Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 14(3)2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35336962

RESUMEN

Salmonella enterica serovar Newport bacteriophage 7-11 shares 41 homologous ORFs with Escherichia coli phage phiEco32, and both phages encode a protein similar to bacterial RNA polymerase promoter specificity σ subunit. Here, we investigated the temporal pattern of 7-11 gene expression during infection and compared it to the previously determined transcription strategy of phiEco32. Using primer extension and in vitro transcription assays, we identified eight promoters recognized by host RNA polymerase holoenzyme containing 7-11 σ subunit SaPh711_gp47. These promoters are characterized by a bipartite consensus, GTAAtg-(16)-aCTA, and are located upstream of late phage genes. While dissimilar from single-element middle and late promoters of phiEco32 recognized by holoenzymes formed by the phi32_gp36 σ factor, the 7-11 late promoters are located at genome positions similar to those of phiEco32 middle and late promoters. Two early 7-11 promoters are recognized by the RNA polymerase holoenzyme containing the host primary σ70 factor. Unlike the case of phiEco32, no shut-off of σ70-dependent transcription is observed during 7-11 infection and there are no middle promoters. These differences can be explained by the fact that phage 7-11 does not encode a homologue of phi32_gp79, an inhibitor of host and early phage transcription and an activator of transcription by the phi32_gp36-holoenzyme.


Asunto(s)
Bacteriófagos , Factor sigma , Bacteriófagos/genética , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación de la Expresión Génica , Holoenzimas/genética , Holoenzimas/metabolismo , Factor sigma/genética , Factor sigma/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
2.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34035168

RESUMEN

For Type I CRISPR-Cas systems, a mode of CRISPR adaptation named priming has been described. Priming allows specific and highly efficient acquisition of new spacers from DNA recognized (primed) by the Cascade-crRNA (CRISPR RNA) effector complex. Recognition of the priming protospacer by Cascade-crRNA serves as a signal for engaging the Cas3 nuclease-helicase required for both interference and primed adaptation, suggesting the existence of a primed adaptation complex (PAC) containing the Cas1-Cas2 adaptation integrase and Cas3. To detect this complex in vivo, we here performed chromatin immunoprecipitation with Cas3-specific and Cas1-specific antibodies using cells undergoing primed adaptation. We found that prespacers are bound by both Cas1 (presumably, as part of the Cas1-Cas2 integrase) and Cas3, implying direct physical association of the interference and adaptation machineries as part of PAC.


Asunto(s)
ADN Helicasas/metabolismo , Endonucleasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Sistemas CRISPR-Cas
3.
Nature ; 589(7841): 306-309, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33208949

RESUMEN

CrAss-like phages are a recently described expansive group of viruses that includes the most abundant virus in the human gut1-3. The genomes of all crAss-like phages encode a large virion-packaged protein2,4 that contains a DFDxD sequence motif, which forms the catalytic site in cellular multisubunit RNA polymerases (RNAPs)5. Here, using Cellulophaga baltica crAss-like phage phi14:2 as a model system, we show that this protein is a DNA-dependent RNAP that is translocated into the host cell along with the phage DNA and transcribes early phage genes. We determined the crystal structure of this 2,180-residue enzyme in a self-inhibited state, which probably occurs before virion packaging. This conformation is attained with the help of a cleft-blocking domain that interacts with the active site and occupies the cavity in which the RNA-DNA hybrid binds. Structurally, phi14:2 RNAP is most similar to eukaryotic RNAPs that are involved in RNA interference6,7, although most of the phi14:2 RNAP structure (nearly 1,600 residues) maps to a new region of the protein fold space. Considering this structural similarity, we propose that eukaryal RNA interference polymerases have their origins in phage, which parallels the emergence of the mitochondrial transcription apparatus8.


Asunto(s)
Bacteriófagos/clasificación , Bacteriófagos/enzimología , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/metabolismo , Flavobacteriaceae/virología , Bacteriófagos/genética , Dominio Catalítico , Sistema Libre de Células , Cristalografía por Rayos X , ADN de Cadena Simple/biosíntesis , ADN de Cadena Simple/genética , ARN Polimerasas Dirigidas por ADN/genética , Evolución Molecular , Regulación Viral de la Expresión Génica , Genes Virales/genética , Modelos Biológicos , Modelos Moleculares , Dominios Proteicos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Interferencia de ARN , Transcripción Genética
4.
Viruses ; 12(9)2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32887488

RESUMEN

In this study, we describe the biological function of the phage-encoded protein RNA polymerase alpha subunit cleavage protein (Rac), a predicted Gcn5-related acetyltransferase encoded by phiKMV-like viruses. These phages encode a single-subunit RNA polymerase for transcription of their late (structure- and lysis-associated) genes, whereas the bacterial RNA polymerase is used at the earlier stages of infection. Rac mediates the inactivation of bacterial transcription by introducing a specific cleavage in the α subunit of the bacterial RNA polymerase. This cleavage occurs within the flexible linker sequence and disconnects the C-terminal domain, required for transcription initiation from most highly active cellular promoters. To achieve this, Rac likely taps into a novel post-translational modification (PTM) mechanism within the host Pseudomonas aeruginosa. From an evolutionary perspective, this novel phage-encoded regulation mechanism confirms the importance of PTMs in the prokaryotic metabolism and represents a new way by which phages can hijack the bacterial host metabolism.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Fagos Pseudomonas/enzimología , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/virología , Proteínas Virales/metabolismo , Acetiltransferasas/genética , Proteínas Bacterianas/genética , ARN Polimerasas Dirigidas por ADN/genética , Interacciones Huésped-Patógeno , Fagos Pseudomonas/genética , Pseudomonas aeruginosa/genética , Transcripción Genética , Proteínas Virales/genética
5.
Nucleic Acids Res ; 48(17): 9787-9803, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32821943

RESUMEN

Type III CRISPR-Cas systems provide immunity to foreign DNA by targeting its transcripts. Target recognition activates RNases and DNases that may either destroy foreign DNA directly or elicit collateral damage inducing death of infected cells. While some Type III systems encode a reverse transcriptase to acquire spacers from foreign transcripts, most contain conventional spacer acquisition machinery found in DNA-targeting systems. We studied Type III spacer acquisition in phage-infected Thermus thermophilus, a bacterium that lacks either a standalone reverse transcriptase or its fusion to spacer integrase Cas1. Cells with spacers targeting a subset of phage transcripts survived the infection, indicating that Type III immunity does not operate through altruistic suicide. In the absence of selection spacers were acquired from both strands of phage DNA, indicating that no mechanism ensuring acquisition of RNA-targeting spacers exists. Spacers that protect the host from the phage demonstrate a very strong strand bias due to positive selection during infection. Phages that escaped Type III interference accumulated deletions of integral number of codons in an essential gene and much longer deletions in a non-essential gene. This and the fact that Type III immunity can be provided by plasmid-borne mini-arrays open ways for genomic manipulation of Thermus phages.


Asunto(s)
Bacteriófagos/fisiología , Sistemas CRISPR-Cas , Thermus thermophilus/genética , Thermus thermophilus/virología , Bacteriófagos/genética , Bacteriófagos/patogenicidad , Genes Bacterianos , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno/genética , ADN Polimerasa Dirigida por ARN/genética
6.
Viruses ; 12(8)2020 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-32722583

RESUMEN

The Escherichia coli bacteriophage T5 has three temporal classes of genes (pre-early, early, and late). All three classes are transcribed by host RNA polymerase (RNAP) containing the σ70 promoter specificity subunit. Molecular mechanisms responsible for the switching of viral transcription from one class to another remain unknown. Here, we find the product of T5 gene 026 (gpT5.026) in RNAP preparations purified from T5-infected cells and demonstrate in vitro its tight binding to E. coli RNAP. While proteins homologous to gpT5.026 are encoded by all T5-related phages, no similarities to proteins with known functions can be detected. GpT5.026 binds to two regions of the RNAP ß subunit and moderately inhibits RNAP interaction with the discriminator region of σ70-dependent promoters. A T5 mutant with disrupted gene 026 is viable, but the host cell lysis phase is prolongated and fewer virus particles are produced. During the mutant phage infection, the number of early transcripts increases, whereas the number of late transcripts decreases. We propose that gpT5.026 is part of the regulatory cascade that orchestrates a switch from early to late bacteriophage T5 transcription.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Colifagos/genética , ARN Polimerasas Dirigidas por ADN/genética , Proteínas Virales/genética , Proteínas Portadoras/aislamiento & purificación , Regulación Viral de la Expresión Génica , Unión Proteica , Transcripción Genética
7.
Proc Natl Acad Sci U S A ; 116(9): 3556-3561, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30737287

RESUMEN

Double-stranded DNA viruses, including bacteriophages and herpesviruses, package their genomes into preformed capsids, using ATP-driven motors. Seeking to advance structural and mechanistic understanding, we established in vitro packaging for a thermostable bacteriophage, P23-45 of Thermus thermophilus Both the unexpanded procapsid and the expanded mature capsid can package DNA in the presence of packaging ATPase over the 20 °C to 70 °C temperature range, with optimum activity at 50 °C to 65 °C. Cryo-EM reconstructions for the mature and immature capsids at 3.7-Å and 4.4-Å resolution, respectively, reveal conformational changes during capsid expansion. Capsomer interactions in the expanded capsid are reinforced by formation of intersubunit ß-sheets with N-terminal segments of auxiliary protein trimers. Unexpectedly, the capsid has T=7 quasi-symmetry, despite the P23-45 genome being twice as large as those of known T=7 phages, in which the DNA is compacted to near-crystalline density. Our data explain this anomaly, showing how the canonical HK97 fold has adapted to double the volume of the capsid, while maintaining its structural integrity. Reconstructions of the procapsid and the expanded capsid defined the structure of the single vertex containing the portal protein. Together with a 1.95-Å resolution crystal structure of the portal protein and DNA packaging assays, these reconstructions indicate that capsid expansion affects the conformation of the portal protein, while still allowing DNA to be packaged. These observations suggest a mechanism by which structural events inside the capsid can be communicated to the outside.


Asunto(s)
Bacteriófagos/ultraestructura , Cápside/ultraestructura , Empaquetamiento del ADN/genética , Virus ADN/ultraestructura , Bacteriófagos/genética , Microscopía por Crioelectrón , Virus ADN/genética , ADN Viral/genética , ADN Viral/ultraestructura , Virión/genética , Virión/ultraestructura , Ensamble de Virus/genética
8.
Nucleic Acids Res ; 46(20): 10810-10826, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30295835

RESUMEN

C-proteins control restriction-modification (R-M) systems' genes transcription to ensure sufficient levels of restriction endonuclease to allow protection from foreign DNA while avoiding its modification by excess methyltransferase. Here, we characterize transcription regulation in C-protein dependent R-M system Kpn2I. The Kpn2I restriction endonuclease gene is transcribed from a constitutive, weak promoter, which, atypically, is C-protein independent. Kpn2I C-protein (C.Kpn2I) binds upstream of the strong methyltransferase gene promoter and inhibits it, likely by preventing the interaction of the RNA polymerase sigma subunit with the -35 consensus element. Diminished transcription from the methyltransferase promoter increases transcription from overlapping divergent C-protein gene promoters. All known C-proteins affect transcription initiation from R-M genes promoters. Uniquely, the C.Kpn2I binding site is located within the coding region of its gene. C.Kpn2I acts as a roadblock stalling elongating RNA polymerase and decreasing production of full-length C.Kpn2I mRNA. Mathematical modeling shows that this unusual mode of regulation leads to the same dynamics of accumulation of R-M gene transcripts as observed in systems where C-proteins act at transcription initiation stage only. Bioinformatics analyses suggest that transcription regulation through binding of C.Kpn2I-like proteins within the coding regions of their genes may be widespread.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endodesoxirribonucleasas/metabolismo , Klebsiella pneumoniae/genética , Transcripción Genética , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Sitios de Unión , Codón Iniciador , Biología Computacional , Desoxirribonucleasa I/metabolismo , Endodesoxirribonucleasas/genética , Escherichia coli/metabolismo , Funciones de Verosimilitud , Filogenia , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Dominios Proteicos , Termodinámica
9.
Nucleic Acids Res ; 45(6): 3297-3307, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28204574

RESUMEN

During primed CRISPR adaptation spacers are preferentially selected from DNA recognized by CRISPR interference machinery, which in the case of Type I CRISPR-Cas systems consists of CRISPR RNA (crRNA) bound effector Cascade complex that locates complementary targets, and Cas3 executor nuclease/helicase. A complex of Cas1 and Cas2 proteins is capable of inserting new spacers in the CRISPR array. Here, we show that in Escherichia coli cells undergoing primed adaptation, spacer-sized fragments of foreign DNA are associated with Cas1. Based on sensitivity to digestion with nucleases, the associated DNA is not in a standard double-stranded state. Spacer-sized fragments are cut from one strand of foreign DNA in Cas1- and Cas3-dependent manner. These fragments are generated from much longer S1-nuclease sensitive fragments of foreign DNA that require Cas3 for their production. We propose that in the course of CRISPR interference Cas3 generates fragments of foreign DNA that are recognized by the Cas1-Cas2 adaptation complex, which excises spacer-sized fragments and channels them for insertion into CRISPR array.


Asunto(s)
Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Endodesoxirribonucleasas/metabolismo , Proteínas de Escherichia coli/metabolismo , ADN/química , ADN/metabolismo , ADN Helicasas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Plásmidos/genética
10.
Nucleic Acids Res ; 45(6): 3580-3590, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28100693

RESUMEN

Bacteriophages and large dsDNA viruses encode sophisticated machinery to translocate their DNA into a preformed empty capsid. An essential part of this machine, the large terminase protein, processes viral DNA into constituent units utilizing its nuclease activity. Crystal structures of the large terminase nuclease from the thermophilic bacteriophage G20c show that it is most similar to the RuvC family of the RNase H-like endonucleases. Like RuvC proteins, the nuclease requires either Mn2+, Mg2+ or Co2+ ions for activity, but is inactive with Zn2+ and Ca2+. High resolution crystal structures of complexes with different metals reveal that in the absence of DNA, only one catalytic metal ion is accommodated in the active site. Binding of the second metal ion may be facilitated by conformational variability, which enables the two catalytic aspartic acids to be brought closer to each other. Structural comparison indicates that in common with the RuvC family, the location of the two catalytic metals differs from other members of the RNase H family. In contrast to a recently proposed mechanism, the available data do not support binding of the two metals at an ultra-short interatomic distance. Thus we postulate that viral terminases cleave DNA by the canonical RuvC-like mechanism.


Asunto(s)
Endodesoxirribonucleasas/química , Metales/química , Proteínas Virales/química , Biocatálisis , Dominio Catalítico , División del ADN , ADN Viral/metabolismo , Endodesoxirribonucleasas/metabolismo , Genoma Viral , Modelos Moleculares , Recombinasas/química , Thermus thermophilus/enzimología , Proteínas Virales/metabolismo , Ensamble de Virus
11.
Proc Natl Acad Sci U S A ; 112(7): 2017-22, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25646468

RESUMEN

Transcription antitermination is a common strategy of gene expression regulation, but only a few transcription antitermination factors have been studied in detail. Here, we dissect the transcription antitermination mechanism of Xanthomonas oryzae virus Xp10 protein p7, which binds host RNA polymerase (RNAP) and regulates both transcription initiation and termination. We show that p7 suppresses intrinsic termination by decreasing RNAP pausing and increasing the transcription complex stability, in cooperation with host-encoded factor NusA. Uniquely, the antitermination activity of p7 depends on the ω subunit of the RNAP core and is modulated by ppGpp. In contrast, the inhibition of transcription initiation by p7 does not require ω but depends on other RNAP sites. Our results suggest that p7, a bifunctional transcription factor, uses distinct mechanisms to control different steps of transcription. We propose that regulatory functions of the ω subunit revealed by our analysis may extend to its homologs in eukaryotic RNAPs.


Asunto(s)
Bacteriófagos/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Regiones Terminadoras Genéticas , Xanthomonas/virología
12.
J Virol ; 88(18): 10501-10, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24965474

RESUMEN

UNLABELLED: Pseudomonas aeruginosa bacteriophage ϕKZ is the type representative of the giant phage genus, which is characterized by unusually large virions and genomes. By unraveling the transcriptional map of the ∼ 280-kb ϕKZ genome to single-nucleotide resolution, we combine 369 ϕKZ genes into 134 operons. Early transcription is initiated from highly conserved AT-rich promoters distributed across the ϕKZ genome and located on the same strand of the genome. Early transcription does not require phage or host protein synthesis. Transcription of middle and late genes is dependent on protein synthesis and mediated by poorly conserved middle and late promoters. Unique to ϕKZ is its ability to complete its infection in the absence of bacterial RNA polymerase (RNAP) enzyme activity. We propose that transcription of the ϕKZ genome is performed by the consecutive action of two ϕKZ-encoded, noncanonical multisubunit RNAPs, one of which is packed within the virion, another being the product of early genes. This unique, rifampin-resistant transcriptional machinery is conserved within the diverse giant phage genus. IMPORTANCE: The data presented in this paper offer, for the first time, insight into the complex transcriptional scheme of giant bacteriophages. We show that Pseudomonas aeruginosa giant phage ϕKZ is able to infect and lyse its host cell and produce phage progeny in the absence of functional bacterial transcriptional machinery. This unique property can be attributed to two phage-encoded putative RNAP enzymes, which contain very distant homologues of bacterial ß and ß'-like RNAP subunits.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteriófagos/crecimiento & desarrollo , ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación Viral de la Expresión Génica , Fagos Pseudomonas/crecimiento & desarrollo , Pseudomonas aeruginosa/enzimología , Proteínas Bacterianas/genética , Bacteriófagos/enzimología , Bacteriófagos/genética , Bacteriófagos/fisiología , ARN Polimerasas Dirigidas por ADN/genética , Genoma Viral , Interacciones Huésped-Patógeno , Fagos Pseudomonas/enzimología , Fagos Pseudomonas/genética , Fagos Pseudomonas/fisiología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/virología , Transcripción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
13.
Virology ; 436(1): 67-74, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23127595

RESUMEN

Escherichia coli bacteriophage T7 is a founding member of a large clade of podoviruses encoding a single-subunit RNA polymerase (RNAP). Phages of the family rely on host RNAP for transcription of early viral genes; viral RNAP transcribes non-early viral genes. T7 and its close relatives encode an inhibitor of host RNAP, the gp2 protein. Gp2 is essential for phage development and ensures that host RNAP does not interfere with viral RNAP transcription at late stages of infection. Here, we identify host RNAP inhibitors encoded by a subset of T7 clade phages related to ϕKMV phage of Pseudomonas aeruginosa. We demonstrate that these proteins are functionally identical to T7 gp2 in vivo and in vitro. The ability of some Pseudomonas phage gp2-like proteins to inhibit RNAP is modulated by N-terminal domains, which are absent from the T7 phage homolog. This finding indicates that Pseudomonas phages may use external or internal cues to initiate inhibition of host RNAP transcription and that gp2-like proteins from these phages may be receptors of these cues.


Asunto(s)
Bacteriófago T7/genética , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Fagos Pseudomonas/genética , Pseudomonas aeruginosa/virología , Proteínas Represoras/genética , Bacteriófago T7/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Inhibidores Enzimáticos/metabolismo , Escherichia coli/virología , Genes Virales , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Fagos Pseudomonas/metabolismo , Proteínas Represoras/metabolismo , Alineación de Secuencia , Transcripción Genética , Proteínas Virales
14.
J Mol Biol ; 416(3): 389-99, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22261232

RESUMEN

Escherichia coli phage phiEco32 encodes two proteins that bind to host RNA polymerase (RNAP): gp79, a novel protein, and gp36, a distant homolog of σ(70) family proteins. Here, we investigated the temporal pattern of phiEco32 and host gene expression during infection. Host transcription shutoff and three distinct bacteriophage temporal gene classes (early, middle, and late) were revealed. A combination of bioinformatic and biochemical approaches allowed identification of phage promoters recognized by a host RNAP holoenzyme containing the σ(70) factor. These promoters are located upstream of early phage genes. A combination of macroarray data, primer extension, and in vitro transcription analyses allowed identification of six promoters recognized by an RNAP holoenzyme containing gp36. These promoters are characterized by a single-consensus element tAATGTAtA and are located upstream of the middle and late phage genes. Curiously, gp79, an inhibitor of host and early phage transcription by σ(70) holoenzyme, activated transcription by the gp36 holoenzyme in vitro.


Asunto(s)
Colifagos/genética , Escherichia coli/virología , Regulación Viral de la Expresión Génica , Secuencia de Bases , Biología Computacional , ARN Polimerasas Dirigidas por ADN/metabolismo , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Factor sigma/metabolismo , Transcripción Genética , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...