Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 52(8): 4659-4675, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38554102

ABSTRACT

RexA and RexB function as an exclusion system that prevents bacteriophage T4rII mutants from growing on Escherichia coli λ phage lysogens. Recent data established that RexA is a non-specific DNA binding protein that can act independently of RexB to bias the λ bistable switch toward the lytic state, preventing conversion back to lysogeny. The molecular interactions underlying these activities are unknown, owing in part to a dearth of structural information. Here, we present the 2.05-Å crystal structure of the λ RexA dimer, which reveals a two-domain architecture with unexpected structural homology to the recombination-associated protein RdgC. Modelling suggests that our structure adopts a closed conformation and would require significant domain rearrangements to facilitate DNA binding. Mutagenesis coupled with electromobility shift assays, limited proteolysis, and double electron-electron spin resonance spectroscopy support a DNA-dependent conformational change. In vivo phenotypes of RexA mutants suggest that DNA binding is not a strict requirement for phage exclusion but may directly contribute to modulation of the bistable switch. We further demonstrate that RexA homologs from other temperate phages also dimerize and bind DNA in vitro. Collectively, these findings advance our mechanistic understanding of Rex functions and provide new evolutionary insights into different aspects of phage biology.


Subject(s)
Bacteriophage lambda , DNA-Binding Proteins , Models, Molecular , Viral Proteins , Bacteriophage lambda/genetics , Crystallography, X-Ray , Viral Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Protein Binding , Protein Multimerization , DNA, Viral/genetics , DNA, Viral/metabolism , Mutation , Lysogeny , Escherichia coli/virology , Escherichia coli/genetics , Escherichia coli/metabolism , DNA/metabolism , DNA/chemistry
2.
Mol Microbiol ; 120(2): 122-140, 2023 08.
Article in English | MEDLINE | ID: mdl-37254295

ABSTRACT

Overcoming lysogenization defect (OLD) proteins are a conserved family of ATP-powered nucleases that function in anti-phage defense. Recent bioinformatic, genetic, and crystallographic studies have yielded new insights into the structure, function, and evolution of these enzymes. Here we review these developments and propose a new classification scheme to categorize OLD homologs that relies on gene neighborhoods, biochemical properties, domain organization, and catalytic machinery. This taxonomy reveals important similarities and differences between family members and provides a blueprint to contextualize future in vivo and in vitro findings. We also detail how OLD nucleases are related to PARIS and Septu anti-phage defense systems and discuss important mechanistic questions that remain unanswered.


Subject(s)
Bacteria , Bacteriophages , Esterases , Bacteriophages/physiology , Bacteria/enzymology , Bacteria/virology , Esterases/chemistry , Exodeoxyribonuclease V , Adenosine Triphosphatases/chemistry
3.
Curr Protoc ; 3(2): e656, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36779782

ABSTRACT

The bacterial chromosome and bacterial plasmids can be engineered in vivo by homologous recombination using either PCR products or synthetic double-stranded DNA (dsDNA) or single-stranded DNA as substrates. Multiple linear dsDNA molecules can be assembled into an intact plasmid. The technology of recombineering is possible because bacteriophage-encoded recombination proteins efficiently recombine sequences with homologies as short as 35 to 50 bases. Recombineering allows DNA sequences to be inserted or deleted without regard to the location of restriction sites and can also be used in combination with CRISPR/Cas targeting systems. © 2023 Wiley Periodicals LLC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA. Basic Protocol: Making electrocompetent cells and transforming with linear DNA Support Protocol 1: Selection/counter-selections for genome engineering Support Protocol 2: Creating and screening for oligo recombinants by PCR Support Protocol 3: Other methods of screening for unselected recombinants Support Protocol 4: Curing recombineering plasmids containing a temperature-sensitive replication function Support Protocol 5: Removal of the prophage by recombineering Alternate Protocol 1: Using CRISPR/Cas9 as a counter-selection following recombineering Alternate Protocol 2: Assembly of linear dsDNA fragments into functional plasmids Alternate Protocol 3: Retrieval of alleles onto a plasmid by gap repair Alternate Protocol 4: Modifying multicopy plasmids with recombineering Support Protocol 6: Screening for unselected plasmid recombinants Alternate Protocol 5: Recombineering with an intact λ prophage Alternate Protocol 6: Targeting an infecting λ phage with the defective prophage strains.


Subject(s)
Escherichia coli , Homologous Recombination , Humans , Escherichia coli/genetics , Genetic Engineering/methods , Plasmids/genetics , Polymerase Chain Reaction
4.
Curr Protoc ; 2(12): e605, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36546891

ABSTRACT

The technology of recombineering, in vivo genetic engineering, was initially developed in Escherichia coli and uses bacteriophage-encoded homologous recombination proteins to efficiently recombine DNA at short homologies (35 to 50 nt). Because the technology is homology driven, genomic DNA can be modified precisely and independently of restriction site location. Recombineering uses linear DNA substrates that are introduced into the cell by electroporation; these can be PCR products, synthetic double-strand DNA (dsDNA), or single-strand DNA (ssDNA). Here we describe the applications, challenges, and factors affecting ssDNA and dsDNA recombineering in a variety of non-model bacteria, both Gram-negative and -positive, and recent breakthroughs in the field. We list different microbes in which the widely used phage λ Red and Rac RecET recombination systems have been used for in vivo genetic engineering. New homologous ssDNA and dsDNA recombineering systems isolated from non-model bacteria are also described. The Basic Protocol outlines a method for ssDNA recombineering in the non-model species of Shewanella. The Alternate Protocol describes the use of CRISPR/Cas as a counter-selection system in conjunction with recombineering to enhance recovery of recombinants. We provide additional background information, pertinent considerations for experimental design, and parameters critical for success. The design of ssDNA oligonucleotides (oligos) and various internet-based tools for oligo selection from genome sequences are also described, as is the use of oligo-mediated recombination. This simple form of genome editing uses only ssDNA oligo(s) and does not require an exogenous recombination system. The information presented here should help researchers identify a recombineering system suitable for their microbe(s) of interest. If no system has been characterized for a specific microbe, researchers can find guidance in developing a recombineering system from scratch. We provide a flowchart of decision-making paths for strategically applying annealase-dependent or oligo-mediated recombination in non-model and undomesticated bacteria. © 2022 Wiley Periodicals LLC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA. Basic Protocol: ssDNA recombineering in Shewanella species Alternate Protocol: ssDNA recombineering coupled to CRISPR/Cas9 in Shewanella species.


Subject(s)
Bacteria , Gene Editing , Humans , Bacteria/genetics , Homologous Recombination , Base Sequence , DNA, Single-Stranded/genetics
5.
Phage (New Rochelle) ; 3(3): 153-164, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36204488

ABSTRACT

The phage λ rexA and rexB genes are expressed from the P RM promoter in λ lysogens along with the cI repressor gene. RexB is also expressed from a second promoter, P LIT, embedded in rexA. The combined expression of rexA and rexB causes Escherichia coli to be more ultraviolet (UV) sensitive. Sensitivity is further increased when RexB levels are reduced by a defect in the P LIT promoter, thus the degree of sensitivity can be modulated by the ratio of RexA/RexB. Expression of the phage λ ren gene rescues this host UV sensitive phenotype; Ren also rescues an aberrant lysis phenotype caused by RexA and RexB. We screened an E. coli two-hybrid library to identify bacterial proteins with which each of these phage proteins physically interact. The results extend previous observations concerning λ Rex exclusion and show the importance of E. coli electron transport and sulfur assimilation pathways for the phage.

6.
Mol Microbiol ; 116(4): 1044-1063, 2021 10.
Article in English | MEDLINE | ID: mdl-34379857

ABSTRACT

The CI and Cro repressors of bacteriophage λ create a bistable switch between lysogenic and lytic growth. In λ lysogens, CI repressor expressed from the PRM promoter blocks expression of the lytic promoters PL and PR to allow stable maintenance of the lysogenic state. When lysogens are induced, CI repressor is inactivated and Cro repressor is expressed from the lytic PR promoter. Cro repressor blocks PRM transcription and CI repressor synthesis to ensure that the lytic state proceeds. RexA and RexB proteins, like CI, are expressed from the PRM promoter in λ lysogens; RexB is also expressed from a second promoter, PLIT , embedded in rexA. Here we show that RexA binds CI repressor and assists the transition from lysogenic to lytic growth, using both intact lysogens and defective prophages with reporter genes under the control of the lytic PL and PR promoters. Once lytic growth begins, if the bistable switch does return to the immune state, RexA expression lessens the probability that it will remain there, thus stabilizing the lytic state and activation of the lytic PL  and PR  promoters. RexB modulates the effect of RexA and may also help establish phage DNA replication as lytic growth ensues.


Subject(s)
Bacteriophage lambda/physiology , DNA Replication , Lysogeny , Repressor Proteins/metabolism , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , DNA, Viral , Gene Expression Regulation, Viral , Genes, Viral , Promoter Regions, Genetic , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism
7.
Mol Microbiol ; 112(6): 1798-1813, 2019 12.
Article in English | MEDLINE | ID: mdl-31545538

ABSTRACT

Genetic elements in the bacteriophage λ immunity region contribute to stable maintenance and synchronous induction of the integrated Escherichia coli prophage. There is a bistable switch between lysogenic and lytic growth that is orchestrated by the CI and Cro repressors acting on the lytic (PL and PR ) and lysogenic (PRM ) promoters, referred to as the Genetic Switch. Other less well-characterized elements in the phage immunity region include the PLIT promoter and the immunity terminator, TIMM . The PLIT promoter is repressed by the bacterial LexA protein in λ lysogens. LexA repressor, like the λ CI repressor, is inactivated during the SOS response to DNA damage, and this regulation ensures that the PLIT promoter and the lytic PL and PR promoters are synchronously activated. Proper RexA and RexB protein levels are critical for the switch from lysogeny to lytic growth. Mutation of PLIT reduces RexB levels relative to RexA, compromising cellular energetics and causing a 10-fold reduction in lytic phage yield. The RexA and RexB proteins interact with themselves and each other in a bacterial two-hybrid system. We also find that the transcription terminator, TIMM , is a Rho-independent, intrinsic terminator. Inactivation of TIMM has minimal effect on λ lysogenization or prophage induction.


Subject(s)
Bacteriophage lambda/genetics , Bacteriophage lambda/physiology , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism , Bacterial Proteins/metabolism , DNA, Viral/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Gene Expression Regulation, Viral/genetics , Genes, Switch/genetics , Genes, Viral/genetics , Lysogeny/genetics , Mutation , Promoter Regions, Genetic/genetics , Repressor Proteins , Serine Endopeptidases/metabolism , Transcription, Genetic , Viral Regulatory and Accessory Proteins , Virus Activation
8.
ACS Synth Biol ; 8(8): 1877-1889, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31277550

ABSTRACT

Dissimilatory metal-reducing bacteria, particularly those from the genus Shewanella, are of importance for bioremediation of metal contaminated sites and sustainable energy production. However, studies on this species have suffered from a lack of effective genetic tools for precise and high throughput genome manipulation. Here we report the development of a highly efficient system based on single-stranded DNA oligonucleotide recombineering coupled with CRISPR/Cas9-mediated counter-selection. Our system uses two plasmids: a sgRNA targeting vector and an editing vector, the latter harboring both Cas9 and the phage recombinase W3 Beta. Following the experimental analysis of Cas9 activity, we demonstrate the ability of this system to efficiently and precisely engineer different Shewanella strains with an average efficiency of >90% among total transformed cells, compared to ≃5% by recombineering alone, and regardless of the gene modified. We also show that different genetic changes can be introduced: mismatches, deletions, and small insertions. Surprisingly, we found that use of CRISPR/Cas9 alone allows selection of recombinase-independent S. oneidensis mutations, albeit at lower efficiency and frequency. With synthesized single-stranded DNA as substrates for homologous recombination and Cas9 as a counter-selectable marker, this new system provides a rapid, scalable, versatile, and scarless tool that will accelerate progress in Shewanella genomic engineering.


Subject(s)
CRISPR-Cas Systems/genetics , Shewanella/enzymology , Shewanella/metabolism , Bacteria/enzymology , Bacteria/genetics , Bacteria/metabolism , CRISPR-Associated Protein 9/genetics , DNA, Single-Stranded/genetics , Electroporation , Gene Editing , Mutation/genetics , Plasmids/genetics , Recombinases/genetics , Recombinases/metabolism , Shewanella/genetics
9.
Sci Rep ; 9(1): 39, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30631105

ABSTRACT

Shewanella oneidensis MR-1 is an invaluable host for the discovery and engineering of pathways important for bioremediation of toxic and radioactive metals and understanding extracellular electron transfer. However, genetic manipulation is challenging due to the lack of genetic tools. Previously, the only reliable method used for introducing DNA into Shewanella spp. at high efficiency was bacterial conjugation, enabling transposon mutagenesis and targeted knockouts using suicide vectors for gene disruptions. Here, we describe development of a robust and simple electroporation method in S. oneidensis that allows an efficiency of ~4.0 x 106 transformants/µg DNA. High transformation efficiency is maintained when cells are frozen for long term storage. In addition, we report a new prophage-mediated genome engineering (recombineering) system using a λ Red Beta homolog from Shewanella sp. W3-18-1. By targeting two different chromosomal alleles, we demonstrate its application for precise genome editing using single strand DNA oligonucleotides and show that an efficiency of ~5% recombinants among total cells can be obtained. This is the first effective and simple strategy for recombination with markerless mutations in S. oneidensis. Continued development of this recombinant technology will advance high-throughput and genome modification efforts to engineer and investigate S. oneidensis and other environmental bacteria.


Subject(s)
Electroporation/methods , Gene Editing/methods , Genetics, Microbial/methods , Oligonucleotides/genetics , Shewanella/genetics , Recombination, Genetic
10.
mBio ; 7(5)2016 09 13.
Article in English | MEDLINE | ID: mdl-27624131

ABSTRACT

UNLABELLED: Recombineering, in vivo genetic engineering with bacteriophage homologous recombination systems, is a powerful technique for making genetic modifications in bacteria. Two systems widely used in Escherichia coli are the Red system from phage λ and RecET from the defective Rac prophage. We investigated the in vivo dependence of recombineering on DNA replication of the recombining substrate using plasmid targets. For λ Red recombination, when DNA replication of a circular target plasmid is prevented, recombination with single-stranded DNA oligonucleotides is greatly reduced compared to that under replicating conditions. For RecET recombination, when DNA replication of the targeted plasmid is prevented, the recombination frequency is also reduced, to a level identical to that seen for the Red system in the absence of replication. The very low level of oligonucleotide recombination observed in the absence of any phage recombination functions is the same in the presence or absence of DNA replication. In contrast, both the Red and RecET systems recombine a nonreplicating linear dimer plasmid with high efficiency to yield a circular monomer. Therefore, the DNA replication requirement is substrate dependent. Our data are consistent with recombination by both the Red and RecET systems occurring predominately by single-strand annealing rather than by strand invasion. IMPORTANCE: Bacteriophage homologous recombination systems are widely used for in vivo genetic engineering in bacteria. Single- or double-stranded linear DNA substrates containing short flanking homologies to chromosome targets are used to generate precise and accurate genetic modifications when introduced into bacteria expressing phage recombinases. Understanding the molecular mechanism of these recombination systems will facilitate improvements in the technology. Here, two phage-specific systems are shown to require exposure of complementary single-strand homologous targets for efficient recombination; these single-strand regions may be created during DNA replication or by single-strand exonuclease digestion of linear duplex DNA. Previously, in vitro studies reported that these recombinases promote the single-strand annealing of two complementary DNAs and also strand invasion of a single DNA strand into duplex DNA to create a three-stranded region. Here, in vivo experiments show that recombinase-mediated annealing of complementary single-stranded DNA is the predominant recombination pathway in E. coli.


Subject(s)
Bacteriophage lambda/genetics , DNA Replication , Escherichia coli/genetics , Escherichia coli/virology , Prophages/genetics , Recombination, Genetic
11.
FEMS Microbiol Lett ; 363(3)2016 Feb.
Article in English | MEDLINE | ID: mdl-26705574

ABSTRACT

We describe a genetic ß-galactoside reporter system using a disk diffusion assay on MacConkey Lactose agar petri plates to monitor maintenance of the bacteriophage λ prophage state and viral induction in Escherichia coli K-12. Evidence is presented that the phage λ major lytic promoters, pL and pR, are activated when cells containing the reporters are exposed to the energy poison carbonyl cyanide m-chlorophenyl hydrazine, CCCP. This uncoupler of oxidative phosphorylation inhibits ATP synthesis by collapsing the proton motive force. Expression of the λ lytic promoters in response to CCCP requires host RecA function and an autocleavable CI repressor, as does SOS induction of the λ prophage that occurs by a DNA damage-dependent pathway. λ Cro function is required for CCCP-mediated activation of the λ lytic promoters. CCCP does not induce an sfi-lacZ SOS reporter.


Subject(s)
Bacteriophage lambda/drug effects , Bacteriophage lambda/physiology , Carbonyl Cyanide m-Chlorophenyl Hydrazone/metabolism , Escherichia coli K12/drug effects , Escherichia coli K12/virology , Lysogeny/drug effects , Virus Activation/drug effects , Artificial Gene Fusion , Genes, Reporter , Promoter Regions, Genetic , Proton-Motive Force/drug effects , beta-Galactosidase/analysis , beta-Galactosidase/genetics
12.
Curr Protoc Mol Biol ; 106: 1.16.1-1.16.39, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24733238

ABSTRACT

The bacterial chromosome and bacterial plasmids can be engineered in vivo by homologous recombination using PCR products and synthetic oligonucleotides as substrates. This is possible because bacteriophage-encoded recombination proteins efficiently recombine sequences with homologies as short as 35 to 50 bases. Recombineering allows DNA sequences to be inserted or deleted without regard to location of restriction sites. This unit first describes preparation of electrocompetent cells expressing the recombineering functions and their transformation with dsDNA or ssDNA. It then presents support protocols that describe several two-step selection/counter-selection methods of making genetic alterations without leaving any unwanted changes in the targeted DNA, and a method for retrieving onto a plasmid a genetic marker (cloning by retrieval) from the Escherichia coli chromosome or a co-electroporated DNA fragment. Additional protocols describe methods to screen for unselected mutations, removal of the defective prophage from recombineering strains, and other useful techniques.


Subject(s)
DNA, Bacterial/genetics , Escherichia coli/genetics , Genetic Engineering/methods , Homologous Recombination , Polymerase Chain Reaction/methods , Humans
13.
PLoS Genet ; 10(3): e1004217, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24651041

ABSTRACT

Assembly of the essential, tubulin-like FtsZ protein into a ring-shaped structure at the nascent division site determines the timing and position of cytokinesis in most bacteria and serves as a scaffold for recruitment of the cell division machinery. Here we report that expression of bacteriophage λ kil, either from a resident phage or from a plasmid, induces filamentation of Escherichia coli cells by rapid inhibition of FtsZ ring formation. Mutant alleles of ftsZ resistant to the Kil protein map to the FtsZ polymer subunit interface, stabilize FtsZ ring assembly, and confer increased resistance to endogenous FtsZ inhibitors, consistent with Kil inhibiting FtsZ assembly. Cells with the normally essential cell division gene zipA deleted (in a modified background) display normal FtsZ rings after kil expression, suggesting that ZipA is required for Kil-mediated inhibition of FtsZ rings in vivo. In support of this model, point mutations in the C-terminal FtsZ-interaction domain of ZipA abrogate Kil activity without discernibly altering FtsZ-ZipA interactions. An affinity-tagged-Kil derivative interacts with both FtsZ and ZipA, and inhibits sedimentation of FtsZ filament bundles in vitro. Together, these data inspire a model in which Kil interacts with FtsZ and ZipA in the cell to prevent FtsZ assembly into a coherent, division-competent ring structure. Phage growth assays show that kil+ phage lyse ∼30% later than kil mutant phage, suggesting that Kil delays lysis, perhaps via its interaction with FtsZ and ZipA.


Subject(s)
Bacterial Proteins/genetics , Bacteriophage lambda/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cytokinesis/genetics , Cytoskeletal Proteins/genetics , Escherichia coli Proteins/genetics , Viral Proteins/genetics , Escherichia coli/genetics , Peptides/genetics , Peptides/metabolism , Protein Binding
14.
Methods Enzymol ; 533: 79-102, 2013.
Article in English | MEDLINE | ID: mdl-24182919

ABSTRACT

A 'gene knockout' or 'knockout' is a mutation that inactivates a gene function. These mutations are very useful for classical genetic studies as well as for modern techniques including functional genomics. In the past, knockouts of bacterial genes were often made by transposon mutagenesis. In this case, laborious screens are required to find a knockout in the gene of interest. Knockouts of other organisms have traditionally been made by first using in vitro genetic engineering to modify genes contained on plasmids or bacterial artificial chromosomes (BACs) and later moving these modified constructs to the organism of interest by cell culture techniques. Other methods utilizing a combination of genetic engineering and in vivo homologous recombination were inefficient at best. Recombineering provides a new way to generate knockout mutations directly on the bacterial chromosome or to modify any plasmid or BAC in vivo as a prelude to making knockouts in other organisms. The constructs are designed to the base pair and are not dependent on suitable restriction sites. A drug cassette can be placed anywhere within a gene or the open reading frame of the gene can be replaced with the drug cassette. Either way, the desired construct is selected for.


Subject(s)
Drug Resistance, Bacterial/genetics , Gene Knockout Techniques/methods , Genetic Engineering/methods , Anti-Bacterial Agents/pharmacology , Bacteriophage lambda/drug effects , Bacteriophage lambda/genetics , Chromosomes, Bacterial , DNA Primers , DNA-Binding Proteins/genetics , Gene Knockout Techniques/instrumentation , Genetic Engineering/instrumentation , Mutation , Plasmids , Polymerase Chain Reaction/methods , Viral Proteins/genetics
15.
Methods Enzymol ; 533: 157-77, 2013.
Article in English | MEDLINE | ID: mdl-24182922

ABSTRACT

Recombineering provides the ability to make rapid, precise, and inexpensive genetic alterations to any DNA sequence, either in the chromosome or cloned onto a vector that replicates in E. coli (or other recombineering-proficient bacteria), and to do so in a highly efficient manner. Complicated genetic constructs that are impossible to make with in vitro genetic engineering can be created in days with recombineering. Recombineering with single-strand DNA (ssDNA) can be used to create single or multiple clustered point mutations, small or large (up to 10kb) deletions, and small (10-20 base) insertions such as sequence tags. Using optimized conditions, point mutations can be made with such high frequencies that they can be found without selection. This technology excels at creating both directed and random mutations.


Subject(s)
Genetic Engineering/methods , Oligonucleotides/genetics , DNA Mismatch Repair/genetics , DNA Primers , DNA, Single-Stranded , Electroporation/instrumentation , Electroporation/methods , Escherichia coli/genetics , Genetic Engineering/instrumentation , Homologous Recombination , Mutation , Point Mutation , Polymerase Chain Reaction/methods
16.
Nucleic Acids Res ; 41(22): e204, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24203710

ABSTRACT

The two-step process of selection and counter-selection is a standard way to enable genetic modification and engineering of bacterial genomes using homologous recombination methods. The tetA and sacB genes are contained in a DNA cassette and confer a novel dual counter-selection system. Expression of tetA confers bacterial resistance to tetracycline (Tc(R)) and also causes sensitivity to the lipophillic chelator fusaric acid; sacB causes sensitivity to sucrose. These two genes are introduced as a joint DNA cassette into Escherichia coli by selection for Tc(R). A medium containing both fusaric acid and sucrose has been developed, in which, coexpression of tetA-sacB is orders of magnitude more sensitive as a counter-selection agent than either gene alone. In conjunction with the homologous recombination methods of recombineering and P1 transduction, this powerful system has been used to select changes in the bacterial genome that cannot be directly detected by other counter-selection systems.


Subject(s)
Antiporters/genetics , Bacterial Proteins/genetics , Escherichia coli/genetics , Genetic Engineering/methods , Hexosyltransferases/genetics , Recombination, Genetic , Transduction, Genetic , Culture Media , Escherichia coli Proteins/genetics , Gene Fusion , Sucrose/metabolism
17.
Mol Microbiol ; 88(5): 906-20, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23634873

ABSTRACT

Synthetic single-strand oligonucleotides (oligos) with homology to genomic DNA have proved to be highly effective for constructing designed mutations in targeted genomes, a process referred to as recombineering. The cellular functions important for this type of homologous recombination have yet to be determined. Towards this end, we have identified Escherichia coli functions that process the recombining oligo and affect bacteriophage λ Red-mediated oligo recombination. To determine the nature of oligo processing during recombination, each oligo contained multiple nucleotide changes: a single base change allowing recombinant selection, and silent changes serving as genetic markers to determine the extent of oligo processing during the recombination. Such oligos were often not incorporated into the host chromosome intact; many were partially degraded in the process of recombination. The position and number of these silent nucleotide changes within the oligo strongly affect both oligo processing and recombination frequency. Exonucleases, especially those associated with DNA Polymerases I and III, affect inheritance of the silent nucleotide changes in the oligos. We demonstrate for the first time that the major DNA polymerases (Pol I and Pol III) and DNA ligase are directly involved with oligo recombination.


Subject(s)
DNA Polymerase III/metabolism , DNA Polymerase I/metabolism , DNA, Bacterial/metabolism , Escherichia coli/enzymology , Oligonucleotides/metabolism , Recombination, Genetic , Bacteriophage lambda/genetics , DNA Ligase ATP , DNA Ligases/metabolism , Escherichia coli/genetics
18.
Plasmid ; 67(2): 148-54, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22252137

ABSTRACT

Circularized regions of the chromosome containing the origin of replication, oriC, can be maintained as autonomous minichromosomes, oriC plasmids. We show that oriC plasmids containing precise, pre-determined segments of the chromosome can be generated by a simple in vivo recombineering technique. We generated two such plasmids carrying fluorescent markers. These were transferred to a recipient strain with a different fluorescent marker near the chromosomal copy of oriC. Thus the fates of the oriC plasmid and chromosomal origins could be followed independently in living cells by fluorescence microscopy. In contrast to a previous report, we show that there is a strong tendency of oriC plasmid copies to accumulate at the cell center as a single or double focus at the plane of cell division. This is not simply due to exclusion from the nucleoid space but rather appears to be a specific recognition and retention of the plasmid by some central-located cell site.


Subject(s)
Chromosome Segregation , Chromosomes, Bacterial , Escherichia coli/genetics , Plasmids/genetics , Recombination, Genetic , Biological Transport , Escherichia coli/metabolism , Gene Order , Open Reading Frames , Plasmids/metabolism
19.
J Mol Biol ; 407(1): 45-59, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21256136

ABSTRACT

Recombination with single-strand DNA oligonucleotides (oligos) in Escherichia coli is an efficient and rapid way to modify replicons in vivo. The generation of nucleotide alteration by oligo recombination provides novel assays for studying cellular processes. Single-strand exonucleases inhibit oligo recombination, and recombination is increased by mutating all four known exonucleases. Increasing oligo concentration or adding nonspecific carrier oligo titrates out the exonucleases. In a model for oligo recombination, λ Beta protein anneals the oligo to complementary single-strand DNA at the replication fork. Mismatches are created, and the methyl-directed mismatch repair (MMR) system acts to eliminate the mismatches inhibiting recombination. Three ways to evade MMR through oligo design include, in addition to the desired change (1) a C·C mismatch 6 bp from that change; (2) four or more adjacent mismatches; or (3) mismatches at four or more consecutive wobble positions. The latter proves useful for making high-frequency changes that alter only the target amino acid sequence and even allows modification of essential genes. Efficient uptake of DNA is important for oligo-mediated recombination. Uptake of oligos or plasmids is dependent on media and is 10,000-fold reduced for cells grown in minimal versus rich medium. Genomewide engineering technologies utilizing recombineering will benefit from both optimized recombination frequencies and a greater understanding of how biological processes such as DNA replication and cell division impact recombinants formed at multiple chromosomal loci. Recombination events at multiple loci in individual cells are described here.


Subject(s)
DNA Repair , DNA Replication , DNA, Single-Stranded/genetics , Escherichia coli/genetics , Oligonucleotides/pharmacology , Recombination, Genetic , Escherichia coli/growth & development , Escherichia coli/metabolism , Plasmids
20.
Nat Protoc ; 4(2): 206-23, 2009.
Article in English | MEDLINE | ID: mdl-19180090

ABSTRACT

Recombineering is an efficient method of in vivo genetic engineering applicable to chromosomal as well as episomal replicons in Escherichia coli. This method circumvents the need for most standard in vitro cloning techniques. Recombineering allows construction of DNA molecules with precise junctions without constraints being imposed by restriction enzyme site location. Bacteriophage homologous recombination proteins catalyze these recombineering reactions using double- and single-stranded linear DNA substrates, so-called targeting constructs, introduced by electroporation. Gene knockouts, deletions and point mutations are readily made, gene tags can be inserted and regions of bacterial artificial chromosomes or the E. coli genome can be subcloned by gene retrieval using recombineering. Most of these constructs can be made within about 1 week's time.


Subject(s)
Escherichia coli/metabolism , Genetic Engineering/methods , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , DNA, Bacterial/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/physiology , Plasmids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...