Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Publication year range
1.
Mol Cell ; 81(7): 1499-1514.e6, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33621478

ABSTRACT

Despite their diverse biochemical characteristics and functions, all DNA-binding proteins share the ability to accurately locate their target sites among the vast excess of non-target DNA. Toward identifying universal mechanisms of the target search, we used single-molecule tracking of 11 diverse DNA-binding proteins in living Escherichia coli. The mobility of these proteins during the target search was dictated by DNA interactions rather than by their molecular weights. By generating cells devoid of all chromosomal DNA, we discovered that the nucleoid is not a physical barrier for protein diffusion but significantly slows the motion of DNA-binding proteins through frequent short-lived DNA interactions. The representative DNA-binding proteins (irrespective of their size, concentration, or function) spend the majority (58%-99%) of their search time bound to DNA and occupy as much as ∼30% of the chromosomal DNA at any time. Chromosome crowding likely has important implications for the function of all DNA-binding proteins.


Subject(s)
DNA, Bacterial/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , DNA, Bacterial/genetics , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics
2.
Mol Cell ; 78(2): 250-260.e5, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32097603

ABSTRACT

Structural maintenance of chromosomes (SMC) complexes organize chromosomes ubiquitously, thereby contributing to their faithful segregation. We demonstrate that under conditions of increased chromosome occupancy of the Escherichia coli SMC complex, MukBEF, the chromosome is organized as a series of loops around a thin (<130 nm) MukBEF axial core, whose length is ∼1,100 times shorter than the chromosomal DNA. The linear order of chromosomal loci is maintained in the axial cores, whose formation requires MukBEF ATP hydrolysis. Axial core structure in non-replicating chromosomes is predominantly linear (1 µm) but becomes circular (1.5 µm) in the absence of MatP because of its failure to displace MukBEF from the 800 kbp replication termination region (ter). Displacement of MukBEF from ter by MatP in wild-type cells directs MukBEF colocalization with the replication origin. We conclude that MukBEF individualizes and compacts the chromosome lengthwise, demonstrating a chromosome organization mechanism similar to condensin in mitotic chromosome formation.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Bacterial/genetics , Escherichia coli Proteins/genetics , Repressor Proteins/genetics , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/ultrastructure , Adenosine Triphosphate/genetics , Chromosomal Proteins, Non-Histone/ultrastructure , Chromosome Segregation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/ultrastructure , Escherichia coli/genetics , Escherichia coli Proteins/ultrastructure , Mitosis/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Replication Origin/genetics , Repressor Proteins/ultrastructure
3.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Article in English | MEDLINE | ID: mdl-34385314

ABSTRACT

Structural maintenance of chromosomes (SMC) complexes contribute to chromosome organization in all domains of life. In Escherichia coli, MukBEF, the functional SMC homolog, promotes spatiotemporal chromosome organization and faithful chromosome segregation. Here, we address the relative contributions of MukBEF and the replication terminus (ter) binding protein, MatP, to chromosome organization-segregation. We show that MukBEF, but not MatP, is required for the normal localization of the origin of replication to midcell and for the establishment of translational symmetry between newly replicated sister chromosomes. Overall, chromosome orientation is normally maintained through division from one generation to the next. Analysis of loci flanking the replication termination region (ter), which demark the ends of the linearly organized portion of the nucleoid, demonstrates that MatP is required for maintenance of chromosome orientation. We show that DNA-bound ß2-processivity clamps, which mark the lagging strands at DNA replication forks, localize to the cell center, independent of replisome location but dependent on MukBEF action, and consistent with translational symmetry of sister chromosomes. Finally, we directly show that the older ("immortal") template DNA strand, propagated from previous generations, is preferentially inherited by the cell forming at the old pole, dependent on MukBEF and MatP. The work further implicates MukBEF and MatP as central players in chromosome organization, segregation, and nonrandom inheritance of genetic material and suggests a general framework for understanding how chromosome conformation and dynamics shape subcellular organization.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation/physiology , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Repressor Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Escherichia coli Proteins/genetics , Gene Deletion , Gene Expression Regulation, Bacterial/physiology
4.
Cell ; 133(1): 90-102, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18394992

ABSTRACT

A prevalent view of DNA replication has been that it is carried out in fixed "replication factories." By tracking the progression of sister replication forks with respect to genetic loci in live Escherichia coli, we show that at initiation replisomes assemble at replication origins irrespective of where the origins are positioned within the cell. Sister replisomes separate and move to opposite cell halves shortly after initiation, migrating outwards as replication proceeds and both returning to midcell as replication termination approaches. DNA polymerase is maintained at stalled replication forks, and over short intervals of time replisomes are more dynamic than genetic loci. The data are inconsistent with models in which replisomes associated with sister forks act within a fixed replication factory. We conclude that independent replication forks follow the path of the compacted chromosomal DNA, with no structure other than DNA anchoring the replisome to any particular cellular region.


Subject(s)
DNA Replication , DNA, Bacterial/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Chromosomes, Bacterial/metabolism , DNA Replication Timing , Replication Origin , Replicon
5.
Mol Cell ; 54(5): 832-43, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24768536

ABSTRACT

In physiological settings, DNA translocases will encounter DNA-bound proteins, which must be dislodged or bypassed to allow continued translocation. FtsK is a bacterial translocase that promotes chromosome dimer resolution and decatenation by activating XerCD-dif recombination. To better understand how translocases act in crowded environments, we used single-molecule imaging to visualize FtsK in real time as it collided with other proteins. We show that FtsK can push, evict, and even bypass DNA-bound proteins. The primary factor dictating the outcome of collisions was the relative affinity of the proteins for their specific binding sites. Importantly, protein-protein interactions between FtsK and XerD help prevent removal of XerCD from DNA by promoting rapid reversal of FtsK. Finally, we demonstrate that RecBCD always overwhelms FtsK when these two motor proteins collide while traveling along the same DNA molecule, indicating that RecBCD is capable of exerting a much greater force than FtsK when translocating along DNA.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/genetics , Membrane Proteins/chemistry , Bacteriophage lambda/genetics , Binding Sites , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Viral/chemistry , Promoter Regions, Genetic , Protein Structure, Tertiary , Protein Transport
6.
Nucleic Acids Res ; 47(18): 9696-9707, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31400115

ABSTRACT

Ubiquitous Structural Maintenance of Chromosomes (SMC) complexes use a proteinaceous ring-shaped architecture to organize and individualize chromosomes, thereby facilitating chromosome segregation. They utilize cycles of adenosine triphosphate (ATP) binding and hydrolysis to transport themselves rapidly with respect to DNA, a process requiring protein conformational changes and multiple DNA contact sites. By analysing changes in the architecture and stoichiometry of the Escherichia coli SMC complex, MukBEF, as a function of nucleotide binding to MukB and subsequent ATP hydrolysis, we demonstrate directly the formation of dimer of MukBEF dimer complexes, dependent on dimeric MukF kleisin. Using truncated and full length MukB, in combination with MukEF, we show that engagement of the MukB ATPase heads on nucleotide binding directs the formation of dimers of heads-engaged dimer complexes. Complex formation requires functional interactions between the C- and N-terminal domains of MukF with the MukB head and neck, respectively, and MukE, which organizes the complexes by stabilizing binding of MukB heads to MukF. In the absence of head engagement, a MukF dimer bound by MukE forms complexes containing only a dimer of MukB. Finally, we demonstrate that cells expressing MukBEF complexes in which MukF is monomeric are Muk-, with the complexes failing to associate with chromosomes.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , Escherichia coli Proteins/genetics , Repressor Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomes/chemistry , Chromosomes/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Protein Binding , Repressor Proteins/chemistry
7.
Nucleic Acids Res ; 47(1): 210-220, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30445553

ABSTRACT

Bacterial DNA gyrase introduces negative supercoils into chromosomal DNA and relaxes positive supercoils introduced by replication and transiently by transcription. Removal of these positive supercoils is essential for replication fork progression and for the overall unlinking of the two duplex DNA strands, as well as for ongoing transcription. To address how gyrase copes with these topological challenges, we used high-speed single-molecule fluorescence imaging in live Escherichia coli cells. We demonstrate that at least 300 gyrase molecules are stably bound to the chromosome at any time, with ∼12 enzymes enriched near each replication fork. Trapping of reaction intermediates with ciprofloxacin revealed complexes undergoing catalysis. Dwell times of ∼2 s were observed for the dispersed gyrase molecules, which we propose maintain steady-state levels of negative supercoiling of the chromosome. In contrast, the dwell time of replisome-proximal molecules was ∼8 s, consistent with these catalyzing processive positive supercoil relaxation in front of the progressing replisome.


Subject(s)
DNA Gyrase/chemistry , DNA, Superhelical/chemistry , DNA-Binding Proteins/chemistry , Escherichia coli/enzymology , Catalysis , DNA Gyrase/genetics , DNA Gyrase/isolation & purification , DNA, Superhelical/genetics , DNA, Superhelical/isolation & purification , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Protein Binding , Single Molecule Imaging
8.
Mol Microbiol ; 111(1): 204-220, 2019 01.
Article in English | MEDLINE | ID: mdl-30318635

ABSTRACT

Although mycobacteria are rod shaped and divide by simple binary fission, their cell cycle exhibits unusual features: unequal cell division producing daughter cells that elongate with different velocities, as well as asymmetric chromosome segregation and positioning throughout the cell cycle. As in other bacteria, mycobacterial chromosomes are segregated by pair of proteins, ParA and ParB. ParA is an ATPase that interacts with nucleoprotein ParB complexes - segrosomes and non-specifically binds the nucleoid. Uniquely in mycobacteria, ParA interacts with a polar protein DivIVA (Wag31), responsible for asymmetric cell elongation, however the biological role of this interaction remained unknown. We hypothesised that this interaction plays a critical role in coordinating chromosome segregation with cell elongation. Using a set of ParA mutants, we determined that disruption of ParA-DNA binding enhanced the interaction between ParA and DivIVA, indicating a competition between the nucleoid and DivIVA for ParA binding. Having identified the ParA mutation that disrupts its recruitment to DivIVA, we found that it led to inefficient segrosomes separation and increased the cell elongation rate. Our results suggest that ParA modulates DivIVA activity. Thus, we demonstrate that the ParA-DivIVA interaction facilitates chromosome segregation and modulates cell elongation.


Subject(s)
Bacterial Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA, Bacterial/metabolism , Mycobacterium smegmatis/cytology , Mycobacterium smegmatis/enzymology , Bacterial Proteins/genetics , Chromosome Segregation , DNA Mutational Analysis , Mycobacterium smegmatis/growth & development
9.
Curr Genet ; 66(5): 895-899, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32300862

ABSTRACT

Structural maintenance of chromosomes (SMC) complexes are ancient and conserved molecular machines that organize chromosomes in all domains of life. We propose that the principles of chromosome folding needed to accommodate DNA inside a cell in an accessible form will follow similar principles in prokaryotes and eukaryotes. However, the exact contributions of SMC complexes to bacterial chromosome organization have been elusive. Recently, it was shown that the SMC homolog, MukBEF, organizes and individualizes the Escherichia coli chromosome by forming a filamentous axial core from which DNA loops emanate, similar to the action of condensin in mitotic chromosome formation. MukBEF action, along with its interaction with the partner protein, MatP, also facilitates chromosome individualization by directing opposite chromosome arms (replichores) to different cell halves. This contrasts with the situation in many other bacteria, where SMC complexes organise chromosomes in a way that the opposite replichores are aligned along the long axis of the cell. We highlight the similarities and differences of SMC complex contributions to chromosome organization in bacteria and eukaryotes, and summarize the current mechanistic understanding of the processes.


Subject(s)
Bacterial Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Bacterial/metabolism , Multiprotein Complexes/metabolism , Bacteria/genetics , Bacteria/metabolism , Chromosomes, Bacterial/genetics , Chromosomes, Bacterial/ultrastructure , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Repressor Proteins/metabolism
10.
Annu Rev Genet ; 46: 121-43, 2012.
Article in English | MEDLINE | ID: mdl-22934648

ABSTRACT

In dividing cells, chromosome duplication once per generation must be coordinated with faithful segregation of newly replicated chromosomes and with cell growth and division. Many of the mechanistic details of bacterial replication elongation are well established. However, an understanding of the complexities of how replication initiation is controlled and coordinated with other cellular processes is emerging only slowly. In contrast to eukaryotes, in which replication and segregation are separate in time, the segregation of most newly replicated bacterial genetic loci occurs sequentially soon after replication. We compare the strategies used by chromosomes and plasmids to ensure their accurate duplication and segregation and discuss how these processes are coordinated spatially and temporally with growth and cell division. We also describe what is known about the three conserved families of ATP-binding proteins that contribute to chromosome segregation and discuss their inter-relationships in a range of disparate bacteria.


Subject(s)
Chromosome Segregation , Chromosomes, Bacterial/genetics , DNA Replication , DNA, Bacterial/genetics , Escherichia coli/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Division , Chromosomes, Bacterial/metabolism , DNA Primase , DNA, Bacterial/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Plasmids/genetics , Plasmids/metabolism , Transcription, Genetic , Translocation, Genetic
11.
Nature ; 506(7487): 249-53, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24362571

ABSTRACT

DNA double-strand break (DSB) repair by homologous recombination has evolved to maintain genetic integrity in all organisms. Although many reactions that occur during homologous recombination are known, it is unclear where, when and how they occur in cells. Here, by using conventional and super-resolution microscopy, we describe the progression of DSB repair in live Escherichia coli. Specifically, we investigate whether homologous recombination can occur efficiently between distant sister loci that have segregated to opposite halves of an E. coli cell. We show that a site-specific DSB in one sister can be repaired efficiently using distant sister homology. After RecBCD processing of the DSB, RecA is recruited to the cut locus, where it nucleates into a bundle that contains many more RecA molecules than can associate with the two single-stranded DNA regions that form at the DSB. Mature bundles extend along the long axis of the cell, in the space between the bulk nucleoid and the inner membrane. Bundle formation is followed by pairing, in which the two ends of the cut locus relocate at the periphery of the nucleoid and together move rapidly towards the homology of the uncut sister. After sister locus pairing, RecA bundles disassemble and proteins that act late in homologous recombination are recruited to give viable recombinants 1-2-generation-time equivalents after formation of the initial DSB. Mutated RecA proteins that do not form bundles are defective in sister pairing and in DSB-induced repair. This work reveals an unanticipated role of RecA bundles in channelling the movement of the DNA DSB ends, thereby facilitating the long-range homology search that occurs before the strand invasion and transfer reactions.


Subject(s)
Chromosome Pairing , Chromosomes, Bacterial , DNA Breaks, Double-Stranded , Escherichia coli Proteins/metabolism , Escherichia coli , Rec A Recombinases/metabolism , Recombinational DNA Repair , Chromosomes, Bacterial/genetics , Chromosomes, Bacterial/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Rec A Recombinases/genetics , Recombinational DNA Repair/genetics
12.
Nature ; 516(7530): 259-262, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25470041

ABSTRACT

In every living organism, cell division requires accurate identification of the division site and placement of the division machinery. In bacteria, this process is traditionally considered to begin with the polymerization of the highly conserved tubulin-like protein FtsZ into a ring that locates precisely at mid-cell. Over the past decades, several systems have been reported to regulate the spatiotemporal assembly and placement of the FtsZ ring. However, the human pathogen Streptococcus pneumoniae, in common with many other organisms, is devoid of these canonical systems and the mechanisms of positioning the division machinery remain unknown. Here we characterize a novel factor that locates at the division site before FtsZ and guides septum positioning in pneumococcus. Mid-cell-anchored protein Z (MapZ) forms ring structures at the cell equator and moves apart as the cell elongates, therefore behaving as a permanent beacon of division sites. MapZ then positions the FtsZ ring through direct protein-protein interactions. MapZ-mediated control differs from previously described systems mostly on the basis of negative regulation of FtsZ assembly. Furthermore, MapZ is an endogenous target of the Ser/Thr kinase StkP, which was recently shown to have a central role in cytokinesis and morphogenesis of S. pneumoniae. We show that both phosphorylated and non-phosphorylated forms of MapZ are required for proper Z-ring formation and dynamics. Altogether, this work uncovers a new mechanism for bacterial cell division that is regulated by phosphorylation and illustrates that nature has evolved a diversity of cell division mechanisms adapted to the different bacterial clades.


Subject(s)
Bacterial Proteins/metabolism , Cytokinesis , Cytoskeletal Proteins/metabolism , Streptococcus pneumoniae/cytology , Streptococcus pneumoniae/metabolism , Bacterial Proteins/genetics , Phosphorylation , Protein Transport , Tubulin/metabolism
13.
Nucleic Acids Res ; 46(4): 1821-1833, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29294118

ABSTRACT

The formation of 3' single-stranded DNA overhangs is a first and essential step during homology-directed repair of double-stranded breaks (DSB) of DNA, a task that in Escherichia coli is performed by RecBCD. While this protein complex has been well characterized through in vitro single-molecule studies, it has remained elusive how end resection proceeds in the crowded and complex environment in live cells. Here, we develop a two-color fluorescent reporter to directly observe the resection of individual inducible DSB sites within live E. coli cells. Real-time imaging shows that RecBCD during end resection degrades DNA with remarkably high speed (∼1.6 kb/s) and high processivity (>∼100 kb). The results show a pronounced asymmetry in the processing of the two DNA ends of a DSB, where much longer stretches of DNA are degraded in the direction of terminus. The microscopy observations are confirmed using quantitative polymerase chain reaction measurements of the DNA degradation. Deletion of the recD gene drastically decreased the length of resection, allowing for recombination with short ectopic plasmid homologies and significantly increasing the efficiency of horizontal gene transfer between strains. We thus visualized and quantified DNA end resection by the RecBCD complex in live cells, recorded DNA-degradation linked to end resection and uncovered a general relationship between the length of end resection and the choice of the homologous recombination template.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Escherichia coli Proteins/metabolism , Exodeoxyribonuclease V/metabolism , DNA, Bacterial/metabolism , Deoxyribonucleases, Type II Site-Specific , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Exodeoxyribonuclease V/genetics , Gene Deletion , Gene Transfer, Horizontal , Luminescent Proteins , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction , Saccharomyces cerevisiae Proteins
14.
Nucleic Acids Res ; 44(8): 3801-10, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27036863

ABSTRACT

Programmable control of the cell cycle has been shown to be a powerful tool in cell-biology studies. Here, we develop a novel system for controlling the bacterial cell cycle, based on binding of CRISPR/dCas9 to the origin-of-replication locus. Initiation of replication of bacterial chromosomes is accurately regulated by the DnaA protein, which promotes the unwinding of DNA at oriC We demonstrate that the binding of CRISPR/dCas9 to any position within origin or replication blocks the initiation of replication. Serial-dilution plating, single-cell fluorescence microscopy, and flow-cytometry experiments show that ongoing rounds of chromosome replication are finished upon CRISPR/dCas9 binding, but no new rounds are initiated. Upon arrest, cells stay metabolically active and accumulate cell mass. We find that elevating the temperature from 37 to 42°C releases the CRISR/dCas9 replication inhibition, and we use this feature to recover cells from the arrest. Our simple and robust method of controlling the bacterial cell cycle is a useful asset for synthetic biology and DNA-replication studies in particular. The inactivation of CRISPR/dCas9 binding at elevated temperatures may furthermore be of wide interest for CRISPR/Cas9 applications in genomic engineering.


Subject(s)
CRISPR-Cas Systems , DNA Replication , Escherichia coli/genetics , CRISPR-Associated Proteins/metabolism , Chromosomes, Bacterial , Clustered Regularly Interspaced Short Palindromic Repeats , Escherichia coli/growth & development , Escherichia coli/metabolism , Plasmids/genetics , Replication Origin , Temperature
15.
Nucleic Acids Res ; 44(13): 6262-73, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27166373

ABSTRACT

Protein-DNA complexes are one of the principal barriers the replisome encounters during replication. One such barrier is the Tus-ter complex, which is a direction dependent barrier for replication fork progression. The details concerning the dynamics of the replisome when encountering these Tus-ter barriers in the cell are poorly understood. By performing quantitative fluorescence microscopy with microfuidics, we investigate the effect on the replisome when encountering these barriers in live Escherichia coli cells. We make use of an E. coli variant that includes only an ectopic origin of replication that is positioned such that one of the two replisomes encounters a Tus-ter barrier before the other replisome. This enables us to single out the effect of encountering a Tus-ter roadblock on an individual replisome. We demonstrate that the replisome remains stably bound after encountering a Tus-ter complex from the non-permissive direction. Furthermore, the replisome is only transiently blocked, and continues replication beyond the barrier. Additionally, we demonstrate that these barriers affect sister chromosome segregation by visualizing specific chromosomal loci in the presence and absence of the Tus protein. These observations demonstrate the resilience of the replication fork to natural barriers and the sensitivity of chromosome alignment to fork progression.


Subject(s)
DNA Replication/genetics , DNA-Binding Proteins/genetics , Escherichia coli Proteins/genetics , Chromosome Segregation/genetics , Chromosomes, Bacterial/genetics , DNA Helicases/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Macromolecular Substances/metabolism
16.
Proc Natl Acad Sci U S A ; 112(37): E5133-41, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26324908

ABSTRACT

The FtsK dsDNA translocase functions in bacterial chromosome unlinking by activating XerCD-dif recombination in the replication terminus region. To analyze FtsK assembly and translocation, and the subsequent activation of XerCD-dif recombination, we extended the tethered fluorophore motion technique, using two spectrally distinct fluorophores to monitor two effective lengths along the same tethered DNA molecule. We observed that FtsK assembled stepwise on DNA into a single hexamer, and began translocation rapidly (∼ 0.25 s). Without extruding DNA loops, single FtsK hexamers approached XerCD-dif and resided there for ∼ 0.5 s irrespective of whether XerCD-dif was synapsed or unsynapsed. FtsK then dissociated, rather than reversing. Infrequently, FtsK activated XerCD-dif recombination when it encountered a preformed synaptic complex, and dissociated before the completion of recombination, consistent with each FtsK-XerCD-dif encounter activating only one round of recombination.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Integrases/metabolism , Membrane Proteins/metabolism , Adenosine Triphosphate/chemistry , Base Sequence , Chromosomes, Bacterial/metabolism , DNA/chemistry , Escherichia coli/genetics , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Likelihood Functions , Molecular Sequence Data , Mutagenesis, Site-Directed , Normal Distribution , Protein Binding , Protein Transport , Recombination, Genetic
17.
Nucleic Acids Res ; 43(16): 7865-77, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26160884

ABSTRACT

Each cell division requires the unwinding of millions of DNA base pairs to allow chromosome duplication and gene transcription. As DNA replication and transcription share the same template, conflicts between both processes are unavoidable and head-on collisions are thought to be particularly problematic. Surprisingly, a recent study reported unperturbed cell cycle progression in Escherichia coli cells with an ectopic replication origin in which highly transcribed rrn operons were forced to be replicated opposite to normal. In this study we have re-generated a similar strain and found the doubling time to be twice that of normal cells. Replication profiles of this background revealed significant deviations in comparison to wild-type profiles, particularly in highly transcribed regions and the termination area. These deviations were alleviated by mutations that either inactivate the termination area or destabilise RNA polymerase complexes and allow their easier displacement by replication forks. Our data demonstrate that head-on replication-transcription conflicts are highly problematic. Indeed, analysis of the replication profile of the previously published E. coli construct revealed a chromosomal rearrangement that alleviates replication-transcription conflicts in an intriguingly simple way. Our data support the idea that avoiding head-on collisions has significantly contributed to shaping the distinct architecture of bacterial chromosomes.


Subject(s)
Chromosomes, Bacterial , DNA Replication , Escherichia coli/genetics , Replication Origin , Transcription, Genetic , Escherichia coli/growth & development
18.
PLoS Genet ; 10(8): e1004504, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25101671

ABSTRACT

Cell division in Escherichia coli starts with assembly of FtsZ protofilaments into a ring-like structure, the Z-ring. Positioning of the Z-ring at midcell is thought to be coordinated by two regulatory systems, nucleoid occlusion and the Min system. In E. coli, nucleoid occlusion is mediated by the SlmA proteins. Here, we address the question of whether there are additional positioning systems that are capable of localizing the E. coli divisome with respect to the cell center. Using quantitative fluorescence imaging we show that slow growing cells lacking functional Min and SlmA nucleoid occlusion systems continue to divide preferentially at midcell. We find that the initial Z-ring assembly occurs over the center of the nucleoid instead of nucleoid-free regions under these conditions. We determine that Z-ring formation begins shortly after the arrival of the Ter macrodomain at the nucleoid center. Removal of either the MatP, ZapB, or ZapA proteins significantly affects the accuracy and precision of Z-ring positioning relative to the nucleoid center in these cells in accordance with the idea that these proteins link the Ter macrodomain and the Z-ring. Interestingly, even in the absence of Min, SlmA, and the putative Ter macrodomain - Z-ring link, there remains a weak midcell positioning bias for the Z-ring. Our work demonstrates that additional Z-ring localization systems are present in E. coli than are known currently. In particular, we identify that the Ter macrodomain acts as a landmark for the Z-ring in the presence of MatP, ZapB and ZapA proteins.


Subject(s)
Carrier Proteins/genetics , Cell Cycle/genetics , Cell Division/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Cell Cycle Proteins/genetics , Cell Polarity/genetics , Chromosomal Proteins, Non-Histone/genetics , Escherichia coli/growth & development , Indoles , Optical Imaging
19.
Proc Natl Acad Sci U S A ; 111(7): 2734-9, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24449860

ABSTRACT

Worldwide dissemination of antibiotic resistance in bacteria is facilitated by plasmids that encode postsegregational killing (PSK) systems. These produce a stable toxin (T) and a labile antitoxin (A) conditioning cell survival to plasmid maintenance, because only this ensures neutralization of toxicity. Shortage of antibiotic alternatives and the link of TA pairs to PSK have stimulated the opinion that premature toxin activation could be used to kill these recalcitrant organisms in the clinic. However, validation of TA pairs as therapeutic targets requires unambiguous understanding of their mode of action, consequences for cell viability, and function in plasmids. Conflicting with widespread notions concerning these issues, we had proposed that the TA pair kis-kid (killing suppressor-killing determinant) might function as a plasmid rescue system and not as a PSK system, but this remained to be validated. Here, we aimed to clarify unsettled mechanistic aspects of Kid activation, and of the effects of this for kis-kid-bearing plasmids and their host cells. We confirm that activation of Kid occurs in cells that are about to lose the toxin-encoding plasmid, and we show that this provokes highly selective restriction of protein outputs that inhibits cell division temporarily, avoiding plasmid loss, and stimulates DNA replication, promoting plasmid rescue. Kis and Kid are conserved in plasmids encoding multiple antibiotic resistance genes, including extended spectrum ß-lactamases, for which therapeutic options are scarce, and our findings advise against the activation of this TA pair to fight pathogens carrying these extrachromosomal DNAs.


Subject(s)
Cell Division/physiology , DNA Replication/physiology , Drug Resistance, Bacterial/physiology , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , R Factors/physiology , Base Sequence , Blotting, Western , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Microscopy, Electron , Molecular Sequence Data , Oligonucleotides/genetics , R Factors/metabolism , Sequence Analysis, DNA
20.
Mol Cell ; 31(4): 498-509, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18722176

ABSTRACT

Dimeric circular chromosomes, formed by recombination between monomer sisters, cannot be segregated to daughter cells at cell division. XerCD site-specific recombination at the Escherichia coli dif site converts these dimers to monomers in a reaction that requires the DNA translocase FtsK. Short DNA sequences, KOPS (GGGNAGGG), which are polarized toward dif in the chromosome, direct FtsK translocation. FtsK interacts with KOPS through a C-terminal winged helix domain gamma. The crystal structure of three FtsKgamma domains bound to 8 bp KOPS DNA demonstrates how three gamma domains recognize KOPS. Using covalently linked dimers of FtsK, we infer that three gamma domains per hexamer are sufficient to recognize KOPS and load FtsK and subsequently activate recombination at dif. During translocation, FtsK fails to recognize an inverted KOPS sequence. Therefore, we propose that KOPS act solely as a loading site for FtsK, resulting in a unidirectionally oriented hexameric motor upon DNA.


Subject(s)
Bacterial Proteins/metabolism , DNA, Bacterial/metabolism , Pseudomonas aeruginosa/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/chemistry , Base Sequence , Biological Assay , Crystallography, X-Ray , Dimerization , Hydrolysis , Models, Biological , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL