Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 322
Filtrar
1.
Virology ; 596: 110101, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38754335

RESUMO

This study characterizes a newly isolated Demerecviridae phage, named vB_SalS_PSa2, belonging to the phage T5 group. The main variations between vB_SalS_PSa2 and T5 concern structural proteins related to morphology and host recognition. vB_SalS_PSa2 is infective to 19 out of the 25 tested Salmonella enterica (including the rare "Sendai" and "Equine" serotypes) and Escherichia coli isolates, most of them being multidrug resistant. vB_SalS_PSa2 displayed good thermal stability (4-60 °C) and broad pH stability (4.0-12.0). It also exhibited antibacterial activity against S. enterica sv. Paratyphi A Enb50 at 4 °C in milk during the whole tested period (5 d), and for 3-6 h at both 25 and 37 °C. Furthermore, vB_SalS_PSa2 was able to inhibit biofilm formation and to show degradation activity on mature biofilms of E. coli K12 and S. enterica sv. Paratyphi Enb50 in both LB and milk. Altogether, these results indicate that phage vB_SalS_PSa2 is a valuable candidate for controlling foodborne S. enterica and E. coli pathogens.


Assuntos
Escherichia coli , Salmonella enterica , Salmonella enterica/virologia , Escherichia coli/virologia , Leite/virologia , Animais , Microbiologia de Alimentos , Genoma Viral , Biofilmes/crescimento & desenvolvimento , Fagos de Salmonella/fisiologia , Fagos de Salmonella/isolamento & purificação , Fagos de Salmonella/classificação , Fagos de Salmonella/genética , Bacteriófagos/fisiologia , Bacteriófagos/genética , Bacteriófagos/classificação , Bacteriófagos/isolamento & purificação , Concentração de Íons de Hidrogênio , Filogenia , Especificidade de Hospedeiro
2.
Biochem Biophys Res Commun ; 712-713: 149915, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38663038

RESUMO

Viral infections pose a significant threat to public health, and the production of interferons represents one of the most critical antiviral innate immune responses of the host. Consequently, the screening and identification of compounds or reagents that induce interferon production are of paramount importance. This study commenced with the cultivation of host bacterium 15,597, followed by the infection of Escherichia coli with the MS2 bacteriophage. Utilizing the J2 capture technique, a class of dsRNA mixtures (MS2+15,597) was isolated from the E. coli infected with the MS2 bacteriophage. Subsequent investigations were conducted on the immunostimulatory activity of the MS2+15,597 mixture. The results indicated that the dsRNA mixtures (MS2+15,597) extracted from E. coli infected with the MS2 bacteriophage possess the capability to activate innate immunity, thereby inducing the production of interferon-ß. These dsRNA mixtures can activate the RIG-I and TLR3 pattern recognition receptors, stimulating the expression of interferon stimulatory factors 3/7, which in turn triggers the NF-κB signaling pathway, culminating in the cellular production of interferon-ß to achieve antiviral effects. This study offers novel insights and strategies for the development of broad-spectrum antiviral drugs, potentially providing new modalities for future antiviral therapies.


Assuntos
Escherichia coli , Levivirus , RNA de Cadeia Dupla , Escherichia coli/virologia , Escherichia coli/genética , Escherichia coli/metabolismo , RNA de Cadeia Dupla/metabolismo , Humanos , Levivirus/genética , Receptor 3 Toll-Like/metabolismo , Receptor 3 Toll-Like/genética , Imunidade Inata , Interferon beta/metabolismo , Interferon beta/genética , NF-kappa B/metabolismo , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/genética , Transdução de Sinais , Fator Regulador 7 de Interferon/metabolismo , Fator Regulador 7 de Interferon/genética , Receptores Imunológicos , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética
3.
Mol Cell ; 83(24): 4600-4613.e6, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38096825

RESUMO

In response to the persistent exposure to phage infection, bacteria have evolved diverse antiviral defense mechanisms. In this study, we report a bacterial two-component defense system consisting of a Sir2 NADase and a HerA helicase. Cryo-electron microscopy reveals that Sir2 and HerA assemble into a ∼1 MDa supramolecular octadecamer. Unexpectedly, this complex exhibits various enzymatic activities, including ATPase, NADase, helicase, and nuclease, which work together in a sophisticated manner to fulfill the antiphage function. Therefore, we name this defense system "Nezha" after a divine warrior in Chinese mythology who employs multiple weapons to defeat enemies. Our findings demonstrate that Nezha could sense phage infections, self-activate to arrest cell growth, eliminate phage genomes, and subsequently deactivate to allow for cell recovery. Collectively, Nezha represents a paradigm of sophisticated and multifaceted strategies bacteria use to defend against viral infections.


Assuntos
Caudovirales , Escherichia coli , Adenosina Trifosfatases , Microscopia Crioeletrônica , DNA Helicases , NAD+ Nucleosidase , Escherichia coli/enzimologia , Escherichia coli/virologia
4.
Mol Cell ; 83(24): 4586-4599.e5, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38096827

RESUMO

SIR2-HerA, a bacterial two-protein anti-phage defense system, induces bacterial death by depleting NAD+ upon phage infection. Biochemical reconstitution of SIR2, HerA, and the SIR2-HerA complex reveals a dynamic assembly process. Unlike other ATPases, HerA can form various oligomers, ranging from dimers to nonamers. When assembled with SIR2, HerA forms a hexamer and converts SIR2 from a nuclease to an NAD+ hydrolase, representing an unexpected regulatory mechanism mediated by protein assembly. Furthermore, high concentrations of ATP can inhibit NAD+ hydrolysis by the SIR2-HerA complex. Cryo-EM structures of the SIR2-HerA complex reveal a giant supramolecular assembly up to 1 MDa, with SIR2 as a dodecamer and HerA as a hexamer, crucial for anti-phage defense. Unexpectedly, the HerA hexamer resembles a spiral staircase and exhibits helicase activities toward dual-forked DNA. Together, we reveal the supramolecular assembly of SIR2-HerA as a unique mechanism for switching enzymatic activities and bolstering anti-phage defense strategies.


Assuntos
Proteínas de Escherichia coli , Escherichia coli , Sirtuínas , Fagos T , Adenosina Trifosfatases/genética , Proteínas de Bactérias/genética , NAD , Sirtuínas/metabolismo , Escherichia coli/enzimologia , Escherichia coli/virologia , Proteínas de Escherichia coli/metabolismo
5.
Nature ; 620(7976): 1054-1062, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37587340

RESUMO

The mechanisms by which viruses hijack the genetic machinery of the cells they infect are of current interest. When bacteriophage T4 infects Escherichia coli, it uses three different adenosine diphosphate (ADP)-ribosyltransferases (ARTs) to reprogram the transcriptional and translational apparatus of the host by ADP-ribosylation using nicotinamide adenine dinucleotide (NAD) as a substrate1,2. NAD has previously been identified as a 5' modification of cellular RNAs3-5. Here we report that the T4 ART ModB accepts not only NAD but also NAD-capped RNA (NAD-RNA) as a substrate and attaches entire RNA chains to acceptor proteins in an 'RNAylation' reaction. ModB specifically RNAylates the ribosomal proteins rS1 and rL2 at defined Arg residues, and selected E. coli and T4 phage RNAs are linked to rS1 in vivo. T4 phages that express an inactive mutant of ModB have a decreased burst size and slowed lysis of E. coli. Our findings reveal a distinct biological role for NAD-RNA, namely the activation of the RNA for enzymatic transfer to proteins. The attachment of specific RNAs to ribosomal proteins might provide a strategy for the phage to modulate the host's translation machinery. This work reveals a direct connection between RNA modification and post-translational protein modification. ARTs have important roles far beyond viral infections6, so RNAylation may have far-reaching implications.


Assuntos
ADP Ribose Transferases , Bacteriófago T4 , Proteínas de Escherichia coli , Escherichia coli , NAD , RNA , Proteínas Virais , ADP Ribose Transferases/metabolismo , Bacteriófago T4/enzimologia , Bacteriófago T4/genética , Bacteriófago T4/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/virologia , NAD/metabolismo , Proteínas Ribossômicas/química , Proteínas Ribossômicas/metabolismo , Proteínas Virais/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , RNA/química , RNA/genética , RNA/metabolismo , Biossíntese de Proteínas , Regulação Bacteriana da Expressão Gênica , Processamento de Proteína Pós-Traducional
6.
J Mol Biol ; 434(21): 167829, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36116540

RESUMO

Enterobacteria phage P1 expresses two types of tail fibre, S and S'. Despite the wide usage of phage P1 for transduction, the host range and the receptor for its alternative S' tail fibre was never determined. Here, a ΔS-cin Δpac E. coli P1 lysogenic strain was generated to allow packaging of phagemid DNA into P1 phage having either S or S' tail fibre. P1(S') could transduce phagemid DNA into Shigella flexneri 2a 2457O, Shigella flexneri 5a M90T and Escherichia coli O3 efficiently. Mutational analysis of the O-antigen assembly genes and LPS inhibition assays indicated that P1(S') transduction requires at least one O-antigen unit. E. coli O111:B4 LPS produced a high neutralising effect against P1(S') transduction, indicating that this E. coli strain could be susceptible to P1(S')-mediated transduction. Mutations in the O-antigen modification genes of S. flexneri 2a 2457O and S. flexneri 5a M90T did not cause significant changes to P1(S') transduction efficiency. A higher transduction efficiency of P1(S') improved the delivery of a cas9 antimicrobial phagemid into both S. flexneri 2457O and M90T. These findings provide novel insights into P1 tropism-switching, by identifying the bacterial strains which are susceptible to P1(S')-mediated transduction, as well as demonstrating its potential for delivering a DNA sequence-specific Cas9 antimicrobial into clinically relevant S. flexneri.


Assuntos
Bacteriófago P1 , Escherichia coli , Antígenos O , Shigella flexneri , Transdução Genética , Proteínas da Cauda Viral , Escherichia coli/genética , Escherichia coli/virologia , Antígenos O/genética , Antígenos O/fisiologia , Shigella flexneri/genética , Shigella flexneri/virologia , Bacteriófago P1/genética , Bacteriófago P1/fisiologia , Proteínas da Cauda Viral/genética
7.
Sci Rep ; 12(1): 941, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35042907

RESUMO

Bacteriophages, simply phages, have long been used as a potential alternative to antibiotics for livestock due to their ability to specifically kill enterotoxigenic Escherichia coli (ETEC), which is a major cause of diarrhea in piglets. However, the control of ETEC infection by phages within intestinal epithelial cells, and their relationship with host immune responses, remain poorly understood. In this study, we evaluated the effect of phage EK99P-1 against ETEC K99-infected porcine intestinal epithelial cell line (IPEC-J2). Phage EK99P-1 prevented ETEC K99-induced barrier disruption by attenuating the increased permeability mediated by the loss of tight junction proteins such as zonula occludens-1 (ZO-1), occludin, and claudin-3. ETEC K99-induced inflammatory responses, such as interleukin (IL)-8 secretion, were decreased by treatment with phage EK99P-1. We used a IPEC-J2/peripheral blood mononuclear cell (PBMC) transwell co-culture system to investigate whether the modulation of barrier disruption and chemokine secretion by phage EK99P-1 in ETEC K99-infected IPEC-J2 would influence immune cells at the site of basolateral. The results showed that phage EK99P-1 reduced the mRNA expression of ETEC K99-induced pro-inflammatory cytokines, IL-1ß and IL-8, from PBMC collected on the basolateral side. Together, these results suggest that phage EK99P-1 prevented ETEC K99-induced barrier dysfunction in IPEC-J2 and alleviated inflammation caused by ETEC K99 infection. Reinforcement of the intestinal barrier, such as regulation of permeability and cytokines, by phage EK99P-1 also modulates the immune cell inflammatory response.


Assuntos
Escherichia coli Enterotoxigênica/virologia , Mucosa Intestinal/metabolismo , Proteínas de Junções Íntimas/metabolismo , Animais , Aderência Bacteriana/fisiologia , Bacteriófagos/genética , Bacteriófagos/metabolismo , Bacteriófagos/patogenicidade , Linhagem Celular , Escherichia coli Enterotoxigênica/genética , Escherichia coli Enterotoxigênica/fisiologia , Células Epiteliais/metabolismo , Escherichia coli/genética , Escherichia coli/fisiologia , Escherichia coli/virologia , Infecções por Escherichia coli/prevenção & controle , Inflamação/metabolismo , Enteropatias/metabolismo , Intestinos , Ocludina/metabolismo , Permeabilidade , Suínos , Junções Íntimas/metabolismo
8.
Nat Commun ; 12(1): 5959, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34645844

RESUMO

The directed evolution of antibodies has yielded important research tools and human therapeutics. The dependence of many antibodies on disulfide bonds for stability has limited the application of continuous evolution technologies to antibodies and other disulfide-containing proteins. Here we describe periplasmic phage-assisted continuous evolution (pPACE), a system for continuous evolution of protein-protein interactions in the disulfide-compatible environment of the E. coli periplasm. We first apply pPACE to rapidly evolve novel noncovalent and covalent interactions between subunits of homodimeric YibK protein and to correct a binding-defective mutant of the anti-GCN4 Ω-graft antibody. We develop an intein-mediated system to select for soluble periplasmic expression in pPACE, leading to an eight-fold increase in soluble expression of the Ω-graft antibody. Finally, we evolve disulfide-containing trastuzumab antibody variants with improved binding to a Her2-like peptide and improved soluble expression. Together, these results demonstrate that pPACE can rapidly optimize proteins containing disulfide bonds, broadening the applicability of continuous evolution.


Assuntos
Evolução Molecular Direcionada/métodos , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Metiltransferases/genética , Periplasma/genética , Isomerases de Dissulfetos de Proteínas/genética , Trastuzumab/genética , Sítios de Ligação , Clonagem Molecular , Colífagos/genética , Colífagos/metabolismo , Dissulfetos/química , Dissulfetos/metabolismo , Escherichia coli/metabolismo , Escherichia coli/virologia , Proteínas de Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Inteínas/genética , Metiltransferases/metabolismo , Modelos Moleculares , Periplasma/metabolismo , Periplasma/virologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Isomerases de Dissulfetos de Proteínas/metabolismo , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trastuzumab/química , Trastuzumab/metabolismo
9.
Nucleic Acids Res ; 49(19): 11257-11273, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34657954

RESUMO

Bacteria have evolved a multitude of systems to prevent invasion by bacteriophages and other mobile genetic elements. Comparative genomics suggests that genes encoding bacterial defence mechanisms are often clustered in 'defence islands', providing a concerted level of protection against a wider range of attackers. However, there is a comparative paucity of information on functional interplay between multiple defence systems. Here, we have functionally characterised a defence island from a multidrug resistant plasmid of the emerging pathogen Escherichia fergusonii. Using a suite of thirty environmentally-isolated coliphages, we demonstrate multi-layered and robust phage protection provided by a plasmid-encoded defence island that expresses both a type I BREX system and the novel GmrSD-family type IV DNA modification-dependent restriction enzyme, BrxU. We present the structure of BrxU to 2.12 Å, the first structure of the GmrSD family of enzymes, and show that BrxU can utilise all common nucleotides and a wide selection of metals to cleave a range of modified DNAs. Additionally, BrxU undergoes a multi-step reaction cycle instigated by an unexpected ATP-dependent shift from an intertwined dimer to monomers. This direct evidence that bacterial defence islands can mediate complementary layers of phage protection enhances our understanding of the ever-expanding nature of phage-bacterial interactions.


Assuntos
Proteínas de Bactérias/química , Colífagos/genética , Enzimas de Restrição-Modificação do DNA/química , Escherichia coli/genética , Escherichia/genética , Plasmídeos/química , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Clonagem Molecular , Colífagos/metabolismo , Cristalografia por Raios X , Enzimas de Restrição-Modificação do DNA/genética , Enzimas de Restrição-Modificação do DNA/metabolismo , DNA Viral/química , DNA Viral/genética , DNA Viral/metabolismo , Escherichia/metabolismo , Escherichia/virologia , Escherichia coli/metabolismo , Escherichia coli/virologia , Expressão Gênica , Ilhas Genômicas , Genômica/métodos , Modelos Moleculares , Plasmídeos/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
10.
Appl Environ Microbiol ; 87(13): e0042321, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33893116

RESUMO

Bacteriophages are the most abundant and diverse biological entities on Earth. Phages exhibit strict host specificity that is largely conferred by adsorption. However, the mechanism underlying this phage host specificity remains poorly understood. In this study, we examined the interaction between outer membrane protein C (OmpC), one of the Escherichia coli receptors, and the long tail fibers of bacteriophage T4. T4 phage uses OmpC of the K-12 strain, but not of the O157 strain, for adsorption, even though OmpCs from the two E. coli strains share 94% homology. We identified amino acids P177 and F182 in loop 4 of the K-12 OmpC as essential for T4 phage adsorption in the copresence of loops 1 and 5. Analyses of phage mutants capable of adsorbing to OmpC mutants demonstrated that amino acids at positions 937 and 942 of the gp37 protein, which is present in the distal tip (DT) region of the T4 long tail fibers, play an important role in adsorption. Furthermore, we created a T4 phage mutant library with artificial modifications in the DT region and isolated and characterized multiple phage mutants capable of adsorbing to OmpC of the O157 strain or lipopolysaccharide of the K-12 strain. These results shed light on the mechanism underlying the phage host specificity mediated by gp37 and OmpC and may be useful in the development of phage therapy via artificial modifications of the DT region of T4 phage. IMPORTANCE Understanding the host specificity of phages will lead to the development of phage therapy. The interaction between outer membrane protein C (OmpC), one of the Escherichia coli receptors, and the gp37 protein present in the distal tip (DT) region of the long tail fibers of T4 bacteriophages largely determines their host specificity. Here, we elucidated the amino acid residues important for the interaction between gp37 and OmpC. This result suggests that the shapes of both proteins at the binding interface play important roles in their interactions, which are likely mediated by multiple residues of both binding partners. Additionally, we successfully isolated multiple phage mutants capable of adsorbing to a variety of E. coli receptors using a mutant T4 phage library with artificial modifications in the DT region, providing a foundation for the alteration of the host specificity.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Bacteriófago T4/patogenicidade , Proteínas de Escherichia coli/metabolismo , Escherichia coli/virologia , Porinas/metabolismo , Proteínas do Envelope Viral/metabolismo , Adsorção , Aminoácidos/química , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Bacteriófago T4/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Especificidade de Hospedeiro , Mutação , Porinas/química , Porinas/genética , Ligação Proteica , Proteínas do Envelope Viral/química
11.
Nat Commun ; 12(1): 1576, 2021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33707432

RESUMO

We apply an oligo-library and machine learning-approach to characterize the sequence and structural determinants of binding of the phage coat proteins (CPs) of bacteriophages MS2 (MCP), PP7 (PCP), and Qß (QCP) to RNA. Using the oligo library, we generate thousands of candidate binding sites for each CP, and screen for binding using a high-throughput dose-response Sort-seq assay (iSort-seq). We then apply a neural network to expand this space of binding sites, which allowed us to identify the critical structural and sequence features for binding of each CP. To verify our model and experimental findings, we design several non-repetitive binding site cassettes and validate their functionality in mammalian cells. We find that the binding of each CP to RNA is characterized by a unique space of sequence and structural determinants, thus providing a more complete description of CP-RNA interaction as compared with previous low-throughput findings. Finally, based on the binding spaces we demonstrate a computational tool for the successful design and rapid synthesis of functional non-repetitive binding-site cassettes.


Assuntos
Allolevivirus/genética , Proteínas do Capsídeo/metabolismo , Escherichia coli/virologia , Levivirus/genética , RNA/metabolismo , Sítios de Ligação Microbiológicos/genética , Sítios de Ligação/genética , Linhagem Celular Tumoral , Escherichia coli/genética , Biblioteca Gênica , Humanos , Aprendizado de Máquina , Plasmídeos/genética
12.
Elife ; 102021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33567250

RESUMO

The first S-adenosyl methionine (SAM) degrading enzyme (SAMase) was discovered in bacteriophage T3, as a counter-defense against the bacterial restriction-modification system, and annotated as a SAM hydrolase forming 5'-methyl-thioadenosine (MTA) and L-homoserine. From environmental phages, we recently discovered three SAMases with barely detectable sequence similarity to T3 SAMase and without homology to proteins of known structure. Here, we present the very first phage SAMase structures, in complex with a substrate analogue and the product MTA. The structure shows a trimer of alpha-beta sandwiches similar to the GlnB-like superfamily, with active sites formed at the trimer interfaces. Quantum-mechanical calculations, thin-layer chromatography, and nuclear magnetic resonance spectroscopy demonstrate that this family of enzymes are not hydrolases but lyases forming MTA and L-homoserine lactone in a unimolecular reaction mechanism. Sequence analysis and in vitro and in vivo mutagenesis support that T3 SAMase belongs to the same structural family and utilizes the same reaction mechanism.


Bacteria can be infected by viruses known as bacteriophages. These viruses inject their genetic material into bacterial cells and use the bacteria's own machinery to build the proteins they need to survive and infect other cells. To protect themselves, bacteria produce a molecule called S-adenosyl methionine, or SAM for short, which deposits marks on the bacteria's DNA. These marks help the bacteria distinguish their own genetic material from the genetic material of foreign invaders: any DNA not bearing the mark from SAM will be immediately broken down by the bacterial cell. This system helps to block many types of bacteriophage infections, but not all. Some bacteriophages carry genes that code for enzymes called SAMases, which can break down SAM, switching off the bacteria's defenses. The most well-known SAMase was first discovered in the 1960s in a bacteriophage called T3. Chemical studies of this SAMase suggested that it works as a 'hydrolase', meaning that it uses water to break SAM apart. New SAMases have since been discovered in bacteriophages from environmental water samples, which, despite being able to degrade SAM, are genetically dissimilar to one another and the SAMase in T3. This brings into question whether these enzymes all use the same mechanism to break SAM down. To gain a better understanding of how these SAMases work, Guo, Söderholm, Kanchugal, Isaksen et al. solved the crystal structure of one of the newly discovered enzymes called Svi3-3. This revealed three copies of the Svi3-3 enzyme join together to form a unit that SAM binds to at the border between two of the enzymes. Computer simulations of this structure suggested that Svi3-3 holds SAM in a position where it cannot interact with water, and that once in the grip of the SAMase, SAM instead reacts with itself and splits into two. Experiments confirmed these predictions for Svi3-3 and the other tested SAMases. Furthermore, the SAMase from bacteriophage T3 was also found to degrade SAM using the same mechanism. This shows that this group of SAMases are not hydrolases as originally thought, but in fact 'lyases': enzymes that break molecules apart without using water. These findings form a starting point for further investigations into how SAM lyases help bacteriophages evade detection. SAM has various different functions in other living organisms, and these lyases could be used to modulate the levels of SAM in future studies investigating its role.


Assuntos
Bacteriófago T3/genética , Liases/genética , Proteínas Virais/genética , Bacteriófago T3/metabolismo , Escherichia coli/virologia , Liases/metabolismo , S-Adenosilmetionina/metabolismo , Proteínas Virais/metabolismo
13.
Methods Mol Biol ; 2211: 3-14, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33336266

RESUMO

Bactofection, a bacterial-mediated form of genetic transfer, is highlighted as an alternative mechanism for gene therapy. A key advantage of this system for immune-reactivity purposes stems from the nature of the bacterial host capable of initiating an immune response by attracting recognition and cellular uptake by antigen-presenting cells (APCs). The approach is also a suitable technique to deliver larger genetic constructs more efficiently as it can transfer plasmids of varying sizes into target mammalian cells. Given these advantages, bacterial vectors are being studied as potential carriers for the delivery of plasmid DNA into target cells to enable expression of heterologous proteins. The bacteria used for bactofection are generally nonpathogenic; however, concerns arise due to the use of a biological agent. To overcome such concerns, enhanced bacterial degradation has been engineered as an attenuation and safety feature for bactofection vectors. In particular, the ΦX174 lysis E (LyE) gene can be repurposed to both minimize bacterial survival within mammalian hosts while also improving overall gene delivery. More specifically, an engineered bacterial vector carrying the LyE gene showed improved gene delivery and safety profiles when tested with murine RAW264.7 macrophage APCs. This chapter outlines steps taken to engineer E. coli for LyE expression as a safer and more effective genetic antigen delivery bactofection vehicle in the context of vaccine utility.


Assuntos
Bacteriófago phi X 174/fisiologia , Escherichia coli/virologia , Expressão Gênica , Transdução Genética , Proteínas Virais/genética , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Hemólise , Interações Hospedeiro-Patógeno , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Modelos Biológicos , Plasmídeos/genética
14.
Methods Mol Biol ; 2225: 77-92, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33108658

RESUMO

Adeno-associated virus (AAV) is a helper-dependent single-stranded DNA parvovirus. Over the years, AAV has become the vector of choice in the gene therapy field due to its safety profile and low immunogenicity. With a carrying capacity of 4.2 kbp, these vectors have demonstrated their clinical value, especially in the field of ophthalmology. Herein we describe methods for the molecular design and packaging of AAV viral vectors. These methods apply to the design of single-stranded or self-complementary AAV vectors.


Assuntos
Clonagem Molecular/métodos , Dependovirus/genética , Engenharia Genética/métodos , Transgenes , Empacotamento do Genoma Viral/genética , Primers do DNA/química , Primers do DNA/metabolismo , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Dependovirus/metabolismo , Escherichia coli/virologia , Terapia Genética/métodos , Humanos , Plasmídeos/química , Plasmídeos/metabolismo , Transdução Genética
15.
Nucleic Acids Res ; 48(21): 12030-12041, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33211866

RESUMO

The CII protein of temperate coliphage 186, like the unrelated CII protein of phage λ, is a transcriptional activator that primes expression of the CI immunity repressor and is critical for efficient establishment of lysogeny. 186-CII is also highly unstable, and we show that in vivo degradation is mediated by both FtsH and RseP. We investigated the role of CII instability by constructing a 186 phage encoding a protease resistant CII. The stabilised-CII phage was defective in the lysis-lysogeny decision: choosing lysogeny with close to 100% frequency after infection, and forming prophages that were defective in entering lytic development after UV treatment. While lysogenic CI concentration was unaffected by CII stabilisation, lysogenic transcription and CI expression was elevated after UV. A stochastic model of the 186 network after infection indicated that an unstable CII allowed a rapid increase in CI expression without a large overshoot of the lysogenic level, suggesting that instability enables a decisive commitment to lysogeny with a rapid attainment of sensitivity to prophage induction.


Assuntos
Proteases Dependentes de ATP/genética , Colífagos/genética , Endopeptidases/genética , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Lisogenia , Proteínas de Membrana/genética , Prófagos/genética , Proteínas Virais/genética , Proteases Dependentes de ATP/metabolismo , Colífagos/crescimento & desenvolvimento , Colífagos/metabolismo , Colífagos/efeitos da radiação , Endopeptidases/metabolismo , Escherichia coli/metabolismo , Escherichia coli/efeitos da radiação , Escherichia coli/virologia , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana/metabolismo , Modelos Estatísticos , Prófagos/crescimento & desenvolvimento , Prófagos/metabolismo , Prófagos/efeitos da radiação , Estabilidade Proteica/efeitos da radiação , Proteólise/efeitos da radiação , Processos Estocásticos , Ativação Transcricional , Raios Ultravioleta , Proteínas Virais/metabolismo
16.
Viruses ; 12(10)2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33036312

RESUMO

Infecting bacteriophage T4 uses a contractile tail structure to breach the envelope of the Escherichia coli host cell. During contraction, the tail tube headed with the "central spike complex" is thought to mechanically puncture the outer membrane. We show here that a purified tip fragment of the central spike complex interacts with periplasmic chaperone PpiD, which is anchored to the cytoplasmic membrane. PpiD may be involved in the penetration of the inner membrane by the T4 injection machinery, resulting in a DNA-conducting channel to translocate the phage DNA into the interior of the cell. Host cells with the ppiD gene deleted showed partial reduction in the plating efficiency of T4, suggesting a supporting role of PpiD to improve the efficiency of the infection process.


Assuntos
Bacteriófago T4/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/virologia , Peptidilprolil Isomerase/metabolismo , Proteínas da Cauda Viral/metabolismo , Membrana Celular/metabolismo , Proteínas de Escherichia coli/genética , Glicosídeo Hidrolases , Peptidilprolil Isomerase/genética , Periplasma/virologia , Proteínas do Envelope Viral/metabolismo , Ligação Viral , Internalização do Vírus
17.
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
18.
Iran J Allergy Asthma Immunol ; 19(2): 149-158, 2020 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-32372628

RESUMO

Purification and preparation of three diagnostic antigens used for the detection of human T-lymphotropic virus (HTLV)-I/-II infection in E.coli are different parts of a multi-step method. In this study, our aim was to design a chimeric protein for the simultaneous detection of HTLV-I and HTLV-II antibodies. Immunodominant B cell linear epitopes of envelope and capsid proteins of HTLV-I/-II were selected and linked together; using a suitable amino acid linker and a chimeric antigen (CA). The codon-optimized synthetic DNA encoding the CA was subcloned into the pGS21aexpression vector and CA expressed as His-GST fused protein in E. coli BL21 (DE3) cells. Then the recombinant CA was purified, using the Ni-NTA (Nickle Nitrilotriacetic acid) affinity chromatography under native conditions. The Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and densitometric scanning results showed that CA accounted for 15% of the total cellular proteins and approximately 50% of the expressed histidine-glutathione s-transferase-chimeric antigen (His-GST-CA) proteins were soluble. The CA was successfully purified in one step with a purity of greater than 90%, which is suitable for antigenicity evaluations. Enzyme-linked immunosorbent assay (ELISA) results showed that the GST fused CA reacted in a concentration-dependent manner with HTLV-I/-II infected sera and was able to distinguish normal serum from HTLV-I/-II infected one with a proper sensitivity. With further validation, CA, as described in the present study could be introduced as a novel reliable, cost-effective and easy alternative for the three separate HTLV-I/-II diagnostic peptide antigens, which is prepared as a fusion with GST.


Assuntos
Antígenos Virais/metabolismo , Epitopos de Linfócito B/metabolismo , Escherichia coli/imunologia , Infecções por HTLV-I/diagnóstico , Infecções por HTLV-II/diagnóstico , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Vírus Linfotrópico T Tipo 2 Humano/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos Antivirais/metabolismo , Antígenos Virais/genética , Biologia Computacional , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito B/genética , Escherichia coli/virologia , Expressão Gênica , Humanos , Transportadores de Ânions Orgânicos/genética , Conformação Proteica , Proteínas Recombinantes de Fusão/genética , Sensibilidade e Especificidade , Sorologia
19.
J Mol Biol ; 432(13): 3771-3789, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32305462

RESUMO

Molecular chaperones maintain cellular protein homeostasis by acting at almost every step in protein biogenesis pathways. The DnaK/HSP70 chaperone has been associated with almost every known essential chaperone functions in bacteria. To act as a bona fide chaperone, DnaK strictly relies on essential co-chaperone partners known as the J-domain proteins (JDPs, DnaJ, Hsp40), which preselect substrate proteins for DnaK, confer its specific cellular localization, and stimulate both its weak ATPase activity and substrate transfer. Remarkably, genome sequencing has revealed the presence of multiple JDP/DnaK chaperone/co-chaperone pairs in a number of bacterial genomes, suggesting that certain pairs have evolved toward more specific functions. In this review, we have used representative sets of bacterial and phage genomes to explore the distribution of JDP/DnaK pairs. Such analysis has revealed an unexpected reservoir of novel bacterial JDPs co-chaperones with very diverse and unexplored function that will be discussed.


Assuntos
Proteínas de Escherichia coli/genética , Escherichia coli/genética , Proteínas de Choque Térmico HSP40/genética , Proteínas de Choque Térmico HSP70/genética , Domínios Proteicos/genética , Adenosina Trifosfatases/genética , Bactérias/virologia , Bacteriófagos/genética , Escherichia coli/virologia , Humanos , Redes e Vias Metabólicas/genética , Chaperonas Moleculares/genética , Biossíntese de Proteínas/genética
20.
Biochem Biophys Res Commun ; 526(2): 485-490, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32238267

RESUMO

Bacteriophage T4 encodes orthologs of the proteins Rad50 (gp46) and Mre11 (gp47), which form a heterotetrameric complex (MR) that participates in the processing of DNA ends for recombination-dependent DNA repair. Crystal and high-resolution cryo-EM structures of Rad50 have revealed DNA binding sites near the dimer interface of Rad50 opposite of Mre11, and near the base of the coiled-coils that extend out from the globular head domain. An analysis of T4-Rad50 using sequenced-based algorithms to identify DNA binding residues predicts that a conserved region of positively charged residues near the C-terminus, distal to the observed binding sites, interacts with DNA. Mutant proteins were generated to test this prediction and their enzymatic and DNA binding activities were evaluated. Consistent with the predictions, the Rad50 C-terminal mutants had reduced affinity for DNA as measured by Rad50 equilibrium DNA binding assays and an increased Km-DNA as determined in MR complex nuclease assays. Moreover, the allosteric activation of ATP hydrolysis activity due to DNA binding was substantially reduced, suggesting that these residues may be involved in the communication between the DNA and ATP binding sites.


Assuntos
Bacteriófago T4/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Virais/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Bacteriófago T4/química , Sítios de Ligação , DNA/metabolismo , Proteínas de Ligação a DNA/química , Escherichia coli/virologia , Exodesoxirribonucleases/química , Exodesoxirribonucleases/metabolismo , Hidrólise , Modelos Moleculares , Ligação Proteica , Proteínas Virais/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA