Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(4): 864-876.e21, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36750095

RESUMEN

A fundamental strategy of eukaryotic antiviral immunity involves the cGAS enzyme, which synthesizes 2',3'-cGAMP and activates the effector STING. Diverse bacteria contain cGAS-like enzymes that produce cyclic oligonucleotides and induce anti-phage activity, known as CBASS. However, this activity has only been demonstrated through heterologous expression. Whether bacteria harboring CBASS antagonize and co-evolve with phages is unknown. Here, we identified an endogenous cGAS-like enzyme in Pseudomonas aeruginosa that generates 3',3'-cGAMP during phage infection, signals to a phospholipase effector, and limits phage replication. In response, phages express an anti-CBASS protein ("Acb2") that forms a hexamer with three 3',3'-cGAMP molecules and reduces phospholipase activity. Acb2 also binds to molecules produced by other bacterial cGAS-like enzymes (3',3'-cUU/UA/UG/AA) and mammalian cGAS (2',3'-cGAMP), suggesting broad inhibition of cGAS-based immunity. Upon Acb2 deletion, CBASS blocks lytic phage replication and lysogenic induction, but rare phages evade CBASS through major capsid gene mutations. Altogether, we demonstrate endogenous CBASS anti-phage function and strategies of CBASS inhibition and evasion.


Asunto(s)
Bacterias , Bacteriófagos , Animales , Bacterias/inmunología , Bacterias/virología , Bacteriófagos/fisiología , Inmunidad , Nucleotidiltransferasas/metabolismo
2.
Cell ; 178(6): 1452-1464.e13, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31474367

RESUMEN

Phages express anti-CRISPR (Acr) proteins to inhibit CRISPR-Cas systems that would otherwise destroy their genomes. Most acr genes are located adjacent to anti-CRISPR-associated (aca) genes, which encode proteins with a helix-turn-helix DNA-binding motif. The conservation of aca genes has served as a signpost for the identification of acr genes, but the function of the proteins encoded by these genes has not been investigated. Here we reveal that an acr-associated promoter drives high levels of acr transcription immediately after phage DNA injection and that Aca proteins subsequently repress this transcription. Without Aca activity, this strong transcription is lethal to a phage. Our results demonstrate how sufficient levels of Acr proteins accumulate early in the infection process to inhibit existing CRISPR-Cas complexes in the host cell. They also imply that the conserved role of Aca proteins is to mitigate the deleterious effects of strong constitutive transcription from acr promoters.


Asunto(s)
Bacteriófagos/genética , Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Proteínas Virales/genética , Proteínas Asociadas a CRISPR/genética , Escherichia coli/virología , Regiones Promotoras Genéticas/genética , Pseudomonas aeruginosa/virología , Factores de Transcripción/genética , Transcripción Genética
3.
Cell ; 174(4): 917-925.e10, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30033364

RESUMEN

Bacteria utilize CRISPR-Cas adaptive immune systems for protection from bacteriophages (phages), and some phages produce anti-CRISPR (Acr) proteins that inhibit immune function. Despite thorough mechanistic and structural information for some Acr proteins, how they are deployed and utilized by a phage during infection is unknown. Here, we show that Acr production does not guarantee phage replication when faced with CRISPR-Cas immunity, but instead, infections fail when phage population numbers fall below a critical threshold. Infections succeed only if a sufficient Acr dose is contributed to a single cell by multiple phage genomes. The production of Acr proteins by phage genomes that fail to replicate leave the cell immunosuppressed, which predisposes the cell for successful infection by other phages in the population. This altruistic mechanism for CRISPR-Cas inhibition demonstrates inter-virus cooperation that may also manifest in other host-parasite interactions.


Asunto(s)
Bacteriófagos/inmunología , Sistemas CRISPR-Cas/inmunología , Interacciones Huésped-Patógeno/inmunología , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/virología , Proteínas Virales/inmunología , Evolución Molecular , Pseudomonas aeruginosa/genética , Proteínas Virales/metabolismo
4.
Cell ; 168(1-2): 150-158.e10, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-28041849

RESUMEN

Bacterial CRISPR-Cas systems utilize sequence-specific RNA-guided nucleases to defend against bacteriophage infection. As a countermeasure, numerous phages are known that produce proteins to block the function of class 1 CRISPR-Cas systems. However, currently no proteins are known to inhibit the widely used class 2 CRISPR-Cas9 system. To find these inhibitors, we searched cas9-containing bacterial genomes for the co-existence of a CRISPR spacer and its target, a potential indicator for CRISPR inhibition. This analysis led to the discovery of four unique type II-A CRISPR-Cas9 inhibitor proteins encoded by Listeria monocytogenes prophages. More than half of L. monocytogenes strains with cas9 contain at least one prophage-encoded inhibitor, suggesting widespread CRISPR-Cas9 inactivation. Two of these inhibitors also blocked the widely used Streptococcus pyogenes Cas9 when assayed in Escherichia coli and human cells. These natural Cas9-specific "anti-CRISPRs" present tools that can be used to regulate the genome engineering activities of CRISPR-Cas9.


Asunto(s)
Bacteriófagos/metabolismo , Sistemas CRISPR-Cas , Endonucleasas/antagonistas & inhibidores , Ingeniería Genética , Listeria monocytogenes/enzimología , Proteínas Bacterianas/antagonistas & inhibidores , Proteína 9 Asociada a CRISPR , Escherichia coli , Células HEK293 , Humanos , Listeria monocytogenes/inmunología , Listeria monocytogenes/virología , Profagos
5.
Cell ; 169(1): 47-57.e11, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28340349

RESUMEN

Genetic conflict between viruses and their hosts drives evolution and genetic innovation. Prokaryotes evolved CRISPR-mediated adaptive immune systems for protection from viral infection, and viruses have evolved diverse anti-CRISPR (Acr) proteins that subvert these immune systems. The adaptive immune system in Pseudomonas aeruginosa (type I-F) relies on a 350 kDa CRISPR RNA (crRNA)-guided surveillance complex (Csy complex) to bind foreign DNA and recruit a trans-acting nuclease for target degradation. Here, we report the cryo-electron microscopy (cryo-EM) structure of the Csy complex bound to two different Acr proteins, AcrF1 and AcrF2, at an average resolution of 3.4 Å. The structure explains the molecular mechanism for immune system suppression, and structure-guided mutations show that the Acr proteins bind to residues essential for crRNA-mediated detection of DNA. Collectively, these data provide a snapshot of an ongoing molecular arms race between viral suppressors and the immune system they target.


Asunto(s)
Bacteriófagos/química , Proteínas Asociadas a CRISPR/química , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/virología , ARN Bacteriano/química , Proteínas Virales/química , Bacteriófagos/clasificación , Bacteriófagos/genética , Microscopía por Crioelectrón , Cristalografía por Rayos X , Vigilancia Inmunológica , Modelos Moleculares , Pseudomonas aeruginosa/genética , ARN Bacteriano/metabolismo , ARN Bacteriano/ultraestructura , Proteínas Virales/ultraestructura
6.
Mol Cell ; 84(2): 375-385.e7, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38103556

RESUMEN

Cyclic-oligonucleotide-based anti-phage signaling system (CBASS) is a common immune system that uses cyclic oligonucleotide signals to limit phage replication. In turn, phages encode anti-CBASS (Acb) proteins such as Acb2, which can sequester some cyclic dinucleotides (CDNs) and limit downstream effector activation. Here, we identified that Acb2 sequesters many CDNs produced by CBASS systems and inhibits stimulator of interferon genes (STING) activity in human cells. Surprisingly, the Acb2 hexamer also binds with high affinity to CBASS cyclic trinucleotides (CTNs) 3'3'3'-cyclic AMP-AMP-AMP and 3'3'3'-cAAG at a distinct site from CDNs. One Acb2 hexamer can simultaneously bind two CTNs and three CDNs. Phage-encoded Acb2 provides protection from type III-C CBASS that uses cA3 signaling molecules. Moreover, phylogenetic analysis of >2,000 Acb2 homologs encoded by diverse phages and prophages revealed that most are expected to bind both CTNs and CDNs. Altogether, Acb2 sequesters nearly all known CBASS signaling molecules through two distinct binding pockets and therefore serves as a broad-spectrum inhibitor of cGAS-based immunity.


Asunto(s)
Bacteriófagos , Nucleótidos Cíclicos , Humanos , Nucleótidos Cíclicos/metabolismo , Bacteriófagos/genética , Bacteriófagos/metabolismo , Filogenia , AMP Cíclico , Oligonucleótidos
7.
Mol Cell ; 82(11): 2161-2166.e3, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35623354

RESUMEN

CRISPR systems are prokaryotic adaptive immune systems that use RNA-guided Cas nucleases to recognize and destroy foreign genetic elements. To overcome CRISPR immunity, bacteriophages have evolved diverse families of anti-CRISPR proteins (Acrs). Recently, Lin et al. (2020) described the discovery and characterization of 7 Acr families (AcrVIA1-7) that inhibit type VI-A CRISPR systems. We detail several inconsistencies that question the results reported in the Lin et al. (2020) study. These include inaccurate bioinformatics analyses and bacterial strains that are impossible to construct. Published strains were provided by the authors, but MS2 bacteriophage plaque assays did not support the published results. We also independently tested the Acr sequences described in the original report, in E. coli and mammalian cells, but did not observe anti-Cas13a activity. Taken together, our data and analyses prompt us to question the claim that AcrVIA1-7 reported in Lin et al. are type VI anti-CRISPR proteins.


Asunto(s)
Bacteriófagos , Proteínas Asociadas a CRISPR , Animales , Bacteriófagos/genética , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Escherichia coli/genética , Escherichia coli/metabolismo , Leptotrichia/genética , Mamíferos/metabolismo , Profagos/genética , Profagos/metabolismo , Ribonucleasas/metabolismo
8.
Mol Cell ; 81(3): 571-583.e6, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33412111

RESUMEN

The arms race between bacteria and phages has led to the evolution of diverse anti-phage defenses, several of which are controlled by quorum-sensing pathways. In this work, we characterize a quorum-sensing anti-activator protein, Aqs1, found in Pseudomonas phage DMS3. We show that Aqs1 inhibits LasR, the master regulator of quorum sensing, and present the crystal structure of the Aqs1-LasR complex. The 69-residue Aqs1 protein also inhibits PilB, the type IV pilus assembly ATPase protein, which blocks superinfection by phages that require the pilus for infection. This study highlights the remarkable ability of small phage proteins to bind multiple host proteins and disrupt key biological pathways. As quorum sensing influences various anti-phage defenses, Aqs1 provides a mechanism by which infecting phages might simultaneously dampen multiple defenses. Because quorum-sensing systems are broadly distributed across bacteria, this mechanism of phage counter-defense may play an important role in phage-host evolutionary dynamics.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteriófagos/metabolismo , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum , Transactivadores/metabolismo , Proteínas Virales/metabolismo , Proteínas Bacterianas/genética , Bacteriófagos/genética , Bacteriófagos/patogenicidad , Fimbrias Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/crecimiento & desarrollo , Piocianina/metabolismo , Transactivadores/genética , Proteínas Virales/genética
9.
Mol Cell ; 73(3): 601-610.e5, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30595438

RESUMEN

CRISPR-Cas immune systems utilize RNA-guided nucleases to protect bacteria from bacteriophage infection. Bacteriophages have in turn evolved inhibitory "anti-CRISPR" (Acr) proteins, including six inhibitors (AcrIIA1-AcrIIA6) that can block DNA cutting and genome editing by type II-A CRISPR-Cas9 enzymes. We show here that AcrIIA2 and its more potent homolog, AcrIIA2b, prevent Cas9 binding to DNA by occluding protein residues required for DNA binding. Cryo-EM-determined structures of AcrIIA2 or AcrIIA2b bound to S. pyogenes Cas9 reveal a mode of competitive inhibition of DNA binding that is distinct from other known Acrs. Differences in the temperature dependence of Cas9 inhibition by AcrIIA2 and AcrIIA2b arise from differences in both inhibitor structure and the local inhibitor-binding environment on Cas9. These findings expand the natural toolbox for regulating CRISPR-Cas9 genome editing temporally, spatially, and conditionally.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas , ADN/metabolismo , Edición Génica/métodos , Fagos Pseudomonas/metabolismo , Pseudomonas aeruginosa/enzimología , ARN Guía de Kinetoplastida/metabolismo , Temperatura , Proteínas Virales/metabolismo , Unión Competitiva , Proteína 9 Asociada a CRISPR/antagonistas & inhibidores , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/ultraestructura , Microscopía por Crioelectrón , ADN/genética , ADN/ultraestructura , Escherichia coli/enzimología , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , Conformación Proteica , Fagos Pseudomonas/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/virología , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/ultraestructura , Relación Estructura-Actividad , Proteínas Virales/genética , Proteínas Virales/ultraestructura
10.
PLoS Biol ; 21(12): e3002431, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38064533

RESUMEN

Bacteriophages encode anti-CRISPR (Acr) proteins that inactivate CRISPR-Cas bacterial immune systems, allowing successful invasion, replication, and prophage integration. Acr proteins inhibit CRISPR-Cas systems using a wide variety of mechanisms. AcrIIA1 is encoded by numerous phages and plasmids, binds specifically to the Cas9 HNH domain, and was the first Acr discovered to inhibit SpyCas9. Here, we report the observation of AcrIIA1-induced degradation of SpyCas9 and SauCas9 in human cell culture, the first example of Acr-induced degradation of CRISPR-Cas nucleases in human cells. AcrIIA1-induced degradation of SpyCas9 is abolished by mutations in AcrIIA1 that break a direct physical interaction between the 2 proteins. Targeted Cas9 protein degradation by AcrIIA1 could modulate Cas9 nuclease activity in human therapies. The small size and specificity of AcrIIA1 could be used in a CRISPR-Cas proteolysis-targeting chimera (PROTAC), providing a tool for developing safe and precise gene editing applications.


Asunto(s)
Bacteriófagos , Sistemas CRISPR-Cas , Humanos , Sistemas CRISPR-Cas/genética , Bacteriófagos/genética , Proteína 9 Asociada a CRISPR/metabolismo , Edición Génica , Lisogenia
11.
Nature ; 577(7789): 244-248, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31819262

RESUMEN

All viruses require strategies to inhibit or evade the immune pathways of cells that they infect. The viruses that infect bacteria, bacteriophages (phages), must avoid immune pathways that target nucleic acids, such as CRISPR-Cas and restriction-modification systems, to replicate efficiently1. Here we show that jumbo phage ΦKZ segregates its DNA from immunity nucleases of its host, Pseudomonas aeruginosa, by constructing a proteinaceous nucleus-like compartment. ΦKZ is resistant to many immunity mechanisms that target DNA in vivo, including two subtypes of CRISPR-Cas3, Cas9, Cas12a and the restriction enzymes HsdRMS and EcoRI. Cas proteins and restriction enzymes are unable to access the phage DNA throughout the infection, but engineering the relocalization of EcoRI inside the compartment enables targeting of the phage and protection of host cells. Moreover, ΦKZ is sensitive to Cas13a-a CRISPR-Cas enzyme that targets RNA-probably owing to phage mRNA localizing to the cytoplasm. Collectively, we propose that Pseudomonas jumbo phages evade a broad spectrum of DNA-targeting nucleases through the assembly of a protein barrier around their genome.


Asunto(s)
Proteínas Asociadas a CRISPR/metabolismo , Fagos Pseudomonas/genética , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/virología , Proteínas Virales/química , Sistemas CRISPR-Cas , ADN Viral/química , Genoma Viral , Fagos Pseudomonas/química
12.
Annu Rev Microbiol ; 74: 21-37, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32503371

RESUMEN

More than 50 protein families have been identified that inhibit CRISPR (clustered regularly interspaced short palindromic repeats)-Cas-mediated adaptive immune systems. Here, we analyze the available anti-CRISPR (Acr) structures and describe common themes and unique mechanisms of stoichiometric and enzymatic suppressors of CRISPR-Cas. Stoichiometric inhibitors often function as molecular decoys of protein-binding partners or nucleic acid targets, while enzymatic suppressors covalently modify Cas ribonucleoprotein complexes or degrade immune signaling molecules. We review mechanistic insights that have been revealed by structures of Acrs, discuss some of the trade-offs associated with each of these strategies, and highlight how Acrs are regulated and deployed in the race to overcome adaptive immunity.


Asunto(s)
Bacteriófagos/genética , Proteína 9 Asociada a CRISPR/inmunología , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/inmunología , Proteínas Virales/genética , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/fisiología , Evolución Molecular , Unión Proteica , Proteínas Virales/inmunología
13.
Nucleic Acids Res ; 51(10): 4995-5005, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37140042

RESUMEN

Bacteria use a diverse arsenal of anti-phage immune systems, including CRISPR-Cas and restriction enzymes. Recent advances in anti-phage system discovery and annotation tools have unearthed many unique systems, often encoded in horizontally transferred defense islands, which can be horizontally transferred. Here, we developed Hidden Markov Models (HMMs) for defense systems and queried microbial genomes on the NCBI database. Out of the 30 species with >200 completely sequenced genomes, our analysis found Pseudomonas aeruginosa exhibits the greatest diversity of anti-phage systems, as measured by Shannon entropy. Using network analysis to identify the common neighbors of anti-phage systems, we identified two core defense hotspot loci (cDHS1 and cDHS2). cDHS1 is up to 224 kb (median: 26 kb) with varied arrangements of more than 30 distinct immune systems across isolates, while cDHS2 has 24 distinct systems (median: 6 kb). Both cDHS regions are occupied in a majority of P. aeruginosa isolates. Most cDHS genes are of unknown function potentially representing new anti-phage systems, which we validated by identifying a novel anti-phage system (Shango) commonly encoded in cDHS1. Identifying core genes flanking immune islands could simplify immune system discovery and may represent popular landing spots for diverse MGEs carrying anti-phage systems.


Asunto(s)
Bacteriófagos , Pseudomonas aeruginosa , Bacteriófagos/genética , Sistemas CRISPR-Cas , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/virología
14.
Nat Chem Biol ; 18(12): 1417-1424, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36163386

RESUMEN

Anti-CRISPR (Acr) proteins are encoded by phages to inactivate CRISPR-Cas systems of bacteria and archaea and are used to enhance the CRISPR toolbox for genome editing. Here we report the structure and mechanism of AcrIF24, an Acr protein that inhibits the type I-F CRISPR-Cas system from Pseudomonas aeruginosa. AcrIF24 is a homodimer that associates with two copies of the surveillance complex (Csy) and prevents the hybridization between CRISPR RNA and target DNA. Furthermore, AcrIF24 functions as an anti-CRISPR-associated (Aca) protein to repress the transcription of the acrIF23-acrIF24 operon. Alone or in complex with Csy, AcrIF24 is capable of binding to the acrIF23-acrIF24 promoter DNA with nanomolar affinity. The structure of a Csy-AcrIF24-promoter DNA complex at 2.7 Å reveals the mechanism for transcriptional suppression. Our results reveal that AcrIF24 functions as an Acr-Aca fusion protein, and they extend understanding of the diverse mechanisms used by Acr proteins.


Asunto(s)
Bacteriófagos , Proteínas Asociadas a CRISPR , Proteínas Asociadas a CRISPR/genética , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo , Sistemas CRISPR-Cas , Bacteriófagos/genética , Bacteriófagos/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo
15.
Nucleic Acids Res ; 50(11): 6414-6422, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35670674

RESUMEN

CRISPR-Cas12a (Cpf1) is a bacterial RNA-guided nuclease that cuts double-stranded DNA (dsDNA) at sites specified by a CRISPR RNA (crRNA) guide. Additional activities have been ascribed to this enzyme in vitro: site-specific (cis) single-stranded DNA (ssDNA) cleavage and indiscriminate (trans) degradation of ssDNA, RNA, and dsDNA after activation by a complementary target. The ability of Cas12a to cleave nucleic acids indiscriminately has been harnessed for many applications, including diagnostics, but it remains unknown if it contributes to bacterial immunity. Here, we provide evidence that cleavage of ssDNA in cis or in trans by Cas12a is insufficient to impact immunity. Using LbCas12a expressed in either Pseudomonas aeruginosa or Escherichia coli, we observed that cleavage of dsDNA targets did not elicit cell death or dormancy, suggesting insignificant levels of collateral damage against host RNA or DNA. Canonical immunity against invasive dsDNA also had no impact on the replicative fitness of co-infecting dsDNA phage, ssDNA phage or plasmid in trans. Lastly, crRNAs complementary to invasive ssDNA did not provide protection, suggesting that ssDNA cleavage does not occur in vivo or is insignificant. Overall, these results suggest that CRISPR-Cas12a immunity predominantly occurs via canonical targeting of dsDNA, and that the other activities do not significantly impact infection outcomes.


Asunto(s)
Proteínas Asociadas a CRISPR , Sistemas CRISPR-Cas , ADN de Cadena Simple , Inmunidad , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo , ADN , ADN de Cadena Simple/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Inmunidad/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , ARN Guía de Kinetoplastida/genética
16.
J Bacteriol ; 205(6): e0002923, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37260386

RESUMEN

Most Pseudomonas aeruginosa strains produce bacteriocins derived from contractile or noncontractile phage tails known as R- and F-type pyocins, respectively. These bacteriocins possess strain-specific bactericidal activity against P. aeruginosa and likely increase evolutionary fitness through intraspecies competition. R-type pyocins have been studied extensively and show promise as alternatives to antibiotics. Although they have similar therapeutic potential, experimental studies on F-type pyocins are limited. Here, we provide a bioinformatic and experimental investigation of F-type pyocins. We introduce a systematic naming scheme for genes found in R- and F-type pyocin operons and identify 15 genes invariably found in strains producing F-type pyocins. Five proteins encoded at the 3' end of the F-type pyocin cluster are divergent in sequence and likely determine bactericidal specificity. We use sequence similarities among these proteins to define eleven distinct F-type pyocin groups, five of which had not been previously described. The five genes encoding the variable proteins associate in two modules that have clearly reassorted independently during the evolution of these operons. These proteins are considerably more diverse than the specificity-determining tail fibers of R-type pyocins, suggesting that F-type pyocins may have emerged earlier. Experimental studies on six F-type pyocin groups show that each displays a distinct spectrum of bactericidal activity. This activity is strongly influenced by the lipopolysaccharide O-antigen type, but other factors also play a role. F-type pyocins appear to kill as efficiently as R-type pyocins. These studies set the stage for the development of F-type pyocins as antibacterial therapeutics. IMPORTANCE Pseudomonas aeruginosa is an opportunistic pathogen that causes antibiotic-resistant infections with high mortality rates, particularly in immunocompromised individuals and cystic fibrosis patients. Due to the increasing frequency of multidrug-resistant P. aeruginosa infections, there is great need for the development of alternative therapeutics. In this study, we investigate one such potential therapeutic: F-type pyocins, which are bacteriocins naturally produced by P. aeruginosa that resemble noncontractile phage tails. We show that they are potent killers of P. aeruginosa and identify their probable bactericidal specificity determinants, which opens up the possibility of engineering them to precisely target strains of pathogenic bacteria. The resemblance of F-type pyocins to well-characterized phage tails will greatly facilitate their development into effective antibacterials.


Asunto(s)
Bacteriocinas , Bacteriófagos , Humanos , Piocinas/farmacología , Pseudomonas aeruginosa/metabolismo , Bacteriocinas/genética , Bacteriocinas/farmacología , Bacteriocinas/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Bacteriófagos/metabolismo
17.
Nat Methods ; 17(5): 471-479, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32203383

RESUMEN

Clustered, regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated (Cas) genes, a diverse family of prokaryotic adaptive immune systems, have emerged as a biotechnological tool and therapeutic. The discovery of protein inhibitors of CRISPR-Cas systems, called anti-CRISPR (Acr) proteins, enables the development of more controllable and precise CRISPR-Cas tools. Here we discuss applications of Acr proteins for post-translational control of CRISPR-Cas systems in prokaryotic and mammalian cells, organisms and ecosystems.


Asunto(s)
Proteínas Asociadas a CRISPR/antagonistas & inhibidores , Sistemas CRISPR-Cas , Inhibidores Enzimáticos/farmacología , Edición Génica , Regulación de la Expresión Génica , Animales , Humanos , Ingeniería de Proteínas
18.
Nat Methods ; 17(12): 1183-1190, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33077967

RESUMEN

CRISPR-Cas technologies have enabled programmable gene editing in eukaryotes and prokaryotes. However, the leading Cas9 and Cas12a enzymes are limited in their ability to make large deletions. Here, we used the processive nuclease Cas3, together with a minimal Type I-C Cascade-based system for targeted genome engineering in bacteria. DNA cleavage guided by a single CRISPR RNA generated large deletions (7-424 kilobases) in Pseudomonas aeruginosa with near-100% efficiency, while Cas9 yielded small deletions and point mutations. Cas3 generated bidirectional deletions originating from the programmed site, which was exploited to reduce the P. aeruginosa genome by 837 kb (13.5%). Large deletion boundaries were efficiently specified by a homology-directed repair template during editing with Cascade-Cas3, but not Cas9. A transferable 'all-in-one' vector was functional in Escherichia coli, Pseudomonas syringae and Klebsiella pneumoniae, and endogenous CRISPR-Cas use was enhanced with an 'anti-anti-CRISPR' strategy. P. aeruginosa Type I-C Cascade-Cas3 (PaeCas3c) facilitates rapid strain manipulation with applications in synthetic biology, genome minimization and the removal of large genomic regions.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , ADN Helicasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Edición Génica/métodos , Ingeniería Genética/métodos , Secuencia de Bases/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Escherichia coli/genética , Genoma Bacteriano/genética , Klebsiella pneumoniae/genética , Pseudomonas aeruginosa/genética , Pseudomonas syringae/genética , Eliminación de Secuencia/genética
19.
Nucleic Acids Res ; 49(4): 2114-2125, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33544853

RESUMEN

Bacteria deploy multiple defenses to prevent mobile genetic element (MGEs) invasion. CRISPR-Cas immune systems use RNA-guided nucleases to target MGEs, which counter with anti-CRISPR (Acr) proteins. Our understanding of the biology and co-evolutionary dynamics of the common Type I-C CRISPR-Cas subtype has lagged because it lacks an in vivo phage-host model system. Here, we show the anti-phage function of a Pseudomonas aeruginosa Type I-C CRISPR-Cas system encoded on a conjugative pKLC102 island, and its Acr-mediated inhibition by distinct MGEs. Seven genes with anti-Type I-C function (acrIC genes) were identified, many with highly acidic amino acid content, including previously described DNA mimic AcrIF2. Four of the acr genes were broad spectrum, also inhibiting I-E or I-F P. aeruginosa CRISPR-Cas subtypes. Dual inhibition comes at a cost, however, as simultaneous expression of Type I-C and I-F systems renders phages expressing the dual inhibitor AcrIF2 more sensitive to targeting. Mutagenesis of numerous acidic residues in AcrIF2 did not impair anti-I-C or anti-I-F function per se but did exacerbate inhibition defects during competition, suggesting that excess negative charge may buffer DNA mimics against competition. Like AcrIF2, five of the Acr proteins block Cascade from binding DNA, while two function downstream, likely preventing Cas3 recruitment or activity. One such inhibitor, AcrIC3, is found in an 'anti-Cas3' cluster within conjugative elements, encoded alongside bona fide Cas3 inhibitors AcrIF3 and AcrIE1. Our findings demonstrate an active battle between an MGE-encoded CRISPR-Cas system and its diverse MGE targets.


Asunto(s)
Sistemas CRISPR-Cas , Secuencias Repetitivas Esparcidas , Pseudomonas aeruginosa/genética , Bacteriófagos/genética , Bacteriófagos/metabolismo , Proteínas Asociadas a CRISPR/metabolismo , División del ADN , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/virología , Proteínas Virales/metabolismo
20.
J Bacteriol ; 204(5): e0010522, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35389256

RESUMEN

Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated (Cas) systems are prokaryotic adaptive immune systems that have been well characterized biochemically, but in vivo spatiotemporal regulation and cell biology remain largely unaddressed. Here, we used fluorescent fusion proteins introduced at the chromosomal CRISPR-Cas locus to study the localization of the type I-F CRISPR-Cas system in Pseudomonas aeruginosa. When lacking a target in the cell, the Cascade complex is broadly nucleoid bound, while Cas3 is diffuse in the cytoplasm. When targeted to an integrated prophage, however, the CRISPR RNA (crRNA)-guided type I-F Cascade complex and a majority of Cas3 molecules in the cell are recruited to a single focus. Nucleoid association of the Csy proteins that form the Cascade complex is crRNA dependent and specifically inhibited by the expression of anti-CRISPR AcrIF2, which blocks protospacer adjacent motif (PAM) binding. The Cas9 nuclease is also nucleoid localized, only when single guide RNA (sgRNA) bound, which is abolished by the PAM-binding inhibitor AcrIIA4. Our findings reveal PAM-dependent nucleoid surveillance and spatiotemporal regulation in type I CRISPR-Cas that separates the nuclease-helicase Cas3 from the crRNA-guided surveillance complex. IMPORTANCE CRISPR-Cas systems, the prokaryotic adaptive immune systems, are largely understood using structural biology, biochemistry, and genetics. How CRISPR-Cas effectors are organized within cells is currently not well understood. By investigating the cell biology of the type I-F CRISPR-Cas system, we show that the surveillance complex, which "patrols" the cell to find targets, is largely nucleoid bound, while Cas3 nuclease is cytoplasmic. Nucleoid localization is also conserved for class 2 CRISPR-Cas single protein effector Cas9. Our observation of differential localization of the surveillance complex and Cas3 reveals a new layer of posttranslational spatiotemporal regulation to prevent autoimmunity.


Asunto(s)
Proteínas Asociadas a CRISPR , Bacterias/metabolismo , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , ADN Helicasas/genética , Endonucleasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA