Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.009
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1373052, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38808067

RESUMEN

Among the Acinetobacter genus, Acinetobacter pittii stands out as an important opportunistic infection causative agent commonly found in hospital settings, which poses a serious threat to human health. Recently, the high prevalence of carbapenem-resistant A. pittii isolates has created significant therapeutic challenges for clinicians. Bacteriophages and their derived enzymes are promising therapeutic alternatives or adjuncts to antibiotics effective against multidrug-resistant bacterial infections. However, studies investigating the depolymerases specific to A. pittii strains are scarce. In this study, we identified and characterized a capsule depolymerase, Dpo27, encoded by the bacteriophage IME-Ap7, which targets A. pittii. A total of 23 clinical isolates of Acinetobacter spp. were identified as A. pittii (21.91%, 23/105), and seven A. pittii strains with various K locus (KL) types (KL14, KL32, KL38, KL111, KL163, KL207, and KL220) were used as host bacteria for phage screening. The lytic phage IME-Ap7 was isolated using A. pittii 7 (KL220) as an indicator bacterium and was observed for depolymerase activity. A putative tail fiber gene encoding a polysaccharide-degrading enzyme (Dpo27) was identified and expressed. The results of the modified single-spot assay showed that both A. pittii 7 and 1492 were sensitive to Dpo27, which was assigned the KL220 type. After incubation with Dpo27, A. pittii strain was susceptible to killing by human serum; moreover, the protein displayed no hemolytic activity against erythrocytes. Furthermore, the protein exhibited sustained activity across a wide pH range (5.0-10.0) and at temperatures between 20 and 50°C. In summary, the identified capsule depolymerase Dpo27 holds promise as an alternative treatment for combating KL220-type A. pittii infections.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter , Bacteriófagos , Glicósido Hidrolasas , Bacteriófagos/genética , Bacteriófagos/enzimología , Bacteriófagos/aislamiento & purificación , Humanos , Acinetobacter/enzimología , Acinetobacter/genética , Acinetobacter/virología , Acinetobacter/efectos de los fármacos , Infecciones por Acinetobacter/microbiología , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Cápsulas Bacterianas/metabolismo , Cápsulas Bacterianas/genética
2.
Viruses ; 16(5)2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38793652

RESUMEN

The genus Acinetobacter comprises both environmental and clinically relevant species associated with hospital-acquired infections. Among them, Acinetobacter baumannii is a critical priority bacterial pathogen, for which the research and development of new strategies for antimicrobial treatment are urgently needed. Acinetobacter spp. produce a variety of structurally diverse capsular polysaccharides (CPSs), which surround the bacterial cells with a thick protective layer. These surface structures are primary receptors for capsule-specific bacteriophages, that is, phages carrying tailspikes with CPS-depolymerizing/modifying activities. Phage tailspike proteins (TSPs) exhibit hydrolase, lyase, or esterase activities toward the corresponding CPSs of a certain structure. In this study, the data on all lytic capsule-specific phages infecting Acinetobacter spp. with genomes deposited in the NCBI GenBank database by January 2024 were summarized. Among the 149 identified TSPs encoded in the genomes of 143 phages, the capsular specificity (K specificity) of 46 proteins has been experimentally determined or predicted previously. The specificity of 63 TSPs toward CPSs, produced by various Acinetobacter K types, was predicted in this study using a bioinformatic analysis. A comprehensive phylogenetic analysis confirmed the prediction and revealed the possibility of the genetic exchange of gene regions corresponding to the CPS-recognizing/degrading parts of different TSPs between morphologically and taxonomically distant groups of capsule-specific Acinetobacter phages.


Asunto(s)
Acinetobacter , Cápsulas Bacterianas , Bacteriófagos , Genoma Viral , Filogenia , Bacteriófagos/genética , Bacteriófagos/enzimología , Bacteriófagos/clasificación , Acinetobacter/virología , Acinetobacter/genética , Acinetobacter/enzimología , Cápsulas Bacterianas/metabolismo , Cápsulas Bacterianas/genética , Proteínas de la Cola de los Virus/genética , Proteínas de la Cola de los Virus/metabolismo , Polisacáridos/metabolismo , Polisacáridos Bacterianos/metabolismo , Polisacáridos Bacterianos/genética , Acinetobacter baumannii/virología , Acinetobacter baumannii/genética , Acinetobacter baumannii/enzimología , Glicósido Hidrolasas
3.
World J Microbiol Biotechnol ; 40(6): 186, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38683213

RESUMEN

The ability of most opportunistic bacteria to form biofilms, coupled with antimicrobial resistance, hinder the efforts to control widespread infections, resulting in high risks of negative outcomes and economic costs. Endolysins are promising compounds that efficiently combat bacteria, including multidrug-resistant strains and biofilms, without a low probability of subsequent emergence of stable endolysin-resistant phenotypes. However, the details of antibiofilm effects of these enzymes are poorly understood. To elucidate the interactions of bacteriophage endolysins LysAm24, LysAp22, LysECD7, and LysSi3 with bacterial films formed by Gram-negative species, we estimated their composition and assessed the endolysins' effects on the most abundant exopolymers in vitro. The obtained data suggests a pronounced efficiency of these lysins against biofilms with high (Klebsiella pneumoniae) and low (Acinetobacter baumannii) matrix contents, or dual-species biofilms, resulting in at least a twofold loss of the biomass. These peptidoglycan hydrolases interacted diversely with protective compounds of biofilms such as extracellular DNA and polyanionic carbohydrates, indicating a spectrum of biofilm-disrupting effects for bacteriolytic phage enzymes. Specifically, we detected disruption of acid exopolysaccharides by LysAp22, strong DNA-binding capacity of LysAm24, both of these interactions for LysECD7, and neither of them for LysSi3.


Asunto(s)
Bacteriófagos , Biopelículas , Endopeptidasas , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Endopeptidasas/metabolismo , Endopeptidasas/farmacología , Endopeptidasas/química , Bacteriófagos/enzimología , Acinetobacter baumannii/efectos de los fármacos , Klebsiella pneumoniae/efectos de los fármacos , Proteínas Virales/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/química
4.
Proc Natl Acad Sci U S A ; 121(19): e2317954121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38683976

RESUMEN

Several microbial genomes lack textbook-defined essential genes. If an essential gene is absent from a genome, then an evolutionarily independent gene of unknown function complements its function. Here, we identified frequent nonhomologous replacement of an essential component of DNA replication initiation, a replicative helicase loader gene, in Vibrionaceae. Our analysis of Vibrionaceae genomes revealed two genes with unknown function, named vdhL1 and vdhL2, that were substantially enriched in genomes without the known helicase-loader genes. These genes showed no sequence similarities to genes with known function but encoded proteins structurally similar with a viral helicase loader. Analyses of genomic syntenies and coevolution with helicase genes suggested that vdhL1/2 encodes a helicase loader. The in vitro assay showed that Vibrio harveyi VdhL1 and Vibrio ezurae VdhL2 promote the helicase activity of DnaB. Furthermore, molecular phylogenetics suggested that vdhL1/2 were derived from phages and replaced an intrinsic helicase loader gene of Vibrionaceae over 20 times. This high replacement frequency implies the host's advantage in acquiring a viral helicase loader gene.


Asunto(s)
ADN Helicasas , Replicación del ADN , Filogenia , Vibrionaceae , Vibrionaceae/genética , Vibrionaceae/enzimología , ADN Helicasas/metabolismo , ADN Helicasas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Bacteriófagos/genética , Bacteriófagos/enzimología , Evolución Molecular , Genoma Bacteriano , AdnB Helicasas/metabolismo , AdnB Helicasas/genética , Vibrio/genética , Vibrio/enzimología
5.
Nat Microbiol ; 9(6): 1579-1592, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38589469

RESUMEN

Prokaryotic antiviral defence systems are frequently toxic for host cells and stringent regulation is required to ensure survival and fitness. These systems must be readily available in case of infection but tightly controlled to prevent activation of an unnecessary cellular response. Here we investigate how the bacterial cyclic oligonucleotide-based antiphage signalling system (CBASS) uses its intrinsic protein modification system to regulate the nucleotide cyclase. By integrating a type II CBASS system from Bacillus cereus into the model organism Bacillus subtilis, we show that the protein-conjugating Cap2 (CBASS associated protein 2) enzyme links the cyclase exclusively to the conserved phage shock protein A (PspA) in the absence of phage. The cyclase-PspA conjugation is reversed by the deconjugating isopeptidase Cap3 (CBASS associated protein 3). We propose a model in which the cyclase is held in an inactive state by conjugation to PspA in the absence of phage, with conjugation released upon infection, priming the cyclase for activation.


Asunto(s)
Bacillus subtilis , Proteínas Bacterianas , Bacillus subtilis/virología , Bacillus subtilis/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Bacillus cereus/virología , Bacillus cereus/enzimología , Bacillus cereus/genética , Bacillus cereus/inmunología , Transducción de Señal , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/enzimología , Liasas de Fósforo-Oxígeno/metabolismo , Liasas de Fósforo-Oxígeno/genética , Regulación Bacteriana de la Expresión Génica
6.
Nat Microbiol ; 9(6): 1566-1578, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38649411

RESUMEN

The cyclic-oligonucleotide-based anti-phage signalling system (CBASS) is a type of innate prokaryotic immune system. Composed of a cyclic GMP-AMP synthase (cGAS) and CBASS-associated proteins, CBASS uses cyclic oligonucleotides to activate antiviral immunity. One major class of CBASS contains a homologue of eukaryotic ubiquitin-conjugating enzymes, which is either an E1-E2 fusion or a single E2. However, the functions of single E2s in CBASS remain elusive. Here, using biochemical, genetic, cryo-electron microscopy and mass spectrometry investigations, we discover that the E2 enzyme from Serratia marcescens regulates cGAS by imitating the ubiquitination cascade. This includes the processing of the cGAS C terminus, conjugation of cGAS to a cysteine residue, ligation of cGAS to a lysine residue, cleavage of the isopeptide bond and poly-cGASylation. The poly-cGASylation activates cGAS to produce cGAMP, which acts as an antiviral signal and leads to cell death. Thus, our findings reveal a unique regulatory role of E2 in CBASS.


Asunto(s)
Nucleotidiltransferasas , Enzimas Ubiquitina-Conjugadoras , Ubiquitinación , Enzimas Ubiquitina-Conjugadoras/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/química , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/química , Transducción de Señal , Nucleótidos Cíclicos/metabolismo , Bacteriófagos/genética , Bacteriófagos/enzimología , Ubiquitina/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Humanos , Microscopía por Crioelectrón , Inmunidad Innata
7.
Microbiol Spectr ; 12(6): e0038224, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38651885

RESUMEN

Integration of carbapenemase gene blaIMP into the chromosome of carbapenem-resistant Acinetobacter baumannii (CRAB) has not been reported. The aim of this study was to explore the genomic characteristics of CRAB AB322 isolated from a Taiwanese patient diagnosed with bacteremia in 2011, whose chromosome harbors blaIMP-19. Disk diffusion and broth microdilution were employed to analyze the antimicrobial susceptibility of AB322 to 14 antimicrobials. Nanopore whole-genome sequencing platform was utilized for AB322 genome sequencing, and conjugation was further performed to investigate the transferability of blaIMP-19 to amikacin-resistant A. baumannii 218 (AB218) and Acinetobacter nosocomialis 254 (AN254). The results showed that AB322 was classified as multidrug-resistant A. baumannii but remained susceptible to ampicillin/sulbactam, colistin, and tigecycline. Whole-genome sequencing revealed the AB322 genome, consisting of a 4,098,985-bp chromosome, a 71,590-bp conjugative plasmid named pAB322-1, and an 8,726-bp plasmid named pAB322-2. Multilocus sequence typing analysis indicated that AB322 belonged to sequence type 1. AB322 chromosome harbored numerous acquired antimicrobial resistance genes, including aph(3')-Ia, aadA1b, aadA1, aac(6')-Ib3, aac (3)-Ia, blaADC-25, blaOXA-69, blaIMP-19, catA1, sul1, and tet(A), conferring resistance to ß-lactams, aminoglycosides, chloramphenicol, sulfamethoxazole, and tetracyclines. Moreover, blaIMP-19 was identified to be situated within class 1 integron In240 and an incomplete PHAGE_Salmon_SJ46_NC_031129 on AB322 chromosome. However, conjugation experiments revealed that blaIMP-19 could not be transferred to AB218 and AN254 in our testing conditions. In conclusion, we first report the presence of chromosomal-integrated blaIMP-19 in CRAB, possibly mediated by integron. The future dissemination of blaIMP-19 among different species, leading to carbapenem resistance dissemination, requires close monitoring. IMPORTANCE: The horizontal transfer of antimicrobial-resistant genes is crucial for the dissemination of resistance, especially as Acinetobacter baumannii has emerged as a clinically significant pathogen. However, in this study, we first report the integration of the blaIMP-19 gene into the chromosome of A. baumannii, and such horizontal transfer may be associated with integron-phage elements. Additionally, it is possible that these DNA fragments carrying antimicrobial-resistant genes could further spread to other pathogens by moving horizontally onto conjugative plasmids.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Antibacterianos , Proteínas Bacterianas , Farmacorresistencia Bacteriana Múltiple , Integrones , Plásmidos , beta-Lactamasas , Acinetobacter baumannii/genética , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/enzimología , Plásmidos/genética , Integrones/genética , Humanos , Infecciones por Acinetobacter/microbiología , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana Múltiple/genética , Taiwán , Pruebas de Sensibilidad Microbiana , Secuenciación Completa del Genoma , Bacteriófagos/genética , Bacteriófagos/enzimología , Cromosomas Bacterianos/genética , Carbapenémicos/farmacología , Tipificación de Secuencias Multilocus , Bacteriemia/microbiología
8.
Adv Sci (Weinh) ; 10(24): e2301694, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37310410

RESUMEN

Nanozymes, featuring intrinsic biocatalytic effects and broad-spectrum antimicrobial properties, are emerging as a novel antibiotic class. However, prevailing bactericidal nanozymes face a challenging dilemma between biofilm penetration and bacterial capture capacity, significantly impeding their antibacterial efficacy. Here, this work introduces a photomodulable bactericidal nanozyme (ICG@hMnOx ), composed of a hollow virus-spiky MnOx nanozyme integrated with indocyanine green, for dually enhanced biofilm penetration and bacterial capture for photothermal-boosted catalytic therapy of bacterial infections. ICG@hMnOx demonstrates an exceptional capability to deeply penetrate biofilms, owing to its pronounced photothermal effect that disrupts the compact structure of biofilms. Simultaneously, the virus-spiky surface significantly enhances the bacterial capture capacity of ICG@hMnOx . This surface acts as a membrane-anchored generator of reactive oxygen species and a glutathione scavenger, facilitating localized photothermal-boosted catalytic bacterial disinfection. Effective treatment of methicillin-resistant Staphylococcus aureus-associated biofilm infections is achieved using ICG@hMnOx , offering an appealing strategy to overcome the longstanding trade-off between biofilm penetration and bacterial capture capacity in antibacterial nanozymes. This work presents a significant advancement in the development of nanozyme-based therapies for combating biofilm-related bacterial infections.


Asunto(s)
Bacteriófagos , Biopelículas , Staphylococcus aureus Resistente a Meticilina , Nanopartículas , Infecciones Estafilocócicas , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Humanos , Biopelículas/efectos de los fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Bacteriófagos/enzimología , Nanopartículas/química , Rayos Láser
9.
Nucleic Acids Res ; 51(13): 6566-6577, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37293959

RESUMEN

Using an amber suppression-based noncanonical amino acid (ncAA) mutagenesis approach, the chemical space in phage display can be significantly expanded for drug discovery. In this work, we demonstrate the development of a novel helper phage, CMa13ile40, for continuous enrichment of amber obligate phage clones and efficient production of ncAA-containing phages. CMa13ile40 was constructed by insertion of a Candidatus Methanomethylophilus alvus pyrrolysyl-tRNA synthetase/PylT gene cassette into a helper phage genome. The novel helper phage allowed for a continuous amber codon enrichment strategy for two different libraries and demonstrated a 100-fold increase in packaging selectivity. CMa13ile40 was then used to create two peptide libraries containing separate ncAAs, Nϵ-tert-butoxycarbonyl-lysine and Nϵ-allyloxycarbonyl-lysine, respectively. These libraries were used to identify peptide ligands that bind to the extracellular domain of ZNRF3. Each selection showed differential enrichment of unique sequences dependent upon the ncAA used. Peptides from both selections were confirmed to have low micromolar affinity for ZNRF3 that was dependent upon the presence of the ncAA used for selection. Our results demonstrate that ncAAs in phages provide unique interactions for identification of unique peptides. As an effective tool for phage display, we believe that CMa13ile40 can be broadly applied to a wide variety of applications.


Asunto(s)
Aminoácidos , Aminoacil-ARNt Sintetasas , Bacteriófagos , Técnicas de Visualización de Superficie Celular , Aminoácidos/química , Aminoacil-ARNt Sintetasas/genética , Aminoacil-ARNt Sintetasas/metabolismo , Bacteriófagos/enzimología , Bacteriófagos/genética , Técnicas de Visualización de Superficie Celular/métodos , Péptidos/metabolismo , Descubrimiento de Drogas
10.
J Virol ; 96(17): e0106322, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-36000841

RESUMEN

Bacteriophages (phages) are an integral part of the human oral microbiome. Their roles in modulating bacterial physiology and shaping microbial communities have been discussed but remain understudied due to limited isolation and characterization of oral phage. Here, we report the isolation of LC001, a lytic phage targeting human oral Schaalia odontolytica (formerly known as Actinomyces odontolyticus) strain XH001. We showed that LC001 attached to and infected surface-grown, but not planktonic, XH001 cells, and it displayed remarkable host specificity at the strain level. Whole-genome sequencing of spontaneous LC001-resistant, surface-grown XH001 mutants revealed that the majority of the mutants carry nonsense or frameshift mutations in XH001 gene APY09_05145 (renamed ltg-1), which encodes a putative lytic transglycosylase (LT). The mutants are defective in LC001 binding, as revealed by direct visualization of the significantly reduced attachment of phage particles to the XH001 spontaneous mutants compared that to the wild type. Meanwhile, targeted deletion of ltg-1 produced a mutant that is defective in LC001 binding and resistant to LC001 infection even as surface-grown cells, while complementation of ltg-1 in the mutant background restored the LC001-sensitive phenotype. Intriguingly, similar expression levels of ltg-1 were observed in surface-grown and planktonic XH001, which displayed LC001-binding and nonbinding phenotypes, respectively. Furthermore, the overexpression of ltg-1 failed to confer an LC001-binding and -sensitive phenotype to planktonic XH001. Thus, our data suggested that rather than directly serving as a phage receptor, ltg-1-encoded LT may increase the accessibility of phage receptor, possibly via its enzymatic activity, by cleaving the peptidoglycan structure for better receptor exposure during peptidoglycan remodeling, a function that can be exploited by LC001 to facilitate infection. IMPORTANCE The evidence for the presence of a diverse and abundant phage population in the host-associated oral microbiome came largely from metagenomic analysis or the observation of virus-like particles within saliva/plaque samples, while the isolation of oral phage and investigation of their interaction with bacterial hosts are limited. Here, we report the isolation of LC001, the first lytic phage targeting oral Schaalia odontolytica. Our study suggested that LC001 may exploit the host bacterium-encoded lytic transglycosylase function to gain access to the receptor, thus facilitating its infection.


Asunto(s)
Actinomycetaceae , Bacteriófagos , Glicosiltransferasas , Actinomycetaceae/enzimología , Actinomycetaceae/virología , Receptores de Bacteriógrafos/metabolismo , Bacteriófagos/enzimología , Bacteriófagos/genética , Bacteriófagos/fisiología , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Especificidad del Huésped , Humanos , Microbiota , Boca/microbiología , Boca/virología , Mutación , Peptidoglicano/metabolismo , Plancton/virología , Proteínas Virales/genética , Proteínas Virales/metabolismo
11.
J Mol Biol ; 434(14): 167670, 2022 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-35671831

RESUMEN

Phages, as well as phage-derived proteins, especially lysins and depolymerases, are intensively studied to become prospective alternatives or supportive antibacterials used alone or in combination. In the common phage therapy approach, the unwanted emergence of phage-resistant variants from the treated bacterial population can be postponed or reduced by the utilization of an effective phage cocktail. In this work, we present a publicly available web tool PhREEPred (Phage Resistance Emergence Prediction) (https://phartner.shinyapps.io/PhREEPred/), which will allow an informed choice of the composition of phage cocktails by predicting the outcome of phage cocktail or phage/depolymerase combination treatments against encapsulated bacterial pathogens given a mutating population that escapes single phage treatment. PhREEPred simulates solutions of our mathematical model calibrated and tested on the experimental Klebsiella pneumoniae setup and Klebsiella-specific lytic phages: K63 type-specific phage KP34 equipped with a capsule-degrading enzyme (KP34p57), capsule-independent myoviruses KP15 and KP27, and recombinant capsule depolymerase KP34p57. The model can calculate the phage-resistance emergence depending on the bacterial growth rate and initial density, the multiplicity of infection, phage latent period, its infectiveness and the cocktail composition, as well as initial depolymerase concentration and activity rate. This model reproduced the experimental results and showed that (i) the phage cocktail of parallelly infecting phages is less effective than the one composed of sequentially infecting phages; (ii) depolymerase can delay or prevent bacterial resistance by unveiling an alternative receptor for initially inactive phages. In our opinion, this customer-friendly web tool will allow for the primary design of the phage cocktail and phage-depolymerase combination effectiveness against encapsulated pathogens.


Asunto(s)
Bacterias , Infecciones Bacterianas , Bacteriólisis , Bacteriófagos , Simulación por Computador , Uso de Internet , Terapia de Fagos , Bacterias/virología , Infecciones Bacterianas/terapia , Bacteriófagos/enzimología , Humanos , Klebsiella pneumoniae/virología , Modelos Teóricos , Estudios Prospectivos
12.
Viruses ; 14(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35215761

RESUMEN

Phages utilize lysis systems to allow the release of newly assembled viral particles that kill the bacterial host. This is also the case for phage AP1, which infects the rice pathogen Acidovorax oryzae. However, how lysis occurs on a molecular level is currently unknown. We performed in silico bioinformatics analyses, which indicated that the lysis cassette contains a holin (HolAP) and endolysin (LysAP), which are encoded by two adjacent genes. Recombinant expression of LysAP caused Escherichia coli lysis, while HolAP arrested growth. Co-expression of both proteins resulted in enhanced lysis activity compared to the individual proteins alone. Interestingly, LysAP contains a C-terminal region transmembrane domain, which is different from most known endolysins where a N-terminal hydrophobic region is found, with the potential to insert into the membrane. We show that the C-terminal transmembrane domain is crucial for protein localization and bacterial lysis in phage AP1. Our study characterizes the new phage lysis cassette and the mechanism to induce cell disruption, giving new insight in the understanding of phage life cycles.


Asunto(s)
Bacteriófagos/genética , Comamonadaceae/virología , Endopeptidasas/metabolismo , Genoma Viral/genética , Secuencia de Aminoácidos , Bacteriólisis , Bacteriófagos/enzimología , Bacteriófagos/fisiología , Biología Computacional , Endopeptidasas/genética , Escherichia coli/virología , Alineación de Secuencia , Proteínas Virales/genética , Proteínas Virales/metabolismo
13.
Int J Mol Sci ; 23(2)2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-35054821

RESUMEN

DNA polymerases are enzymes capable of synthesizing DNA. They are involved in replication of genomes of all cellular organisms as well as in processes of DNA repair and genetic recombination. However, DNA polymerases can also be encoded by viruses, including bacteriophages, and such enzymes are involved in viral DNA replication. DNA synthesizing enzymes are grouped in several families according to their structures and functions. Nevertheless, there are examples of bacteriophage-encoded DNA polymerases which are significantly different from other known enzymes capable of catalyzing synthesis of DNA. These differences are both structural and functional, indicating a huge biodiversity of bacteriophages and specific properties of their enzymes which had to evolve under certain conditions, selecting unusual properties of the enzymes which are nonetheless crucial for survival of these viruses, propagating as special kinds of obligatory parasites. In this review, we present a brief overview on DNA polymerases, and then we discuss unusual properties of different bacteriophage-encoded enzymes, such as those able to initiate DNA synthesis using the protein-priming mechanisms or even start this process without any primer, as well as able to incorporate untypical nucleotides. Apart from being extremely interesting examples of biochemical biodiversity, bacteriophage-encoded DNA polymerases can also be useful tools in genetic engineering and biotechnology.


Asunto(s)
Bacteriófagos/fisiología , ADN Polimerasa Dirigida por ADN/metabolismo , Bacteriófagos/enzimología , Ingeniería Genética , Proteínas Virales/metabolismo , Replicación Viral
14.
Nucleic Acids Res ; 50(15): 8401-8417, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-35066583

RESUMEN

PLEs (phage-inducible chromosomal island-like elements) are phage parasites integrated into the chromosome of epidemic Vibrio cholerae. In response to infection by its viral host ICP1, PLE excises, replicates and hijacks ICP1 structural components for transduction. Through an unknown mechanism, PLE prevents ICP1 from transitioning to rolling circle replication (RCR), a prerequisite for efficient packaging of the viral genome. Here, we characterize a PLE-encoded nuclease, NixI, that blocks phage development likely by nicking ICP1's genome as it transitions to RCR. NixI-dependent cleavage sites appear in ICP1's genome during infection of PLE(+) V. cholerae. Purified NixI demonstrates in vitro nuclease activity specifically for sites in ICP1's genome and we identify a motif that is necessary for NixI-mediated cleavage. Importantly, NixI is sufficient to limit ICP1 genome replication and eliminate progeny production, representing the most inhibitory PLE-encoded mechanism revealed to date. We identify distant NixI homologs in an expanded family of putative phage parasites in vibrios that lack nucleotide homology to PLEs but nonetheless share genomic synteny with PLEs. More generally, our results reveal a previously unknown mechanism deployed by phage parasites to limit packaging of their viral hosts' genome and highlight the prominent role of nuclease effectors as weapons in the arms race between antagonizing genomes.


Parasites of viruses, often referred to as satellites, are found in all domains of life and have been co-opted for host defense across diverse virus-host systems multiple independent times. This study describes the mechanism by which such an element prevents a bacterial virus (a 'phage') from otherwise infecting Vibrio cholera and related bacteria. The study is of broad interest to investigators with interests in phage-host interactions and microbial genetics.


Asunto(s)
Bacteriófagos , Desoxirribonucleasa I/metabolismo , Vibrio cholerae , Animales , Bacteriófagos/enzimología , Bacteriófagos/fisiología , Genoma Viral , Vibrio cholerae/genética , Vibrio cholerae/virología , Replicación Viral
15.
Nature ; 602(7895): 142-147, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35082445

RESUMEN

Public databases contain a planetary collection of nucleic acid sequences, but their systematic exploration has been inhibited by a lack of efficient methods for searching this corpus, which (at the time of writing) exceeds 20 petabases and is growing exponentially1. Here we developed a cloud computing infrastructure, Serratus, to enable ultra-high-throughput sequence alignment at the petabase scale. We searched 5.7 million biologically diverse samples (10.2 petabases) for the hallmark gene RNA-dependent RNA polymerase and identified well over 105 novel RNA viruses, thereby expanding the number of known species by roughly an order of magnitude. We characterized novel viruses related to coronaviruses, hepatitis delta virus and huge phages, respectively, and analysed their environmental reservoirs. To catalyse the ongoing revolution of viral discovery, we established a free and comprehensive database of these data and tools. Expanding the known sequence diversity of viruses can reveal the evolutionary origins of emerging pathogens and improve pathogen surveillance for the anticipation and mitigation of future pandemics.


Asunto(s)
Nube Computacional , Bases de Datos Genéticas , Virus ARN/genética , Virus ARN/aislamiento & purificación , Alineación de Secuencia/métodos , Virología/métodos , Viroma/genética , Animales , Archivos , Bacteriófagos/enzimología , Bacteriófagos/genética , Biodiversidad , Coronavirus/clasificación , Coronavirus/enzimología , Coronavirus/genética , Evolución Molecular , Virus de la Hepatitis Delta/enzimología , Virus de la Hepatitis Delta/genética , Humanos , Modelos Moleculares , Virus ARN/clasificación , Virus ARN/enzimología , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Programas Informáticos
16.
J Mol Biol ; 434(5): 167420, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-34954237

RESUMEN

Phages, plasmids, and other mobile genetic elements express inhibitors of CRISPR-Cas immune systems, known as anti-CRISPR proteins, to protect themselves from targeted destruction. These anti-CRISPR proteins have been shown to function through very diverse mechanisms. In this work we investigate the activity of an anti-CRISPR isolated from a prophage in Haemophilus parainfluenzae that blocks CRISPR-Cas9 DNA cleavage activity. We determine the three-dimensional crystal structure of AcrIIC4Hpa and show that it binds to the Cas9 Recognition Domain. This binding does not prevent the Cas9-anti-CRISPR complex from interacting with target DNA but does inhibit DNA cleavage. AcrIIC4Hpa likely acts by blocking the conformational changes that allow the HNH and RuvC endonuclease domains to contact the DNA sites to be nicked.


Asunto(s)
Bacteriófagos , Proteína 9 Asociada a CRISPR , División del ADN , Haemophilus parainfluenzae , Proteínas Virales , Bacteriófagos/enzimología , Proteína 9 Asociada a CRISPR/antagonistas & inhibidores , Proteína 9 Asociada a CRISPR/química , Haemophilus parainfluenzae/virología , Profagos/enzimología , Dominios Proteicos , Proteínas Virales/química , Proteínas Virales/metabolismo
17.
Biochem J ; 479(2): 207-223, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-34935873

RESUMEN

Bacteriophage endolysins degrade peptidoglycan and have been identified as antibacterial candidates to combat antimicrobial resistance. Considering the catalytic and structural diversity of endolysins, there is a paucity of structural data to inform how these enzymes work at the molecular level - key data that is needed to realize the potential of endolysin-based antibacterial agents. Here, we determine the atomic structure and define the enzymatic function of Escherichia coli O157:H7 phage FTEBc1 endolysin, LysT84. Bioinformatic analysis reveals that LysT84 is a modular endolysin, which is unusual for Gram-negative endolysins, comprising a peptidoglycan binding domain and an enzymatic domain. The crystal structure of LysT84 (2.99 Å) revealed a mostly α-helical protein with two domains connected by a linker region but packed together. LysT84 was determined to be a monomer in solution using analytical ultracentrifugation. Small-angle X-ray scattering data revealed that LysT84 is a flexible protein but does not have the expected bimodal P(r) function of a multidomain protein, suggesting that the domains of LysT84 pack closely creating a globular protein as seen in the crystal structure. Structural analysis reveals two key glutamate residues positioned on either side of the active site cavity; mutagenesis demonstrating these residues are critical for peptidoglycan degradation. Molecular dynamic simulations suggest that the enzymatically active domain is dynamic, allowing the appropriate positioning of these catalytic residues for hydrolysis of the ß(1-4) bond. Overall, our study defines the structural basis for peptidoglycan degradation by LysT84 which supports rational engineering of related endolysins into effective antibacterial agents.


Asunto(s)
Antibacterianos/química , Bacteriófagos/enzimología , Endopeptidasas/química , Escherichia coli O157/virología , Proteínas Virales/química , Antibacterianos/metabolismo , Biocatálisis , Dominio Catalítico , Pared Celular/metabolismo , Biología Computacional/métodos , Cristalización , Endopeptidasas/metabolismo , Ácido Glutámico/química , Hidrólisis , Simulación de Dinámica Molecular , Peptidoglicano/metabolismo , Conformación Proteica en Hélice alfa , Dominios Proteicos , Proteínas Virales/metabolismo
18.
Viruses ; 13(12)2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34960683

RESUMEN

Phage-derived therapies comprise phage therapy and the use of phage-derived proteins as anti-bacterial therapy. Bacteriophages are natural viruses that target specific bacteria. They were proposed to be used to treat bacterial infections in the 1920s, before the discovery and widespread over-commercialized use of antibiotics. Phage therapy was totally abandoned in Western countries, whereas it is still used in Poland, Georgia and Russia. We review here the history of phage therapy by focusing on bone and joint infection, and on the development of phage therapy in France in this indication. We discuss the rationale of its use in bacterial infection and show the feasibility of phage therapy in the 2020s, based on several patients with complex bone and joint infection who recently received phages as compassionate therapy. Although the status of phage therapy remains to be clarified by health care authorities, obtaining pharmaceutical-grade therapeutic phages (i.e., following good manufacturing practice guidelines or being "GMP-like") targeting bacterial species of concern is essential. Moreover, multidisciplinary clinical expertise has to determine what could be the relevant indications to perform clinical trials. Finally "phage therapy 2.0" has to integrate the following steps: (i) follow the status of phage therapy, that is not settled and defined; (ii) develop in each country a close relationship with the national health care authority; (iii) develop industrial-academic partnerships; (iv) create academic reference centers; (v) identify relevant clinical indications; (vi) use GMP/GMP-like phages with guaranteed quality bioproduction; (vii) start as salvage therapy; (vii) combine with antibiotics and adequate surgery; and (viii) perform clinical trials, to finally (ix) demonstrate in which clinical settings phage therapy provides benefit. Phage-derived proteins such as peptidoglycan hydrolases, polysaccharide depolymerases or lysins are enzymes that also have anti-biofilm activity. In contrast to phages, their development has to follow the classical process of medicinal products. Phage therapy and phage-derived products also have a huge potential to treat biofilm-associated bacterial diseases, and this is of crucial importance in the worldwide spread of antimicrobial resistance.


Asunto(s)
Infecciones Bacterianas/terapia , Enfermedades Óseas Infecciosas/terapia , Artropatías/terapia , Terapia de Fagos , Infecciones Relacionadas con Prótesis/terapia , Proteínas Virales/uso terapéutico , Antibacterianos/uso terapéutico , Artritis Infecciosa/terapia , Bacteriófagos/enzimología , Bacteriófagos/fisiología , Ensayos de Uso Compasivo , Humanos , Osteomielitis/terapia , Terapia de Fagos/normas , Proteínas Virales/metabolismo
19.
Viruses ; 13(12)2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34960808

RESUMEN

Ralstonia solanacearum is the causative agent of bacterial wilt, one of the most destructive plant diseases. While chemical control has an environmental impact, biological control strategies can allow sustainable agrosystems. Three lytic bacteriophages (phages) of R. solanacearum with biocontrol capacity in environmental water and plants were isolated from river water in Europe but not fully analysed, their genomic characterization being fundamental to understand their biology. In this work, the phage genomes were sequenced and subjected to bioinformatic analysis. The morphology was also observed by electron microscopy. Phylogenetic analyses were performed with a selection of phages able to infect R. solanacearum and the closely related phytopathogenic species R. pseudosolanacearum. The results indicated that the genomes of vRsoP-WF2, vRsoP-WM2 and vRsoP-WR2 range from 40,688 to 41,158 bp with almost 59% GC-contents, 52 ORFs in vRsoP-WF2 and vRsoP-WM2, and 53 in vRsoP-WR2 but, with only 22 or 23 predicted proteins with functional homologs in databases. Among them, two lysins and one exopolysaccharide (EPS) depolymerase, this type of depolymerase being identified in R. solanacearum phages for the first time. These three European phages belong to the same novel species within the Gyeongsanvirus, Autographiviridae family (formerly Podoviridae). These genomic data will contribute to a better understanding of the abilities of these phages to damage host cells and, consequently, to an improvement in the biological control of R. solanacearum.


Asunto(s)
Bacteriófagos/genética , Genoma Viral , Glicósido Hidrolasas/metabolismo , Control Biológico de Vectores/métodos , Ralstonia solanacearum/virología , Bacteriófagos/clasificación , Bacteriófagos/enzimología , Bacteriófagos/ultraestructura , Sistemas de Lectura Abierta , Filogenia , Virión/ultraestructura
20.
Viruses ; 13(10)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34696380

RESUMEN

Most endolysins of dsDNA phages are exported by a holin-dependent mechanism, while in some cases endolysins are exported via a holin-independent mechanism. However, it is still unclear whether the same endolysins can be exported by both holin-dependent and holin-independent mechanisms. This study investigated the lysis system of OP2-like phage X2 infecting Xanthomonas oryzae pv. oryzae, causing devastating bacterial leaf blight disease in rice. Based on bioinformatics and protein biochemistry methods, we show that phage X2 employs the classic "holin-endolysin" lysis system. The endolysin acts on the cell envelope and exhibits antibacterial effects in vitro, while the holin facilitates the release of the protein into the periplasm. We also characterized the role of the transmembrane domain (TMD) in the translocation of the endolysin across the inner membrane. We found that the TMD facilitated the translocation of the endolysin via the Sec secretion system. The holin increases the efficiency of protein release, leading to faster and more efficient lysis. Interestingly, in E. coli, the expression of either holin or endolysin with TMDs resulted in the formation of long rod shaped cells. We conclude that the TMD of X2-Lys plays a dual role: One is the transmembrane transport while the other is the inhibition of cell division, resulting in larger cells and thus in a higher number of released viruses per cell.


Asunto(s)
Bacteriófagos/enzimología , Bacteriófagos/genética , Endopeptidasas/genética , Proteínas Virales/genética , Xanthomonas/virología , Bacteriólisis , Bacteriófagos/clasificación , Endopeptidasas/metabolismo , Escherichia coli/genética , Filogenia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA