Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Microbiol Spectr ; 10(6): e0304322, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36374016

ABSTRACT

Heterogeneous environments such as the chronically infected cystic fibrosis lung drive the diversification of Pseudomonas aeruginosa populations into, e.g., mucoid, alginate-overproducing isolates or small-colony variants (SCVs). In this study, we performed extensive genome and transcriptome profiling on a clinical SCV isolate that exhibited high cyclic diguanylate (c-di-GMP) levels and a mucoid phenotype. We observed a delayed, stepwise decrease of the high levels of c-di-GMP as well as alginate gene expression upon passaging the SCV under noninducing, rich medium growth conditions over 7 days. Upon prolonged passaging, this lagging reduction of the high c-di-GMP levels under noninducing planktonic conditions (reminiscent of a hysteretic response) was followed by a phenotypic switch to a large-colony morphology, which could be linked to mutations in the Gac/Rsm signaling pathway. Complementation of the Gac/Rsm signaling-negative large-colony variants with a functional GacSA system restored the SCV colony morphotype but was not able to restore the high c-di-GMP levels of the SCV. Our data thus suggest that expression of the SCV colony morphotype and modulation of c-di-GMP levels are genetically separable and follow different evolutionary paths. The delayed switching of c-di-GMP levels in response to fluctuating environmental conditions might provide a unique opportunity to include a time dimension to close the gap between short-term phenotypic and long-term genetic adaptation to biofilm-associated growth conditions. IMPORTANCE Extreme environments, such as those encountered during an infection process in the human host, make effective bacterial adaptation inevitable. While bacteria adapt individually by activating stress responses, long-term adaptation of bacterial communities to challenging conditions can be achieved via genetic fixation of favorable traits. In this study, we describe a two-pronged bacterial stress resistance strategy in the opportunistic pathogen Pseudomonas aeruginosa. We show that the production of adjusted elevated c-di-GMP levels, which drive protected biofilm-associated phenotypes in vivo, resembles a stable hysteretic response which prevents unwanted frequent switching. Cellular hysteresis might provide a link between individual adaptability and evolutionary adaptation to ensure the evolutionary persistence of host-adapted stress response strategies.


Subject(s)
Bacterial Proteins , Pseudomonas aeruginosa , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Pseudomonas aeruginosa/metabolism , Cyclic GMP/metabolism , Biofilms , Signal Transduction/physiology , Alginates/metabolism , Gene Expression Regulation, Bacterial
2.
Adv Exp Med Biol ; 1386: 303-323, 2022.
Article in English | MEDLINE | ID: mdl-36258077

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that causes life-devastating acute as well as chronic biofilm-associated infections with limited treatment options. Its success is largely due to its remarkable adaptability. P. aeruginosa uses different long- and short-term adaptive mechanisms to increase its fitness, both at the population level through genetic diversification and at the individual cell level by adapting gene expression. These adapted gene expression profiles can be fixed by the accumulation of patho-adaptive mutations. The latter are often found in transcriptional regulators and lead to rewiring of the regulatory network to promote survival at the infected host site. In this chapter, we review recent developments in transcriptional profiling and explain how these provide new insights into the establishment and maintenance of P. aeruginosa infections. We illustrate what can be learned from the application of advanced RNA-seq technology, such as ex vivo RNA-seq, host-pathogen crosstalk (dual RNA-seq), or recording of transcriptional heterogeneity within a bacterial population (single-cell RNA-seq). In addition, we discuss how large transcriptome datasets from a variety of clinical isolates can be used to gain an expanded understanding of bacterial adaptation during the infection process. Global genotype-phenotype correlation studies provide a unique opportunity to discover new evolutionary pathways of infection-related phenotypes and led to the discovery of different strategies of the pathogen P. aeruginosa to build a biofilm. Insights gained from large-scale, multi-layered functional -omics approaches will continue to contribute to a more comprehensive understanding of P. aeruginosa adaptation to the host habitat and promises to pave the way for novel strategies to combat recalcitrant infections.


Subject(s)
Pseudomonas Infections , Humans , Pseudomonas Infections/genetics , Pseudomonas Infections/complications , Pseudomonas Infections/microbiology , Gene Expression Profiling , Pseudomonas aeruginosa/genetics , Biofilms , Phenotype
3.
Front Cell Infect Microbiol ; 12: 851784, 2022.
Article in English | MEDLINE | ID: mdl-35295755

ABSTRACT

Biofilm-associated bacteria exhibit profound changes in bacterial physiology. They thrive in the environment but also in the human host in protected sessile communities. Antimicrobial therapy usually fails, despite the absence of genotypic resistance, and it is commonly accepted that biofilm-grown bacteria are up to 1,000-fold more resistant than planktonic cells. We are only at the beginning to understand the reasons for biofilm recalcitrance, and systematic approaches to describe biofilm-induced tolerance phenotypes are lacking. In this study, we investigated a large and highly diverse collection of 352 clinical Pseudomonas aeruginosa isolates for their antimicrobial susceptibility profiles under biofilm growth conditions towards the antibiotics ciprofloxacin, tobramycin, and colistin. We discovered characteristic patterns of drug-specific killing activity and detected conditional tolerance levels far lower (in the range of the minimal inhibitory concentration (MIC)), but also far higher (up to 16,000-fold increase compared to planktonic cells) than generally believed. This extremely broad distribution of biofilm-induced tolerance phenotypes across the clinical isolates was greatly influenced by the choice of the antibiotic. We furthermore describe cross-tolerance against ciprofloxacin and tobramycin, but not colistin, and observed an additive activity between biofilm-induced tolerance and genetically determined resistance. This became less evident when the biofilm-grown cells were exposed to very high antibiotic concentrations. Although much more remains to be learned on the molecular mechanisms underlying biofilm-induced tolerance, our data on intra-species variations in tolerance profiles provide valuable new insights. Furthermore, our observation that colistin appears to act independently of the tolerance mechanisms of individual clinical strains could make colistin a valuable therapeutic option in chronic biofilm-associated infections characterized by the presence of particularly tolerant strains.


Subject(s)
Pseudomonas Infections , Pseudomonas aeruginosa , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms , Humans , Microbial Sensitivity Tests , Plankton , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Tobramycin/pharmacology
4.
NPJ Biofilms Microbiomes ; 8(1): 6, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35165270

ABSTRACT

The overall success of a pathogenic microbe depends on its ability to efficiently adapt to challenging conditions in the human host. Long-term evolution experiments track and predict adaptive trajectories and have contributed significantly to our understanding of the driving forces of bacterial adaptation. In this study, we conducted a cross-sectional study instead of long-term longitudinal evolution experiments. We analyzed the transcriptional profiles as well as genomic sequence variations of a large number of clinical Pseudomonas aeruginosa isolates that have been recovered from different infected human sites. Convergent changes in gene expression patterns were found in different groups of clinical isolates. The majority of repeatedly observed expression patterns could be attributed to a defective lasR gene, which encodes the major quorum-sensing regulator LasR. Strikingly, the gene expression pattern of the lasR-defective strains appeared to reflect a transcriptional response that evolves in a direction consistent with growth within a biofilm. In a process of genetic assimilation, lasR-deficient P. aeruginosa isolates appear to constitutively express a biofilm-adapted transcriptional profile and no longer require a respective environmental trigger. Our results demonstrate that profiling the functional consequences of pathoadaptive mutations in clinical isolates reveals long-term evolutionary pathways and may explain the success of lasR mutants in the opportunistic pathogen P. aeruginosa in a clinical context.


Subject(s)
Pseudomonas aeruginosa , Transcriptome , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms , Cross-Sectional Studies , Humans , Pseudomonas aeruginosa/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
6.
Nat Microbiol ; 6(2): 151-156, 2021 02.
Article in English | MEDLINE | ID: mdl-33398098

ABSTRACT

Biofilms are microbial communities that represent a highly abundant form of microbial life on Earth. Inside biofilms, phenotypic and genotypic variations occur in three-dimensional space and time; microscopy and quantitative image analysis are therefore crucial for elucidating their functions. Here, we present BiofilmQ-a comprehensive image cytometry software tool for the automated and high-throughput quantification, analysis and visualization of numerous biofilm-internal and whole-biofilm properties in three-dimensional space and time.


Subject(s)
Biofilms , Image Cytometry/methods , Imaging, Three-Dimensional/methods , Microbiota , Software , Bacteria/cytology , Bacteria/genetics , Bacteria/growth & development , Spatio-Temporal Analysis
7.
Article in English | MEDLINE | ID: mdl-31934344

ABSTRACT

Studying parallel evolution of similar traits in independent within-species lineages provides an opportunity to address evolutionary predictability of molecular changes underlying adaptation. In this study, we monitored biofilm forming capabilities, motility, and virulence phenotypes of a plethora of phylogenetically diverse clinical isolates of the opportunistic pathogen Pseudomonas aeruginosa. We also recorded biofilm-specific and planktonic transcriptional responses. We found that P. aeruginosa isolates could be stratified based on the production of distinct organismal traits. Three major biofilm phenotypes, which shared motility and virulence phenotypes, were produced repeatedly in several isolates, indicating that the phenotypes evolved via parallel or convergent evolution. Of note, while we found a restricted general response to the biofilm environment, the individual groups of biofilm phenotypes reproduced biofilm transcriptional profiles that included the expression of well-known biofilm features, such as surface adhesive structures and extracellular matrix components. Our results provide insights into distinct ways to make a biofilm and indicate that genetic adaptations can modulate multiple pathways for biofilm development that are followed by several independent clinical isolates. Uncovering core regulatory pathways that drive biofilm-associated growth and tolerance towards environmental stressors promises to give clues to host and environmental interactions and could provide useful targets for new clinical interventions.


Subject(s)
Biofilms/growth & development , Gene Expression Profiling/methods , Plankton/microbiology , Plant Proteins/genetics , Pseudomonas aeruginosa/physiology , A549 Cells , Bacterial Adhesion , Evolution, Molecular , Gene Expression Regulation, Bacterial , Humans , Phenotype , Phylogeny , Sequence Analysis, RNA , Virulence
8.
Cells ; 8(10)2019 09 23.
Article in English | MEDLINE | ID: mdl-31547513

ABSTRACT

Comparative genomics has greatly facilitated the identification of shared as well as unique features among individual cells or tissues, and thus offers the potential to find disease markers. While proteomics is recognized for its potential to generate quantitative maps of protein expression, comparative proteomics in bacteria has been largely restricted to the comparison of single cell lines or mutant strains. In this study, we used a data independent acquisition (DIA) technique, which enables global protein quantification of large sample cohorts, to record the proteome profiles of overall 27 whole genome sequenced and transcriptionally profiled clinical isolates of the opportunistic pathogen Pseudomonas aeruginosa. Analysis of the proteome profiles across the 27 clinical isolates grown under planktonic and biofilm growth conditions led to the identification of a core biofilm-associated protein profile. Furthermore, we found that protein-to-mRNA ratios between different P. aeruginosa strains are well correlated, indicating conserved patterns of post-transcriptional regulation. Uncovering core regulatory pathways, which drive biofilm formation and associated antibiotic tolerance in bacterial pathogens, promise to give clues to interactions between bacterial species and their environment and could provide useful targets for new clinical interventions to combat biofilm-associated infections.


Subject(s)
Bacterial Proteins/analysis , Biofilms/growth & development , Proteome/analysis , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Genomics , Mass Spectrometry/methods , Microbial Sensitivity Tests , Proteomics/methods , Pseudomonas aeruginosa/chemistry , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...