Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 393
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(27): e2407832121, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38935560

RESUMEN

In 1967, in this journal, Evelyn Witkin proposed the existence of a coordinated DNA damage response in Escherichia coli, which later came to be called the "SOS response." We revisited this response using the replication inhibitor azidothymidine (AZT) and RNA-Seq analysis and identified several features. We confirm the induction of classic Save our ship (SOS) loci and identify several genes, including many of the pyrimidine pathway, that have not been previously demonstrated to be DNA damage-inducible. Despite a strong dependence on LexA, these genes lack LexA boxes and their regulation by LexA is likely to be indirect via unknown factors. We show that the transcription factor "stringent starvation protein" SspA is as important as LexA in the regulation of AZT-induced genes and that the genes activated by SspA change dramatically after AZT exposure. Our experiments identify additional LexA-independent DNA damage inducible genes, including 22 small RNA genes, some of which appear to activated by SspA. Motility and chemotaxis genes are strongly down-regulated by AZT, possibly as a result of one of more of the small RNAs or other transcription factors such as AppY and GadE, whose expression is elevated by AZT. Genes controlling the iron siderophore, enterobactin, and iron homeostasis are also strongly induced, independent of LexA. We confirm that IraD antiadaptor protein is induced independent of LexA and that a second antiadaptor, IraM is likewise strongly AZT-inducible, independent of LexA, suggesting that RpoS stabilization via these antiadaptor proteins is an integral part of replication stress tolerance.


Asunto(s)
Daño del ADN , Replicación del ADN , Proteínas de Escherichia coli , Escherichia coli , Regulación Bacteriana de la Expresión Génica , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Replicación del ADN/efectos de los fármacos , Respuesta SOS en Genética/efectos de los fármacos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Serina Endopeptidasas
2.
Int J Mol Sci ; 25(11)2024 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-38892328

RESUMEN

Curcumin is a natural compound that is considered safe and may have potential health benefits; however, its poor stability and water insolubility limit its therapeutic applications. Different strategies aim to increase its water solubility. Here, we tested the compound PVP-curcumin as a photosensitizer for antimicrobial photodynamic therapy (aPDT) as well as its potential to act as an adjuvant in antibiotic drug therapy. Gram-negative E. coli K12 and Gram-positive S. capitis were subjected to aPDT using various PVP-curcumin concentrations (1-200 µg/mL) and 475 nm blue light (7.5-45 J/cm2). Additionally, results were compared to aPDT using 415 nm blue light. Gene expression of recA and umuC were analyzed via RT-qPCR to assess effects on the bacterial SOS response. Further, the potentiation of Ciprofloxacin by PVP-curcumin was investigated, as well as its potential to prevent the emergence of antibiotic resistance. Both bacterial strains were efficiently reduced when irradiated with 415 nm blue light (2.2 J/cm2) and 10 µg/mL curcumin. Using 475 nm blue light, bacterial reduction followed a biphasic effect with higher efficacy in S. capitis compared to E. coli K12. PVP-curcumin decreased recA expression but had limited effect regarding enhancing antibiotic treatment or impeding resistance development. PVP-curcumin demonstrated effectiveness as a photosensitizer against both Gram-positive and Gram-negative bacteria but did not modulate the bacterial SOS response.


Asunto(s)
Antibacterianos , Ciprofloxacina , Curcumina , Fármacos Fotosensibilizantes , Rec A Recombinasas , Curcumina/farmacología , Fármacos Fotosensibilizantes/farmacología , Rec A Recombinasas/metabolismo , Rec A Recombinasas/genética , Ciprofloxacina/farmacología , Antibacterianos/farmacología , Fotoquimioterapia/métodos , Respuesta SOS en Genética/efectos de los fármacos , Escherichia coli K12/efectos de los fármacos , Escherichia coli K12/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Povidona/química , Povidona/farmacología , Pruebas de Sensibilidad Microbiana , Escherichia coli/efectos de los fármacos , Luz , Proteínas de Unión al ADN
3.
mBio ; 15(6): e0050424, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38757974

RESUMEN

A recent demonstration of synergy between a temperate phage and the antibiotic ciprofloxacin suggested a scalable approach to exploiting temperate phages in therapy, termed temperate phage-antibiotic synergy, which specifically interacted with the lysis-lysogeny decision. To determine whether this would hold true across antibiotics, we challenged Escherichia coli with the phage HK97 and a set of 13 antibiotics spanning seven classes. As expected, given the conserved induction pathway, we observed synergy with classes of drugs known to induce an SOS response: a sulfa drug, other quinolones, and mitomycin C. While some ß-lactams exhibited synergy, this appeared to be traditional phage-antibiotic synergy, with no effect on the lysis-lysogeny decision. Curiously, we observed a potent synergy with antibiotics not known to induce the SOS response: protein synthesis inhibitors gentamicin, kanamycin, tetracycline, and azithromycin. The synergy results in an eightfold reduction in the effective minimum inhibitory concentration of gentamicin, complete eradication of the bacteria, and, when administered at sub-optimal doses, drastically decreases the frequency of lysogens emerging from the combined challenge. However, lysogens exhibit no increased sensitivity to the antibiotic; synergy was maintained in the absence of RecA; and the antibiotic reduced the initial frequency of lysogeny rather than selecting against formed lysogens. Our results confirm that SOS-inducing antibiotics broadly result in temperate-phage-specific synergy, but that other antibiotics can interact with temperate phages specifically and result in synergy. This is the first report of a means of chemically blocking entry into lysogeny, providing a new means for manipulating the key lysis-lysogeny decision.IMPORTANCEThe lysis-lysogeny decision is made by most bacterial viruses (bacteriophages, phages), determining whether to kill their host or go dormant within it. With over half of the bacteria containing phages waiting to wake, this is one of the most important behaviors in all of biology. These phages are also considered unusable for therapy because of this behavior. In this paper, we show that many antibiotics bias this behavior to "wake" the dormant phages, forcing them to kill their host, but some also prevent dormancy in the first place. These will be important tools to study this critical decision point and may enable the therapeutic use of these phages.


Asunto(s)
Antibacterianos , Escherichia coli , Lisogenia , Antibacterianos/farmacología , Escherichia coli/virología , Escherichia coli/efectos de los fármacos , Respuesta SOS en Genética/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Colifagos/fisiología , Colifagos/efectos de los fármacos , Sinergismo Farmacológico , Bacteriófagos/fisiología , Bacteriófagos/efectos de los fármacos , Mitomicina/farmacología
4.
Drug Resist Updat ; 75: 101087, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38678745

RESUMEN

In recent years, new evidence has shown that the SOS response plays an important role in the response to antimicrobials, with involvement in the generation of clinical resistance. Here we evaluate the impact of heterogeneous expression of the SOS response in clinical isolates of Escherichia coli on response to the fluoroquinolone, ciprofloxacin. In silico analysis of whole genome sequencing data showed remarkable sequence conservation of the SOS response regulators, RecA and LexA. Despite the genetic homogeneity, our results revealed a marked differential heterogeneity in SOS response activation, both at population and single-cell level, among clinical isolates of E. coli in the presence of subinhibitory concentrations of ciprofloxacin. Four main stages of SOS response activation were identified and correlated with cell filamentation. Interestingly, there was a correlation between clinical isolates with higher expression of the SOS response and further progression to resistance. This heterogeneity in response to DNA damage repair (mediated by the SOS response) and induced by antimicrobial agents could be a new factor with implications for bacterial evolution and survival contributing to the generation of antimicrobial resistance.


Asunto(s)
Antibacterianos , Ciprofloxacina , Proteínas de Escherichia coli , Escherichia coli , Pruebas de Sensibilidad Microbiana , Rec A Recombinasas , Respuesta SOS en Genética , Respuesta SOS en Genética/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Ciprofloxacina/farmacología , Humanos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Antibacterianos/farmacología , Rec A Recombinasas/genética , Rec A Recombinasas/metabolismo , Farmacorresistencia Bacteriana/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Daño del ADN/efectos de los fármacos , Secuenciación Completa del Genoma , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/tratamiento farmacológico , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Adaptación Fisiológica , Reparación del ADN/efectos de los fármacos , Proteínas de Unión al ADN
5.
J Hazard Mater ; 471: 134257, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38636236

RESUMEN

The widespread use of disinfectants during the global response to the 2019 coronavirus pandemic has increased the co-occurrence of disinfection byproducts (DBPs) and antibiotic resistance genes (ARGs). Although DBPs pose major threats to public health globally, there is limited knowledge regarding their biological effects on ARGs. This study aimed to investigate the effects of two inorganic DBPs (chlorite and bromate) on the conjugative transfer of RP4 plasmid among Escherichia coli strains at environmentally relevant concentrations. Interestingly, the frequency of conjugative transfer was initially inhibited when the exposure time to chlorite or bromate was less than 24 h. However, this inhibition transformed into promotion when the exposure time was extended to 36 h. Short exposures to chlorite or bromate were shown to impede the electron transport chain, resulting in an ATP shortage and subsequently inhibiting conjugative transfer. Consequently, this stimulates the overproduction of reactive oxygen species (ROS) and activation of the SOS response. Upon prolonged exposure, the resurgent energy supply promoted conjugative transfer. These findings offer novel and valuable insights into the effects of environmentally relevant concentrations of inorganic DBPs on the conjugative transfer of ARGs, thereby providing a theoretical basis for the management of DBPs.


Asunto(s)
Bromatos , Cloruros , Escherichia coli , Estrés Oxidativo , Plásmidos , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Bromatos/toxicidad , Plásmidos/genética , Cloruros/farmacología , Desinfectantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Conjugación Genética/efectos de los fármacos , Farmacorresistencia Microbiana/genética , Farmacorresistencia Bacteriana/genética , Farmacorresistencia Bacteriana/efectos de los fármacos , Respuesta SOS en Genética/efectos de los fármacos
6.
Antimicrob Agents Chemother ; 68(5): e0146223, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38534113

RESUMEN

Although the mechanistic connections between SOS-induced mutagenesis and antibiotic resistance are well established, our current understanding of the impact of SOS response levels, recovery durations, and transcription/translation activities on mutagenesis remains relatively limited. In this study, when bacterial cells were exposed to mutagens like ultraviolet light for defined time intervals, a compelling connection between the rate of mutagenesis and the RecA-mediated SOS response levels became evident. Our observations also indicate that mutagenesis primarily occurs during the subsequent recovery phase following the removal of the mutagenic agent. When transcription/translation was inhibited or energy molecules were depleted at the onset of treatment or during the early recovery phase, there was a noticeable decrease in SOS response activation and mutagenesis. However, targeting these processes later in the recovery phase does not have the same effect in reducing mutagenesis, suggesting that the timing of inhibiting transcription/translation or depleting energy molecules is crucial for their efficacy in reducing mutagenesis. Active transcription, translation, and energy availability within the framework of SOS response and DNA repair mechanisms appear to be conserved attributes, supported by their consistent manifestation across diverse conditions, including the use of distinct mutagens such as fluoroquinolones and various bacterial strains.


Asunto(s)
Escherichia coli , Mutagénesis , Rec A Recombinasas , Respuesta SOS en Genética , Rayos Ultravioleta , Respuesta SOS en Genética/efectos de los fármacos , Respuesta SOS en Genética/genética , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Rec A Recombinasas/genética , Rec A Recombinasas/metabolismo , Antibacterianos/farmacología , Reparación del ADN , Mutágenos/farmacología , Proteínas de Escherichia coli/genética , Farmacorresistencia Bacteriana/genética , Transcripción Genética
7.
Nature ; 603(7900): 315-320, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35197633

RESUMEN

Colibactin is a chemically unstable small-molecule genotoxin that is produced by several different bacteria, including members of the human gut microbiome1,2. Although the biological activity of colibactin has been extensively investigated in mammalian systems3, little is known about its effects on other microorganisms. Here we show that colibactin targets bacteria that contain prophages, and induces lytic development through the bacterial SOS response. DNA, added exogenously, protects bacteria from colibactin, as does expressing a colibactin resistance protein (ClbS) in non-colibactin-producing cells. The prophage-inducing effects that we observe apply broadly across different phage-bacteria systems and in complex communities. Finally, we identify bacteria that have colibactin resistance genes but lack colibactin biosynthetic genes. Many of these bacteria are infected with predicted prophages, and we show that the expression of their ClbS homologues provides immunity from colibactin-triggered induction. Our study reveals a mechanism by which colibactin production could affect microbiomes and highlights a role for microbial natural products in influencing population-level events such as phage outbreaks.


Asunto(s)
Bacterias , Toxinas Bacterianas , Péptidos , Policétidos , Profagos , Activación Viral , Bacterias/efectos de los fármacos , Bacterias/virología , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacología , Bacteriólisis/efectos de los fármacos , Interacciones Microbianas/efectos de los fármacos , Péptidos/metabolismo , Péptidos/farmacología , Policétidos/metabolismo , Policétidos/farmacología , Profagos/efectos de los fármacos , Profagos/fisiología , Respuesta SOS en Genética/efectos de los fármacos , Activación Viral/efectos de los fármacos
8.
Nat Commun ; 13(1): 672, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115506

RESUMEN

Microbial biocontainment is an essential goal for engineering safe, next-generation living therapeutics. However, the genetic stability of biocontainment circuits, including kill switches, is a challenge that must be addressed. Kill switches are among the most difficult circuits to maintain due to the strong selection pressure they impart, leading to high potential for evolution of escape mutant populations. Here we engineer two CRISPR-based kill switches in the probiotic Escherichia coli Nissle 1917, a single-input chemical-responsive switch and a 2-input chemical- and temperature-responsive switch. We employ parallel strategies to address kill switch stability, including functional redundancy within the circuit, modulation of the SOS response, antibiotic-independent plasmid maintenance, and provision of intra-niche competition by a closely related strain. We demonstrate that strains harboring either kill switch can be selectively and efficiently killed inside the murine gut, while strains harboring the 2-input switch are additionally killed upon excretion. Leveraging redundant strategies, we demonstrate robust biocontainment of our kill switch strains and provide a template for future kill switch development.


Asunto(s)
Sistemas CRISPR-Cas/genética , Escherichia coli/genética , Ingeniería Genética/métodos , Probióticos/metabolismo , Animales , Antibacterianos/farmacología , Escherichia coli/metabolismo , Escherichia coli/fisiología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ratones Endogámicos C57BL , Viabilidad Microbiana/efectos de los fármacos , Viabilidad Microbiana/genética , Probióticos/farmacología , Respuesta SOS en Genética/efectos de los fármacos , Respuesta SOS en Genética/genética , Estreptomicina/farmacología , Temperatura , Tetraciclinas/farmacología
9.
Elife ; 112022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35037621

RESUMEN

The plasmid-mediated quinolone resistance (PMQR) genes have been shown to promote high-level bacterial resistance to fluoroquinolone antibiotics, potentially leading to clinical treatment failures. In Escherichia coli, sub-minimum inhibitory concentrations (sub-MICs) of the widely used fluoroquinolones are known to induce the SOS response. Interestingly, the expression of several PMQR qnr genes is controlled by the SOS master regulator, LexA. During the characterization of a small qnrD-plasmid carried in E. coli, we observed that the aminoglycosides become able to induce the SOS response in this species, thus leading to the elevated transcription of qnrD. Our findings show that the induction of the SOS response is due to nitric oxide (NO) accumulation in the presence of sub-MIC of aminoglycosides. We demonstrated that the NO accumulation is driven by two plasmid genes, ORF3 and ORF4, whose products act at two levels. ORF3 encodes a putative flavin adenine dinucleotide (FAD)-binding oxidoreductase which helps NO synthesis, while ORF4 codes for a putative fumarate and nitrate reductase (FNR)-type transcription factor, related to an O2-responsive regulator of hmp expression, able to repress the Hmp-mediated NO detoxification pathway of E. coli. Thus, this discovery, that other major classes of antibiotics may induce the SOS response could have worthwhile implications for antibiotic stewardship efforts in preventing the emergence of resistance.


Asunto(s)
Aminoglicósidos/farmacología , Farmacorresistencia Bacteriana/genética , Escherichia coli , Plásmidos/genética , Respuesta SOS en Genética/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/fisiología , Óxido Nítrico/metabolismo , Quinolonas
10.
Cells ; 10(11)2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34831448

RESUMEN

Antibiotics are well known drugs which, when present above certain concentrations, are able to inhibit the growth of certain bacteria. However, a growing body of evidence shows that even when present at lower doses (subMIC, for sub-minimal inhibitory concentration), unable to inhibit or affect microbial growth, antibiotics work as signaling molecules, affect gene expression and trigger important bacterial stress responses. However, how subMIC antibiotic signaling interplays with other well-known signaling networks in bacteria (and the consequences of such interplay) is not well understood. In this work, through transcriptomic and genetic approaches, we have explored how quorum-sensing (QS) proficiency of V. cholerae affects this pathogen's response to subMIC doses of the aminoglycoside tobramycin (TOB). We show that the transcriptomic signature of V. cholerae in response to subMIC TOB depends highly on the presence of QS master regulator HapR. In parallel, we show that subMIC doses of TOB are able to negatively interfere with the AI-2/LuxS QS network of V. cholerae, which seems critical for survival to aminoglycoside treatment and TOB-mediated induction of SOS response in this species. This interplay between QS and aminoglycosides suggests that targeting QS signaling may be a strategy to enhance aminoglycoside efficacy in V. cholerae.


Asunto(s)
Aminoglicósidos/farmacología , Viabilidad Microbiana , Percepción de Quorum/efectos de los fármacos , Vibrio cholerae/fisiología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Viabilidad Microbiana/efectos de los fármacos , Mutación/genética , Percepción de Quorum/genética , Respuesta SOS en Genética/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tobramicina/farmacología , Transcriptoma/genética , Vibrio cholerae/efectos de los fármacos , Vibrio cholerae/genética , Vibrio cholerae/crecimiento & desarrollo
11.
Toxins (Basel) ; 13(7)2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34201801

RESUMEN

Several classes of non-antibiotic drugs, including psychoactive drugs, proton-pump inhibitors (PPIs), non-steroidal anti-inflammatory drugs (NSAIDs), and others, appear to have strong antimicrobial properties. We considered whether psychoactive drugs induce the SOS response in E. coli bacteria and, consequently, induce Shiga toxins in Shiga-toxigenic E. coli (STEC). We measured the induction of an SOS response using a recA-lacZ E. coli reporter strain, as RecA is an early, reliable, and quantifiable marker for activation of the SOS stress response pathway. We also measured the production and release of Shiga toxin 2 (Stx2) from a classic E. coli O157:H7 strain, derived from a food-borne outbreak due to spinach. Some, but not all, serotonin selective reuptake inhibitors (SSRIs) and antipsychotic drugs induced an SOS response. The use of SSRIs is widespread and increasing; thus, the use of these antidepressants could account for some cases of hemolytic-uremic syndrome due to STEC and is not attributable to antibiotic administration. SSRIs could have detrimental effects on the normal intestinal microbiome in humans. In addition, as SSRIs are resistant to environmental breakdown, they could have effects on microbial communities, including aquatic ecosystems, long after they have left the human body.


Asunto(s)
Antipsicóticos/farmacología , Respuesta SOS en Genética/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Toxina Shiga II/metabolismo , Escherichia coli Shiga-Toxigénica/efectos de los fármacos , Escherichia coli Shiga-Toxigénica/genética , Escherichia coli Shiga-Toxigénica/metabolismo , beta-Galactosidasa/genética
12.
Biomolecules ; 11(6)2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-34072355

RESUMEN

A broad spectrum of volatile organic compounds' (VOCs') biological activities has attracted significant scientific interest, but their mechanisms of action remain little understood. The mechanism of action of two VOCs-the cyclic monoterpenes (-)-limonene and (+)-α-pinene-on bacteria was studied in this work. We used genetically engineered Escherichia coli bioluminescent strains harboring stress-responsive promoters (responsive to oxidative stress, DNA damage, SOS response, protein damage, heatshock, membrane damage) fused to the luxCDABE genes of Photorhabdus luminescens. We showed that (-)-limonene induces the PkatG and PsoxS promoters due to the formation of reactive oxygen species and, as a result, causes damage to DNA (SOSresponse), proteins (heat shock), and membrane (increases its permeability). The experimental data indicate that the action of (-)-limonene at high concentrations and prolonged incubation time makes degrading processes in cells irreversible. The effect of (+)-α-pinene is much weaker: it induces only heat shock in the bacteria. Moreover, we showed for the first time that (-)-limonene completely inhibits the DnaKJE-ClpB bichaperone-dependent refolding of heat-inactivated bacterial luciferase in both E. coli wild type and mutant ΔibpB strains. (+)-α-Pinene partially inhibits refolding only in ΔibpB mutant strain.


Asunto(s)
Proteínas Bacterianas , Monoterpenos Bicíclicos , Daño del ADN , ADN Bacteriano , Escherichia coli K12 , Limoneno , Respuesta SOS en Genética/efectos de los fármacos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Monoterpenos Bicíclicos/química , Monoterpenos Bicíclicos/metabolismo , Monoterpenos Bicíclicos/farmacología , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Limoneno/química , Limoneno/metabolismo , Limoneno/farmacología , Photorhabdus/genética
13.
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
14.
Cells ; 10(4)2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33921732

RESUMEN

A bioinformatic search for LexA boxes, combined with transcriptomic detection of loci responsive to DNA damage, identified 48 members of the SOS regulon in the genome of Salmonella enterica serovar Typhimurium. Single cell analysis using fluorescent fusions revealed that heterogeneous expression is a common trait of SOS response genes, with formation of SOSOFF and SOSON subpopulations. Phenotypic cell variants formed in the absence of external DNA damage show gene expression patterns that are mainly determined by the position and the heterology index of the LexA box. SOS induction upon DNA damage produces SOSOFF and SOSON subpopulations that contain live and dead cells. The nature and concentration of the DNA damaging agent and the time of exposure are major factors that influence the population structure upon SOS induction. An analogy can thus be drawn between the SOS response and other bacterial stress responses that produce phenotypic cell variants.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Respuesta SOS en Genética , Salmonella typhimurium/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Cromosomas Bacterianos/genética , Daño del ADN/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Sitios Genéticos , Ácido Nalidíxico/farmacología , Respuesta SOS en Genética/efectos de los fármacos , Salmonella typhimurium/efectos de los fármacos , Análisis de la Célula Individual
15.
Future Med Chem ; 13(2): 143-155, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33410707

RESUMEN

Antimicrobial resistance is a pressing threat to global health, with multidrug-resistant pathogens becoming increasingly prevalent. The bacterial SOS pathway functions in response to DNA damage that occurs during infection, initiating several pro-survival and resistance mechanisms, such as DNA repair and hypermutation. This makes SOS pathway components potential targets that may combat drug-resistant pathogens and decrease resistance emergence. This review discusses the mechanism of the SOS pathway; the structure and function of potential targets AddAB, RecBCD, RecA and LexA; and efforts to develop selective small-molecule inhibitors of these proteins. These inhibitors may serve as valuable tools for target validation and provide the foundations for desperately needed novel antibacterial therapeutics.


Asunto(s)
Antibacterianos/química , Inhibidores Enzimáticos/química , Respuesta SOS en Genética/efectos de los fármacos , Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Reparación del ADN/efectos de los fármacos , Farmacorresistencia Bacteriana , Inhibidores Enzimáticos/farmacología , Exodesoxirribonucleasa V/antagonistas & inhibidores , Exodesoxirribonucleasa V/genética , Exodesoxirribonucleasas/antagonistas & inhibidores , Exodesoxirribonucleasas/genética , Regulación de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Rec A Recombinasas/antagonistas & inhibidores , Rec A Recombinasas/genética , Serina Endopeptidasas/genética , Transducción de Señal , Relación Estructura-Actividad
16.
Artículo en Inglés | MEDLINE | ID: mdl-32660820

RESUMEN

DNA is exposed to the attack of several exogenous agents that modify its chemical structure, so cells must repair those changes in order to survive. Alkylating agents introduce methyl or ethyl groups in most of the cyclic or exocyclic nitrogen atoms of the ring and exocyclic oxygen available in DNA bases producing damage that can induce the SOS response in Escherichia coli and many other bacteria. Likewise, ultraviolet light produces mainly cyclobutane pyrimidine dimers that arrest the progression of the replication fork and triggers such response. The need of some enzymes (such as RecO, ExoI and RecJ) in processing injuries produced by gamma radiation prior the induction of the SOS response has been reported before. In the present work, several repair-defective strains of E. coli were treated with methyl methanesulfonate, ethyl methanesulfonate, mitomycin C or ultraviolet light. Both survival and SOS induction (by means of the Chromotest) were tested. Our results indicate that the participation of these genes depends on the type of injury caused by a genotoxin on DNA.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Mutágenos/farmacología , Respuesta SOS en Genética/efectos de los fármacos , Respuesta SOS en Genética/genética , Alquilantes/farmacología , Proteínas Bacterianas/genética , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Metanosulfonato de Etilo/farmacología , Metilmetanosulfonato/farmacología , Mitomicina/farmacología , Dímeros de Pirimidina/farmacología , Rayos Ultravioleta/efectos adversos
17.
Microbiology (Reading) ; 166(8): 785-793, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32579098

RESUMEN

Salmonella enterica subspecies enterica serovar Typhimurium (S. Typhimurium) definitive phage type 104 (DT104), S. enterica subspecies enterica serovar Worthington (S. Worthington) and S. bongori produce ArtA and ArtB (ArtAB) toxin homologues, which catalyse ADP-ribosylation of pertussis toxin-sensitive G protein. ArtAB gene (artAB) is encoded on prophage in DT104 and its expression is induced by mitomycin C (MTC) and hydrogen peroxide (H2O2) that trigger the bacterial SOS response. Although the genetic regulatory mechanism associated with artAB expression is not characterized, it is thought to be associated with prophage induction, which occurs when the RecA-mediated SOS response is triggered. Here we show that subinhibitory concentration of quinolone antibiotics that are SOS-inducing agents, also induce ArtAB production in these Salmonella strains. Both MTC and fluoroquinolone antibiotics such as enrofloxacin-induced artA and recA transcription and artAB-encoding prophage (ArtAB-prophage) in DT104 and S. Worthington. However, in S. bongori, which harbours artAB genes on incomplete prophage, artA transcription was induced by MTC and enrofloxacin, but prophage induction was not observed. Taken together, these results suggest that SOS response followed by induction of artAB transcription is essential for ArtAB production. H2O2-mediated induction of ArtAB prophage and efficient production of ArtAB was observed in DT104 but not in S. Worthington and S. bongori. Therefore, induction of artAB expression with H2O2 is strain-specific, and the mode of action of H2O2 as an SOS-inducing agent might be different from those of MTC and quinolone antibiotics.


Asunto(s)
ADP Ribosa Transferasas/genética , Antibacterianos/farmacología , Toxinas Bacterianas/genética , Respuesta SOS en Genética/efectos de los fármacos , Salmonella enterica/efectos de los fármacos , Salmonella/efectos de los fármacos , ADP Ribosa Transferasas/metabolismo , Toxinas Bacterianas/metabolismo , Peróxido de Hidrógeno/farmacología , Mitomicina/farmacología , Profagos/efectos de los fármacos , Profagos/genética , Quinolonas/farmacología , Rec A Recombinasas/genética , Respuesta SOS en Genética/genética , Salmonella/genética , Fagos de Salmonella/efectos de los fármacos , Fagos de Salmonella/genética , Salmonella enterica/genética , Especificidad de la Especie , Transcripción Genética/efectos de los fármacos
18.
J Bacteriol ; 202(2)2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31548275

RESUMEN

All organisms regulate cell cycle progression by coordinating cell division with DNA replication status. In eukaryotes, DNA damage or problems with replication fork progression induce the DNA damage response (DDR), causing cyclin-dependent kinases to remain active, preventing further cell cycle progression until replication and repair are complete. In bacteria, cell division is coordinated with chromosome segregation, preventing cell division ring formation over the nucleoid in a process termed nucleoid occlusion. In addition to nucleoid occlusion, bacteria induce the SOS response after replication forks encounter DNA damage or impediments that slow or block their progression. During SOS induction, Escherichia coli expresses a cytoplasmic protein, SulA, that inhibits cell division by directly binding FtsZ. After the SOS response is turned off, SulA is degraded by Lon protease, allowing for cell division to resume. Recently, it has become clear that SulA is restricted to bacteria closely related to E. coli and that most bacteria enforce the DNA damage checkpoint by expressing a small integral membrane protein. Resumption of cell division is then mediated by membrane-bound proteases that cleave the cell division inhibitor. Further, many bacterial cells have mechanisms to inhibit cell division that are regulated independently from the canonical LexA-mediated SOS response. In this review, we discuss several pathways used by bacteria to prevent cell division from occurring when genome instability is detected or before the chromosome has been fully replicated and segregated.


Asunto(s)
División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Replicación del ADN/efectos de los fármacos , Replicación del ADN/efectos de la radiación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bleomicina/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , División Celular/genética , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Replicación del ADN/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/efectos de la radiación , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteasa La/genética , Proteasa La/metabolismo , Radiación Ionizante , Respuesta SOS en Genética/efectos de los fármacos , Respuesta SOS en Genética/genética , Respuesta SOS en Genética/efectos de la radiación
19.
Int J Med Microbiol ; 310(1): 151361, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31640923

RESUMEN

Urinary tract infections are one of the most common bacterial infections and a major public health problem. The predominant causative agents are uropathogenic Escherichia coli. These strains differ from commensal E. coli by the presence of additional horizontally acquired chromosomal material, so-called pathogenicity islands, which encode traits that promote efficient bacterial colonization of the urinary tract. Uropathogenic model strain E. coli 536 possesses six archetypal pathogenicity islands. Bacteriophage-like integrases encoded by each pathogenicity island contribute to island instability. To learn more about the stability of these six islands and factors controlling their stability we constructed two chromosomal reporter systems for the measurement of island loss, as well as for the measurement of the promoter activity of the six island-associated integrase genes at the population level. We used these reporter gene modules to analyze the role of SOS response in island instability. Tests with subinhibitory concentrations of different antibiotics, including many drugs commonly used for the treatment of urinary tract infection, indicated that only SOS response-inducing antibiotics led to an increased loss of islands which was always associated with an increase in the bacterial subpopulations showing high integrase promoter activity. This suggests that island excision correlates with the expression of the cognate integrase. Our reporter modules are valuable tools to investigate the impact of various growth conditions on genome plasticity. Furthermore, a better understanding of the conditions, which affect bacterial integrase expression may open ways to specifically manipulate the genome content of bacterial pathogens by increasing pathogenicity island deletion rates in infecting or colonizing bacteria, thus leading to the attenuation of bacterial pathogens.


Asunto(s)
Antibacterianos/farmacología , Genoma Bacteriano , Islas Genómicas/genética , Integrasas/genética , Respuesta SOS en Genética/efectos de los fármacos , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/genética , Humanos , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/enzimología
20.
Life Sci ; 241: 117116, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31790690

RESUMEN

AIMS: LexA protein is a transcriptional repressor which regulates the expression of more than 60 genes belonging to the SOS global regulatory network activated by damages to bacterial DNA. Considering its role in bacteria, LexA represents a key target to counteract bacterial resistance: the possibility to modulate SOS response through the inhibition of LexA autoproteolysis may lead to reduced drug susceptibility and acquisition of resistance in bacteria. In our study we investigated boron-containing compounds as potential inhibitors of LexA self-cleavage. MAIN METHODS: The inhibition of LexA self-cleavage was evaluated by following the variation of the first-order rate constant by LC-MS at several concentrations of inhibitors. In silico analysis was applied to predict the binding orientations assumed by the inhibitors in the protein active site, upon covalent binding to the catalytic Ser-119. Bacterial filamentation assay was used to confirm the ability of (3-aminophenyl)boronic acid to interfere with SOS induced activation. KEY FINDINGS: Boron-containing compounds act as inhibitors of LexA self-cleavage, as also confirmed by molecular modelling where the compounds interact with the catalytic Ser-119, via the formation of an acyl-enzyme intermediate. A new equation for the description of the inhibition potency in an autoproteolytic enzyme is also disclosed. Bacterial filamentation assays strongly support the interference of our compounds with the SOS response activation through inhibition of septum formation. SIGNIFICANCE: The obtained results demonstrated that phenylboronic compounds could be exploited in a hit-to-lead optimization process toward effective LexA self-cleavage inhibitors. They would sustain the rehabilitation in therapy of several dismissed antibiotics.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Compuestos de Boro/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Antibacterianos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Compuestos de Boro/química , Farmacorresistencia Bacteriana/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Levofloxacino/farmacología , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Respuesta SOS en Genética/efectos de los fármacos , Serina Endopeptidasas/química , Serina Endopeptidasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...