Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 46(15): 7701-7715, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-29982635

RESUMEN

Chromosome duplication initiates via the assembly of replication forks at defined origins. Forks proceed in opposite directions until they fuse with a converging fork. Recent work highlights that fork fusions are highly choreographed both in pro- and eukaryotic cells. The circular Escherichia coli chromosome is replicated from a single origin (oriC), and a single fork fusion takes place in a specialised termination area opposite oriC that establishes a fork trap mediated by Tus protein bound at ter sequences that allows forks to enter but not leave. Here we further define the molecular details of fork fusions and the role of RecG helicase in replication termination. Our data support the idea that fork fusions have the potential to trigger local re-replication of the already replicated DNA. In ΔrecG cells this potential is realised in a substantial fraction of cells and is dramatically elevated when one fork is trapped for some time before the converging fork arrives. They also support the idea that the termination area evolved to contain such over-replication and we propose that the stable arrest of replication forks at ter/Tus complexes is an important feature that limits the likelihood of problems arising as replication terminates.


Asunto(s)
Cromosomas Bacterianos/genética , Replicación del ADN/genética , ADN Bacteriano/genética , Proteínas de Escherichia coli/genética , Escherichia coli/genética , ADN Bacteriano/química , ADN Bacteriano/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Modelos Genéticos , Mutación , Conformación de Ácido Nucleico , Origen de Réplica/genética
2.
Nucleic Acids Res ; 45(5): 2571-2584, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-27956500

RESUMEN

Conflicts between replication and transcription challenge chromosome duplication. Escherichia coli replisome movement along transcribed DNA is promoted by Rep and UvrD accessory helicases with Δrep ΔuvrD cells being inviable under rapid growth conditions. We have discovered that mutations in a tRNA gene, aspT, in an aminoacyl tRNA synthetase, AspRS, and in a translation factor needed for efficient proline-proline bond formation, EF-P, suppress Δrep ΔuvrD lethality. Thus replication-transcription conflicts can be alleviated by the partial sacrifice of a mechanism that reduces replicative barriers, namely translating ribosomes that reduce RNA polymerase backtracking. Suppression depends on RelA-directed synthesis of (p)ppGpp, a signalling molecule that reduces replication-transcription conflicts, with RelA activation requiring ribosomal pausing. Levels of (p)ppGpp in these suppressors also correlate inversely with the need for Rho activity, an RNA translocase that can bind to emerging transcripts and displace transcription complexes. These data illustrate the fine balance between different mechanisms in facilitating gene expression and genome duplication and demonstrate that accessory helicases are a major determinant of this balance. This balance is also critical for other aspects of bacterial survival: the mutations identified here increase persistence indicating that similar mutations could arise in naturally occurring bacterial populations facing antibiotic challenge.


Asunto(s)
Replicación del ADN , Escherichia coli/genética , Genoma Bacteriano , Extensión de la Cadena Peptídica de Translación , ADN Helicasas/genética , Escherichia coli/enzimología , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Mutación , ARN de Transferencia de Aspártico/genética , Supresión Genética , Aminoacilación de ARN de Transferencia
3.
Genes (Basel) ; 7(8)2016 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-27483323

RESUMEN

Accessory replicative helicases aid the primary replicative helicase in duplicating protein-bound DNA, especially transcribed DNA. Recombination enzymes also aid genome duplication by facilitating the repair of DNA lesions via strand exchange and also processing of blocked fork DNA to generate structures onto which the replisome can be reloaded. There is significant interplay between accessory helicases and recombination enzymes in both bacteria and lower eukaryotes but how these replication repair systems interact to ensure efficient genome duplication remains unclear. Here, we demonstrate that the DNA content defects of Escherichia coli cells lacking the strand exchange protein RecA are driven primarily by conflicts between replication and transcription, as is the case in cells lacking the accessory helicase Rep. However, in contrast to Rep, neither RecA nor RecBCD, the helicase/exonuclease that loads RecA onto dsDNA ends, is important for maintaining rapid chromosome duplication. Furthermore, RecA and RecBCD together can sustain viability in the absence of accessory replicative helicases but only when transcriptional barriers to replication are suppressed by an RNA polymerase mutation. Our data indicate that the minimisation of replisome pausing by accessory helicases has a more significant impact on successful completion of chromosome duplication than recombination-directed fork repair.

4.
Curr Genet ; 62(4): 827-840, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27038615

RESUMEN

The RecG protein of Escherichia coli is a double-stranded DNA translocase that unwinds a variety of branched substrates in vitro. Although initially associated with homologous recombination and DNA repair, studies of cells lacking RecG over the past 25 years have led to the suggestion that the protein might be multi-functional and associated with a number of additional cellular processes, including initiation of origin-independent DNA replication, the rescue of stalled or damaged replication forks, replication restart, stationary phase or stress-induced 'adaptive' mutations and most recently, naïve adaptation in CRISPR-Cas immunity. Here we discuss the possibility that many of the phenotypes of recG mutant cells that have led to this conclusion may stem from a single defect, namely the failure to prevent re-replication of the chromosome. We also present data indicating that this failure does indeed contribute substantially to the much-reduced recovery of recombinants in conjugational crosses with strains lacking both RecG and the RuvABC Holliday junction resolvase.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Cromosomas Bacterianos , Daño del ADN , Replicación del ADN , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Genotipo , Recombinación Homóloga , Mutación
5.
Nucleic Acids Res ; 42(9): 5702-14, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24692661

RESUMEN

RecG is a DNA translocase encoded by most species of bacteria. The Escherichia coli protein targets branched DNA substrates and drives the unwinding and rewinding of DNA strands. Its ability to remodel replication forks and to genetically interact with PriA protein have led to the idea that it plays an important role in securing faithful genome duplication. Here we report that RecG co-localises with sites of DNA replication and identify conserved arginine and tryptophan residues near its C-terminus that are needed for this localisation. We establish that the extreme C-terminus, which is not resolved in the crystal structure, is vital for DNA unwinding but not for DNA binding. Substituting an alanine for a highly conserved tyrosine near the very end results in a substantial reduction in the ability to unwind replication fork and Holliday junction structures but has no effect on substrate affinity. Deleting or substituting the terminal alanine causes an even greater reduction in unwinding activity, which is somewhat surprising as this residue is not uniformly present in closely related RecG proteins. More significantly, the extreme C-terminal mutations have little effect on localisation. Mutations that do prevent localisation result in only a slight reduction in the capacity for DNA repair.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/metabolismo , Daño del ADN , Replicación del ADN , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/química , Genes Bacterianos , Genes Letales , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas
6.
Nature ; 500(7464): 608-11, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23892781

RESUMEN

Chromosome duplication normally initiates through the assembly of replication fork complexes at defined origins. DNA synthesis by any one fork is thought to cease when it meets another travelling in the opposite direction, at which stage the replication machinery may simply dissociate before the nascent strands are finally ligated. But what actually happens is not clear. Here we present evidence consistent with the idea that every fork collision has the potential to threaten genomic integrity. In Escherichia coli this threat is kept at bay by RecG DNA translocase and by single-strand DNA exonucleases. Without RecG, replication initiates where forks meet through a replisome assembly mechanism normally associated with fork repair, replication restart and recombination, establishing new forks with the potential to sustain cell growth and division without an active origin. This potential is realized when roadblocks to fork progression are reduced or eliminated. It relies on the chromosome being circular, reinforcing the idea that replication initiation is triggered repeatedly by fork collision. The results reported raise the question of whether replication fork collisions have pathogenic potential for organisms that exploit several origins to replicate each chromosome.


Asunto(s)
Cromosomas Bacterianos/metabolismo , Replicación del ADN , Escherichia coli/genética , Origen de Réplica , Proteínas Bacterianas/metabolismo , Bacteriófagos/genética , Bacteriófagos/fisiología , Cromosomas Bacterianos/genética , ADN Helicasas/metabolismo , ADN Bacteriano , ADN Circular , ADN de Cadena Simple , Proteínas de Unión al ADN/metabolismo , Escherichia coli/enzimología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/virología , Proteínas de Escherichia coli/metabolismo , Exodesoxirribonucleasa V/metabolismo , Exonucleasas/metabolismo , Marcadores Genéticos/genética , Inestabilidad Genómica
7.
Proc Natl Acad Sci U S A ; 110(18): 7252-7, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23589869

RESUMEN

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.


Asunto(s)
Replicación del ADN , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Muerte Celular , ADN Helicasas/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Complejos Multienzimáticos/metabolismo , Nucleoproteínas/metabolismo , Unión Proteica , Transcripción Genética
8.
Mol Microbiol ; 86(3): 675-91, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22957744

RESUMEN

RecG is a DNA translocase that helps to maintain genomic integrity. Initial studies suggested a role in promoting recombination, a possibility consistent with synergism between recG and ruv null alleles and reinforced when the protein was shown to unwind Holliday junctions. In this article we describe novel suppressors of recG and show that the pathology seen without RecG is suppressed on reducing or eliminating PriB, a component of the PriA system for replisome assembly and replication restart. Suppression is conditional, depending on additional mutations that modify ribosomal subunit S6 or one of three subunits of RNA polymerase. The latter suppress phenotypes associated with deletion of priB, enabling the deletion to suppress recG. They include alleles likely to disrupt interactions with transcription anti-terminator, NusA. Deleting priB has a different effect in ruv strains. It provokes abortive recombination and compromises DNA repair in a manner consistent with PriB being required to limit exposure of recombinogenic ssDNA. This synergism is reduced by the RNA polymerase mutations identified. Taken together, the results reveal that RecG curbs a potentially negative effect of proteins that direct replication fork assembly at sites removed from the normal origin, a facility needed to resolve conflicts between replication and transcription.


Asunto(s)
Proteínas Bacterianas/genética , Daño del ADN , ADN Helicasas/genética , Proteínas de Unión al ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , Endodesoxirribonucleasas/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Proteínas Bacterianas/metabolismo , ADN Helicasas/metabolismo , Reparación del ADN , Replicación del ADN , Proteínas de Unión al ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Endodesoxirribonucleasas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Mutación , Recombinación Genética , Ribosomas/genética , Ribosomas/metabolismo
9.
BMC Microbiol ; 12: 26, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22373098

RESUMEN

BACKGROUND: Manipulations of the DNA double helix during replication, transcription and other nucleic acid processing cause a change of DNA topology, which results in torsional stress. This stress is relaxed by DNA topoisomerases, a class of enzymes present in all domains of life. Negatively supercoiled DNA is relaxed by type IA topoisomerases that are widespread in bacteria, archaea and eukaryotes. In Escherichia coli there is conflicting data about viability of ΔtopA cells lacking topoisomerase I. RESULTS: In this study we sought to clarify whether E. coli cells lacking topoisomerase I are viable by using a plasmid-based lethality assay that allowed us to investigate the phenotype of ΔtopA cells without the presence of any compensatory mutations. Our results show that cells lacking topoisomerase I show an extreme growth defect and cannot be cultured without the accumulation of compensatory mutations. This growth defect can be partially suppressed by overexpression of topoisomerase III, the other type IA topoisomerase in E. coli, suggesting that the accumulation of torsional stress is, at least partially, responsible for the lethality of ΔtopA cells. The absence of RNase HI strongly exacerbates the phenotype of cells lacking topoisomerase I, which supports the idea that the processing of RNA:DNA hybrids is vitally important in ΔtopA cells. However, we did not observe suppression of the ΔtopA phenotype by increasing the level of R-loop processing enzymes, such as RNase HI or RecG. CONCLUSIONS: Our data show unambiguously that E. coli cells are not viable in the absence of DNA topoisomerase I without the presence of compensatory mutations. Furthermore, our data suggest that the accumulation of R-loops is not the primary reason for the severe growth defect of cells lacking topoisomerase I, which is in contrast to the current literature. Potential reasons for this discrepancy are discussed.


Asunto(s)
ADN-Topoisomerasas de Tipo I/deficiencia , Escherichia coli/enzimología , Escherichia coli/fisiología , Eliminación de Gen , Viabilidad Microbiana , ADN-Topoisomerasas de Tipo I/genética , ADN-Topoisomerasas de Tipo I/metabolismo , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Expresión Génica , Ribonucleasa H/genética , Ribonucleasa H/metabolismo , Supresión Genética
10.
Nucleic Acids Res ; 39(3): 949-57, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20923786

RESUMEN

Genome duplication requires accessory helicases to displace proteins ahead of advancing replication forks. Escherichia coli contains three helicases, Rep, UvrD and DinG, that might promote replication of protein-bound DNA. One of these helicases, Rep, also interacts with the replicative helicase DnaB. We demonstrate that Rep is the only putative accessory helicase whose absence results in an increased chromosome duplication time. We show also that the interaction between Rep and DnaB is required for Rep to maintain rapid genome duplication. Furthermore, this Rep-DnaB interaction is critical in minimizing the need for both recombinational processing of blocked replication forks and replisome reassembly, indicating that colocalization of Rep and DnaB minimizes stalling and subsequent inactivation of replication forks. These data indicate that E. coli contains only one helicase that acts as an accessory motor at the fork in wild-type cells, that such an activity is critical for the maintenance of rapid genome duplication and that colocalization with the replisome is crucial for this function. Given that the only other characterized accessory motor, Saccharomyces cerevisiae Rrm3p, associates physically with the replisome, our demonstration of the functional importance of such an association indicates that colocalization may be a conserved feature of accessory replicative motors.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , AdnB Helicasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Complejos Multienzimáticos/metabolismo , ADN Helicasas/análisis , AdnB Helicasas/análisis , Escherichia coli/enzimología , Proteínas de Escherichia coli/análisis , Genoma Bacteriano , Transcripción Genética
11.
Genetics ; 186(2): 473-92, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20647503

RESUMEN

Replication of the Escherichia coli chromosome usually initiates at a single origin (oriC) under control of DnaA. Two forks are established and move away in opposite directions. Replication is completed when these meet in a broadly defined terminus area half way around the circular chromosome. RecG appears to consolidate this arrangement by unwinding D-loops and R-loops that PriA might otherwise exploit to initiate replication at other sites. It has been suggested that without RecG such replication generates 3' flaps as the additional forks collide and displace nascent leading strands, providing yet more potential targets for PriA. Here we show that, to stay alive, cells must have either RecG or a 3' single-stranded DNA (ssDNA) exonuclease, which can be exonuclease I, exonuclease VII, or SbcCD. Cells lacking all three nucleases are inviable without RecG. They also need RecA recombinase and a Holliday junction resolvase to survive rapid growth, but SOS induction, although elevated, is not required. Additional requirements for Rep and UvrD are identified and linked with defects in DNA mismatch repair and with the ability to cope with conflicts between replication and transcription, respectively. Eliminating PriA helicase activity removes the requirement for RecG. The data are consistent with RecG and ssDNA exonucleases acting to limit PriA-mediated re-replication of the chromosome and the consequent generation of linear DNA branches that provoke recombination and delay chromosome segregation.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Exodesoxirribonucleasas/metabolismo , Cromosomas Bacterianos/metabolismo , ADN Helicasas/genética , Reparación de la Incompatibilidad de ADN , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Escherichia coli/citología , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/genética , Exonucleasas/metabolismo , Resolvasas de Unión Holliday/genética , Resolvasas de Unión Holliday/metabolismo , Recombinación Genética
12.
Nucleic Acids Res ; 38(19): 6433-46, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20525790

RESUMEN

The DNA-binding protein RdgC has been identified as an inhibitor of RecA-mediated homologous recombination in Escherichia coli. In Neisseria species, RdgC also has a role in virulence-associated antigenic variation. We have previously solved the crystal structure of the E. coli RdgC protein and shown it to form a toroidal dimer. In this study, we have conducted a mutational analysis of residues proposed to mediate interactions at the dimer interfaces. We demonstrate that destabilizing either interface has a serious effect on in vivo function, even though a stable complex with circular DNA was still observed. We conclude that tight binding is required for inhibition of RecA activity. We also investigated the role of the RdgC finger domain, and demonstrate that it plays a crucial role in the binding of circular DNA. Together, these data allow us to propose a model for how RdgC loads onto DNA. We discuss how RdgC might inhibit RecA-mediated strand exchange, and how RdgC might be displaced by other DNA metabolism enzymes such as polymerases and helicases.


Asunto(s)
ADN Circular/metabolismo , Proteínas de Escherichia coli/química , Sitios de Unión , ADN Helicasas/genética , Dimerización , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Eliminación de Gen , Modelos Moleculares , Mutación , Plásmidos/metabolismo , Estructura Terciaria de Proteína
13.
Genetics ; 185(1): 23-37, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20157002

RESUMEN

RuvABC and RecG are thought to provide alternative pathways for the late stages of recombination in Escherichia coli. Inactivation of both blocks the recovery of recombinants in genetic crosses. RuvABC resolves Holliday junctions, with RuvAB driving branch migration and RuvC catalyzing junction cleavage. RecG also drives branch migration, but no nuclease has been identified that might act with RecG to cleave junctions, apart from RusA, which is not normally expressed. We searched for an alternative nuclease using a synthetic lethality assay to screen for mutations causing inviability in the absence of RuvC, on the premise that a strain without any ability to cut junctions might be inviable. All the mutations identified mapped to polA, dam, or uvrD. None of these genes encodes a nuclease that cleaves Holliday junctions. Probing the reason for the inviability using the RusA Holliday junction resolvase provided strong evidence in each case that the RecG pathway is very ineffective at removing junctions and indicated that a nuclease component most probably does not exist. It also revealed new suppressors of recG, which were located to the ssb gene. Taken together with the results from the synthetic lethality assays, the properties of the mutant SSB proteins provide evidence that, rather than promoting recombination, a major function of RecG is to curb potentially pathological replication initiated via PriA protein at sites remote from oriC.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Resolvasas de Unión Holliday/metabolismo , Recombinación Genética/genética , Sustitución de Aminoácidos/genética , Proteínas Bacterianas/metabolismo , Cromosomas Bacterianos/metabolismo , ADN Helicasas/metabolismo , Reparación del ADN , Replicación del ADN , ADN Bacteriano/metabolismo , Endodesoxirribonucleasas/metabolismo , Escherichia coli/citología , Viabilidad Microbiana , Modelos Biológicos , Supresión Genética/genética
14.
DNA Repair (Amst) ; 9(3): 210-23, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20093100

RESUMEN

The RecG protein of Escherichia coli is a double-stranded DNA translocase that unwinds a variety of branched DNAs in vitro, including Holliday junctions, replication forks, D-loops and R-loops. Coupled with the reported pleiotropy of recG mutations, this broad range of potential targets has made it hard to pin down what the protein does in vivo, though roles in recombination and replication fork repair have been suggested. However, recent studies suggest that RecG provides a more general defence against pathological DNA replication. We have postulated that this is achieved through the ability of RecG to eliminate substrates that the replication restart protein, PriA, could otherwise exploit to re-replicate the chromosome. Without RecG, PriA triggers a cascade of events that interfere with the duplication and segregation of chromosomes. Here we review the studies that led us to this idea and to conclude that RecG may be both a specialist activity and a general guardian of the genome.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Helicasas/metabolismo , Genoma Bacteriano , Animales , Proteínas Bacterianas/química , ADN Helicasas/química , Replicación del ADN , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Humanos , Recombinación Genética
15.
Mol Cell ; 36(4): 654-66, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19941825

RESUMEN

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.


Asunto(s)
ADN Helicasas/metabolismo , ADN Bacteriano/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Motoras Moleculares/metabolismo , Complejos Multienzimáticos/metabolismo , Medios de Cultivo , Replicación del ADN , AdnB Helicasas/metabolismo , Escherichia coli/citología , Escherichia coli/enzimología , Escherichia coli/crecimiento & desarrollo , Prueba de Complementación Genética , Mutación/genética , Nucleoproteínas/metabolismo , Unión Proteica , Supresión Genética , Transcripción Genética
16.
DNA Repair (Amst) ; 8(12): 1434-43, 2009 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-19846353

RESUMEN

RecN is a highly conserved, SMC-like protein in bacteria. It plays an important role in the repair of DNA double-strand breaks and is therefore a key factor in maintaining genome integrity. The insolubility of Escherichia coli RecN has limited efforts to unravel its function. We overcame this limitation by replacing the resident coding sequence with that of Haemophilus influenzae RecN. The heterologous construct expresses Haemophilus RecN from the SOS-inducible E. coli promoter. The hybrid gene is fully functional, promoting survival after I-SceI induced DNA breakage, gamma irradiation or exposure to mitomycin C as effectively as the native gene, indicating that the repair activity is conserved between these two species. H. influenzae RecN is quite soluble, even when expressed at high levels, and is readily purified. Its analysis by ionisation-mass spectrometry, gel filtration and glutaraldehyde crosslinking indicates that it is probably a dimer under physiological conditions, although a higher multimer cannot be excluded. The purified protein displays a weak ATPase activity that is essential for its DNA repair function in vivo. However, no DNA-binding activity was detected, which contrasts with RecN from Bacillus subtilis. RecN proteins from Aquifex aeolicus and Bacteriodes fragilis also proved soluble. Neither binds DNA, but the Aquifex RecN has weak ATPase activity. Our findings support studies indicating that RecN, and the SOS response in general, behave differently in E. coli and B. subtilis. The hybrid recN reported provides new opportunities to study the genetics and biochemistry of how RecN operates in E. coli.


Asunto(s)
Proteínas Bacterianas/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Enzimas de Restricción del ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Enzimas de Restricción del ADN/química , Enzimas de Restricción del ADN/genética , Escherichia coli/química , Datos de Secuencia Molecular , Multimerización de Proteína , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Solubilidad
17.
Mol Microbiol ; 74(4): 940-55, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19818016

RESUMEN

Duplication and transmission of chromosomes require precise control of chromosome replication and segregation. Here we present evidence that RecG is a major factor influencing these processes in bacteria. We show that the extensive DnaA-independent stable DNA replication observed without RecG can lead to replication of any area of the chromosome. This replication is further elevated following irradiation with UV light and appears to be perpetuated by secondary events that continue long after the elimination of UV lesions. The resulting pathological cascade is associated with an increased number of replication forks traversing the chromosome, sometimes with extensive regional amplification of the chromosome, and with the accumulation of highly branched DNA intermediates containing few Holliday junctions. We propose that the cascade is triggered by replication fork collisions that generate 3' single-strand DNA flaps, providing sites for PriA to initiate re-replication of the DNA and thus to generate linear duplexes that provoke recombination, allowing priming of even further replication. Our results shed light on why termination of replication in bacteria is normally limited to a single encounter of two forks and carefully orchestrated within a restricted area, and explain how a system of multiple forks and random termination can operate in eukaryotes.


Asunto(s)
Segregación Cromosómica , Cromosomas Bacterianos/metabolismo , Replicación del ADN , Proteínas de Escherichia coli/fisiología , Escherichia coli/enzimología , Escherichia coli/fisiología , ADN Cruciforme/metabolismo , Escherichia coli/efectos de la radiación , Proteínas de Escherichia coli/genética , Rayos Ultravioleta
18.
Mol Microbiol ; 73(3): 352-66, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19538444

RESUMEN

Little is known about what happens when forks meet to complete DNA replication in any organism. In this study we present data suggesting that the collision of replication forks is a potential threat to genomic stability. We demonstrate that Escherichia coli cells lacking RecG helicase suffer major defects in chromosome replication following UV irradiation, and that this is associated with high levels of DNA synthesis initiated independently of the initiator protein DnaA. This UV-induced stable DNA replication is dependent on PriA helicase and continues long after UV-induced lesions have been excised. We suggest UV irradiation triggers the assembly of new replication forks, leading to multiple fork collisions outside the terminus area. Such collisions may generate branched DNAs that serve to establish further new forks, resulting in uncontrolled DNA amplification. We propose that RecG reduces the likelihood of this pathological cascade being set in motion by reducing initiation of replication at D- and R-loops, and other structures generated as a result of fork collisions. Our results shed light on why replication initiation in bacteria is limited to a single origin and why termination is carefully orchestrated to a single event within a restricted area each cell cycle.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , División Celular , Cromosomas Bacterianos/genética , Daño del ADN , ADN Helicasas/genética , Replicación del ADN/efectos de la radiación , ADN Bacteriano/biosíntesis , Escherichia coli/citología , Escherichia coli/metabolismo , Escherichia coli/efectos de la radiación , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Rayos Ultravioleta
19.
DNA Repair (Amst) ; 7(9): 1517-30, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18606573

RESUMEN

DNA double-strand breaks threaten the stability of the genome, and yet are induced deliberately during meiosis in order to provoke homologous recombination and generate the crossovers needed to promote faithful chromosome transmission. Crossovers are secured via biased resolution of the double Holliday junction intermediates formed when both ends of the broken chromosome engage an intact homologue. To investigate whether the enzymes catalysing branch migration and resolution of Holliday junctions are directed to favour production of either crossover or noncrossover products, we engineered a genetic system based on DNA breakage induced by the I-SceI endonuclease to detect analogous exchanges in Escherichia coli where the enzymology of recombination is more fully understood. Analysis of the recombinants generated revealed approximately equal numbers of crossover and noncrossover products, regardless of whether repair is mediated via RecG, RuvABC, or the RusA resolvase. We conclude that there little or no control of crossing over at the level of Holliday junction resolution. Thus, if similar resolvases function during meiosis, additional factors must act to bias recombination in favour of crossover progeny.


Asunto(s)
Intercambio Genético , Roturas del ADN de Doble Cadena , Reparación del ADN , Resolvasas de Unión Holliday/fisiología , Proteínas Bacterianas/fisiología , ADN Helicasas , Endodesoxirribonucleasas/fisiología , Proteínas de Escherichia coli/fisiología , Exodesoxirribonucleasas/fisiología
20.
DNA Repair (Amst) ; 7(9): 1589-602, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18644471

RESUMEN

In dividing cells, the stalling of replication fork complexes by impediments to DNA unwinding or by template imperfections that block synthesis by the polymerase subunits is a serious threat to genomic integrity and cell viability. What happens to stalled forks depends on the nature of the offending obstacle. In UV-irradiated Escherichia coli cells DNA synthesis is delayed for a considerable period, during which forks undergo extensive processing before replication can resume. Thus, restart depends on factors needed to load the replicative helicase, indicating that the replisome may have dissociated. It also requires the RecFOR proteins, which are known to load RecA recombinase on single-stranded DNA, implying that template strands are exposed. To gain a further understanding of how UV irradiation affects replication and how replication resumes after a block, we used fluorescence microscopy and BrdU or radioisotope labelling to examine chromosome replication and cell cycle progression. Our studies confirm that RecFOR promote efficient reactivation of stalled forks and demonstrate that they are also needed for productive replication initiated at the origin, or triggered elsewhere by damage to the DNA. Although delayed, all modes of replication do recover in the absence of these proteins, but nascent DNA strands are degraded more extensively by RecJ exonuclease. However, these strands are also degraded in the presence of RecFOR when restart is blocked by other means, indicating that RecA loading is not sufficient to stabilise and protect the fork. This is consistent with the idea that RecA actively promotes restart. Thus, in contrast to eukaryotic cells, there may be no factor in bacterial cells acting specifically to stabilise stalled forks. Instead, nascent strands may be protected by the simple expedient of promoting restart. We also report that the efficiency of fork reactivation is not affected in polB mutants.


Asunto(s)
ADN Polimerasa II/fisiología , Reparación del ADN , Replicación del ADN , Escherichia coli/genética , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/genética , Escherichia coli/efectos de la radiación , Proteínas de Escherichia coli/genética , Mutación , Rec A Recombinasas/genética , Origen de Réplica , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA