Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(37): e2123092119, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36067314

RESUMO

Levels of the cellular dNTPs, the direct precursors for DNA synthesis, are important for DNA replication fidelity, cell cycle control, and resistance against viruses. Escherichia coli encodes a dGTPase (2'-deoxyguanosine-5'-triphosphate [dGTP] triphosphohydrolase [dGTPase]; dgt gene, Dgt) that establishes the normal dGTP level required for accurate DNA replication but also plays a role in protecting E. coli against bacteriophage T7 infection by limiting the dGTP required for viral DNA replication. T7 counteracts Dgt using an inhibitor, the gene 1.2 product (Gp1.2). This interaction is a useful model system for studying the ongoing evolutionary virus/host "arms race." We determined the structure of Gp1.2 by NMR spectroscopy and solved high-resolution cryo-electron microscopy structures of the Dgt-Gp1.2 complex also including either dGTP substrate or GTP coinhibitor bound in the active site. These structures reveal the mechanism by which Gp1.2 inhibits Dgt and indicate that Gp1.2 preferentially binds the GTP-bound form of Dgt. Biochemical assays reveal that the two inhibitors use different modes of inhibition and bind to Dgt in combination to yield enhanced inhibition. We thus propose an in vivo inhibition model wherein the Dgt-Gp1.2 complex equilibrates with GTP to fully inactivate Dgt, limiting dGTP hydrolysis and preserving the dGTP pool for viral DNA replication.


Assuntos
Bacteriófago T7 , Proteínas de Escherichia coli , Escherichia coli , GTP Fosfo-Hidrolases , Guanosina Trifosfato , Proteínas Virais , Bacteriófago T7/fisiologia , Microscopia Crioeletrônica , Replicação do DNA , DNA Viral/metabolismo , Escherichia coli/enzimologia , Escherichia coli/virologia , Proteínas de Escherichia coli/química , GTP Fosfo-Hidrolases/metabolismo , Guanosina Trifosfato/metabolismo , Conformação Proteica , Proteínas Virais/química , Replicação Viral
2.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34417311

RESUMO

In most bacteriophages, genome transport across bacterial envelopes is carried out by the tail machinery. In viruses of the Podoviridae family, in which the tail is not long enough to traverse the bacterial wall, it has been postulated that viral core proteins assembled inside the viral head are translocated and reassembled into a tube within the periplasm that extends the tail channel. Bacteriophage T7 infects Escherichia coli, and despite extensive studies, the precise mechanism by which its genome is translocated remains unknown. Using cryo-electron microscopy, we have resolved the structure of two different assemblies of the T7 DNA translocation complex composed of the core proteins gp15 and gp16. Gp15 alone forms a partially folded hexamer, which is further assembled upon interaction with gp16 into a tubular structure, forming a channel that could allow DNA passage. The structure of the gp15-gp16 complex also shows the location within gp16 of a canonical transglycosylase motif involved in the degradation of the bacterial peptidoglycan layer. This complex docks well in the tail extension structure found in the periplasm of T7-infected bacteria and matches the sixfold symmetry of the phage tail. In such cases, gp15 and gp16 that are initially present in the T7 capsid eightfold-symmetric core would change their oligomeric state upon reassembly in the periplasm. Altogether, these results allow us to propose a model for the assembly of the core translocation complex in the periplasm, which furthers understanding of the molecular mechanism involved in the release of T7 viral DNA into the bacterial cytoplasm.


Assuntos
Bacteriófago T7/fisiologia , DNA Viral/fisiologia , Translocação Genética , Proteínas do Core Viral/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Bacteriófago T7/genética , Microscopia Crioeletrônica , Regulação Viral da Expressão Gênica , Processamento de Imagem Assistida por Computador , Microscopia Eletrônica , Modelos Moleculares , Morfolinos , Conformação Proteica , Proteínas do Core Viral/genética
3.
Chembiochem ; 22(18): 2805-2813, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34240805

RESUMO

Over the past decades, starting from crude cell extracts, a variety of successful preparation protocols and optimized reaction conditions have been established for the production of cell-free gene expression systems. One of the crucial steps during the preparation of cell extract-based expression systems is the cell lysis procedure itself, which largely determines the quality of the active components of the extract. Here we evaluate the utility of an E. coli cell extract, which was prepared using a combination of lysozyme incubation and a gentle sonication step. As quality measure, we demonstrate the cell-free expression of YFP at concentrations up to 0.6 mg/mL. In addition, we produced and assembled T7 bacteriophages up to a titer of 108  PFU/mL. State-of-the-art quantitative proteomics was used to compare the produced extracts with each other and with a commercial extract. The differences in protein composition were surprisingly small between lysozyme-assisted sonication (LAS) extracts, but we observed an increase in the release of DNA-binding proteins for increasing numbers of sonication cycles. Proteins taking part in carbohydrate metabolism, glycolysis, amino acid and nucleotide related pathways were found to be more abundant in the LAS extract, while proteins related to RNA modification and processing, DNA modification and replication, transcription regulation, initiation, termination and the TCA cycle were found enriched in the commercial extract.


Assuntos
Bacteriófago T7/fisiologia , Escherichia coli/metabolismo , Expressão Gênica , Muramidase/metabolismo , Proteoma/análise , Proteômica/métodos , Escherichia coli/química , Espectrometria de Massas , Análise de Componente Principal , Sonicação , Montagem de Vírus
4.
Microbiol Spectr ; 9(1): e0014121, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34259542

RESUMO

Lytic bacteriophages are expected as effective tools to control infectious bacteria in human and pathogenic or spoilage bacteria in foods. Leaderless bacteriocins (LLBs) are simple bacteriocins produced by Gram-positive bacteria. LLBs do not possess an N-terminal leader peptide in the precursor, which means that they are active immediately after translation. In this study, we constructed a novel antimicrobial agent, an LLB-producing phage (LLB-phage), by genetic engineering to introduce the LLB structural gene into the lytic phage genome. To this end, lnqQ (structure gene of an LLB, lacticin Q) and trxA, an essential gene for T7 phage genome replication, were integrated in tandem into T7 phage genome using homologous recombination in Escherichia coli host strain. The recombinant lnqQ-T7 phage was isolated by a screening method using ΔtrxA host strain. lnqQ-T7 phage formed a clear halo in agar plates containing both E. coli and lacticin Q-susceptible Bacillus coagulans, indicating that lnqQ-T7 phage could produce a significant amount of lacticin Q. Lacticin Q production did not exert a significant effect on the lytic cycle of T7 phage. In fact, the production of lacticin Q enhanced T7 phage lytic activity and helped to prevent the emergence of bacterial populations resistant against this phage. These results serve as a proof of principle for LLB-phages. There are different types of LLBs and phages, meaning that in the future, it may be possible to produce any number of LLB-phages which can be designed to efficiently control different types of bacterial contamination in different settings. IMPORTANCE We demonstrated that we could combine LLB and phage to construct promising novel antimicrobial agents, LLB-phage. The first LLB-phage, lnqQ-T7 phage, can control the growth of both the Gram-negative host strain and neighboring Gram-positive bacteria while preventing the emergence of phage resistance in the host strain. There are several different types of LLBs and phages, suggesting that we may be able to design a battery of LLB-phages by selecting novel combinations of LLBs and phages. These constructs could be tailored to control various bacterial contaminations and infectious diseases.


Assuntos
Bacteriocinas/genética , Bacteriófago T7/genética , Escherichia coli/virologia , Bactérias Gram-Positivas/virologia , Bacteriocinas/metabolismo , Bacteriófago T7/fisiologia , Escherichia coli/fisiologia , Engenharia Genética , Bactérias Gram-Positivas/fisiologia , Replicação Viral
5.
J Virol ; 95(18): e0092021, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34191582

RESUMO

K1 capsule-specific phages of Escherichia coli have been reported in recent years, but the molecular mechanism involved in host recognition of these phages remains unknown. In this study, the interactions between PNJ1809-36, a new K1-specific phage, and its host bacterium, E. coli DE058, were investigated. A transposon mutation library was used to screen for receptor-related genes. Gene deletion, lysis curve determination, plaque formation test, adsorption assay, and inhibition assay of phage by lipopolysaccharide (LPS) showed that capsular polysaccharide (CPS) was the first receptor for the initial adsorption of PNJ1809-36 to E. coli DE058 and that LPS was a secondary receptor for the irreversible binding of the phage. The penultimate galactose in the outer core was identified as the specific binding region on LPS. Through antibody blocking assay, fluorescence labeling and high-performance gel permeation chromatography, the tail protein ORF261 of phage PNJ1809-36 was identified as the receptor-binding protein on CPS. Given these findings, we propose a model for the recognition process of phage PNJ1809-36 on E. coli DE058: the phage PNJ1809-36 tail protein ORF261 recognizes and adsorbs to the K1 capsule, and then the K1 capsule is partially degraded, exposing the active site of LPS which is recognized by phage PNJ1809-36. This model provides insight into the molecular mechanisms between K1-specific phages and their host bacteria. IMPORTANCE It has been speculated that CPS is the main receptor of K1-specific phages belonging to Siphoviridae. In recent years, a new type of K1-specific phage belonging to Myoviridae has been reported, but its host recognition mechanisms remain unknown. Here, we studied the interactions between PNJ1809-36, a new type of K1 phage, and its host bacterium, E. coli DE058. Our research showed that the phage initially adsorbed to the K1 capsule mediated by ORF261 and then bound to the penultimate galactose of LPS to begin the infection process.


Assuntos
Antígenos de Bactérias/metabolismo , Cápsulas Bacterianas/metabolismo , Bacteriófago T7/fisiologia , Escherichia coli/metabolismo , Lipopolissacarídeos/metabolismo , Polissacarídeos Bacterianos/metabolismo , Sequência de Aminoácidos , Escherichia coli/virologia , Homologia de Sequência de Aminoácidos
6.
Methods Mol Biol ; 2281: 323-332, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33847969

RESUMO

The single-stranded DNA-binding protein gp2.5 of bacteriophage T7 plays myriad functions in the replication of phage genomes. In addition to interacting with ssDNA, gp2.5 binds to the T7 DNA polymerase and primase/helicase proteins, regulating their enzymatic activities. Here we describe in vitro methods to examine the effects of gp2.5 on primer synthesis and extension by the T7 replisome.


Assuntos
Bacteriófago T7/fisiologia , Primers do DNA/síntese química , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Virais/metabolismo , Primers do DNA/genética , Replicação do DNA , DNA Viral/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Replicação Viral
7.
ACS Synth Biol ; 10(3): 459-465, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33577295

RESUMO

Cell-free systems allow interference with gene expression processes without requiring elaborate genetic engineering procedures. This makes it ideally suited for rapid prototyping of synthetic biological parts. Inspired by nature's strategies for the control of gene expression via short antisense RNA molecules, we here investigated the use of small DNA (sDNA) for translational inhibition in the context of cell-free protein expression. We designed sDNA molecules to be complementary to the ribosome binding site (RBS) and the downstream coding sequence of targeted mRNA molecules. Depending on sDNA concentration and the promoter used for transcription of the mRNA, this resulted in a reduction of gene expression of targeted genes by up to 50-fold. We applied the cell-free sDNA technique (CF-sDNA) to modulate cell-free gene expression from the native T7 phage genome by suppressing the production of the major capsid protein of the phage. This resulted in a reduced phage titer, but at the same time drastically improved cell-free replication of the phage genome, which we utilized to amplify the T7 genome by more than 15 000-fold in a droplet-based serial dilution experiment. Our simple antisense sDNA approach extends the possibilities to exert translational control in cell-free expression systems, which should prove useful for cell-free prototyping of native phage genomes and also cell-free phage manipulation.


Assuntos
Bacteriófago T7/fisiologia , DNA Antissenso/metabolismo , Inativação Gênica , Genoma Viral , Bacteriófago T7/genética , Proteínas do Capsídeo/antagonistas & inibidores , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Sistema Livre de Células , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Regiões Promotoras Genéticas , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Transcrição Gênica , Replicação Viral
8.
Nature ; 589(7840): 120-124, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32937646

RESUMO

Viperin is an interferon-induced cellular protein that is conserved in animals1. It has previously been shown to inhibit the replication of multiple viruses by producing the ribonucleotide 3'-deoxy-3',4'-didehydro (ddh)-cytidine triphosphate (ddhCTP), which acts as a chain terminator for viral RNA polymerase2. Here we show that eukaryotic viperin originated from a clade of bacterial and archaeal proteins that protect against phage infection. Prokaryotic viperins produce a set of modified ribonucleotides that include ddhCTP, ddh-guanosine triphosphate (ddhGTP) and ddh-uridine triphosphate (ddhUTP). We further show that prokaryotic viperins protect against T7 phage infection by inhibiting viral polymerase-dependent transcription, suggesting that it has an antiviral mechanism of action similar to that of animal viperin. Our results reveal a class of potential natural antiviral compounds produced by bacterial immune systems.


Assuntos
Antivirais/metabolismo , Proteínas Arqueais/metabolismo , Proteínas de Bactérias/metabolismo , Bacteriófago T7/imunologia , Evolução Molecular , Células Procarióticas/metabolismo , Proteínas/metabolismo , Antivirais/imunologia , Proteínas Arqueais/química , Bactérias/imunologia , Bactérias/metabolismo , Bactérias/virologia , Proteínas de Bactérias/química , Bacteriófago T7/enzimologia , Bacteriófago T7/fisiologia , DNA Polimerase Dirigida por DNA/metabolismo , Humanos , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Células Procarióticas/imunologia , Células Procarióticas/virologia , Proteínas/química , Proteínas/genética , Ribonucleotídeos/biossíntese , Ribonucleotídeos/química , Ribonucleotídeos/metabolismo , Transcrição Gênica/efeitos dos fármacos
9.
J Agric Food Chem ; 68(39): 10920-10927, 2020 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-32845633

RESUMO

To reduce the use of conventional chemical pesticides, naturally occurring biopesticides such as bacteriophages have emerged as a promising solution, but effectiveness of these biopesticides can be limited because of their UV and desiccation instability. This study developed a biopolymer formulation to improve the phage stability, enhance the antimicrobial activity of phages, and prevent bacterial contaminations on a leaf surface in the presence of UV-A. The mixture of microscale polydopamine (PDA) particles with whey protein isolate (WPI)-glycerol formulation was effective for enhancing the stability of T7 phages in spraying solution and on a model leaf surface during 4 h exposure to UV-A and 1 h exposure to the simulated sunlight, respectively. The T7 phages incorporated with the biopolymer formulation effectively improved the antimicrobial activity of phages, as exhibited by greater than 2.8 log reduction in model bacteria Escherichia coli BL21 and also illustrated by significant potential of this formulation to prevent bacterial contamination and colonization of the plant surface. In summary, this study illustrates that phages combined with a biopolymer formulation can be an effective approach for a field deployable biocontrol solution of bacterial contamination in the agricultural environment.


Assuntos
Antibiose , Bacteriófago T7/fisiologia , Bacteriófago T7/efeitos da radiação , Escherichia coli/virologia , Doenças das Plantas/virologia , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/fisiologia , Doenças das Plantas/prevenção & controle , Plantas/microbiologia , Plantas/virologia , Raios Ultravioleta
10.
Nucleic Acids Res ; 48(10): 5397-5406, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32338761

RESUMO

BREX (for BacteRiophage EXclusion) is a superfamily of common bacterial and archaeal defence systems active against diverse bacteriophages. While the mechanism of BREX defence is currently unknown, self versus non-self differentiation requires methylation of specific asymmetric sites in host DNA by BrxX (PglX) methyltransferase. Here, we report that T7 bacteriophage Ocr, a DNA mimic protein that protects the phage from the defensive action of type I restriction-modification systems, is also active against BREX. In contrast to the wild-type phage, which is resistant to BREX defence, T7 lacking Ocr is strongly inhibited by BREX, and its ability to overcome the defence could be complemented by Ocr provided in trans. We further show that Ocr physically associates with BrxX methyltransferase. Although BREX+ cells overproducing Ocr have partially methylated BREX sites, their viability is unaffected. The result suggests that, similar to its action against type I R-M systems, Ocr associates with as yet unidentified BREX system complexes containing BrxX and neutralizes their ability to both methylate and exclude incoming phage DNA.


Assuntos
Bacteriófago T7/fisiologia , Proteínas Virais/metabolismo , Bacteriófago T7/genética , Metilação de DNA , Metilases de Modificação do DNA/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/virologia , Plasmídeos , Proteínas Virais/genética
11.
ISME J ; 14(1): 123-134, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31578469

RESUMO

Bacteriophage shape the composition and function of microbial communities. Yet it remains difficult to predict the effect of phage on microbial interactions. Specifically, little is known about how phage influence mutualisms in networks of cross-feeding bacteria. We mathematically modeled the impacts of phage in a synthetic microbial community in which Escherichia coli and Salmonella enterica exchange essential metabolites. In this model, independent phage attack of either species was sufficient to temporarily inhibit both members of the mutualism; however, the evolution of phage resistance facilitated yields similar to those observed in the absence of phage. In laboratory experiments, attack of S. enterica with P22vir phage followed these modeling expectations of delayed community growth with little change in the final yield of bacteria. In contrast, when E. coli was attacked with T7 phage, S. enterica, the nonhost species, reached higher yields compared with no-phage controls. T7 infection increased nonhost yield by releasing consumable cell debris, and by driving evolution of partially resistant E. coli that secreted more carbon. Our results demonstrate that phage can have extensive indirect effects in microbial communities, that the nature of these indirect effects depends on metabolic and evolutionary mechanisms, and that knowing the degree of evolved resistance leads to qualitatively different predictions of bacterial community dynamics in response to phage attack.


Assuntos
Bacteriófago T7/fisiologia , Fagos de Salmonella/fisiologia , Simbiose , Escherichia coli/metabolismo , Escherichia coli/virologia , Salmonella enterica/metabolismo , Salmonella enterica/virologia
12.
Biochemistry ; 58(45): 4466-4479, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31659895

RESUMO

Recent structural studies of the bacteriophage T7 DNA replication system have shed light on how multiple proteins assemble to copy two antiparallel DNA strands. In T7, acidic C-terminal tails of both the primase-helicase and single-stranded DNA binding protein bind to two basic patches on the DNA polymerase to aid in replisome assembly, processivity, and coordinated DNA synthesis. Although these electrostatic interactions are essential for DNA replication, the molecular details for how these tails bind the polymerase are unknown. We have determined an X-ray crystal structure of the T7 DNA polymerase bound to both a primer/template DNA and a peptide that mimics the C-terminal tail of the primase-helicase. The structure reveals that the essential C-terminal phenylalanine of the tail binds to a hydrophobic pocket that is surrounded by positive charge on the surface of the polymerase. We show that alterations of polymerase residues that engage the tail lead to defects in viral replication. In the structure, we also observe dTTP bound in the exonuclease active site and stacked against tryptophan 160. Using both primer/extension assays and high-throughput sequencing, we show how mutations in the exonuclease active site lead to defects in mismatch repair and an increase in the level of mutagenesis of the T7 genome. Finally, using small-angle X-ray scattering, we provide the first solution structures of a complex between the single-stranded DNA binding protein and the DNA polymerase and show how a single-stranded DNA binding protein dimer engages both one and two copies of DNA polymerase.


Assuntos
Bacteriófago T7/fisiologia , DNA Polimerase Dirigida por DNA/química , Proteínas Virais/química , Bacteriófago T7/química , Domínio Catalítico , Cristalografia por Raios X , DNA Viral/química , DNA Viral/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Ligação Proteica , Eletricidade Estática , Proteínas Virais/metabolismo , Replicação Viral
13.
BMC Res Notes ; 12(1): 560, 2019 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-31488211

RESUMO

OBJECTIVE: Our immediate objective is to determine whether infectivity of lytic podophage T3 has a relatively high persistence in the blood of a mouse, as suggested by previous data. Secondarily, we determine whether the T3 surface has changed during this mouse passage. The surface is characterized by native agarose gel electrophoresis (AGE). Beyond our current data, the long-term objective is optimization of phages chosen for therapy of all bacteremias and associated sepsis. RESULTS: We find that the persistence of T3 in mouse blood is higher by over an order of magnitude than the previously reported persistence of (1) lysogenic phages lambda and P22, and (2) lytic phage T7, a T3 relative. We explain these differences via the lysogenic character of lambda and P22, and the physical properties of T7. For the future, we propose testing a new, AGE-based strategy for rapidly screening for high-persistence, lytic, environmental podophages that have phage therapy-promoting physical properties.


Assuntos
Bacteriemia/terapia , Bacteriófago T3/fisiologia , Terapia por Fagos/métodos , Sepse/terapia , Animais , Bacteriemia/sangue , Bacteriólise , Bacteriófago T7/fisiologia , Feminino , Camundongos Endogâmicos C57BL , Sepse/sangue
14.
Anal Bioanal Chem ; 411(27): 7273-7279, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31511947

RESUMO

Reporter phage systems have emerged as a promising technology for the detection of bacteria in foods and water. However, the sensitivity of these assays is often limited by the concentration of the expressed reporter as well as matrix interferences associated with the sample. In this study, bacteriophage T7 was engineered to overexpress mutated alkaline phosphatase fused to a carbohydrate-binding module (ALP*-CBM) following infection of E. coli to enable colorimetric detection in a model system. Magnetic cellulose particles were employed to separate and concentrate the overexpressed ALP*-CBM in bacterial lysate. Infection of E. coli with the engineered phage resulted in a limit of quantitation of 1.2 × 105 CFU, equating to 1.2 × 103 CFU/mL in 3.5 h when using a colorimetric assay and 100 mL sample volume. When employing an enrichment step, < 101 CFU/mL could be visually detected from a 100 mL sample volume within 8 h. These results suggest that affinity tag modified enzymes coupled with a material support can provide a simple and effective means to improve signal sensitivity of phage-based assays. Graphical abstract.


Assuntos
Bacteriófago T7/fisiologia , Colorimetria/métodos , Escherichia coli/isolamento & purificação , Genes Reporter , Fosfatase Alcalina/genética , Contagem de Colônia Microbiana , Engenharia Genética/métodos
15.
Nat Commun ; 10(1): 3746, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31431626

RESUMO

Double-stranded DNA bacteriophages package their genome at high pressure inside a procapsid through the portal, an oligomeric ring protein located at a unique capsid vertex. Once the DNA has been packaged, the tail components assemble on the portal to render the mature infective virion. The tail tightly seals the ejection conduit until infection, when its interaction with the host membrane triggers the opening of the channel and the viral genome is delivered to the host cell. Using high-resolution cryo-electron microscopy and X-ray crystallography, here we describe various structures of the T7 bacteriophage portal and fiber-less tail complex, which suggest a possible mechanism for DNA retention and ejection: a portal closed conformation temporarily retains the genome before the tail is assembled, whereas an open portal is found in the tail. Moreover, a fold including a seven-bladed ß-propeller domain is described for the nozzle tail protein.


Assuntos
Bacteriófago T7/fisiologia , Proteínas do Capsídeo/ultraestrutura , Capsídeo/ultraestrutura , Empacotamento do DNA , Modelos Moleculares , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica , Cristalografia por Raios X , DNA Viral/metabolismo , Domínios Proteicos
16.
Science ; 363(6429)2019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30679383

RESUMO

Visualization in atomic detail of the replisome that performs concerted leading- and lagging-DNA strand synthesis at a replication fork has not been reported. Using bacteriophage T7 as a model system, we determined cryo-electron microscopy structures up to 3.2-angstroms resolution of helicase translocating along DNA and of helicase-polymerase-primase complexes engaging in synthesis of both DNA strands. Each domain of the spiral-shaped hexameric helicase translocates sequentially hand-over-hand along a single-stranded DNA coil, akin to the way AAA+ ATPases (adenosine triphosphatases) unfold peptides. Two lagging-strand polymerases are attached to the primase, ready for Okazaki fragment synthesis in tandem. A ß hairpin from the leading-strand polymerase separates two parental DNA strands into a T-shaped fork, thus enabling the closely coupled helicase to advance perpendicular to the downstream DNA duplex. These structures reveal the molecular organization and operating principles of a replisome.


Assuntos
Bacteriófago T7/enzimologia , Bacteriófago T7/fisiologia , DNA Helicases/química , DNA Primase/química , DNA Polimerase Dirigida por DNA/química , Proteínas Virais/química , Replicação Viral , Microscopia Crioeletrônica , Domínios Proteicos
17.
Sci Rep ; 9(1): 620, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30679452

RESUMO

Bacteriophages (phages) or viruses that specifically infect bacteria have widely been studied as biocontrol agents against animal and plant bacterial diseases. They offer many advantages compared to antibiotics. The American Foulbrood (AFB) is a bacterial disease affecting honeybee larvae caused by Paenibacillus larvae. Phages can be very significant in fighting it mostly due to European restrictions to the use of antibiotics in beekeeping. New phages able to control P. larvae in hives have already been reported with satisfactory results. However, the efficacy and feasibility of administering phages indirectly to larvae through their adult workers only by providing phages in bees' feeders has never been evaluated. This strategy is considered herein the most feasible as far as hive management is concerned. This in vivo study investigated the ability of a phage to reach larvae in an infective state after oral administration to honeybees. The screening (by direct PFU count) and quantification (by quantitative PCR) of the phage in bee organs and in larvae after ingestion allowed us to conclude that despite 104 phages reaching larvae only an average of 32 were available to control the spread of the disease. The fast inactivation of many phages in royal jelly could compromise this therapeutic approach. The protection of phages from hive-derived conditions should be thus considered in further developments for AFB treatment.


Assuntos
Bacteriófago T7/fisiologia , Bacteriófago T7/patogenicidade , Abelhas/virologia , Larva/virologia , Animais , Escherichia coli/virologia , Imunofluorescência , Paenibacillus larvae/virologia
18.
J Biol Phys ; 44(2): 225-235, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29654426

RESUMO

We use the nano-dissection capabilities of atomic force microscopy to induce structural alterations on individual virus capsids in liquid milieu. We fracture the protein shells either with single nanoindentations or by increasing the tip-sample interaction force in amplitude modulation dynamic mode. The normal behavior is that these cracks persist in time. However, in very rare occasions they self-recuperate to retrieve apparently unaltered virus particles. In this work, we show the topographical evolution of three of these exceptional events occurring in T7 bacteriophage capsids. Our data show that single nanoindentation produces a local recoverable fracture that corresponds to the deepening of a capsomer. In contrast, imaging in dynamic mode induced cracks that separate the virus morphological subunits. In both cases, the breakage patterns follow intratrimeric loci.


Assuntos
Bacteriófago T7/metabolismo , Microscopia de Força Atômica , Bacteriófago T7/fisiologia , Fenômenos Biomecânicos , Capsídeo/química , Capsídeo/metabolismo , Fatores de Tempo , Vírion/química , Vírion/metabolismo
19.
Nat Commun ; 9(1): 695, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29449554

RESUMO

Introduction of synthetic circuits into microbes creates competition between circuit and host genes for shared cellular resources, such as ribosomes. This can lead to the emergence of unwanted coupling between the expression of different circuit genes, complicating the design process and potentially leading to circuit failure. By expressing a synthetic 16S rRNA with altered specificity, we can partition the ribosome pool into host-specific and circuit-specific activities. We show mathematically and experimentally that the effects of resource competition can be alleviated by targeting genes to different ribosomal pools. This division of labour can be used to increase flux through a metabolic pathway. We develop a model of cell physiology which is able to capture these observations and use it to design a dynamic resource allocation controller. When implemented, this controller acts to decouple genes by increasing orthogonal ribosome production as the demand for translational resources by a synthetic circuit increases.


Assuntos
Expressão Gênica , Redes Reguladoras de Genes/genética , RNA Ribossômico 16S/genética , Ribossomos/genética , Algoritmos , Bacteriófago T7/genética , Bacteriófago T7/fisiologia , Escherichia coli/genética , Escherichia coli/virologia , Interações Hospedeiro-Patógeno/genética , Modelos Genéticos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Ribossômico 16S/metabolismo , Ribossomos/metabolismo
20.
Nanoscale ; 10(4): 1898-1904, 2018 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-29318247

RESUMO

The foremost event of bacteriophage infection is the ejection of genomic material into the host bacterium after virus binding to surface receptor sites. How ejection is triggered is yet unknown. Here we show, in single mature T7 phage particles, that tapping the capsid wall with an oscillating atomic-force-microscope cantilever triggers rapid DNA ejection via the tail complex. The triggering rate increases exponentially as a function of force, following transition-state theory, across an activation barrier of 23 kcal mol-1 at 1.2 nm along the reaction coordinate. The conformation of the ejected DNA molecule revealed that it had been exposed to a propulsive force. This force, arising from intra-capsid pressure, assists in initiating the ejection process and the transfer of DNA across spatial dimensions beyond that of the virion. Chemical immobilization of the tail fibers also resulted in enhanced DNA ejection, suggesting that the triggering process might involve a conformational switch that can be mechanically activated either by external forces or via the tail-fiber complex.


Assuntos
Bacteriófago T7/fisiologia , Capsídeo , DNA Viral , Microscopia de Força Atômica , Vibração , Vírion
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA