemistry , Nucleosomes/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
11.
Epigenetics ; 9(5): 693-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24565939

ABSTRACT

The HepA-related protein (HARP/SMARCAL1) is an ATP-dependent annealing helicase that is capable of rewinding DNA structures that are stably unwound due to binding of the single-stranded DNA (ssDNA)-binding protein Replication Protein A (RPA). HARP has been implicated in maintaining genome integrity through its role in DNA replication and repair, two processes that generate RPA-coated ssDNA. In addition, mutations in HARP cause a rare disease known as Schimke immuno-osseous dysplasia. In this study, we purified HARP containing complexes with the goal of identifying the predominant factors that stably associate with HARP. We found that HARP preferentially interacts with RPA molecules that are bound to the DNA-dependent protein kinase (DNA-PK). We also found that RPA is phosphorylated by DNA-PK in vitro, while the RPA-HARP complexes are not. Our results suggest that, in addition to its annealing helicase activity, which eliminates the natural binding substrate for RPA, HARP blocks the phosphorylation of RPA by DNA-PK.


Subject(s)
DNA Helicases/isolation & purification , DNA-Activated Protein Kinase/isolation & purification , Nuclear Proteins/isolation & purification , Replication Protein A/isolation & purification , DNA Helicases/metabolism , DNA, Single-Stranded/metabolism , DNA-Activated Protein Kinase/metabolism , HeLa Cells , Humans , Nuclear Proteins/metabolism , Phosphorylation , Replication Protein A/metabolism
12.
J Biol Chem ; 288(10): 7096-104, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23349465

ABSTRACT

MeCP2 is an abundant methyl-cytosine-guanine (CG)-binding protein and transcriptional repressor. We developed a biochemical system that exhibits CG methylation-specific transcriptional repression by purified human MeCP2. MeCP2 represses transcription by histone deacetylase (HDAC)-dependent and HDAC-independent mechanisms. Our system appears to recreate the HDAC-independent component of MeCP2-mediated repression and occurs via inhibition of the assembly of transcription preinitiation complexes. At a ratio of approximately one molecule of MeCP2 per two methyl-CG dinucleotides, as found in mammalian neurons, the magnitude of methylation-specific repression was greater than 10-fold. Notably, the HDAC inhibitor trichostatin A had no effect on MeCP2-mediated repression with either naked DNA or chromatin templates. We designed a CG-deficient core promoter that is resistant to MeCP2-mediated repression when placed in a plasmid lacking CG dinucleotides. By using this CG-deficient reporter as a reference, we found that eight CG dinucleotides in the core promoter region are sufficient for strong methylation-specific repression by MeCP2. In contrast, MeCP2 does not repress a construct with 13 CG dinucleotides located ∼1.7 kbp upstream of the promoter. Furthermore, by analysis of C-terminally truncated MeCP2 proteins, we found that binding of MeCP2 to methyl-CG dinucleotides is not sufficient for transcriptional repression. Hence, MeCP2-mediated repression is not due to the simple steric blockage of the transcriptional machinery. These experiments suggest that MeCP2 can function as a global methyl-CG-specific, HDAC-independent repressor. This HDAC-independent mechanism of MeCP2-mediated repression may be important in cells, such as mammalian neurons, that have high levels of CG methylation and MeCP2.


Subject(s)
Gene Expression Regulation , Histone Deacetylases/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Transcription, Genetic/genetics , Chromatin/genetics , Chromatin/metabolism , DNA/genetics , DNA/metabolism , DNA Methylation , Dinucleoside Phosphates/genetics , Electrophoretic Mobility Shift Assay , HeLa Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Methyl-CpG-Binding Protein 2/genetics , Mutation , Promoter Regions, Genetic/genetics , Protein Binding , Transcription Initiation, Genetic/drug effects , Transcription, Genetic/drug effects
13.
Rare Dis ; 1: e24735, 2013.
Article in English | MEDLINE | ID: mdl-25002993
14.
Curr Opin Genet Dev ; 21(2): 214-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21324673

ABSTRACT

The proper resolution of branched DNA molecules, which arise during processes such as DNA replication, DNA repair, and transcription, is critical for the maintenance of the genome. Disruption of this process can lead to genome instability and cancer progression. In this review, we describe recent progress on several interesting and biologically important enzymes that act upon different types of branched DNA substrates.


Subject(s)
DNA Helicases/metabolism , DNA Replication , DNA/metabolism , Animals , DNA Repair , Humans , Substrate Specificity
15.
Proc Natl Acad Sci U S A ; 107(49): 20970-3, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21078962

ABSTRACT

The structure and integrity of DNA is of considerable biological and biomedical importance, and it is therefore critical to identify and to characterize enzymes that alter DNA structure. DNA helicases are ATP-driven motor proteins that unwind DNA. Conversely, HepA-related protein (HARP) protein (also known as SMARCAL1 and DNA-dependent ATPase A) is an annealing helicase that rewinds DNA in an ATP-dependent manner. To date, HARP is the only known annealing helicase. Here we report the identification of a second annealing helicase, which we term AH2, for annealing helicase 2. Like HARP, AH2 catalyzes the ATP-dependent rewinding of replication protein A (RPA)-bound complementary single-stranded DNA, but does not exhibit any detectable helicase activity. Unlike HARP, however, AH2 lacks a conserved RPA-binding domain and does not interact with RPA. In addition, AH2 contains an HNH motif, which is commonly found in bacteria and fungi and is often associated with nuclease activity. AH2 appears to be the only vertebrate protein with an HNH motif. Contrary to expectations, purified AH2 does not exhibit nuclease activity, but it remains possible that AH2 contains a latent nuclease that is activated under specific conditions. These structural and functional differences between AH2 and HARP suggest that different annealing helicases have distinct functions in the cell.


Subject(s)
DNA Helicases/physiology , Amino Acid Motifs , Catalysis , DNA Helicases/chemistry , DNA, Single-Stranded/metabolism , Endonucleases , Humans , Molecular Motor Proteins , Protein Binding , Replication Protein A/metabolism
16.
Genes Dev ; 23(20): 2400-4, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19793863

ABSTRACT

HepA-related protein (HARP) (also known as SMARCAL1) is an ATP-driven annealing helicase that catalyzes the formation of dsDNA from complementary Replication protein A (RPA)-bound ssDNA. Here we find that HARP contains a conserved N-terminal motif that is necessary and sufficient for binding to RPA. This RPA-binding motif is not required for annealing helicase activity, but is essential for the recruitment of HARP to sites of laser-induced DNA damage. These findings suggest that the interaction of HARP with RPA increases the concentration of annealing helicase activity in the vicinity of ssDNA regions to facilitate processes such as DNA repair.


Subject(s)
DNA Helicases/metabolism , DNA Repair/physiology , Replication Protein A/metabolism , Amino Acid Motifs , Animals , DNA Damage , DNA Helicases/chemistry , HeLa Cells , Humans , Mice , Protein Binding , Protein Stability
17.
Mol Cell ; 34(5): 620-6, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524541

ABSTRACT

The high-mobility group N (HMGN) proteins are abundant nonhistone chromosomal proteins that bind specifically to nucleosomes at two high-affinity sites. Here we report that purified recombinant human HMGN1 (HMG14) and HMGN2 (HMG17) potently repress ATP-dependent chromatin remodeling by four different molecular motor proteins. In contrast, mutant HMGN proteins with double Ser-to-Glu mutations in their nucleosome-binding domains are unable to inhibit chromatin remodeling. The HMGN-mediated repression of chromatin remodeling is reversible and dynamic. With the ACF chromatin remodeling factor, HMGN2 does not directly inhibit the ATPase activity but rather appears to reduce the affinity of the factor to chromatin. These findings suggest that HMGN proteins serve as a counterbalance to the action of the many ATP-dependent chromatin remodeling activities in the nucleus.


Subject(s)
Chromatin Assembly and Disassembly/physiology , HMGN1 Protein/physiology , HMGN2 Protein/physiology , Nucleosomes/metabolism , Recombinant Proteins/metabolism , Adenosine Triphosphate/metabolism , Chromatin/metabolism , DNA Helicases/metabolism , HMGN1 Protein/genetics , HMGN1 Protein/metabolism , HMGN2 Protein/genetics , HMGN2 Protein/metabolism , Humans , Molecular Motor Proteins/physiology , Mutation , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism
18.
Science ; 322(5902): 748-50, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18974355

ABSTRACT

DNA-dependent adenosine triphosphatases (ATPases) participate in a broad range of biological processes including transcription, DNA repair, and chromatin dynamics. Mutations in the HepA-related protein (HARP) ATPase are responsible for Schimke immuno-osseous dysplasia (SIOD), but the function of the protein is unknown. We found that HARP is an ATP-dependent annealing helicase that rewinds single-stranded DNA bubbles that are stably bound by replication protein A. Other related ATPases, including the DNA translocase Rad54, did not exhibit annealing helicase activity. Analysis of mutant HARP proteins suggests that SIOD is caused by a deficiency in annealing helicase activity. Moreover, the pleiotropy of HARP mutations is consistent with the function of HARP as an annealing helicase that acts throughout the genome to oppose the action of DNA-unwinding activities in the nucleus.


Subject(s)
Adenosine Triphosphate/metabolism , DNA Helicases/metabolism , DNA, Single-Stranded/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , DNA Helicases/chemistry , DNA Helicases/deficiency , DNA Helicases/genetics , DNA, Single-Stranded/chemistry , DNA-Binding Proteins , Humans , Immunologic Deficiency Syndromes/genetics , Mutant Proteins/metabolism , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Osteochondrodysplasias/genetics , Replication Protein A/metabolism , Syndrome
19.
Mol Cell Biol ; 27(22): 7991-8002, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17846119

ABSTRACT

The insulator element at the 5' end of the chicken beta-globin locus acts as a barrier, protecting transgenes against silencing effects of adjacent heterochromatin. We showed earlier that the transcription factor USF1 binds within the insulator and that this site is important for generating in adjacent nucleosomes histone modifications associated with active chromatin and, by inference, with barrier function. To understand the mechanism of USF1 action, we have characterized USF1-containing complexes. USF1 interacts directly with the histone H4R3-specific methyltransferase PRMT1. USF1, PRMT1, and the histone acetyltransferases (HATs) PCAF and SRC-1 form a complex with both H4R3 histone methyltransferase and HAT activities. Small interfering RNA downregulation of USF1 results in localized loss of H4R3 methylation, and other histone modifications associated with euchromatin, at the insulator. A dominant negative peptide that interferes with USF1 binding to DNA causes silencing of an insulated reporter construct, indicating abolition of barrier function. These results show that USF1 plays a direct role in maintaining the barrier, supporting a model in which the insulator works as a barrier by maintaining a local environment of active chromatin.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation , Histones/metabolism , Transcription Factors/metabolism , Upstream Stimulatory Factors/metabolism , Animals , Cell Line , Chickens , Chromatin/chemistry , Erythrocytes/chemistry , Erythrocytes/metabolism , Gene Silencing , Histones/chemistry , Nucleic Acid Conformation , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , RNA Polymerase II/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics , Upstream Stimulatory Factors/genetics , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
20.
Proc Natl Acad Sci U S A ; 101(23): 8620-4, 2004 Jun 08.
Article in English | MEDLINE | ID: mdl-15169959

ABSTRACT

The protein CTCF plays an essential role in the action of a widely distributed class of vertebrate enhancer-blocking insulators, of which the first example was found in a DNA sequence element, HS4, at the 5' end of the chicken beta-globin locus. HS4 contains a binding site for CTCF that is necessary and sufficient for insulator action. Purification of CTCF has revealed that it interacts with proteins involved in subnuclear architecture, notably nucleophosmin, a 38-kDa nucleolar phosphoprotein that is concentrated in nuclear matrix preparations. In this report we show that both CTCF and the HS4 insulator element are incorporated in the matrix; HS4 incorporation depends on the presence of an intact CTCF-binding site. However the DNA sequence in the neighborhood of HS4 is not like that of canonical matrix attachment regions, and its incorporation into the matrix fraction is not sensitive to ribonuclease, suggesting that the insulator is a distinct matrix-associated element.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/genetics , DNA/metabolism , Globins/genetics , Repressor Proteins/metabolism , Animals , Base Sequence , Binding Sites/genetics , CCCTC-Binding Factor , Chickens , Erythrocytes/metabolism , In Vitro Techniques , Locus Control Region , Molecular Sequence Data , Nuclear Matrix/metabolism , Nuclear Proteins/metabolism , Nucleoplasmins , Phosphoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...