Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.764
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 175(5): 1380-1392.e14, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30343895

RESUMEN

ADP-ribosylation of proteins can profoundly impact their function and serves as an effective mechanism by which bacterial toxins impair eukaryotic cell processes. Here, we report the discovery that bacteria also employ ADP-ribosylating toxins against each other during interspecies competition. We demonstrate that one such toxin from Serratia proteamaculans interrupts the division of competing cells by modifying the essential bacterial tubulin-like protein, FtsZ, adjacent to its protomer interface, blocking its capacity to polymerize. The structure of the toxin in complex with its immunity determinant revealed two distinct modes of inhibition: active site occlusion and enzymatic removal of ADP-ribose modifications. We show that each is sufficient to support toxin immunity; however, the latter additionally provides unprecedented broad protection against non-cognate ADP-ribosylating effectors. Our findings reveal how an interbacterial arms race has produced a unique solution for safeguarding the integrity of bacterial cell division machinery against inactivating post-translational modifications.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Proteínas del Citoesqueleto/metabolismo , N-Glicosil Hidrolasas/metabolismo , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/genética , ADP-Ribosilación , Adenosina Difosfato/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Dominio Catalítico , Proteínas del Citoesqueleto/antagonistas & inhibidores , Escherichia coli/crecimiento & desarrollo , Escherichia coli/inmunología , Escherichia coli/metabolismo , Humanos , Mutagénesis Sitio-Dirigida , N-Glicosil Hidrolasas/química , N-Glicosil Hidrolasas/genética , Estructura Terciaria de Proteína , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Alineación de Secuencia , Serratia/metabolismo , Imagen de Lapso de Tiempo
2.
Mol Cell ; 83(2): 165-166, 2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36669478

RESUMEN

Zhang et al.1 reveal a previously unknown route to toxin activation whereby bacteriophage capsid proteins bind the antitoxin domain of the CapRel fused toxin-antitoxin system, triggering translational inhibition via pyrophosporylation of tRNAs and culminating in abortive infection-mediated phage resistance.


Asunto(s)
Antitoxinas , Toxinas Bacterianas , Bacteriófagos , Sistemas Toxina-Antitoxina , Bacteriófagos/metabolismo , Proteínas de la Cápside/genética , Cápside/metabolismo , Bacterias/metabolismo , Antitoxinas/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo
3.
Nature ; 629(8010): 165-173, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38632398

RESUMEN

Streptomyces are a genus of ubiquitous soil bacteria from which the majority of clinically utilized antibiotics derive1. The production of these antibacterial molecules reflects the relentless competition Streptomyces engage in with other bacteria, including other Streptomyces species1,2. Here we show that in addition to small-molecule antibiotics, Streptomyces produce and secrete antibacterial protein complexes that feature a large, degenerate repeat-containing polymorphic toxin protein. A cryo-electron microscopy structure of these particles reveals an extended stalk topped by a ringed crown comprising the toxin repeats scaffolding five lectin-tipped spokes, which led us to name them umbrella particles. Streptomyces coelicolor encodes three umbrella particles with distinct toxin and lectin composition. Notably, supernatant containing these toxins specifically and potently inhibits the growth of select Streptomyces species from among a diverse collection of bacteria screened. For one target, Streptomyces griseus, inhibition relies on a single toxin and that intoxication manifests as rapid cessation of vegetative hyphal growth. Our data show that Streptomyces umbrella particles mediate competition among vegetative mycelia of related species, a function distinct from small-molecule antibiotics, which are produced at the onset of reproductive growth and act broadly3,4. Sequence analyses suggest that this role of umbrella particles extends beyond Streptomyces, as we identified umbrella loci in nearly 1,000 species across Actinobacteria.


Asunto(s)
Antibiosis , Proteínas Bacterianas , Toxinas Bacterianas , Streptomyces , Antibacterianos/biosíntesis , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Antibiosis/efectos de los fármacos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Proteínas Bacterianas/ultraestructura , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacología , Microscopía por Crioelectrón , Lectinas/química , Lectinas/genética , Lectinas/metabolismo , Lectinas/ultraestructura , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Streptomyces/química , Streptomyces/efectos de los fármacos , Streptomyces/genética , Streptomyces/crecimiento & desarrollo , Streptomyces coelicolor/química , Streptomyces coelicolor/genética , Streptomyces coelicolor/metabolismo , Streptomyces griseus/efectos de los fármacos , Streptomyces griseus/genética , Streptomyces griseus/crecimiento & desarrollo , Streptomyces griseus/metabolismo
4.
Cell ; 159(6): 1290-9, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25480294

RESUMEN

Salmonella Typhi is an exclusive human pathogen that causes typhoid fever. Typhoid toxin is a S. Typhi virulence factor that can reproduce most of the typhoid fever symptoms in experimental animals. Toxicity depends on toxin binding to terminally sialylated glycans on surface glycoproteins. Human glycans are unusual because of the lack of CMAH, which in other mammals converts N-acetylneuraminic acid (Neu5Ac) to N-glycolylneuraminic acid (Neu5Gc). Here, we report that typhoid toxin binds to and is toxic toward cells expressing glycans terminated in Neu5Ac (expressed by humans) over glycans terminated in Neu5Gc (expressed by other mammals). Mice constitutively expressing CMAH thus displaying Neu5Gc in all tissues are resistant to typhoid toxin. The atomic structure of typhoid toxin bound to Neu5Ac reveals the structural bases for its binding specificity. These findings provide insight into the molecular bases for Salmonella Typhi's host specificity and may help the development of therapies for typhoid fever.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Glicoproteínas de Membrana/química , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/metabolismo , Salmonella typhi/química , Animales , Toxinas Bacterianas/genética , Línea Celular , Células Cultivadas , Cristalografía por Rayos X , Especificidad del Huésped , Humanos , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Ácidos Neuramínicos/metabolismo , Pan troglodytes , Salmonella typhi/patogenicidad , Fiebre Tifoidea/microbiología
5.
Mol Cell ; 81(15): 3160-3170.e9, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34174184

RESUMEN

RelA-SpoT Homolog (RSH) enzymes control bacterial physiology through synthesis and degradation of the nucleotide alarmone (p)ppGpp. We recently discovered multiple families of small alarmone synthetase (SAS) RSH acting as toxins of toxin-antitoxin (TA) modules, with the FaRel subfamily of toxSAS abrogating bacterial growth by producing an analog of (p)ppGpp, (pp)pApp. Here we probe the mechanism of growth arrest used by four experimentally unexplored subfamilies of toxSAS: FaRel2, PhRel, PhRel2, and CapRel. Surprisingly, all these toxins specifically inhibit protein synthesis. To do so, they transfer a pyrophosphate moiety from ATP to the tRNA 3' CCA. The modification inhibits both tRNA aminoacylation and the sensing of cellular amino acid starvation by the ribosome-associated RSH RelA. Conversely, we show that some small alarmone hydrolase (SAH) RSH enzymes can reverse the pyrophosphorylation of tRNA to counter the growth inhibition by toxSAS. Collectively, we establish RSHs as RNA-modifying enzymes.


Asunto(s)
Toxinas Bacterianas/metabolismo , Guanosina Pentafosfato/metabolismo , Ligasas/metabolismo , ARN de Transferencia/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/farmacología , Bacilos Grampositivos Asporogénicos/química , Bacilos Grampositivos Asporogénicos/metabolismo , Guanosina Pentafosfato/química , Ligasas/química , Ligasas/genética , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/fisiología , Inhibidores de la Síntesis de la Proteína/farmacología , Pirofosfatasas , Ribosomas/metabolismo
6.
Mol Cell ; 80(6): 955-970.e7, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33290744

RESUMEN

Prokaryotic toxin-antitoxin (TA) systems are composed of a toxin capable of interfering with key cellular processes and its neutralizing antidote, the antitoxin. Here, we focus on the HEPN-MNT TA system encoded in the vicinity of a subtype I-D CRISPR-Cas system in the cyanobacterium Aphanizomenon flos-aquae. We show that HEPN acts as a toxic RNase, which cleaves off 4 nt from the 3' end in a subset of tRNAs, thereby interfering with translation. Surprisingly, we find that the MNT (minimal nucleotidyltransferase) antitoxin inhibits HEPN RNase through covalent di-AMPylation (diadenylylation) of a conserved tyrosine residue, Y109, in the active site loop. Furthermore, we present crystallographic snapshots of the di-AMPylation reaction at different stages that explain the mechanism of HEPN RNase inactivation. Finally, we propose that the HEPN-MNT system functions as a cellular ATP sensor that monitors ATP homeostasis and, at low ATP levels, releases active HEPN toxin.


Asunto(s)
Antitoxinas/genética , Toxinas Bacterianas/genética , Ribonucleasas/genética , Sistemas Toxina-Antitoxina/genética , Adenosina Monofosfato/genética , Antídotos/química , Antitoxinas/metabolismo , Aphanizomenon/química , Aphanizomenon/genética , Sistemas CRISPR-Cas/genética , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Ribonucleasas/metabolismo , Tirosina/genética
7.
Annu Rev Microbiol ; 76: 21-43, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35395167

RESUMEN

Toxin-antitoxin (TA) systems are ubiquitous genetic elements in bacteria that consist of a growth-inhibiting toxin and its cognate antitoxin. These systems are prevalent in bacterial chromosomes, plasmids, and phage genomes, but individual systems are not highly conserved, even among closely related strains. The biological functions of TA systems have been controversial and enigmatic, although a handful of these systems have been shown to defend bacteria against their viral predators, bacteriophages. Additionally, their patterns of conservation-ubiquitous, but rapidly acquired and lost from genomes-as well as the co-occurrence of some TA systems with known phage defense elements are suggestive of a broader role in mediating phage defense. Here, we review the existing evidence for phage defense mediated by TA systems, highlighting how toxins are activated by phage infection and how toxins disrupt phage replication. We also discuss phage-encoded systems that counteract TA systems, underscoring the ongoing coevolutionary battle between bacteria and phage. We anticipate that TA systems will continue to emerge as central players in the innate immunity of bacteria against phage.


Asunto(s)
Antitoxinas , Toxinas Bacterianas , Bacteriófagos , Sistemas Toxina-Antitoxina , Antitoxinas/genética , Antitoxinas/farmacología , Bacterias/genética , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Bacteriófagos/genética , Plásmidos , Sistemas Toxina-Antitoxina/genética
8.
Nature ; 593(7858): 261-265, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33911281

RESUMEN

Several enteric pathogens can gain specific metabolic advantages over other members of the microbiota by inducing host pathology and inflammation. The pathogen Clostridium difficile is responsible for a toxin-mediated colitis that causes 450,000 infections and 15,000 deaths in the United States each year1; however, the molecular mechanisms by which C. difficile benefits from this pathology remain unclear. To understand how the metabolism of C. difficile adapts to the inflammatory conditions that its toxins induce, here we use RNA sequencing to define, in a mouse model, the metabolic states of wild-type C. difficile and of an isogenic mutant that lacks toxins. By combining bacterial and mouse genetics, we demonstrate that C. difficile uses sorbitol derived from both diet and host. Host-derived sorbitol is produced by the enzyme aldose reductase, which is expressed by diverse immune cells and is upregulated during inflammation-including during toxin-mediated disease induced by C. difficile. This work highlights a mechanism by which C. difficile can use a host-derived nutrient that is generated during toxin-induced disease by an enzyme that has not previously been associated with infection.


Asunto(s)
Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/metabolismo , Infecciones por Clostridium/microbiología , Interacciones Huésped-Patógeno , Sorbitol/metabolismo , Aldehído Reductasa/metabolismo , Animales , Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/genética , Clostridioides difficile/genética , Infecciones por Clostridium/enzimología , Colitis/enzimología , Colitis/metabolismo , Colitis/microbiología , Femenino , Regulación Bacteriana de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación
9.
Mol Cell ; 75(5): 1031-1042.e4, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31327636

RESUMEN

Every bacterial population harbors a small subpopulation of so-called persisters that are transiently antibiotic tolerant. These persisters are associated with the recalcitrance of chronic infections because they can recolonize the host after antibiotic removal. Although several effectors have been described to induce persistence, persister cell awakening is poorly understood. We previously reported that the toxin HokB induces persistence via pore formation, resulting in membrane depolarization and ATP leakage. We now delineate mechanisms responsible for the awakening of HokB-induced persisters. We show that HokB dimerization by the oxidoreductase DsbA is essential for pore formation and peptide stability. Pores are disassembled via DsbC-mediated monomerization, which targets HokB for DegQ-mediated degradation. Finally, pore disassembly allows membrane repolarization by the electron transport chain, supporting cells to resume growth. These results provide a detailed view of both the formation and awakening of HokB-induced persister cells.


Asunto(s)
Toxinas Bacterianas/metabolismo , Membrana Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Potenciales de la Membrana/fisiología , Proteolisis , Serina Endopeptidasas/metabolismo , Toxinas Bacterianas/genética , Membrana Celular/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Serina Endopeptidasas/genética
10.
Mol Cell ; 73(6): 1282-1291.e8, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30792174

RESUMEN

Toxin-antitoxin (TA) systems regulate fundamental cellular processes in bacteria and represent potential therapeutic targets. We report a new RES-Xre TA system in multiple human pathogens, including Mycobacterium tuberculosis. The toxin, MbcT, is bactericidal unless neutralized by its antitoxin MbcA. To investigate the mechanism, we solved the 1.8 Å-resolution crystal structure of the MbcTA complex. We found that MbcT resembles secreted NAD+-dependent bacterial exotoxins, such as diphtheria toxin. Indeed, MbcT catalyzes NAD+ degradation in vitro and in vivo. Unexpectedly, the reaction is stimulated by inorganic phosphate, and our data reveal that MbcT is a NAD+ phosphorylase. In the absence of MbcA, MbcT triggers rapid M. tuberculosis cell death, which reduces mycobacterial survival in macrophages and prolongs the survival of infected mice. Our study expands the molecular activities employed by bacterial TA modules and uncovers a new class of enzymes that could be exploited to treat tuberculosis and other infectious diseases.


Asunto(s)
Antitoxinas/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Macrófagos/microbiología , Mycobacterium tuberculosis/enzimología , Fosforilasas/metabolismo , Sistemas Toxina-Antitoxina , Tuberculosis/microbiología , Animales , Antibióticos Antituberculosos/farmacología , Antitoxinas/química , Antitoxinas/genética , Carga Bacteriana , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Humanos , Cinética , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Viabilidad Microbiana , Modelos Moleculares , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/patogenicidad , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , NAD/metabolismo , Fosforilasas/química , Fosforilasas/genética , Conformación Proteica , Sistemas Toxina-Antitoxina/genética , Tuberculosis/tratamiento farmacológico
11.
Proc Natl Acad Sci U S A ; 121(27): e2403063121, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38935561

RESUMEN

Type I toxin-antitoxin systems (T1TAs) are bipartite bacterial loci encoding a growth-inhibitory toxin and an antitoxin small RNA (sRNA). In many of these systems, the transcribed toxin mRNA is translationally inactive, but becomes translation-competent upon ribonucleolytic processing. The antitoxin sRNA targets the processed mRNA to inhibit its translation. This two-level control mechanism prevents cotranscriptional translation of the toxin and allows its synthesis only when the antitoxin is absent. Contrary to this, we found that the timP mRNA of the timPR T1TA locus does not undergo enzymatic processing. Instead, the full-length timP transcript is both translationally active and can be targeted by the antitoxin TimR. Thus, tight control in this system relies on a noncanonical mechanism. Based on the results from in vitro binding assays, RNA structure probing, and cell-free translation experiments, we suggest that timP mRNA adopts mutually exclusive structural conformations. The active form uniquely possesses an RNA pseudoknot structure which is essential for translation initiation. TimR preferentially binds to the active conformation, which leads to pseudoknot destabilization and inhibited translation. Based on this, we propose a model in which "structural processing" of timP mRNA enables tight inhibition by TimR in nonpermissive conditions, and TimP synthesis only upon TimR depletion.


Asunto(s)
Conformación de Ácido Nucleico , Biosíntesis de Proteínas , ARN Bacteriano , ARN Mensajero , Sistemas Toxina-Antitoxina , Sistemas Toxina-Antitoxina/genética , ARN Bacteriano/metabolismo , ARN Bacteriano/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Antitoxinas/metabolismo , Antitoxinas/genética , Escherichia coli/metabolismo , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica
12.
Nature ; 583(7817): 631-637, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32641830

RESUMEN

Bacterial toxins represent a vast reservoir of biochemical diversity that can be repurposed for biomedical applications. Such proteins include a group of predicted interbacterial toxins of the deaminase superfamily, members of which have found application in gene-editing techniques1,2. Because previously described cytidine deaminases operate on single-stranded nucleic acids3, their use in base editing requires the unwinding of double-stranded DNA (dsDNA)-for example by a CRISPR-Cas9 system. Base editing within mitochondrial DNA (mtDNA), however, has thus far been hindered by challenges associated with the delivery of guide RNA into the mitochondria4. As a consequence, manipulation of mtDNA to date has been limited to the targeted destruction of the mitochondrial genome by designer nucleases9,10.Here we describe an interbacterial toxin, which we name DddA, that catalyses the deamination of cytidines within dsDNA. We engineered split-DddA halves that are non-toxic and inactive until brought together on target DNA by adjacently bound programmable DNA-binding proteins. Fusions of the split-DddA halves, transcription activator-like effector array proteins, and a uracil glycosylase inhibitor resulted in RNA-free DddA-derived cytosine base editors (DdCBEs) that catalyse C•G-to-T•A conversions in human mtDNA with high target specificity and product purity. We used DdCBEs to model a disease-associated mtDNA mutation in human cells, resulting in changes in respiration rates and oxidative phosphorylation. CRISPR-free DdCBEs enable the precise manipulation of mtDNA, rather than the elimination of mtDNA copies that results from its cleavage by targeted nucleases, with broad implications for the study and potential treatment of mitochondrial disorders.


Asunto(s)
Toxinas Bacterianas/metabolismo , Citidina Desaminasa/metabolismo , ADN Mitocondrial/genética , Edición Génica/métodos , Genes Mitocondriales/genética , Mitocondrias/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Secuencia de Bases , Burkholderia cenocepacia/enzimología , Burkholderia cenocepacia/genética , Respiración de la Célula/genética , Citidina/metabolismo , Citidina Desaminasa/química , Citidina Desaminasa/genética , Genoma Mitocondrial/genética , Células HEK293 , Humanos , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/terapia , Mutación , Fosforilación Oxidativa , Ingeniería de Proteínas , ARN Guía de Kinetoplastida/genética , Especificidad por Sustrato , Sistemas de Secreción Tipo VI/metabolismo
13.
Mol Cell ; 70(5): 768-784, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29398446

RESUMEN

Bacterial toxin-antitoxin (TA) modules are abundant genetic elements that encode a toxin protein capable of inhibiting cell growth and an antitoxin that counteracts the toxin. The majority of toxins are enzymes that interfere with translation or DNA replication, but a wide variety of molecular activities and cellular targets have been described. Antitoxins are proteins or RNAs that often control their cognate toxins through direct interactions and, in conjunction with other signaling elements, through transcriptional and translational regulation of TA module expression. Three major biological functions of TA modules have been discovered, post-segregational killing ("plasmid addiction"), abortive infection (bacteriophage immunity through altruistic suicide), and persister formation (antibiotic tolerance through dormancy). In this review, we summarize the current state of the field and highlight how multiple levels of regulation shape the conditions of toxin activation to achieve the different biological functions of TA modules.


Asunto(s)
Antitoxinas/metabolismo , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , ARN Bacteriano/metabolismo , Antitoxinas/química , Antitoxinas/genética , Bacterias/genética , Bacterias/inmunología , Bacterias/patogenicidad , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Evolución Molecular , Regulación Bacteriana de la Expresión Génica , Inmunidad Innata , Viabilidad Microbiana , Modelos Moleculares , Conformación de Ácido Nucleico , Conformación Proteica , Procesamiento Postranscripcional del ARN , ARN Bacteriano/química , ARN Bacteriano/genética , Relación Estructura-Actividad , Transcripción Genética
14.
Nucleic Acids Res ; 52(4): 1847-1859, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38224456

RESUMEN

Toxin-antitoxin (TA) systems are small selfish genetic modules that increase vertical stability of their replicons. They have long been thought to stabilize plasmids by killing cells that fail to inherit a plasmid copy through a phenomenon called post-segregational killing (PSK) or addiction. While this model has been widely accepted, no direct observation of PSK was reported in the literature. Here, we devised a system that enables visualization of plasmid loss and PSK at the single-cell level using meganuclease-driven plasmid curing. Using the ccd system, we show that cells deprived of a ccd-encoding plasmid show hallmarks of DNA damage, i.e. filamentation and induction of the SOS response. Activation of ccd triggered cell death in most plasmid-free segregants, although some intoxicated cells were able to resume growth, showing that PSK-induced damage can be repaired in a SOS-dependent manner. Damage induced by ccd activates resident lambdoid prophages, which potentiate the killing effect of ccd. The loss of a model plasmid containing TA systems encoding toxins presenting various molecular mechanisms induced different morphological changes, growth arrest and loss of viability. Our experimental setup enables further studies of TA-induced phenotypes and suggests that PSK is a general mechanism for plasmid stabilization by TA systems.


Asunto(s)
Bacterias , Plásmidos , Sistemas Toxina-Antitoxina , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Plásmidos/genética , Bacterias/genética
15.
Nucleic Acids Res ; 52(10): 5987-6001, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38485701

RESUMEN

Mycobacterium tuberculosis transfer RNA (tRNA) terminal nucleotidyltransferase toxin, MenT3, incorporates nucleotides at the 3'-CCA end of tRNAs, blocking their aminoacylation and inhibiting protein synthesis. Here, we show that MenT3 most effectively adds CMPs to the 3'-CCA end of tRNA. The crystal structure of MenT3 in complex with CTP reveals a CTP-specific nucleotide-binding pocket. The 4-NH2 and the N3 and O2 atoms of cytosine in CTP form hydrogen bonds with the main-chain carbonyl oxygen of P120 and the side chain of R238, respectively. MenT3 expression in Escherichia coli selectively reduces the levels of seryl-tRNASers, indicating specific inactivation of tRNASers by MenT3. Consistently, MenT3 incorporates CMPs into tRNASer most efficiently, among the tested E. coli tRNA species. The longer variable loop unique to class II tRNASers is crucial for efficient CMP incorporation into tRNASer by MenT3. Replacing the variable loop of E. coli tRNAAla with the longer variable loop of M. tuberculosis tRNASer enables MenT3 to incorporate CMPs into the chimeric tRNAAla. The N-terminal positively charged region of MenT3 is required for CMP incorporation into tRNASer. A docking model of tRNA onto MenT3 suggests that an interaction between the N-terminal region and the longer variable loop of tRNASer facilitates tRNA substrate selection.


Asunto(s)
Mycobacterium tuberculosis , ARN de Transferencia , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/enzimología , Especificidad por Sustrato , ARN de Transferencia/metabolismo , ARN de Transferencia/química , Escherichia coli/genética , Escherichia coli/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Modelos Moleculares , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Citidina/química , Citidina/metabolismo , Sitios de Unión , Cristalografía por Rayos X , ARN Nucleotidiltransferasas/metabolismo , ARN Nucleotidiltransferasas/química , ARN Nucleotidiltransferasas/genética
16.
Trends Biochem Sci ; 46(12): 953-959, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34429235

RESUMEN

Large clostridial toxins (LCTs) are a family of six homologous disease-causing proteins characterised by their large size (>200 kDa) and conserved multidomain architectures. Using their central translocation and receptor-binding domain (T domain), LCTs bind host cell receptors and translocate their upstream glycosyltransferase and cysteine protease domain across the endosomal membrane and into the cytosol. The recent discovery of hundreds of LCT-like T domains in diverse genomic contexts and domain architectures from bacteria other than clostridia has provided significant new insights into the enigmatic process of LCT translocation, but also has put the definition of what constitutes an LCT into question. In this opinion article, we discuss how these findings have expanded our understanding of LCT translocation and reshaped the scope of the LCT family.


Asunto(s)
Toxinas Bacterianas , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Membranas Intracelulares/metabolismo , Dominios Proteicos
17.
Bioinformatics ; 40(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38885365

RESUMEN

MOTIVATION: ADP-ribosylation is a critical modification involved in regulating diverse cellular processes, including chromatin structure regulation, RNA transcription, and cell death. Bacterial ADP-ribosyltransferase toxins (bARTTs) serve as potent virulence factors that orchestrate the manipulation of host cell functions to facilitate bacterial pathogenesis. Despite their pivotal role, the bioinformatic identification of novel bARTTs poses a formidable challenge due to limited verified data and the inherent sequence diversity among bARTT members. RESULTS: We proposed a deep learning-based model, ARTNet, specifically engineered to predict bARTTs from bacterial genomes. Initially, we introduced an effective data augmentation method to address the issue of data scarcity in training ARTNet. Subsequently, we employed a data optimization strategy by utilizing ART-related domain subsequences instead of the primary full sequences, thereby significantly enhancing the performance of ARTNet. ARTNet achieved a Matthew's correlation coefficient (MCC) of 0.9351 and an F1-score (macro) of 0.9666 on repeated independent test datasets, outperforming three other deep learning models and six traditional machine learning models in terms of time efficiency and accuracy. Furthermore, we empirically demonstrated the ability of ARTNet to predict novel bARTTs across domain superfamilies without sequence similarity. We anticipate that ARTNet will greatly facilitate the screening and identification of novel bARTTs from bacterial genomes. AVAILABILITY AND IMPLEMENTATION: ARTNet is publicly accessible at http://www.mgc.ac.cn/ARTNet/. The source code of ARTNet is freely available at https://github.com/zhengdd0422/ARTNet/.


Asunto(s)
ADP Ribosa Transferasas , Biología Computacional , Aprendizaje Profundo , ADP Ribosa Transferasas/metabolismo , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/genética , Biología Computacional/métodos , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Genoma Bacteriano , Bacterias/genética
18.
PLoS Pathog ; 19(10): e1011496, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37871122

RESUMEN

Clostridioides difficile is a leading cause of antibiotic-associated diarrhea and nosocomial infection in the United States. The symptoms of C. difficile infection (CDI) are associated with the production of two homologous protein toxins, TcdA and TcdB. The toxins are considered bona fide targets for clinical diagnosis as well as the development of novel prevention and therapeutic strategies. While there are extensive studies that document these efforts, there are several gaps in knowledge that could benefit from the creation of new research tools. First, we now appreciate that while TcdA sequences are conserved, TcdB sequences can vary across the span of circulating clinical isolates. An understanding of the TcdA and TcdB epitopes that drive broadly neutralizing antibody responses could advance the effort to identify safe and effective toxin-protein chimeras and fragments for vaccine development. Further, an understanding of TcdA and TcdB concentration changes in vivo can guide research into how host and microbiome-focused interventions affect the virulence potential of C. difficile. We have developed a panel of alpaca-derived nanobodies that bind specific structural and functional domains of TcdA and TcdB. We note that many of the potent neutralizers of TcdA bind epitopes within the delivery domain, a finding that could reflect roles of the delivery domain in receptor binding and/or the conserved role of pore-formation in the delivery of the toxin enzyme domains to the cytosol. In contrast, neutralizing epitopes for TcdB were found in multiple domains. The nanobodies were also used for the creation of sandwich ELISA assays that allow for quantitation of TcdA and/or TcdB in vitro and in the cecal and fecal contents of infected mice. We anticipate these reagents and assays will allow researchers to monitor the dynamics of TcdA and TcdB production over time, and the impact of various experimental interventions on toxin production in vivo.


Asunto(s)
Toxinas Bacterianas , Clostridioides difficile , Anticuerpos de Dominio Único , Animales , Ratones , Toxinas Bacterianas/genética , Toxinas Bacterianas/química , Enterotoxinas/genética , Clostridioides difficile/genética , Clostridioides difficile/metabolismo , Epítopos/metabolismo , Proteínas Bacterianas/metabolismo
19.
Mol Syst Biol ; 20(8): 859-879, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39069594

RESUMEN

Secretion systems play a crucial role in microbe-microbe or host-microbe interactions. Among these systems, the extracellular contractile injection system (eCIS) is a unique bacterial and archaeal extracellular secretion system that injects protein toxins into target organisms. However, the specific proteins that eCISs inject into target cells and their functions remain largely unknown. Here, we developed a machine learning classifier to identify eCIS-associated toxins (EATs). The classifier combines genetic and biochemical features to identify EATs. We also developed a score for the eCIS N-terminal signal peptide to predict EAT loading. Using the classifier we classified 2,194 genes from 950 genomes as putative EATs. We validated four new EATs, EAT14-17, showing toxicity in bacterial and eukaryotic cells, and identified residues of their respective active sites that are critical for toxicity. Finally, we show that EAT14 inhibits mitogenic signaling in human cells. Our study provides insights into the diversity and functions of EATs and demonstrates machine learning capability of identifying novel toxins. The toxins can be employed in various applications dependently or independently of eCIS.


Asunto(s)
Aprendizaje Automático , Humanos , Toxinas Bacterianas/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
20.
Annu Rev Microbiol ; 74: 497-520, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32680451

RESUMEN

All bacteria must compete for growth niches and other limited environmental resources. These existential battles are waged at several levels, but one common strategy entails the transfer of growth-inhibitory protein toxins between competing cells. These antibacterial effectors are invariably encoded with immunity proteins that protect cells from intoxication by neighboring siblings. Several effector classes have been described, each designed to breach the cell envelope of target bacteria. Although effector architectures and export pathways tend to be clade specific, phylogenetically distant species often deploy closely related toxin domains. Thus, diverse competition systems are linked through a common reservoir of toxin-immunity pairs that is shared via horizontal gene transfer. These toxin-immunity protein pairs are extraordinarily diverse in sequence, and this polymorphism underpins an important mechanism of self/nonself discrimination in bacteria. This review focuses on the structures, functions, and delivery mechanisms of polymorphic toxin effectors that mediate bacterial competition.


Asunto(s)
Bacterias/inmunología , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Transferencia de Gen Horizontal , Interacciones Microbianas , Bacterias/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Sistemas de Secreción Tipo VI/genética , Sistemas de Secreción Tipo VI/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA