Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.027
Filtrar
Más filtros

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 ; 184(23): 5728-5739.e16, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34644530

RESUMEN

The cyclic pyrimidines 3',5'-cyclic cytidine monophosphate (cCMP) and 3',5'-cyclic uridine monophosphate (cUMP) have been reported in multiple organisms and cell types. As opposed to the cyclic nucleotides 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP), which are second messenger molecules with well-established regulatory roles across all domains of life, the biological role of cyclic pyrimidines has remained unclear. Here we report that cCMP and cUMP are second messengers functioning in bacterial immunity against viruses. We discovered a family of bacterial pyrimidine cyclase enzymes that specifically synthesize cCMP and cUMP following phage infection and demonstrate that these molecules activate immune effectors that execute an antiviral response. A crystal structure of a uridylate cyclase enzyme from this family explains the molecular mechanism of selectivity for pyrimidines as cyclization substrates. Defense systems encoding pyrimidine cyclases, denoted here Pycsar (pyrimidine cyclase system for antiphage resistance), are widespread in prokaryotes. Our results assign clear biological function to cCMP and cUMP as immunity signaling molecules in bacteria.


Asunto(s)
Bacterias/inmunología , Bacterias/virología , Bacteriófagos/fisiología , CMP Cíclico/metabolismo , Nucleótidos Cíclicos/metabolismo , Uridina Monofosfato/metabolismo , Secuencia de Aminoácidos , Bacterias/genética , Burkholderia/enzimología , CMP Cíclico/química , Ciclización , Escherichia coli/enzimología , Modelos Moleculares , Mutación/genética , Nucleótidos Cíclicos/química , Liasas de Fósforo-Oxígeno/química , Liasas de Fósforo-Oxígeno/metabolismo , Pirimidinas/metabolismo , Uridina Monofosfato/química
3.
Cell ; 183(6): 1551-1561.e12, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33157039

RESUMEN

Retrons are bacterial genetic elements comprised of a reverse transcriptase (RT) and a non-coding RNA (ncRNA). The RT uses the ncRNA as template, generating a chimeric RNA/DNA molecule in which the RNA and DNA components are covalently linked. Although retrons were discovered three decades ago, their function remained unknown. We report that retrons function as anti-phage defense systems. The defensive unit is composed of three components: the RT, the ncRNA, and an effector protein. We examined multiple retron systems and show that they confer defense against a broad range of phages via abortive infection. Focusing on retron Ec48, we show evidence that it "guards" RecBCD, a complex with central anti-phage functions in bacteria. Inhibition of RecBCD by phage proteins activates the retron, leading to abortive infection and cell death. Thus, the Ec48 retron forms a second line of defense that is triggered if the first lines of defense have collapsed.


Asunto(s)
Bacterias/genética , Bacterias/inmunología , Bacteriófagos/fisiología , ARN no Traducido/genética , ADN Polimerasa Dirigida por ARN/genética , Bacterias/virología , Islas de CpG/genética , ADN/metabolismo , Escherichia coli/genética , Escherichia coli/inmunología , Escherichia coli/virología , Proteínas de Escherichia coli/metabolismo , Filogenia
4.
Cell ; 172(6): 1239-1259, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29522745

RESUMEN

In bacteria and archaea, clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated (Cas) proteins constitute an adaptive immune system against phages and other foreign genetic elements. Here, we review the biology of the diverse CRISPR-Cas systems and the major progress achieved in recent years in understanding the underlying mechanisms of the three stages of CRISPR-Cas immunity: adaptation, crRNA biogenesis, and interference. The ecology and regulation of CRISPR-Cas in the context of phage infection, the roles of these systems beyond immunity, and the open questions that propel the field forward are also discussed.


Asunto(s)
Bacterias/genética , Bacteriófagos/genética , Biología/tendencias , Sistemas CRISPR-Cas , Inmunidad Adaptativa/genética , Bacterias/virología , Bacteriófagos/fisiología , Regulación Bacteriana de la Expresión Génica , Modelos Genéticos , Transducción de Señal/genética
5.
Cell ; 172(6): 1260-1270, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29522746

RESUMEN

Bacteriophages, discovered about a century ago, have been pivotal as models for understanding the fundamental principles of molecular biology. While interest in phage biology declined after the phage "golden era," key recent developments, including advances in phage genomics, microscopy, and the discovery of the CRISPR-Cas anti-phage defense system, have sparked a renaissance in phage research in the past decade. This review highlights recently discovered unexpected complexities in phage biology, describes a new arsenal of phage genes that help them overcome bacterial defenses, and discusses advances toward documentation of the phage biodiversity on a global scale.


Asunto(s)
Bacteriófagos/genética , Biología/tendencias , Genoma Viral/genética , Genómica/tendencias , Biología Molecular/tendencias , Bacterias/genética , Bacterias/virología , Bacteriófagos/fisiología , Sistemas CRISPR-Cas , Variación Genética , Genómica/métodos , Lisogenia/genética , Modelos Genéticos
6.
Mol Cell ; 84(18): 3378-3380, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39303679

RESUMEN

In two recent studies in Nature, Hör et al.1 and Chambers et al.2 report that ubiquitin-like conjugation in bacteria antagonizes phage replication.


Asunto(s)
Ubiquitinación , Ubiquitina/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Bacteriófagos/metabolismo , Bacteriófagos/fisiología , Replicación Viral , Bacterias/metabolismo , Bacterias/genética , Bacterias/virología
7.
Nat Rev Genet ; 25(11): 785-802, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38886215

RESUMEN

Dense packaging of genomic DNA is crucial for organismal survival, as DNA length always far exceeds the dimensions of the cells that contain it. Organisms, therefore, use sophisticated machineries to package their genomes. These systems range across kingdoms from a single ultra-powerful rotary motor that spools the DNA into a bacteriophage head, to hundreds of thousands of relatively weak molecular motors that coordinate the compaction of mitotic chromosomes in eukaryotic cells. Recent technological advances, such as DNA proximity-based sequencing approaches, polymer modelling and in vitro reconstitution of DNA loop extrusion, have shed light on the biological mechanisms driving DNA organization in different systems. Here, we discuss DNA packaging in bacteriophage, bacteria and eukaryotic cells, which, despite their extreme variation in size, structure and genomic content, all rely on the action of molecular motors to package their genomes.


Asunto(s)
Bacteriófagos , Empaquetamiento del ADN , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/metabolismo , Humanos , Proteínas Motoras Moleculares/metabolismo , Proteínas Motoras Moleculares/genética , Cromosomas Humanos/metabolismo , Cromosomas Humanos/genética , ADN Viral/genética , ADN Viral/metabolismo
8.
Nature ; 631(8022): 850-856, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39020165

RESUMEN

Several immune pathways in humans conjugate ubiquitin-like proteins to virus and host molecules as a means of antiviral defence1-5. Here we studied an antiphage defence system in bacteria, comprising a ubiquitin-like protein, ubiquitin-conjugating enzymes E1 and E2, and a deubiquitinase. We show that during phage infection, this system specifically conjugates the ubiquitin-like protein to the phage central tail fibre, a protein at the tip of the tail that is essential for tail assembly as well as for recognition of the target host receptor. Following infection, cells encoding this defence system release a mixture of partially assembled, tailless phage particles and fully assembled phages in which the central tail fibre is obstructed by the covalently attached ubiquitin-like protein. These phages show severely impaired infectivity, explaining how the defence system protects the bacterial population from the spread of phage infection. Our findings demonstrate that conjugation of ubiquitin-like proteins is an antiviral strategy conserved across the tree of life.


Asunto(s)
Proteínas Bacterianas , Bacteriófagos , Enzimas Desubicuitinizantes , Escherichia coli , Enzimas Ubiquitina-Conjugadoras , Ubiquitinas , Ensamble de Virus , Bacteriófagos/química , Bacteriófagos/metabolismo , Bacteriófagos/patogenicidad , Bacteriófagos/fisiología , Enzimas Desubicuitinizantes/metabolismo , Escherichia coli/enzimología , Escherichia coli/metabolismo , Escherichia coli/virología , Enzimas Activadoras de Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinas/metabolismo , Proteínas de la Cola de los Virus/metabolismo , Proteínas de la Cola de los Virus/química , Proteínas Bacterianas/metabolismo , Evolución Molecular , Secuencia Conservada
9.
Nature ; 632(8026): 877-884, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38987595

RESUMEN

Microbiome research is now demonstrating a growing number of bacterial strains and genes that affect our health1. Although CRISPR-derived tools have shown great success in editing disease-driving genes in human cells2, we currently lack the tools to achieve comparable success for bacterial targets in situ. Here we engineer a phage-derived particle to deliver a base editor and modify Escherichia coli colonizing the mouse gut. Editing of a ß-lactamase gene in a model E. coli strain resulted in a median editing efficiency of 93% of the target bacterial population with a single dose. Edited bacteria were stably maintained in the mouse gut for at least 42 days following treatment. This was achieved using a non-replicative DNA vector, preventing maintenance and dissemination of the payload. We then leveraged this approach to edit several genes of therapeutic relevance in E. coli and Klebsiella pneumoniae strains in vitro and demonstrate in situ editing of a gene involved in the production of curli in a pathogenic E. coli strain. Our work demonstrates the feasibility of modifying bacteria directly in the gut, offering a new avenue to investigate the function of bacterial genes and opening the door to the design of new microbiome-targeted therapies.


Asunto(s)
Sistemas CRISPR-Cas , Escherichia coli , Microbioma Gastrointestinal , Tracto Gastrointestinal , Edición Génica , Animales , Femenino , Ratones , Bacteriófagos/genética , Bacteriófagos/fisiología , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Sistemas CRISPR-Cas/genética , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/patogenicidad , Escherichia coli/fisiología , Escherichia coli/virología , Microbioma Gastrointestinal/genética , Tracto Gastrointestinal/microbiología , Edición Génica/métodos , Genes Bacterianos/genética , Vectores Genéticos/genética , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/virología , Factores de Tiempo
10.
Cell ; 157(1): 142-50, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24679532

RESUMEN

The virome contains the most abundant and fastest mutating genetic elements on Earth. The mammalian virome is constituted of viruses that infect host cells, virus-derived elements in our chromosomes, and viruses that infect the broad array of other types of organisms that inhabit us. Virome interactions with the host cannot be encompassed by a monotheistic view of viruses as pathogens. Instead, the genetic and transcriptional identity of mammals is defined in part by our coevolved virome, a concept with profound implications for understanding health and disease.


Asunto(s)
Bacteriófagos/fisiología , Mamíferos/virología , Microbiota , Virus/genética , Animales , Bacterias/virología , Bacteriófagos/clasificación , Bacteriófagos/genética , Interacciones Huésped-Patógeno , Humanos , Mamíferos/genética , Mamíferos/inmunología , Virus/clasificación , Virus/metabolismo
11.
Nature ; 617(7961): 581-591, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37165188

RESUMEN

The spatiotemporal structure of the human microbiome1,2, proteome3 and metabolome4,5 reflects and determines regional intestinal physiology and may have implications for disease6. Yet, little is known about the distribution of microorganisms, their environment and their biochemical activity in the gut because of reliance on stool samples and limited access to only some regions of the gut using endoscopy in fasting or sedated individuals7. To address these deficiencies, we developed an ingestible device that collects samples from multiple regions of the human intestinal tract during normal digestion. Collection of 240 intestinal samples from 15 healthy individuals using the device and subsequent multi-omics analyses identified significant differences between bacteria, phages, host proteins and metabolites in the intestines versus stool. Certain microbial taxa were differentially enriched and prophage induction was more prevalent in the intestines than in stool. The host proteome and bile acid profiles varied along the intestines and were highly distinct from those of stool. Correlations between gradients in bile acid concentrations and microbial abundance predicted species that altered the bile acid pool through deconjugation. Furthermore, microbially conjugated bile acid concentrations exhibited amino acid-dependent trends that were not apparent in stool. Overall, non-invasive, longitudinal profiling of microorganisms, proteins and bile acids along the intestinal tract under physiological conditions can help elucidate the roles of the gut microbiome and metabolome in human physiology and disease.


Asunto(s)
Ácidos y Sales Biliares , Microbioma Gastrointestinal , Intestinos , Metaboloma , Proteoma , Humanos , Ácidos y Sales Biliares/metabolismo , Microbioma Gastrointestinal/fisiología , Proteoma/metabolismo , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacteriófagos/aislamiento & purificación , Bacteriófagos/fisiología , Heces/química , Heces/microbiología , Heces/virología , Intestinos/química , Intestinos/metabolismo , Intestinos/microbiología , Intestinos/fisiología , Intestinos/virología , Digestión/fisiología
12.
Nature ; 608(7922): 429-435, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35922510

RESUMEN

Bacteria encode myriad defences that target the genomes of infecting bacteriophage, including restriction-modification and CRISPR-Cas systems1. In response, one family of large bacteriophages uses a nucleus-like compartment to protect its replicating genomes by excluding host defence factors2-4. However, the principal composition and structure of this compartment remain unknown. Here we find that the bacteriophage nuclear shell assembles primarily from one protein, which we name chimallin (ChmA). Combining cryo-electron tomography of nuclear shells in bacteriophage-infected cells and cryo-electron microscopy of a minimal chimallin compartment in vitro, we show that chimallin self-assembles as a flexible sheet into closed micrometre-scale compartments. The architecture and assembly dynamics of the chimallin shell suggest mechanisms for its nucleation and growth, and its role as a scaffold for phage-encoded factors mediating macromolecular transport, cytoskeletal interactions, and viral maturation.


Asunto(s)
Bacterias , Bacteriófagos , Compartimento Celular , Proteínas Virales , Ensamble de Virus , Bacterias/citología , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/virología , Bacteriófagos/química , Bacteriófagos/inmunología , Bacteriófagos/fisiología , Bacteriófagos/ultraestructura , Microscopía por Crioelectrón , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Virales/ultraestructura
13.
Nature ; 605(7910): 522-526, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35395152

RESUMEN

The cyclic oligonucleotide-based antiphage signalling system (CBASS) and the pyrimidine cyclase system for antiphage resistance (Pycsar) are antiphage defence systems in diverse bacteria that use cyclic nucleotide signals to induce cell death and prevent viral propagation1,2. Phages use several strategies to defeat host CRISPR and restriction-modification systems3-10, but no mechanisms are known to evade CBASS and Pycsar immunity. Here we show that phages encode anti-CBASS (Acb) and anti-Pycsar (Apyc) proteins that counteract defence by specifically degrading cyclic nucleotide signals that activate host immunity. Using a biochemical screen of 57 phages in Escherichia coli and Bacillus subtilis, we discover Acb1 from phage T4 and Apyc1 from phage SBSphiJ as founding members of distinct families of immune evasion proteins. Crystal structures of Acb1 in complex with 3'3'-cyclic GMP-AMP define a mechanism of metal-independent hydrolysis 3' of adenosine bases, enabling broad recognition and degradation of cyclic dinucleotide and trinucleotide CBASS signals. Structures of Apyc1 reveal a metal-dependent cyclic NMP phosphodiesterase that uses relaxed specificity to target Pycsar cyclic pyrimidine mononucleotide signals. We show that Acb1 and Apyc1 block downstream effector activation and protect from CBASS and Pycsar defence in vivo. Active Acb1 and Apyc1 enzymes are conserved in phylogenetically diverse phages, demonstrating that cleavage of host cyclic nucleotide signals is a key strategy of immune evasion in phage biology.


Asunto(s)
Bacteriófagos , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Bacteriófago T4/metabolismo , Bacteriófagos/fisiología , Sistemas CRISPR-Cas/genética , Endonucleasas/metabolismo , Escherichia coli/metabolismo , Nucleótidos Cíclicos/metabolismo , Oligonucleótidos , Pirimidinas/metabolismo
14.
PLoS Biol ; 22(4): e3002346, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38648198

RESUMEN

Where there are bacteria, there will be bacteriophages. These viruses are known to be important players in shaping the wider microbial community in which they are embedded, with potential implications for human health. On the other hand, bacteria possess a range of distinct immune mechanisms that provide protection against bacteriophages, including the mutation or complete loss of the phage receptor, and CRISPR-Cas adaptive immunity. While our previous work showed how a microbial community may impact phage resistance evolution, little is known about the inverse, namely how interactions between phages and these different phage resistance mechanisms affect the wider microbial community in which they are embedded. Here, we conducted a 10-day, fully factorial evolution experiment to examine how phage impact the structure and dynamics of an artificial four-species bacterial community that includes either Pseudomonas aeruginosa wild-type or an isogenic mutant unable to evolve phage resistance through CRISPR-Cas. Additionally, we used mathematical modelling to explore the ecological interactions underlying full community behaviour, as well as to identify general principles governing the impacts of phage on community dynamics. Our results show that the microbial community structure is drastically altered by the addition of phage, with Acinetobacter baumannii becoming the dominant species and P. aeruginosa being driven nearly extinct, whereas P. aeruginosa outcompetes the other species in the absence of phage. Moreover, we find that a P. aeruginosa strain with the ability to evolve CRISPR-based resistance generally does better when in the presence of A. baumannii, but that this benefit is largely lost over time as phage is driven extinct. Finally, we show that pairwise data alone is insufficient when modelling our microbial community, both with and without phage, highlighting the importance of higher order interactions in governing multispecies dynamics in complex communities. Combined, our data clearly illustrate how phage targeting a dominant species allows for the competitive release of the strongest competitor while also contributing to community diversity maintenance and potentially preventing the reinvasion of the target species, and underline the importance of mapping community composition before therapeutically applying phage.


Asunto(s)
Bacteriófagos , Sistemas CRISPR-Cas , Microbiota , Pseudomonas aeruginosa , Bacteriófagos/fisiología , Bacteriófagos/genética , Pseudomonas aeruginosa/virología , Acinetobacter baumannii/virología , Mutación , Bacterias/virología , Bacterias/genética
15.
PLoS Biol ; 22(4): e3002566, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38652717

RESUMEN

Phage therapy is a therapeutic approach to treat multidrug-resistant (MDR) infections that employs lytic bacteriophages (phages) to eliminate bacteria. Despite the abundant evidence for its success as an antimicrobial in Eastern Europe, there is scarce data regarding its effects on the human host. Here, we aimed to understand how lytic phages interact with cells of the airway epithelium, the tissue site that is colonized by bacterial biofilms in numerous chronic respiratory disorders. Using a panel of Pseudomonas aeruginosa phages and human airway epithelial cells (AECs) derived from a person with cystic fibrosis (CF), we determined that interactions between phages and epithelial cells depend on specific phage properties as well as physiochemical features of the microenvironment. Although poor at internalizing phages, the airway epithelium responds to phage exposure by changing its transcriptional profile and secreting antiviral and proinflammatory cytokines that correlate with specific phage families. Overall, our findings indicate that mammalian responses to phages are heterogenous and could potentially alter the way that respiratory local defenses aid in bacterial clearance during phage therapy. Thus, besides phage receptor specificity in a particular bacterial isolate, the criteria to select lytic phages for therapy should be expanded to include mammalian cell responses.


Asunto(s)
Fibrosis Quística , Citocinas , Células Epiteliales , Pseudomonas aeruginosa , Humanos , Pseudomonas aeruginosa/virología , Células Epiteliales/virología , Células Epiteliales/metabolismo , Células Epiteliales/inmunología , Citocinas/metabolismo , Fibrosis Quística/terapia , Fibrosis Quística/inmunología , Fibrosis Quística/metabolismo , Terapia de Fagos , Bacteriófagos/fisiología , Bacteriófagos/genética , Mucosa Respiratoria/virología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/inmunología , Infecciones por Pseudomonas/terapia , Infecciones por Pseudomonas/inmunología , Fagos Pseudomonas/metabolismo , Biopelículas
16.
Cell ; 149(7): 1488-99, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22726436

RESUMEN

Tubulins are essential for the reproduction of many eukaryotic viruses, but historically, bacteriophage were assumed not to require a cytoskeleton. Here, we identify a tubulin-like protein, PhuZ, from bacteriophage 201φ2-1 and show that it forms filaments in vivo and in vitro. The PhuZ structure has a conserved tubulin fold, with an unusual, extended C terminus that we demonstrate to be critical for polymerization in vitro and in vivo. Longitudinal packing in the crystal lattice mimics packing observed by EM of in-vitro-formed filaments, indicating how interactions between the C terminus and the following monomer drive polymerization. PhuZ forms a filamentous array that is required for positioning phage DNA within the bacterial cell. Correct positioning to the cell center and optimal phage reproduction only occur when the PhuZ filament is dynamic. Thus, we show that PhuZ assembles a spindle-like array that functions analogously to the microtubule-based spindles of eukaryotes.


Asunto(s)
Bacteriófagos/fisiología , Pseudomonas/virología , Tubulina (Proteína)/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Citoesqueleto/metabolismo , ADN Viral/metabolismo , Guanosina Difosfato/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Pseudomonas/citología , Alineación de Secuencia , Tubulina (Proteína)/química , Tubulina (Proteína)/genética , Proteínas Virales/química , Proteínas Virales/genética
17.
Cell ; 150(4): 792-802, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22901809

RESUMEN

The DNA uptake competence (Com) system of the intracellular bacterial pathogen Listeria monocytogenes is considered nonfunctional. There are no known conditions for DNA transformation, and the Com master activator gene, comK, is interrupted by a temperate prophage. Here, we show that the L. monocytogenes Com system is required during infection to promote bacterial escape from macrophage phagosomes in a manner that is independent of DNA uptake. Further, we find that regulation of the Com system relies on the formation of a functional comK gene via prophage excision. Prophage excision is specifically induced during intracellular growth, primarily within phagosomes, yet, in contrast to classic prophage induction, progeny virions are not produced. This study presents the characterization of an active prophage that serves as a genetic switch to modulate the virulence of its bacterial host in the course of infection.


Asunto(s)
Proteínas Bacterianas/genética , Bacteriófagos/fisiología , Listeria/patogenicidad , Listeria/virología , Macrófagos/inmunología , Macrófagos/microbiología , Fagosomas/microbiología , Activación Viral , Animales , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Células Cultivadas , Femenino , Listeria/genética , Listeria/inmunología , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Alineación de Secuencia
18.
Nature ; 590(7847): 624-629, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33461211

RESUMEN

In the type III CRISPR-Cas immune response of prokaryotes, infection triggers the production of cyclic oligoadenylates that bind and activate proteins that contain a CARF domain1,2. Many type III loci are associated with proteins in which the CRISPR-associated Rossman fold (CARF) domain is fused to a restriction  endonuclease-like domain3,4. However, with the exception of the well-characterized Csm6 and Csx1 ribonucleases5,6, whether and how these inducible effectors provide defence is not known. Here we investigated a type III CRISPR accessory protein, which we name cyclic-oligoadenylate-activated single-stranded ribonuclease and single-stranded deoxyribonuclease 1 (Card1). Card1 forms a symmetrical dimer that has a large central cavity between its CRISPR-associated Rossmann fold and restriction endonuclease domains that binds cyclic tetra-adenylate. The binding of ligand results in a conformational change comprising the rotation of individual monomers relative to each other to form a more compact dimeric scaffold, in which a manganese cation coordinates the catalytic residues and activates the cleavage of single-stranded-but not double-stranded-nucleic acids (both DNA and RNA). In vivo, activation of Card1 induces dormancy of the infected hosts to provide immunity against phage infection and plasmids. Our results highlight the diversity of strategies used in CRISPR systems to provide immunity.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Sistemas CRISPR-Cas/inmunología , ADN de Cadena Simple/metabolismo , Desoxirribonucleasas/metabolismo , Endorribonucleasas/metabolismo , Oligorribonucleótidos/metabolismo , ARN/metabolismo , Staphylococcus/enzimología , Staphylococcus/inmunología , Nucleótidos de Adenina/inmunología , Adenosina Trifosfato/metabolismo , Bacteriófagos/inmunología , Bacteriófagos/fisiología , Biocatálisis , Dominio Catalítico , Desoxirribonucleasas/química , Desoxirribonucleasas/genética , Endorribonucleasas/química , Endorribonucleasas/genética , Activación Enzimática , Ligandos , Manganeso/química , Manganeso/metabolismo , Modelos Moleculares , Oligorribonucleótidos/inmunología , Plásmidos/genética , Plásmidos/metabolismo , Multimerización de Proteína , Rotación , Staphylococcus/crecimiento & desarrollo , Staphylococcus/virología , Especificidad por Sustrato
19.
Nature ; 592(7855): 611-615, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33828299

RESUMEN

Horizontal gene transfer and mutation are the two major drivers of microbial evolution that enable bacteria to adapt to fluctuating environmental stressors1. Clustered, regularly interspaced, short palindromic repeats (CRISPR) systems use RNA-guided nucleases to direct sequence-specific destruction of the genomes of mobile genetic elements that mediate horizontal gene transfer, such as conjugative plasmids2 and bacteriophages3, thus limiting the extent to which bacteria can evolve by this mechanism. A subset of CRISPR systems also exhibit non-specific degradation of DNA4,5; however, whether and how this feature affects the host has not yet been examined. Here we show that the non-specific DNase activity of the staphylococcal type III-A CRISPR-Cas system increases mutations in the host and accelerates the generation of antibiotic resistance in Staphylococcus aureus and Staphylococcus epidermidis. These mutations require the induction of the SOS response to DNA damage and display a distinct pattern. Our results demonstrate that by differentially affecting both mechanisms that generate genetic diversity, type III-A CRISPR systems can modulate the evolution of the bacterial host.


Asunto(s)
Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/inmunología , Mutagénesis , Mutación , Staphylococcus/genética , Antibacterianos/farmacología , Bacteriófagos/clasificación , Bacteriófagos/fisiología , Proteínas Asociadas a CRISPR/metabolismo , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Desoxirribonucleasas/metabolismo , Farmacorresistencia Microbiana/efectos de los fármacos , Respuesta SOS en Genética/efectos de los fármacos , Staphylococcus/efectos de los fármacos , Staphylococcus/inmunología , Staphylococcus/virología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/virología , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/virología , Factores de Tiempo
20.
Mol Cell ; 75(5): 1020-1030.e4, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31350119

RESUMEN

Phage-inducible chromosomal islands (PICIs) represent a novel and universal class of mobile genetic elements, which have broad impact on bacterial virulence. In spite of their relevance, how the Gram-negative PICIs hijack the phage machinery for their own specific packaging and how they block phage reproduction remains to be determined. Using genetic and structural analyses, we solve the mystery here by showing that the Gram-negative PICIs encode a protein that simultaneously performs these processes. This protein, which we have named Rpp (for redirecting phage packaging), interacts with the phage terminase small subunit, forming a heterocomplex. This complex is unable to recognize the phage DNA, blocking phage packaging, but specifically binds to the PICI genome, promoting PICI packaging. Our studies reveal the mechanism of action that allows PICI dissemination in nature, introducing a new paradigm in the understanding of the biology of pathogenicity islands and therefore of bacterial pathogen evolution.


Asunto(s)
Bacteriófagos/fisiología , ADN Viral/metabolismo , Escherichia coli/virología , Islas Genómicas , Ensamble de Virus/fisiología , ADN Viral/genética , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA