Abstract

Pseudomonas aeruginosa is an opportunistic pathogen that thrives in diverse environments and causes a variety of human infections. Pseudomonas aeruginosa AG1 (PaeAG1) is a high-risk sequence type 111 (ST-111) strain isolated from a Costa Rican hospital in 2010. PaeAG1 has both blaVIM-2 and blaIMP-18 genes encoding for metallo-β-lactamases, and it is resistant to β-lactams (including carbapenems), aminoglycosides, and fluoroquinolones. Ciprofloxacin (CIP) is an antibiotic commonly used to treat P. aeruginosa infections, and it is known to produce DNA damage, triggering a complex molecular response. In order to evaluate the effects of a sub-inhibitory CIP concentration on PaeAG1, growth curves using increasing CIP concentrations were compared. We then measured gene expression using RNA-Seq at three time points (0, 2.5 and 5 h) after CIP exposure to identify the transcriptomic determinants of the response (i.e. hub genes, gene clusters and enriched pathways). Changes in expression were determined using differential expression analysis and network analysis using a top–down systems biology approach. A hybrid model using database-based and co-expression analysis approaches was implemented to predict gene–gene interactions. We observed a reduction of the growth curve rate as the sub-inhibitory CIP concentrations were increased. In the transcriptomic analysis, we detected that over time CIP treatment resulted in the differential expression of 518 genes, showing a complex impact at the molecular level. The transcriptomic determinants were 14 hub genes, multiple gene clusters at different levels (associated to hub genes or as co-expression modules) and 15 enriched pathways. Down-regulation of genes implicated in several metabolism pathways, virulence elements and ribosomal activity was observed. In contrast, amino acid catabolism, RpoS factor, proteases, and phenazines genes were up-regulated. Remarkably, > 80 resident-phage genes were up-regulated after CIP treatment, which was validated at phenomic level using a phage plaque assay. Thus, reduction of the growth curve rate and increasing phage induction was evidenced as the CIP concentrations were increased. In summary, transcriptomic and network analyses, as well as the growth curves and phage plaque assays provide evidence that PaeAG1 presents a complex, concentration-dependent response to sub-inhibitory CIP exposure, showing pleiotropic effects at the systems level. Manipulation of these determinants, such as phage genes, could be used to gain more insights about the regulation of responses in PaeAG1 as well as the identification of possible therapeutic targets. To our knowledge, this is the first report of the transcriptomic analysis of CIP response in a ST-111 high-risk P. aeruginosa strain, in particular using a top-down systems biology approach.

Highlights

  • More insights about the regulation of responses in Pseudomonas aeruginosa AG1 (PaeAG1) as well as the identification of possible therapeutic targets

  • Pseudomonas aeruginosa AG1 (PaeAG1) is a multiresistant high-risk sequence type 111 (ST-111) strain (GenBank CP045739)[5]. It was isolated from a Costa Rican hospital and it was the first report of an isolate of P. aeruginosa carrying both blaVIM-2 and blaIMP-18 genes encoding for metallo-β-lactamases enzymes, located in two independent i­ntegrons[5,6]

  • For the case of 50.0 μg/mL, the growth was drastically impaired and no exponential growth was observed. These results indicate that higher CIP concentrations have a stronger effect on the growth rate, even for sub-inhibitory concentrations ­(MICCiprofloxacin 32 μg/mL)

Read more

Summary

Introduction

More insights about the regulation of responses in PaeAG1 as well as the identification of possible therapeutic targets. PaeAG1 is resistant to β-lactams (including carbapenems), aminoglycosides, and fluoroquinolones, being only sensitive to colistin In addition to this multidrug-resistant feature, as in other P. aeruginosa strains, the ability to colonize nosocomial environments makes this strain a high-risk c­ lone[7]. Antibiotic resistance is a major threat to public health because it compromises the administration of appropriate antibiotic therapy, and reduces the therapeutic options to treat infections, increasing patient morbidity and ­mortality[9,10] This situation is aggravated by the emergence of strains resistant to multiple a­ ntibiotics[11], limitation of the knowledge of interactions with pathogens and mechanisms of action of antimicrobial agents, and development of new a­ ntibiotics[12]. Regulatory networks of transcriptional responses to DNA damage involves DNA repair enzymes, and diverse proteins with roles in cell division, metabolism modulation, genetic rearrangements and exchange, mutation, and virulence factor ­production[19]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call