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.
PLoS One ; 8(10): e78141, 2013.
Article in English | MEDLINE | ID: mdl-24147116

ABSTRACT

UvrD-like helicases play diverse roles in DNA replication, repair and recombination pathways. An emerging body of evidence suggests that their different cellular functions are directed by interactions with partner proteins that target unwinding activity to appropriate substrates. Recent studies in E. coli have shown that UvrD can act as an accessory replicative helicase that resolves conflicts between the replisome and transcription complexes, but the mechanism is not understood. Here we show that the UvrD homologue PcrA interacts physically with B. subtilis RNA polymerase, and that an equivalent interaction is conserved in E. coli where UvrD, but not the closely related helicase Rep, also interacts with RNA polymerase. The PcrA-RNAP interaction is direct and independent of nucleic acids or additional mediator proteins. A disordered but highly conserved C-terminal region of PcrA, which distinguishes PcrA/UvrD from otherwise related enzymes such as Rep, is both necessary and sufficient for interaction with RNA polymerase.


Subject(s)
Bacterial Proteins/metabolism , DNA Helicases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Bacterial Proteins/genetics , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Replication/genetics , DNA Replication/physiology , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Protein Binding
2.
Proc Natl Acad Sci U S A ; 110(18): 7252-7, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23589869

ABSTRACT

Replication fork pausing drives genome instability, because any loss of paused replisome activity creates a requirement for reloading of the replication machinery, a potentially mutagenic process. Despite this importance, the relative contributions to fork pausing of different replicative barriers remain unknown. We show here that Deinococcus radiodurans RecD2 helicase inactivates Escherichia coli replisomes that are paused but still functional in vitro, preventing continued fork movement upon barrier removal or bypass, but does not inactivate elongating forks. Using RecD2 to probe replisome pausing in vivo, we demonstrate that most pausing events do not lead to replisome inactivation, that transcription complexes are the primary sources of this pausing, and that an accessory replicative helicase is critical for minimizing the frequency and/or duration of replisome pauses. These findings reveal the hidden potential for replisome inactivation, and hence genome instability, inside cells. They also demonstrate that efficient chromosome duplication requires mechanisms that aid resumption of replication by paused replisomes, especially those halted by protein-DNA barriers such as transcription complexes.


Subject(s)
DNA Replication , DNA, Bacterial/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Cell Death , DNA Helicases/metabolism , DNA-Directed DNA Polymerase/metabolism , Multienzyme Complexes/metabolism , Nucleoproteins/metabolism , Protein Binding , Transcription, Genetic
3.
Mol Cell ; 36(4): 654-66, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19941825

ABSTRACT

Nucleoprotein complexes present challenges to genome stability by acting as potent blocks to replication. One attractive model of how such conflicts are resolved is direct targeting of blocked forks by helicases with the ability to displace the blocking protein-DNA complex. We show that Rep and UvrD each promote movement of E. coli replisomes blocked by nucleoprotein complexes in vitro, that such an activity is required to clear protein blocks (primarily transcription complexes) in vivo, and that a polarity of translocation opposite that of the replicative helicase is critical for this activity. However, these two helicases are not equivalent. Rep but not UvrD interacts physically and functionally with the replicative helicase. In contrast, UvrD likely provides a general means of protein-DNA complex turnover during replication, repair, and recombination. Rep and UvrD therefore provide two contrasting solutions as to how organisms may promote replication of protein-bound DNA.


Subject(s)
DNA Helicases/metabolism , DNA, Bacterial/metabolism , DNA-Directed DNA Polymerase/metabolism , Escherichia coli Proteins/metabolism , Molecular Motor Proteins/metabolism , Multienzyme Complexes/metabolism , Culture Media , DNA Replication , DnaB Helicases/metabolism , Escherichia coli/cytology , Escherichia coli/enzymology , Escherichia coli/growth & development , Genetic Complementation Test , Mutation/genetics , Nucleoproteins/metabolism , Protein Binding , Suppression, Genetic , Transcription, Genetic
4.
DNA Repair (Amst) ; 8(11): 1300-10, 2009 Nov 02.
Article in English | MEDLINE | ID: mdl-19762288

ABSTRACT

During nucleotide excision repair (NER) in bacteria the UvrC nuclease and the short oligonucleotide that contains the DNA lesion are removed from the post-incision complex by UvrD, a superfamily 1A helicase. Helicases are frequently regulated by interactions with partner proteins, and immunoprecipitation experiments have previously indicated that UvrD interacts with UvrB, a component of the post-incision complex. We examined this interaction using 2-hybrid analysis and surface plasmon resonance spectroscopy, and found that the N-terminal domain and the unstructured region at the C-terminus of UvrD interact with UvrB. We analysed the properties of a truncated UvrD protein that lacked the unstructured C-terminal region and found that it showed a diminished affinity for single-stranded DNA, but retained the ability to displace both UvrC and the lesion-containing oligonucleotide from a post-incision nucleotide excision repair complex. The interaction of the C-terminal region of UvrD with UvrB is therefore not an essential feature of the mechanism by which UvrD disassembles the post-incision complex during NER. In further experiments we showed that PcrA helicase from Bacillus stearothermophilus can also displace UvrC and the excised oligonucleotide from a post-incision NER complex, which supports the idea that PcrA performs a UvrD-like function during NER in gram-positive organisms.


Subject(s)
DNA Helicases/metabolism , DNA Repair , DNA/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Adenosine Triphosphatases/metabolism , DNA/chemistry , DNA Helicases/chemistry , DNA Helicases/genetics , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , Substrate Specificity
5.
J Biol Chem ; 284(14): 9612-23, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19208629

ABSTRACT

Helicases play critical roles in all aspects of nucleic acid metabolism by catalyzing the remodeling of DNA and RNA structures. UvrD is an abundant helicase in Escherichia coli with well characterized functions in mismatch and nucleotide excision repair and a possible role in displacement of proteins such as RecA from single-stranded DNA. The mismatch repair protein MutL is known to stimulate UvrD. Here we show that the nucleotide excision repair proteins UvrA and UvrB can together stimulate UvrD-catalyzed unwinding of a range of DNA substrates containing strand discontinuities, including forked DNA substrates. The stimulation is specific for UvrD, as UvrAB failed to stimulate Rep helicase, a UvrD homologue. Moreover, although UvrAB can promote limited strand displacement, stimulation of UvrD did not require the strand displacement function of UvrAB. We conclude that UvrAB, like MutL, modulate UvrD helicase activity. This stimulation likely plays a role in DNA strand and protein displacement by UvrD in nucleotide excision repair. Promotion of UvrD-catalyzed unwinding of nicked duplexes by UvrAB may also explain the need for UvrAB and UvrD in Okazaki fragment processing in cells lacking DNA polymerase I. More generally, these data support the idea that helicase activity is regulated in vivo, with helicases acting as part of multisubunit complexes rather than in isolation.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Adenosine Triphosphatases/genetics , Biocatalysis , DNA/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Substrate Specificity
6.
J Mol Biol ; 381(2): 249-55, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18602646

ABSTRACT

There are many barriers that replication forks must overcome in order to duplicate a genome in vivo. These barriers include damage to the template DNA and proteins bound to this template. If replication is halted by such a block, then the block must be either removed or bypassed for replication to continue. If continuation of replication employs the original fork, avoiding the need to reload the replication apparatus, then the blocked replisome must retain functionality. In vivo studies of Escherichia coli replication forks suggest that replication forks blocked by protein-DNA complexes retain the ability to resume replication upon removal of the block for several hours. Here we tested the functional stability of replication forks reconstituted in vitro and blocked by lac repressor-operator complexes. Once a fork comes to a halt at such a block, it cannot continue subsequently to translocate through the block until addition of IPTG induces repressor dissociation. However, the ability to resume replication is retained only for 4-6 min regardless of the topological state of the template DNA. Comparison of our in vitro data with previous in vivo data suggests that either accessory factors that stabilise blocked forks are present in vivo or the apparent stability of blocked forks in vivo is due to continual reloading of the replication apparatus at the site of the block.


Subject(s)
DNA Replication , DNA, Bacterial/metabolism , Escherichia coli Proteins/metabolism , DNA, Bacterial/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Models, Biological , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Operator Regions, Genetic/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
7.
J Mol Biol ; 358(1): 46-56, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16516228

ABSTRACT

The RecA family of recombinases (RecA, Rad51, RadA and UvsX) catalyse strand-exchange between homologous DNA molecules by utilising conserved DNA-binding modules and a common core ATPase domain. RadB was identified in archaea as a Rad51-like protein on the basis of conserved ATPase sequences. However, RadB does not catalyse strand exchange and does not turn over ATP efficiently. RadB does bind DNA, and here we report a triplet of residues (Lys-His-Arg) that is highly conserved at the RadB C terminus, and is crucial for DNA binding. This is consistent with the motif forming a "basic patch" of highly conserved residues identified in an atomic structure of RadB from Thermococcus kodakaraensis. As the triplet motif is conserved at the C terminus of XRCC2 also, a mammalian Rad51-paralogue, we present a phylogenetic analysis that clarifies the relationship between RadB, Rad51-paralogues and recombinases. We investigate interactions between RadB and ATP using genetics and biochemistry; ATP binding by RadB is needed to promote survival of Haloferax volcanii after UV irradiation, and ATP, but not other NTPs, induces pronounced conformational change in RadB. This is the first genetic analysis of radB, and establishes its importance for maintaining genome stability in archaea. ATP-induced conformational change in RadB may explain previous reports that RadB controls Holliday junction resolution by Hjc, depending on the presence or the absence of ATP.


Subject(s)
Adenosine Triphosphate/metabolism , Archaea , Archaeal Proteins/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , DNA/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Archaeal Proteins/chemistry , DNA Damage , DNA Repair/radiation effects , DNA-Binding Proteins/chemistry , Evolution, Molecular , Haloferax volcanii/cytology , Haloferax volcanii/radiation effects , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Conformation , Recombination, Genetic , Ultraviolet Rays
8.
Nucleic Acids Res ; 33(11): 3678-90, 2005.
Article in English | MEDLINE | ID: mdl-15994460

ABSTRACT

Mutations in mammalian and Drosophila Hel308 and PolQ paralogues cause genome instability but their helicase functions are mysterious. By in vivo and in vitro analysis, we show that Hel308 from archaea (Hel308a) may act at stalled replication forks. Introducing hel308a into Escherichia coli dnaE strains that conditionally accumulate stalled forks caused synthetic lethality, an effect indistinguishable from E.coli RecQ. Further analysis in vivo indicated that the effect of hel308a is exerted independently of homologous recombination. The minimal biochemical properties of Hel308a protein were the same as human Hel308. We describe how helicase actions of Hel308a at fork structures lead specifically to displacement of lagging strands. The invading strand of D-loops is also targeted. Using archaeal Hel308, we propose models of action for the helicase domain of PolQ, promoting loading of the translesion polymerase domain. We speculate that removal of lagging strands at stalled forks by Hel308 promotes the formation of initiation zones, priming restart of lagging strand synthesis.


Subject(s)
Archaeal Proteins/metabolism , DNA Helicases/metabolism , DNA Replication , Archaeal Proteins/classification , DNA/chemistry , DNA/metabolism , DNA Helicases/classification , DNA Polymerase III/genetics , DNA, Cruciform/metabolism , Models, Genetic , Point Mutation
9.
Nucleic Acids Res ; 32(21): 6176-86, 2004.
Article in English | MEDLINE | ID: mdl-15570068

ABSTRACT

We have identified a novel structure-specific nuclease in highly fractionated extracts of the thermophilic archaeon Methanothermobacter thermautotrophicus (Mth). The 71 kDa protein product of open reading frame mth1090 is a nuclease with ATPase activity, which we call Nar71 (Nuclease-ATPase in Repair, 71 kDa). The nar71 gene is located in a gene neighbourhood proposed by genomics to encode a novel DNA repair system conserved in thermophiles. The biochemical characterization of Nar71 presented here is the first analysis from within this neighbourhood, and it supports the insight from genomics. Nuclease activity of Nar71 is specific for 3' flaps and flayed duplexes, targeting single-stranded DNA (ssDNA) regions. This activity requires Mg2+ or Mn2+ and is greatly reduced in ATP. In ATP, Nar71 displaces ssDNA, also with high specificity for 3' flap and flayed duplex DNA. Strand displacement is weak compared with nuclease activity, but in ATPS it is abolished, suggesting that Nar71 couples ATP hydrolysis to DNA strand separation. ATPase assays confirmed that Nar71 is stimulated by ssDNA, though not double-stranded DNA. Mutation of Lys-117 in Nar71 abolished ATPase and nuclease activity, and we describe a separation-of-function mutant (K68A) that has lost ATPase activity but retains nuclease activity. A model of possible Nar71 function in DNA repair is presented.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Repair , Deoxyribonucleases/metabolism , Methanobacteriaceae/enzymology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Amino Acid Substitution , Catalysis , DNA, Single-Stranded/metabolism , Deoxyribonucleases/chemistry , Deoxyribonucleases/genetics , Methanobacteriaceae/genetics , Molecular Sequence Data , Open Reading Frames
SELECTION OF CITATIONS
SEARCH DETAIL
...