Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 344
Filter
Add more filters

Publication year range
1.
Cell ; 159(4): 869-83, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25417162

ABSTRACT

X chromosome inactivation (XCI) depends on the long noncoding RNA Xist and its recruitment of Polycomb Repressive Complex 2 (PRC2). PRC2 is also targeted to other sites throughout the genome to effect transcriptional repression. Using XCI as a model, we apply an unbiased proteomics approach to isolate Xist and PRC2 regulators and identified ATRX. ATRX unexpectedly functions as a high-affinity RNA-binding protein that directly interacts with RepA/Xist RNA to promote loading of PRC2 in vivo. Without ATRX, PRC2 cannot load onto Xist RNA nor spread in cis along the X chromosome. Moreover, epigenomic profiling reveals that genome-wide targeting of PRC2 depends on ATRX, as loss of ATRX leads to spatial redistribution of PRC2 and derepression of Polycomb responsive genes. Thus, ATRX is a required specificity determinant for PRC2 targeting and function.


Subject(s)
DNA Helicases/metabolism , Nuclear Proteins/metabolism , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/metabolism , X Chromosome Inactivation , Animals , DNA Helicases/isolation & purification , Embryonic Stem Cells/metabolism , Female , Male , Mice , Nuclear Proteins/isolation & purification , X-linked Nuclear Protein
2.
Nature ; 606(7912): 204-210, 2022 06.
Article in English | MEDLINE | ID: mdl-35585232

ABSTRACT

Chromosome replication is performed by a complex and intricate ensemble of proteins termed the replisome, where the DNA polymerases Polδ and Polε, DNA polymerase α-primase (Polα) and accessory proteins including AND-1, CLASPIN and TIMELESS-TIPIN (respectively known as Ctf4, Mrc1 and Tof1-Csm3 in Saccharomyces cerevisiae) are organized around the CDC45-MCM-GINS (CMG) replicative helicase1-7. Because a functional human replisome has not been reconstituted from purified proteins, how these factors contribute to human DNA replication and whether additional proteins are required for optimal DNA synthesis are poorly understood. Here we report the biochemical reconstitution of human replisomes that perform fast and efficient DNA replication using 11 purified human replication factors made from 43 polypeptides. Polε, but not Polδ, is crucial for optimal leading-strand synthesis. Unexpectedly, Polε-mediated leading-strand replication is highly dependent on the sliding-clamp processivity factor PCNA and the alternative clamp loader complex CTF18-RFC. We show how CLASPIN and TIMELESS-TIPIN contribute to replisome progression and demonstrate that, in contrast to the budding yeast replisome8, AND-1 directly augments leading-strand replication. Moreover, although AND-1 binds to Polα9,10, the interaction is dispensable for lagging-strand replication, indicating that Polα is functionally recruited via an AND-1-independent mechanism for priming in the human replisome. Collectively, our work reveals how the human replisome achieves fast and efficient leading-strand and lagging-strand DNA replication, and provides a powerful system for future studies of the human replisome and its interactions with other DNA metabolic processes.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase , Multienzyme Complexes , DNA/biosynthesis , DNA Helicases/isolation & purification , DNA Helicases/metabolism , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/isolation & purification , Humans , Multienzyme Complexes/chemistry , Multienzyme Complexes/isolation & purification , Time Factors
3.
Cell ; 132(6): 945-57, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18358808

ABSTRACT

Telomerase is a multisubunit ribonucleoprotein (RNP) complex that adds telomere repeats to the ends of chromosomes. Three essential telomerase components have been identified thus far: the telomerase reverse transcriptase (TERT), the telomerase RNA component (TERC), and the TERC-binding protein dyskerin. Few other proteins are known to be required for human telomerase function, limiting our understanding of both telomerase regulation and mechanisms of telomerase action. Here, we identify the ATPases pontin and reptin as telomerase components through affinity purification of TERT from human cells. Pontin interacts directly with both TERT and dyskerin, and the amount of TERT bound to pontin and reptin peaks in S phase, evidence for cell-cycle-dependent regulation of TERT. Depletion of pontin and reptin markedly impairs telomerase RNP accumulation, indicating an essential role in telomerase assembly. These findings reveal an unanticipated requirement for additional enzymes in telomerase biogenesis and suggest alternative approaches for inhibiting telomerase in cancer.


Subject(s)
Carrier Proteins/chemistry , DNA Helicases/chemistry , Telomerase/chemistry , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/isolation & purification , Adenosine Triphosphatases/metabolism , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Chromatography, Affinity , DNA Helicases/isolation & purification , DNA Helicases/metabolism , HeLa Cells , Holoenzymes/chemistry , Holoenzymes/metabolism , Humans , Models, Biological , Models, Molecular , Nuclear Proteins/metabolism , RNA/metabolism , S Phase , Telomerase/metabolism , Telomere/metabolism
4.
Nucleic Acids Res ; 48(14): 7991-8005, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32621607

ABSTRACT

DNA2 is an essential enzyme involved in DNA replication and repair in eukaryotes. In a search for homologues of this protein, we identified and characterised Geobacillus stearothermophilus Bad, a bacterial DNA helicase-nuclease with similarity to human DNA2. We show that Bad contains an Fe-S cluster and identify four cysteine residues that are likely to co-ordinate the cluster by analogy to DNA2. The purified enzyme specifically recognises ss-dsDNA junctions and possesses ssDNA-dependent ATPase, ssDNA binding, ssDNA endonuclease, 5' to 3' ssDNA translocase and 5' to 3' helicase activity. Single molecule analysis reveals that Bad is a processive DNA motor capable of moving along DNA for distances of >4 kb at a rate of ∼200 bp per second at room temperature. Interestingly, as reported for the homologous human and yeast DNA2 proteins, the DNA unwinding activity of Bad is cryptic and can be unmasked by inactivating the intrinsic nuclease activity. Strikingly, our experiments show that the enzyme loops DNA while translocating, which is an emerging feature of processive DNA unwinding enzymes. The bacterial Bad enzymes will provide an excellent model system for understanding the biochemical properties of DNA2-like helicase-nucleases and DNA looping motor proteins in general.


Subject(s)
Bacterial Proteins/metabolism , DNA Helicases/metabolism , DNA, Single-Stranded/metabolism , Deoxyribonuclease I/metabolism , Geobacillus stearothermophilus/enzymology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/isolation & purification , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , DNA , DNA Helicases/chemistry , DNA Helicases/isolation & purification , Deoxyribonuclease I/chemistry , Deoxyribonuclease I/isolation & purification
5.
Biochem Biophys Res Commun ; 521(1): 212-219, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31635808

ABSTRACT

Liver sinusoidal endothelial cells play a key role maintaining the hepatic homeostasis, the disruption of which is associated with such end-stage liver diseases as hepatocellular carcinoma and cirrhosis. In the present study we investigated the role of brahma-related gene 1 (BRG1), a chromatin remodeling protein, in regulating endothelial transcription and the implication in liver fibrosis. We report that endothelial-specific deletion of BRG1 in mice attenuated liver fibrosis induced by injection with thioacetamide (TAA). Coincidently, alleviation of liver fibrosis as a result of endothelial BRG1 deletion was accompanied by an up-regulation of eNOS activity and NO bioavailability. In cultured endothelial cells, exposure to lipopolysaccharide (LPS) suppressed eNOS activity whereas BRG1 depletion with small interfering RNA restored eNOS-dependent NO production. Further analysis revealed that BRG1 was recruited to the caveolin-1 (CAV1) promoter by Sp1 and activated transcription of CAV1, which in turn inhibited eNOS activity. Mechanistically, BRG1 interacted with the H3K4 trimethyltransferase MLL1 to modulate H3K4 trimethylation surrounding the CAV1 promoter thereby contributing to LPS-induced CAV1 activation. In conclusion, our data unveil a novel role for BRG1 in the regulation of endothelial function and liver fibrosis.


Subject(s)
DNA Helicases/metabolism , Endothelial Cells/metabolism , Fibrosis/metabolism , Liver/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , DNA Helicases/deficiency , DNA Helicases/isolation & purification , Fibrosis/chemically induced , Humans , Liver/drug effects , Mice , Nitric Oxide/analysis , Nuclear Proteins/deficiency , Nuclear Proteins/isolation & purification , Thioacetamide , Transcription Factors/deficiency , Transcription Factors/isolation & purification
6.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29976672

ABSTRACT

Herpes simplex virus 1 (HSV-1) UL51 is a phosphoprotein that functions in the final envelopment in the cytoplasm and viral cell-cell spread, leading to efficient viral replication in cell cultures. To clarify the mechanism by which UL51 is regulated in HSV-1-infected cells, we focused on the phosphorylation of UL51. Mass spectrometry analysis of purified UL51 identified five phosphorylation sites in UL51. Alanine replacement of one of the identified phosphorylation sites in UL51, serine 184 (Ser-184), but not the other identified phosphorylation sites, significantly reduced viral replication and cell-cell spread in HaCaT cells. This mutation induced membranous invaginations adjacent to the nuclear membrane, the accumulation of primary enveloped virions in the invaginations and perinuclear space, and mislocalized UL34 and UL31 in punctate structures at the nuclear membrane; however, it had no effect on final envelopment in the cytoplasm of HaCaT cells. Of note, the alanine mutation in UL51 Ser-184 significantly reduced the mortality of mice following ocular infection. Phosphomimetic mutation in UL51 Ser-184 partly restored the wild-type phenotype in cell cultures and in mice. Based on these results, we concluded that some UL51 functions are specifically regulated by phosphorylation at Ser-184 and that this regulation is critical for HSV-1 replication in cell cultures and pathogenicity in vivoIMPORTANCE HSV-1 UL51 is conserved in all members of the Herpesviridae family. This viral protein is phosphorylated and functions in viral cell-cell spread and cytoplasmic virion maturation in HSV-1-infected cells. Although the downstream effects of HSV-1 UL51 have been clarified, there is a lack of information on how this viral protein is regulated as well as the significance of the phosphorylation of this protein in HSV-1-infected cells. In this study, we show that the phosphorylation of UL51 at Ser-184 promotes viral replication, cell-cell spread, and nuclear egress in cell cultures and viral pathogenicity in mice. This is the first report to identify the mechanism by which UL51 is regulated as well as the significance of UL51 phosphorylation in HSV-1 infection. Our study may provide insights into the regulatory mechanisms of other herpesviral UL51 homologs.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/physiology , DNA Primase/chemistry , DNA Primase/physiology , Herpesvirus 1, Human/pathogenicity , Viral Proteins/chemistry , Viral Proteins/physiology , Virus Release , Virus Replication , Active Transport, Cell Nucleus , Animals , Cell Line , Chlorocebus aethiops , DNA Helicases/genetics , DNA Helicases/isolation & purification , DNA Primase/genetics , DNA Primase/isolation & purification , Eye/virology , HEK293 Cells , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Humans , Mice , Phosphorylation , Protein Serine-Threonine Kinases , Vero Cells , Viral Proteins/genetics , Viral Proteins/isolation & purification , Virion/physiology , Virulence , Virus Assembly
7.
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
8.
Methods ; 108: 1-3, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27565743

ABSTRACT

In this special Methods collection on DNA helicases, I have solicited articles from leading experts in the field with a priority to gather a defined series of papers on highly relevant topics that encompass biological, biochemical, and biophysical aspects of helicase function. The experimental approaches described provide an opportunity for both new and more experienced scientists to use the information for the design of their own investigations. The reader will find detailed methods for single-molecule studies, novel biochemical experiments, genetic analyses, and cell biological assays in a variety of systems with an emphasis placed on state-of-the-art techniques to measure helicase function. Contributing authors were strongly encouraged to provide a carefully constructed description of the methods employed so that others might use this information in a manner that will be useful for their own particular application and helicase of interest. This special issue of Methods dedicated to DNA helicases offers readers a treasure chest of unique experimental approaches and protocols focused on rapidly developing techniques that are useful for studying both in vivo and in vitro aspects of helicase function.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/genetics , DNA/genetics , DNA/chemistry , DNA Helicases/isolation & purification
9.
Methods ; 105: 90-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27038745

ABSTRACT

Recent advances in high-throughput single-molecule magnetic tweezers have paved the way for obtaining information on individual molecules as well as ensemble-averaged behavior in a single assay. Here we describe how to design robust high-throughput magnetic tweezers assays that specifically require application of high forces (>20pN) for prolonged periods of time (>1000s). We elaborate on the strengths and limitations of the typical construct types that can be used and provide a step-by-step guide towards a high tether yield assay based on two examples. Firstly, we discuss a DNA hairpin assay where force-induced strand separation triggers a tight interaction between DNA-binding protein Tus and its binding site Ter, where forces up to 90pN for hundreds of seconds were required to dissociate Tus from Ter. Secondly, we show how the LTag helicase of Simian virus 40 unwinds dsDNA, where a load of 36pN optimizes the assay readout. The approaches detailed here provide guidelines for the high-throughput, quantitative study of a wide range of DNA-protein interactions.


Subject(s)
DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , High-Throughput Screening Assays/methods , Single Molecule Imaging/methods , DNA/chemistry , DNA Helicases/isolation & purification , DNA-Binding Proteins/genetics , Optical Tweezers , Simian virus 40/enzymology
10.
Methods ; 108: 14-23, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27223403

ABSTRACT

DNA helicases participate in virtually all aspects of cellular DNA metabolism by using ATP-fueled directional translocation along the DNA molecule to unwind DNA duplexes, dismantle nucleoprotein complexes, and remove non-canonical DNA structures. Post-translational modifications and helicase interacting partners are often viewed as determining factors in controlling the switch between bona fide helicase activity and other functions of the enzyme that do not involve duplex separation. The bottleneck in developing a mechanistic understanding of human helicases and their control by post-translational modifications is obtaining sufficient quantities of the modified helicase for traditional structure-functional analyses and biochemical reconstitutions. This limitation can be overcome by single-molecule analysis, where several hundred surface-tethered molecules are sufficient to obtain a complete kinetic and thermodynamic description of the helicase-mediated substrate binding and rearrangement. Synthetic oligonucleotides site-specifically labeled with Cy3 and Cy5 fluorophores can be used to create a variety of DNA substrates that can be used to characterize DNA binding, as well as helicase translocation and duplex unwinding activities. This chapter describes "single-molecule sorting", a robust experimental approach to simultaneously quantify, and distinguish the activities of helicases carrying their native post-translational modifications. Using this technique, a DNA helicase of interest can be produced and biotinylated in human cells to enable surface-tethering for the single-molecule studies by total internal reflection fluorescence microscopy. The pool of helicases extracted from the cells is expected to contain a mixture of post-translationally modified and unmodified enzymes, and the contributions from either population can be monitored separately, but in the same experiment providing a direct route to evaluating the effect of a given modification.


Subject(s)
DNA Helicases/isolation & purification , DNA-Binding Proteins/isolation & purification , Flow Cytometry/methods , Single Molecule Imaging/methods , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/genetics , DNA/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Oligonucleotides/chemical synthesis , Oligonucleotides/genetics
11.
Nucleic Acids Res ; 43(18): 8942-54, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26384418

ABSTRACT

ScPif1 DNA helicase is the prototypical member of a 5'-to-3' helicase superfamily conserved from bacteria to human and plays various roles in the maintenance of genomic homeostasis. While many studies have been performed with eukaryotic Pif1 helicases, including yeast and human Pif1 proteins, the potential functions and biochemical properties of prokaryotic Pif1 helicases remain largely unknown. Here, we report the expression, purification and biochemical analysis of Pif1 helicase from Bacteroides sp. 3_1_23 (BsPif1). BsPif1 binds to a large panel of DNA substrates and, in particular, efficiently unwinds partial duplex DNAs with 5'-overhang, fork-like substrates, D-loop and flap-like substrates, suggesting that BsPif1 may act at stalled DNA replication forks and enhance Okazaki fragment maturation. Like its eukaryotic homologues, BsPif1 resolves R-loop structures and unwinds DNA-RNA hybrids. Furthermore, BsPif1 efficiently unfolds G-quadruplexes and disrupts nucleoprotein complexes. Altogether, these results highlight that prokaryotic Pif1 helicases may resolve common issues that arise during DNA transactions. Interestingly, we found that BsPif1 is different from yeast Pif1, but resembles more human Pif1 with regard to substrate specificity, helicase activity and mode of action. These findings are discussed in the context of the possible functions of prokaryotic Pif1 helicases in vivo.


Subject(s)
Bacterial Proteins/metabolism , Bacteroides/enzymology , DNA Helicases/metabolism , DNA/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , DNA/chemistry , DNA Helicases/chemistry , DNA Helicases/isolation & purification , G-Quadruplexes , Substrate Specificity
12.
Proc Natl Acad Sci U S A ; 111(43): 15390-5, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25313033

ABSTRACT

DNA replication in eukaryotes is asymmetric, with separate DNA polymerases (Pol) dedicated to bulk synthesis of the leading and lagging strands. Pol α/primase initiates primers on both strands that are extended by Pol ε on the leading strand and by Pol δ on the lagging strand. The CMG (Cdc45-MCM-GINS) helicase surrounds the leading strand and is proposed to recruit Pol ε for leading-strand synthesis, but to date a direct interaction between CMG and Pol ε has not been demonstrated. While purifying CMG helicase overexpressed in yeast, we detected a functional complex between CMG and native Pol ε. Using pure CMG and Pol ε, we reconstituted a stable 15-subunit CMG-Pol ε complex and showed that it is a functional polymerase-helicase on a model replication fork in vitro. On its own, the Pol2 catalytic subunit of Pol ε is inefficient in CMG-dependent replication, but addition of the Dpb2 protein subunit of Pol ε, known to bind the Psf1 protein subunit of CMG, allows stable synthesis with CMG. Dpb2 does not affect Pol δ function with CMG, and thus we propose that the connection between Dpb2 and CMG helps to stabilize Pol ε on the leading strand as part of a 15-subunit leading-strand holoenzyme we refer to as CMGE. Direct binding between Pol ε and CMG provides an explanation for specific targeting of Pol ε to the leading strand and provides clear mechanistic evidence for how strand asymmetry is maintained in eukaryotes.


Subject(s)
DNA Polymerase II/metabolism , DNA Replication , Holoenzymes/metabolism , Protein Subunits/metabolism , Saccharomyces cerevisiae/enzymology , Chromatography, Gel , DNA Helicases/isolation & purification , DNA Helicases/metabolism , DNA, Circular/metabolism , Models, Biological , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , Time Factors
13.
Malar J ; 15(1): 526, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27809838

ABSTRACT

BACKGROUND: Malaria is one of the most serious and widespread parasitic diseases affecting humans. Because of the spread of resistance in both parasites and the mosquito vectors to anti-malarial drugs and insecticides, controlling the spread of malaria is becoming difficult. Thus, identifying new drug targets is urgently needed. Helicases play key roles in a wide range of cellular activities involving DNA and RNA transactions, making them attractive anti-malarial drug targets. METHODS: ATP-dependent DNA helicase gene (PfRuvB3) of Plasmodium falciparum strain K1, a chloroquine and pyrimethamine-resistant strain, was inserted into pQE-TriSystem His-Strep 2 vector, heterologously expressed and affinity purified. Identity of recombinant PfRuvB3 was confirmed by western blotting coupled with tandem mass spectrometry. Helicase and ATPase activities were characterized as well as co-factors required for optimal function. RESULTS: Recombinant PfRuvB3 has molecular size of 59 kDa, showing both DNA helicase and ATPase activities. Its helicase activity is dependent on divalent cations (Cu2+, Mg2+, Ni+2 or Zn+2) and ATP or dATP but is inhibited by high NaCl concentration (>100 mM). PfPuvB3 is unable to act on blunt-ended duplex DNA, but manifests ATPase activity in the presence of either single- or double-stranded DNA. PfRuvB3.is inhibited by doxorubicin, daunorubicin and netropsin, known DNA helicase inhibitors. CONCLUSIONS: Purified recombinant PfRuvB3 contains both DNA helicase and ATPase activities. Differences in properties of RuvB between the malaria parasite obtained from the study and human host provide an avenue leading to the development of novel drugs targeting specifically the malaria form of RuvB family of DNA helicases.


Subject(s)
DNA Helicases/metabolism , Plasmodium falciparum/enzymology , Recombinant Proteins/metabolism , Blotting, Western , Cations, Divalent/metabolism , Cloning, Molecular , Coenzymes/analysis , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Helicases/isolation & purification , Enzyme Inhibitors/analysis , Gene Expression , Metals/metabolism , Molecular Weight , Plasmodium falciparum/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Substrate Specificity , Tandem Mass Spectrometry
14.
Mol Cell ; 32(3): 313-24, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18995830

ABSTRACT

The Fanconi anemia (FA) pathway is implicated in DNA repair and cancer predisposition. Central to this pathway is the FA core complex, which is targeted to chromatin by FANCM and FAAP24 following replication stress. Here we show that FANCM and FAAP24 interact with the checkpoint protein HCLK2 independently of the FA core complex. In addition to defects in FA pathway activation, downregulation of FANCM or FAAP24 also compromises ATR/Chk1-mediated checkpoint signaling, leading to defective Chk1, p53, and FANCE phosphorylation; 53BP1 focus formation; and Cdc25A degradation. As a result, FANCM and FAAP24 deficiency results in increased endogenous DNA damage and a failure to efficiently invoke cell-cycle checkpoint responses. Moreover, we find that the DNA translocase activity of FANCM, which is dispensable for FA pathway activation, is required for its role in ATR/Chk1 signaling. Our data suggest that DNA damage recognition and remodeling activities of FANCM and FAAP24 cooperate with ATR/Chk1 to promote efficient activation of DNA damage checkpoints.


Subject(s)
DNA Damage , DNA Helicases/genetics , DNA Repair , DNA Replication , DNA-Binding Proteins/genetics , Fanconi Anemia/genetics , Cell Line , DNA Helicases/deficiency , DNA Helicases/isolation & purification , DNA Helicases/metabolism , DNA, Single-Stranded/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group Proteins , Genome , HeLa Cells , Humans , Kidney , Mitosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/isolation & purification , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/isolation & purification , Protein-Tyrosine Kinases/metabolism , S Phase , Signal Transduction/genetics , Signal Transduction/physiology
15.
Nucleic Acids Res ; 41(2): 1029-46, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23175612

ABSTRACT

Pif1 helicase plays various roles in the maintenance of nuclear and mitochondrial genome integrity in most eukaryotes. Here, we used a proteomics approach called isotopic differentiation of interactions as random or targeted to identify specific protein complexes of Saccharomyces cerevisiae Pif1. We identified a stable association between Pif1 and a mitochondrial SSB, Rim1. In vitro co-precipitation experiments using recombinant proteins indicated a direct interaction between Pif1 and Rim1. Fluorescently labeled Rim1 was titrated with Pif1 resulting in an increase in anisotropy and a K(d) value of 0.69 µM. Deletion mutagenesis revealed that the OB-fold domain and the C-terminal tail of Rim1 are both involved in interaction with Pif1. However, a Rim1 C-terminal truncation (Rim1ΔC18) exhibited a nearly 4-fold higher K(d) value. Rim1 stimulated Pif1 DNA helicase activity by 4- to 5-fold, whereas Rim1ΔC18 stimulated Pif1 by 2-fold. Hence, two regions of Rim1, the OB-fold domain and the C-terminal domain, interact with Pif1. One of these interactions occurs through the N-terminal domain of Pif1 because a deletion mutant of Pif1 (Pif1ΔN) retained interaction with Rim1 but did not exhibit stimulation of helicase activity. In light of our in vivo and in vitro data, and previous work, it is likely that the Rim1-Pif1 interaction plays a role in coordination of their functions in mtDNA metabolism.


Subject(s)
DNA Helicases/metabolism , Mitochondrial Proteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Binding Sites , DNA Helicases/chemistry , DNA Helicases/isolation & purification , DNA-Binding Proteins/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Repressor Proteins/chemistry , Repressor Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/isolation & purification , Sequence Deletion
16.
Plant J ; 76(1): 115-27, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23808500

ABSTRACT

To overcome the salinity-induced loss of crop yield, a salinity-tolerant trait is required. The SUV3 helicase is involved in the regulation of RNA surveillance and turnover in mitochondria, but the helicase activity of plant SUV3 and its role in abiotic stress tolerance have not been reported so far. Here we report that the Oryza sativa (rice) SUV3 protein exhibits DNA and RNA helicase, and ATPase activities. Furthermore, we report that SUV3 is induced in rice seedlings in response to high levels of salt. Its expression, driven by a constitutive cauliflower mosaic virus 35S promoter in IR64 transgenic rice plants, confers salinity tolerance. The T1 and T2 sense transgenic lines showed tolerance to high salinity and fully matured without any loss in yields. The T2 transgenic lines also showed tolerance to drought stress. These results suggest that the introduced trait is functional and stable in transgenic rice plants. The rice SUV3 sense transgenic lines showed lesser lipid peroxidation, electrolyte leakage and H2 O2 production, along with higher activities of antioxidant enzymes under salinity stress, as compared with wild type, vector control and antisense transgenic lines. These results suggest the existence of an efficient antioxidant defence system to cope with salinity-induced oxidative damage. Overall, this study reports that plant SUV3 exhibits DNA and RNA helicase and ATPase activities, and provides direct evidence of its function in imparting salinity stress tolerance without yield loss. The possible mechanism could be that OsSUV3 helicase functions in salinity stress tolerance by improving photosynthesis and antioxidant machinery in transgenic rice.


Subject(s)
DEAD-box RNA Helicases/metabolism , Oryza/enzymology , Plant Proteins/metabolism , Sodium Chloride/pharmacology , Stress, Physiological , Antioxidants/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/isolation & purification , DNA Helicases/genetics , DNA Helicases/isolation & purification , DNA Helicases/metabolism , Droughts , Gene Expression Regulation, Plant , Lipid Peroxidation , Oryza/genetics , Oryza/growth & development , Oryza/physiology , Oxidative Stress , Photosynthesis , Plant Proteins/genetics , Plant Proteins/isolation & purification , Plants, Genetically Modified , RNA Helicases/genetics , RNA Helicases/isolation & purification , RNA Helicases/metabolism , Salinity , Salt Tolerance , Seedlings/enzymology , Seedlings/genetics , Seedlings/growth & development , Seedlings/physiology
17.
Plant Mol Biol ; 85(6): 639-51, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24908423

ABSTRACT

DEAD-box helicases play essential role in DNA and RNA metabolism such as replication, repair, recombination, transcription, translation, ribosome biogenesis and splicing which regulate plant growth and development. The presence of helicases in the stress-induced ORFs identified by cDNA microarray indicates that helicases might be playing an important role in stabilizing growth in plants under stress. p68 DEAD-box helicase has been identified and characterized from animal systems but the properties and functions of plant p68 are poorly understood. In this study, the identification, purification and characterization of recombinant p68 from Pisum sativum (Psp68) is presented. Psp68 possesses all the characteristic motifs like DEAD-box ATP-binding and helicase C terminal motifs and is structurally similar to human p68 homologue. Psp68 exhibits ATPase activity in the presence of both DNA and RNA and it binds to DNA as well as RNA. It contains the characteristic RNA helicase activity. Interestingly Psp68 also shows the unique DNA helicase activity, which is bipolar in nature (unwinds DNA in both the 5'-3' and 3'-5' directions). The Km values of Psp68 for ATPase are 0.5126 and 0.9142 mM in the presence of DNA and RNA, respectively. The Km values of Psp68 are 1.6129 and 1.14 nM for DNA helicase and RNA helicase, respectively. The unique properties of Psp68 suggest that it could be a multifunctional protein involved in different aspect of DNA and RNA metabolism. This discovery should make an important contribution to better understanding of nucleic acids metabolism plants.


Subject(s)
DEAD-box RNA Helicases/physiology , DNA Helicases/physiology , Pisum sativum/enzymology , Plant Proteins/physiology , Amino Acid Sequence , Cloning, Molecular , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/isolation & purification , DNA Helicases/chemistry , DNA Helicases/isolation & purification , Models, Molecular , Molecular Sequence Data , Plant Proteins/chemistry , Plant Proteins/isolation & purification , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment
18.
BMC Plant Biol ; 14: 283, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25311683

ABSTRACT

BACKGROUND: Helicases play crucial role in almost all the nucleic acid metabolism including replication, repair, recombination, transcription, translation, ribosome biogenesis and splicing and these processes regulate plant growth and development. It is suggested that helicases play essential roles in stabilizing growth in plants under stress because their presence in the stress-induced ORFs has been identified. Moreover in a recent study we have reported that SUV3 helicase from Oryza sativa (OsSUV3) functions in salinity stress tolerance in transgenic rice by improving the antioxidant machinery. SUV3 helicase has been identified and characterized from yeast and human systems but the properties and functions of plant SUV3 are poorly understood. RESULTS: In this study, the purification and extensive characterization of recombinant OsSUV3 protein (67 kDa) is presented. OsSUV3 binds to DNA and RNA and exhibits DNA as well as RNA-dependent ATPase activities. It also contains the characteristic DNA and RNA helicase activity. OsSUV3 can use mainly ATP or dATP as energy source for the unwinding activity and it cannot unwind the blunt-end duplex DNA substrate. It is interesting to note that OsSUV3 unwinds DNA in both the 5'-3' and 3'-5 directions and thus its activity is bipolar in vitro. The Km values of OsSUV3 are 0.51 nM and 0.95 nM for DNA helicase and RNA helicase, respectively. CONCLUSIONS: This study is the first direct evidence to show the bipolar DNA helicase activity of OsSUV3 protein. The unique properties of OsSUV3 including its dual helicase activity imply that it could be a multifunctional protein involved in biologically significant process of DNA and RNA metabolisms. These results should make significant contribution towards better understanding of SUV3 protein in plants.


Subject(s)
DNA Helicases/metabolism , Oryza/enzymology , RNA Helicases/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , DNA Helicases/genetics , DNA Helicases/isolation & purification , Deoxyadenine Nucleotides/metabolism , Oryza/genetics , Oryza/growth & development , Plant Proteins/genetics , Plant Proteins/isolation & purification , Plant Proteins/metabolism , Protein Binding , RNA Helicases/genetics , RNA Helicases/isolation & purification , Recombinant Proteins , Salinity
19.
Eukaryot Cell ; 11(4): 417-29, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22286094

ABSTRACT

The Saccharomyces cerevisiae SEN1 gene codes for a nuclear, ATP-dependent helicase which is embedded in a complex network of protein-protein interactions. Pleiotropic phenotypes of mutations in SEN1 suggest that Sen1 functions in many nuclear processes, including transcription termination, DNA repair, and RNA processing. Sen1, along with termination factors Nrd1 and Nab3, is required for the termination of noncoding RNA transcripts, but Sen1 is associated during transcription with coding and noncoding genes. Sen1 and Nrd1 both interact directly with Nab3, as well as with the C-terminal domain (CTD) of Rpb1, the largest subunit of RNA polymerase II. It has been proposed that Sen1, Nab3, and Nrd1 form a complex that associates with Rpb1 through an interaction between Nrd1 and the Ser5-phosphorylated (Ser5-P) CTD. To further study the relationship between the termination factors and Rpb1, we used two-hybrid analysis and immunoprecipitation to characterize sen1-R302W, a mutation that impairs an interaction between Sen1 and the Ser2-phosphorylated CTD. Chromatin immunoprecipitation indicates that the impairment of the interaction between Sen1 and Ser2-P causes the reduced occupancy of mutant Sen1 across the entire length of noncoding genes. For protein-coding genes, mutant Sen1 occupancy is reduced early and late in transcription but is similar to that of the wild type across most of the coding region. The combined data suggest a handoff model in which proteins differentially transfer from the Ser5- to the Ser2-phosphorylated CTD to promote the termination of noncoding transcripts or other cotranscriptional events for protein-coding genes.


Subject(s)
DNA Helicases/metabolism , Nuclear Proteins/metabolism , RNA Helicases/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Substitution , DNA Helicases/genetics , DNA Helicases/isolation & purification , Gene Expression Regulation, Fungal , Multiprotein Complexes/isolation & purification , Multiprotein Complexes/metabolism , Nuclear Proteins/isolation & purification , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , RNA Helicases/genetics , RNA Helicases/isolation & purification , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/isolation & purification , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/isolation & purification , Transcription, Genetic , Two-Hybrid System Techniques
20.
Nat Genet ; 17(1): 100-3, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9288107

ABSTRACT

Werner syndrome (WS) is an uncommon autosomal recessive disorder characterized by premature aging. The clinical manifestations of WS, including atherosclerosis and osteoporosis, appear early in adulthood, and death in the fourth to sixth decade commonly ensues from myocardial infarction or cancer. In accord with the aging phenotype, cells from WS patients have a reduced replicative life span in culture. Genomic instability is observed at the cytogenetic level in the form of chromosome breaks and translocations and at the molecular level by multiple large deletions. The Werner syndrome gene (WRN) has recently been cloned. The predicted product is a 1,432-amino-acid protein whose central domain is homologous to members of the RecQ family of DNA helicases. Such homology does not necessarily mean that WRN encodes an active helicase. For example, the Saccharomyces cerevisiae RAD26 gene protein and the human transcription-repair coupling factor CSB (Cockayne syndrome 8) are highly homologous to known helicases, yet neither encodes an active helicase. Moreover, the Bloom's syndrome gene (BLM), discovered before WRN, is also homologous to the RecQ family of DNA helicases, though we still await demonstration that it encodes an active helicase. Here we report that the WS protein does indeed catalyze DNA unwinding.


Subject(s)
DNA Helicases/genetics , Point Mutation , Werner Syndrome/enzymology , Werner Syndrome/genetics , Adult , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , DNA Helicases/isolation & purification , DNA Helicases/metabolism , Humans , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spodoptera , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL