1
|
Stine W, Akiyama T, Weiss D, Kim M. Lineage-dependent variations in single-cell antibiotic susceptibility reveal the selective inheritance of phenotypic resistance in bacteria. Nat Commun 2025; 16:4655. [PMID: 40389422 PMCID: PMC12089280 DOI: 10.1038/s41467-025-59807-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/06/2025] [Indexed: 05/21/2025] Open
Abstract
Genetically identical bacterial cells often exhibit heterogeneous responses to antibiotics - some survive, others die. Here, we show that this heterogeneity propagates across generations to give rise to phenotypic resistance. Using real-time single-cell tracking, we exposed Escherichia coli to the β-lactam cefsulodin at its clinical breakpoint concentration and analyzed cell fate within genealogical trees statistically. Cell survival was strongly correlated among family members, driving the selective enrichment of robust lineages within an otherwise susceptible population. Our genealogical population model identified heritable phenotypic resistance as a key factor underlying this enrichment, which was validated experimentally. Comparing enrichment dynamics between the wild-type and a tolC knock-out strain, deficient in multidrug efflux, uncovered nuanced changes that increased the intergenerational memory of phenotypic resistance. Our findings provide evidence for heritable phenotypic resistance and demonstrate how its propagation through cell-to-cell heterogeneity enables the survival of minority cells within isogenic populations.
Collapse
Affiliation(s)
- Wesley Stine
- Department of Physics, Emory University, Atlanta, GA, USA
| | - Tatsuya Akiyama
- Department of Physics, Emory University, Atlanta, GA, USA
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, USA
| | - David Weiss
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, USA
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
- Antibiotic Research Center, Emory University, Atlanta, GA, USA
| | - Minsu Kim
- Department of Physics, Emory University, Atlanta, GA, USA.
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, USA.
- Antibiotic Research Center, Emory University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Olayé J, Bouzidi H, Aristov A, Barizien A, Gutiérrez Ramos S, Baroud C, Bansaye V. Estimation of the lifetime distribution from fluctuations in Bellman-Harris processes. J Math Biol 2025; 90:56. [PMID: 40327121 DOI: 10.1007/s00285-025-02219-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 05/07/2025]
Abstract
The growth of populations without interactions can often be modeled by branching processes where each individual evolves independently and with the same law. In Bellman-Harris processes, each individual lives a random time and is then replaced by a random number of offspring. We are interested in the estimation of the parameters of this model. Our motivation comes from the estimation of cell division time and we focus on Gamma distribution for lifetime and binary reproduction. The mean of the lifetime is closely related to the growth rate of the population. Going farther and describing lifetime variability from fixed time observations is a challenging task, due to the complexity of the fluctuations of non-Markovian branching processes. Using fine results on these fluctuations, we describe two time-asymptotic regimes and explain how to discriminate between them and estimate the parameters. Then, we consider simulations and biological data to validate and discuss our method. It allows to determine single-cell parameters from time-resolved measurements of populations without the need to track each individual or to know the details of the initial condition. The results can be extended to more general branching processes.
Collapse
Affiliation(s)
- Jules Olayé
- CMAP, INRIA, École Polytechnique, Institut Polytechnique de Paris, 91120, Palaiseau, France.
| | - Hala Bouzidi
- ENSTA Paris, Institut Polytechnique de Paris, 91120, Palaiseau, France
| | - Andrey Aristov
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, 75015, Paris, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91120, Palaiseau, France
| | - Antoine Barizien
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, 75015, Paris, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91120, Palaiseau, France
| | - Salomé Gutiérrez Ramos
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, 75015, Paris, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91120, Palaiseau, France
| | - Charles Baroud
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, 75015, Paris, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91120, Palaiseau, France
| | - Vincent Bansaye
- CMAP, INRIA, École Polytechnique, Institut Polytechnique de Paris, 91120, Palaiseau, France
| |
Collapse
|
3
|
Turbant F, Lewandowska N, Bloch S, Wien F, Chauvet H, Węgrzyn G, Arluison V. Hfq influences ciprofloxacin accumulation in Escherichia coli independently of ompC and ompF post-transcriptional regulation. J Appl Genet 2025; 66:449-457. [PMID: 39920540 DOI: 10.1007/s13353-025-00945-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/09/2025]
Abstract
The antibiotic resistance of pathogenic bacteria is currently one of the major problems in medicine, and finding novel antibacterial agents is one of the most difficult tasks in the field of biomedical sciences. Studies on such tasks can be successful only if genetic and molecular mechanisms leading to antibiotic resistance/sensitivity are understood. Previous reports indicated that the bacterial protein Hfq, discovered as an RNA chaperone but subsequently demonstrated to play also other functions in cells, is involved in the mechanisms of the response of bacterial cells to antibiotics. Recently, it was found that Hfq dysfunction resulted in more effective accumulation of an antibiotic ciprofloxacin in Escherichia coli cells irrespective of the presence or absence of the AcrB efflux pump. However, small RNA-mediated impairment of expression of the ompF gene, which encodes a porin involved in antibiotics influx, reversed the effects of the absence of Hfq on the antibiotic accumulation. This led to the hypothesis that Hfq might influence ciprofloxacin accumulation in the manner independent on its RNA chaperone function, as this protein might also influence cellular membrane structure and functions. Here, we demonstrate that in ompC and ompF mutants of E. coli, accumulation of ciprofloxacin is significantly impaired in the absence of Hfq or its C-terminal domain. These results corroborate the above-mentioned hypothesis on a sRNA-independent mechanism of Hfq-mediated modulation of the antibiotic transmembrane transport. Since fluoroquinolones use both protein- and lipid-mediated pathways to cross the outer membrane, Hfq may influence both processes. This possibility will be discussed herein.
Collapse
Affiliation(s)
- Florian Turbant
- Synchrotron SOLEIL, L Orme Des Merisiers, Saint Aubin BP48, 91192, Gif-Sur-Yvette, France
- Laboratoire Léon Brillouin LLB, UMR12, CEA CNRS, CEA Saclay, 91191, Gif-Sur-Yvette, France
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Natalia Lewandowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Sylwia Bloch
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Frank Wien
- Synchrotron SOLEIL, L Orme Des Merisiers, Saint Aubin BP48, 91192, Gif-Sur-Yvette, France
| | - Hugo Chauvet
- Synchrotron SOLEIL, L Orme Des Merisiers, Saint Aubin BP48, 91192, Gif-Sur-Yvette, France
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland.
| | - Véronique Arluison
- Laboratoire Léon Brillouin LLB, UMR12, CEA CNRS, CEA Saclay, 91191, Gif-Sur-Yvette, France.
- UFR SDV, Université Paris Cité, 75013, Paris, France.
| |
Collapse
|
4
|
Fruet C, Müller EL, Loverdo C, Bitbol AF. Spatial structure facilitates evolutionary rescue by drug resistance. PLoS Comput Biol 2025; 21:e1012861. [PMID: 40179127 PMCID: PMC11967957 DOI: 10.1371/journal.pcbi.1012861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/09/2025] [Indexed: 04/05/2025] Open
Abstract
Bacterial populations often have complex spatial structures, which can impact their evolution. Here, we study how spatial structure affects the evolution of antibiotic resistance in a bacterial population. We consider a minimal model of spatially structured populations where all demes (i.e., subpopulations) are identical and connected to each other by identical migration rates. We show that spatial structure can facilitate the survival of a bacterial population to antibiotic treatment, starting from a sensitive inoculum. Specifically, the bacterial population can be rescued if antibiotic resistant mutants appear and are present when drug is added, and spatial structure can impact the fate of these mutants and the probability that they are present. Indeed, the probability of fixation of neutral or deleterious mutations providing drug resistance is increased in smaller populations. This promotes local fixation of resistant mutants in the structured population, which facilitates evolutionary rescue by drug resistance in the rare mutation regime. Once the population is rescued by resistance, migrations allow resistant mutants to spread in all demes. Our main result that spatial structure facilitates evolutionary rescue by antibiotic resistance extends to more complex spatial structures, and to the case where there are resistant mutants in the inoculum.
Collapse
Affiliation(s)
- Cecilia Fruet
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| | - Ella Linxia Müller
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| | - Claude Loverdo
- Sorbonne Université, CNRS,Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), Paris,France
| | - Anne-Florence Bitbol
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
5
|
Hu Z, Wood KB. Deciphering population-level response under spatial drug heterogeneity on microhabitat structures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638200. [PMID: 40027692 PMCID: PMC11870443 DOI: 10.1101/2025.02.13.638200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Bacteria and cancer cells live in a spatially heterogeneous environment, where migration shapes the microhabitat structures critical for colonization and metastasis. The interplay between growth, migration, and microhabitat structure complicates the prediction of population responses to drugs, such as clearance or sustained growth, posing a longstanding challenge. Here, we disentangle growth-migration dynamics and identify that population decline is determined by two decoupled terms: a spatial growth variation term and a microhabitat structure term. Notably, the microhabitat structure term can be interpreted as a dynamic-related centrality measure. For fixed spatial drug arrangements, we show that interpreting these centralities reveals how different network structures, even with identical edge densities, microhabitat numbers, and spatial heterogeneity, can lead to distinct population-level responses. Increasing edge density shifts the population response from growth to clearance, supporting an inversed centrality-connectivity relationship, and mirroring the effects of higher migration rates. Furthermore, we derive a sufficient condition for robust population decline across various spatial growth rate arrangements, regardless of spatial-temporal fluctuations induced by drugs. Additionally, we demonstrate that varying the maximum growth-to-death ratio, determined by drug-bacteria interactions, can lead to distinct population decline profiles and a minimal decline phase emerges. These findings address key challenges in predicting population-level responses and provide insights into divergent clinical outcomes under identical drug dosages. This work may offer a new method of interpreting treatment dynamics and potential approaches for optimizing spatially explicit drug dosing strategies.
Collapse
|
6
|
Bloch S, Węgrzyn G, Arluison V. The Role of the Hfq Protein in Bacterial Resistance to Antibiotics: A Narrative Review. Microorganisms 2025; 13:364. [PMID: 40005731 PMCID: PMC11858733 DOI: 10.3390/microorganisms13020364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The antibiotic resistance of pathogenic microorganisms is currently one of most major medical problems, causing a few million deaths every year worldwide due to untreatable bacterial infections. Unfortunately, the prognosis is even worse, as over 8 million deaths associated with antibiotic resistance are expected to occur in 2050 if no new effective antibacterial treatments are discovered. The Hfq protein has been discovered as a bacterial RNA chaperone. However, subsequent studies have indicated that this small protein (composed of 102 amino acid residues in Escherichia coli) has more activities, including binding to DNA and influencing its compaction, interaction with biological membranes, formation of amyloid-like structures, and others. Although Hfq is known to participate in many cellular processes, perhaps surprisingly, only reports from recent years have demonstrated its role in bacterial antibiotic resistance. The aim of this narrative review is to discuss how can Hfq affects antibiotic resistance in bacteria and propose how this knowledge may facilitate developing new therapeutic strategies against pathogenic bacteria. We indicate that the mechanisms by which the Hfq protein modulates the response of bacterial cells to antibiotics are quite different, from the regulation of the expression of genes coding for proteins directly involved in antibiotic transportation or action, through direct effects on membranes, to controlling the replication or transposition of mobile genetic elements bearing antibiotic resistance genes. Therefore, we suggest that Hfq could be considered a potential target for novel antimicrobial compounds. We also discuss difficulties in developing such drugs, but since Hfq appears to be a promising target for drugs that may enhance the efficacy of antibiotics, we propose that works on such potential therapeutics are encouraged.
Collapse
Affiliation(s)
- Sylwia Bloch
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland;
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland;
| | - Véronique Arluison
- Laboratoire Léon Brillouin, UMR 12 CEA/CNRS, Bâtiment 563, Site de Saclay, 91191 Gif-sur-Yvette, France
- UFR Science Du Vivant, Université Paris Cité, 35 Rue Hélène Brion, 75013 Paris, France
| |
Collapse
|
7
|
Verdon N, Popescu O, Titmuss S, Allen RJ. Habitat fragmentation enhances microbial collective defence. J R Soc Interface 2025; 22:20240611. [PMID: 39933594 PMCID: PMC11813583 DOI: 10.1098/rsif.2024.0611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/22/2024] [Accepted: 12/19/2024] [Indexed: 02/13/2025] Open
Abstract
Microbes often inhabit complex, spatially partitioned environments such as host tissue or soil, but the effects of habitat fragmentation on microbial ecology and infection dynamics are poorly understood. Here, we investigate how habitat fragmentation impacts a prevalent microbial collective defence mechanism: enzymatic degradation of an environmental toxin. Using a theoretical model, we predict that habitat fragmentation can strongly enhance the collective benefits of enzymatic toxin degradation. For the example of [Formula: see text]-lactamase-producing bacteria that mount a collective defence by degrading a [Formula: see text]-lactam antibiotic, we find that realistic levels of habitat fragmentation can allow a population to survive antibiotic doses that greatly exceed those required to kill a non-fragmented population. This 'habitat-fragmentation rescue' is a stochastic effect that originates from variation in bacterial density among different subpopulations and demographic noise. We also study the contrasting case of collective enzymatic foraging, where enzyme activity releases nutrients from the environment; here we find that increasing habitat fragmentation decreases the lag time for population growth but does not change the ecological outcome. Taken together, this work predicts that stochastic effects arising from habitat fragmentation can greatly enhance the effectiveness of microbial collective defence via enzymatic toxin degradation.
Collapse
Affiliation(s)
- Nia Verdon
- Theoretical Microbial Ecology, Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
| | - Ofelia Popescu
- School of Physics and Astronomy, University of Edinburgh, James Clerk Maxwell Building, Peter Guthrie Tait Road, EdinburghEH9 3FD, UK
| | - Simon Titmuss
- School of Physics and Astronomy, University of Edinburgh, James Clerk Maxwell Building, Peter Guthrie Tait Road, EdinburghEH9 3FD, UK
| | - Rosalind J. Allen
- Theoretical Microbial Ecology, Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
- School of Physics and Astronomy, University of Edinburgh, James Clerk Maxwell Building, Peter Guthrie Tait Road, EdinburghEH9 3FD, UK
| |
Collapse
|
8
|
Parab L, Romeyer Dherbey J, Rivera N, Schwarz M, Gallie J, Bertels F. Chloramphenicol and gentamicin reduce the evolution of resistance to phage ΦX174 by suppressing a subset of E. coli LPS mutants. PLoS Biol 2025; 23:e3002952. [PMID: 39841243 PMCID: PMC11753469 DOI: 10.1371/journal.pbio.3002952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 01/23/2025] Open
Abstract
Bacteriophages infect gram-negative bacteria by attaching to molecules present on the bacterial surface, often lipopolysaccharides (LPS). Modification of LPS can lead to resistance to phage infection. In addition, LPS modifications can impact antibiotic susceptibility, allowing for phage-antibiotic synergism. The evolutionary mechanism(s) behind such synergistic interactions remain largely unclear. Here, we show that the presence of antibiotics can affect the evolution of resistance to phage infection, using phage ΦX174 and Escherichia coli C. We use a collection of 34 E. coli C LPS strains, each of which is resistant to ΦX174, and has either a "rough" or "deep rough" LPS phenotype. Growth of the bacterial strains with the deep rough phenotype is inhibited at low concentrations of chloramphenicol and, to a much lesser degree, gentamicin. Treating E. coli C wild type with ΦX174 and chloramphenicol eliminates the emergence of mutants with the deep rough phenotype, and thereby slows the evolution of resistance to phage infection. At slightly lower chloramphenicol concentrations, phage resistance rates are similar to those observed at high concentrations; yet, we show that the diversity of possible mutants is much larger than at higher chloramphenicol concentrations. These data suggest that specific antibiotic concentrations can lead to synergistic phage-antibiotic interactions that disappear at higher antibiotic concentrations. Overall, we show that the change in survival of various ΦX174-resistant E. coli C mutants in the presence of antibiotics can explain the observed phage-antibiotic synergism.
Collapse
Affiliation(s)
- Lavisha Parab
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Jordan Romeyer Dherbey
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Norma Rivera
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Michael Schwarz
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Jenna Gallie
- Microbial Evolutionary Dynamics Group, Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Frederic Bertels
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| |
Collapse
|
9
|
Jacobs S, Boccarella G, van den Berg P, Van Dijck P, Carolus H. Unlocking the potential of experimental evolution to study drug resistance in pathogenic fungi. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:48. [PMID: 39843963 PMCID: PMC11721431 DOI: 10.1038/s44259-024-00064-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/15/2024] [Indexed: 01/24/2025]
Abstract
Exploring the dynamics and molecular mechanisms of antimicrobial drug resistance provides critical insights for developing effective strategies to combat it. This review highlights the potential of experimental evolution methods to study resistance in pathogenic fungi, drawing on insights from bacteriology and innovative approaches in mycology. We emphasize the versatility of experimental evolution in replicating clinical and environmental scenarios and propose that incorporating evolutionary modelling can enhance our understanding of antifungal resistance evolution. We advocate for a broader application of experimental evolution in medical mycology to improve our still limited understanding of drug resistance in fungi.
Collapse
Affiliation(s)
- Stef Jacobs
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Giorgio Boccarella
- Evolutionary Modelling Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Pieter van den Berg
- Evolutionary Modelling Group, Department of Biology, KU Leuven, Leuven, Belgium
- Evolutionary Modelling Group, Department of Microbial and Molecular Systems, KU Leuven, Leuven, Belgium
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium
- KU Leuven One Health Institute, KU Leuven, Leuven, Belgium
| | - Hans Carolus
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
10
|
Hu Z, Wu Y, Freire T, Gjini E, Wood K. Linking spatial drug heterogeneity to microbial growth dynamics in theory and experiment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624783. [PMID: 39605592 PMCID: PMC11601811 DOI: 10.1101/2024.11.21.624783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Diffusion and migration play pivotal roles in microbial communities - shaping, for example, colonization in new environments and the maintenance of spatial structures of biodiversity. While previous research has extensively studied free diffusion, such as range expansion, there remains a gap in understanding the effects of biologically or physically deleterious confined environments. In this study, we examine the interplay between migration and spatial drug heterogeneity within an experimental meta-community of E. faecalis, a Gram-positive opportunistic pathogen. When the community is confined to spatially-extended habitats ('islands') bordered by deleterious conditions, we find that the population level response depends on the trade-off between the growth rate within the island and the rate of transfer into regions with harsher conditions, a phenomenon we explore by modulating antibiotic concentration within the island. In heterogeneous islands, composed of spatially patterned patches that support varying levels of growth, the population's fate depends critically on the specific spatial arrangement of these patches - the same spatially averaged growth rate leads to diverging responses. These results are qualitatively captured by simple simulations, and analytical expressions which we derive using first-order perturbation approximations to reaction-diffusion models with explicit spatial dependence. Among all possible spatial arrangements, our theoretical and experimental findings reveal that the arrangement with the highest growth rates at the center most effectively mitigates population decline, while the arrangement with the lowest growth rates at the center is the least effective. Extending this approach to more complex experimental communities with varied spatial structures, such as a ring-structured community, further validates the impact of spatial drug arrangement. Our findings suggest new approaches to interpreting diverging clinical outcomes when applying identical drug doses and inform the possible optimization of spatially-explicit dosing strategies.
Collapse
Affiliation(s)
- Zhijian Hu
- Department of Biophysics, University of Michigan, Ann Arbor, USA
- Department of Mathematics, University of Michigan, Ann Arbor, USA
- Center for the Study of Complex Systems, University of Michigan, Ann Arbor, USA
| | - Yuzhen Wu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, USA
| | - Tomas Freire
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Erida Gjini
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Kevin Wood
- Department of Biophysics, University of Michigan, Ann Arbor, USA
- Center for the Study of Complex Systems, University of Michigan, Ann Arbor, USA
- Department of Physics, University of Michigan, Ann Arbor, USA
| |
Collapse
|
11
|
Tavaddod S, Dawson A, Allen RJ. Bacterial aggregation triggered by low-level antibiotic-mediated lysis. NPJ Biofilms Microbiomes 2024; 10:90. [PMID: 39327479 PMCID: PMC11427687 DOI: 10.1038/s41522-024-00553-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 08/20/2024] [Indexed: 09/28/2024] Open
Abstract
Suspended bacterial aggregates play a central role in ocean biogeochemistry, industrial processes and probably many clinical infections - yet the factors that trigger aggregation remain poorly understood, as does the relationship between suspended aggregates and surface-attached biofilms. Here we show that very low doses of cell-wall targeting antibiotic, far below the minimal inhibitory concentration, can trigger aggregation of Escherichia coli cells. This occurs when a few cells lyse, releasing extracellular DNA - thus, cell-to-cell variability in antibiotic response leads to population-level aggregation. Although lysis-triggered aggregation echoes known trigger mechanisms for surface-attached biofilms, these aggregates may have different ecological implications since they do not show increased biofilm-forming potential or increased antibiotic resistance. Our work contributes to understanding the nature of bacterial aggregates and the factors that trigger their formation, and the possible consequences of widespread low-dose antibiotic exposure in the environment and in the body.
Collapse
Affiliation(s)
- Sharareh Tavaddod
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Angela Dawson
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Rosalind J Allen
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK.
- Theoretical Microbial Ecology, Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
12
|
Pedreira A, Vázquez JA, Romanenko A, García MR. Design and Validation of a PLC-Controlled Morbidostat for Investigating Bacterial Drug Resistance. Bioengineering (Basel) 2024; 11:815. [PMID: 39199773 PMCID: PMC11351851 DOI: 10.3390/bioengineering11080815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
During adaptive laboratory evolution experiments, any unexpected interruption in data monitoring or control could lead to the loss of valuable experimental data and compromise the integrity of the entire experiment. Most homemade mini-bioreactors are built employing microcontrollers such as Arduino. Although affordable, these platforms lack the robustness of the programmable logic controller (PLC), which enhances the safety and robustness of the control process. Here, we describe the design and validation of a PLC-controlled morbidostat, an innovative automated continuous-culture mini-bioreactor specifically created to study the evolutionary pathways to drug resistance in microorganisms. This morbidostat includes several improvements, both at the hardware and software level, for better online monitoring and a more robust operation. The device was validated employing Escherichia coli, exploring its adaptive evolution in the presence of didecyldimethylammonium chloride (DDAC), a quaternary ammonium compound widely used for its antimicrobial properties. E. coli was subjected to increasing concentrations of DDAC over 3 days. Our results demonstrated a significant increase in DDAC susceptibility, with evolved populations exhibiting substantial changes in their growth after exposure.
Collapse
Affiliation(s)
- Adrián Pedreira
- Biosystems and Bioprocess Engineering Group (Bio2Eng), Spanish National Research Council (IIM-CSIC), Rúa Eduardo Cabello 6, 36208 Vigo, Spain;
- Group of Recycling and Valorization of Waste Materials (REVAL), Spanish National Research Council (IIM-CSIC), Rúa Eduardo Cabello 6, 36208 Vigo, Spain;
| | - José A. Vázquez
- Group of Recycling and Valorization of Waste Materials (REVAL), Spanish National Research Council (IIM-CSIC), Rúa Eduardo Cabello 6, 36208 Vigo, Spain;
| | | | - Míriam R. García
- Biosystems and Bioprocess Engineering Group (Bio2Eng), Spanish National Research Council (IIM-CSIC), Rúa Eduardo Cabello 6, 36208 Vigo, Spain;
| |
Collapse
|
13
|
Nyhoegen C, Bonhoeffer S, Uecker H. The many dimensions of combination therapy: How to combine antibiotics to limit resistance evolution. Evol Appl 2024; 17:e13764. [PMID: 39100751 PMCID: PMC11297101 DOI: 10.1111/eva.13764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/30/2024] [Accepted: 07/14/2024] [Indexed: 08/06/2024] Open
Abstract
In combination therapy, bacteria are challenged with two or more antibiotics simultaneously. Ideally, separate mutations are required to adapt to each of them, which is a priori expected to hinder the evolution of full resistance. Yet, the success of this strategy ultimately depends on how well the combination controls the growth of bacteria with and without resistance mutations. To design a combination treatment, we need to choose drugs and their doses and decide how many drugs get mixed. Which combinations are good? To answer this question, we set up a stochastic pharmacodynamic model and determine the probability to successfully eradicate a bacterial population. We consider bacteriostatic and two types of bactericidal drugs-those that kill independent of replication and those that kill during replication. To establish results for a null model, we consider non-interacting drugs and implement the two most common models for drug independence-Loewe additivity and Bliss independence. Our results show that combination therapy is almost always better in limiting the evolution of resistance than administering just one drug, even though we keep the total drug dose constant for a 'fair' comparison. Yet, exceptions exist for drugs with steep dose-response curves. Combining a bacteriostatic and a bactericidal drug which can kill non-replicating cells is particularly beneficial. Our results suggest that a 50:50 drug ratio-even if not always optimal-is usually a good and safe choice. Applying three or four drugs is beneficial for treatment of strains with large mutation rates but adding more drugs otherwise only provides a marginal benefit or even a disadvantage. By systematically addressing key elements of treatment design, our study provides a basis for future models which take further factors into account. It also highlights conceptual challenges with translating the traditional concepts of drug independence to the single-cell level.
Collapse
Affiliation(s)
- Christin Nyhoegen
- Research Group Stochastic Evolutionary Dynamics, Department of Theoretical BiologyMax Planck Institute for Evolutionary BiologyPlonGermany
| | - Sebastian Bonhoeffer
- Department of Environmental Systems Science, Institute of Integrative BiologyETH ZurichZurichSwitzerland
| | - Hildegard Uecker
- Research Group Stochastic Evolutionary Dynamics, Department of Theoretical BiologyMax Planck Institute for Evolutionary BiologyPlonGermany
| |
Collapse
|
14
|
Le Quellec L, Aristov A, Gutiérrez Ramos S, Amselem G, Bos J, Baharoglu Z, Mazel D, Baroud CN. Measuring single-cell susceptibility to antibiotics within monoclonal bacterial populations. PLoS One 2024; 19:e0303630. [PMID: 39088440 PMCID: PMC11293721 DOI: 10.1371/journal.pone.0303630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/30/2024] [Indexed: 08/03/2024] Open
Abstract
The emergence of new resistant bacterial strains is a worldwide challenge. A resistant bacterial population can emerge from a single cell that acquires resistance or persistence. Hence, new ways of tackling the mechanism of antibiotic response, such as single cell studies are required. It is necessary to see what happens at the single cell level, in order to understand what happens at the population level. To date, linking the heterogeneity of single-cell susceptibility to the population-scale response to antibiotics remains challenging due to the trade-offs between the resolution and the field of view. Here we present a platform that measures the ability of individual E. coli cells to form small colonies at different ciprofloxacin concentrations, by using anchored microfluidic drops and an image and data analysis pipelines. The microfluidic results are benchmarked against classical microbiology measurements of antibiotic susceptibility, showing an agreement between the pooled microfluidic chip and replated bulk measurements. Further, the experimental likelihood of a single cell to form a colony is used to provide a probabilistic antibiotic susceptibility curve. In addition to the probabilistic viewpoint, the microfluidic format enables the characterization of morphological features over time for a large number of individual cells. This pipeline can be used to compare the response of different bacterial strains to antibiotics with different action mechanisms.
Collapse
Affiliation(s)
- Lena Le Quellec
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, Paris, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Andrey Aristov
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, Paris, France
| | - Salomé Gutiérrez Ramos
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, Paris, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Gabriel Amselem
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, Paris, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Julia Bos
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Bacterial Genome Plasticity Unit, Paris, France
| | - Zeynep Baharoglu
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Bacterial Genome Plasticity Unit, Paris, France
| | - Didier Mazel
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Bacterial Genome Plasticity Unit, Paris, France
| | - Charles N. Baroud
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, Paris, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
15
|
Kratz JC, Banerjee S. Gene Expression Tradeoffs Determine Bacterial Survival and Adaptation to Antibiotic Stress. PRX LIFE 2024; 2:013010. [PMID: 39449977 PMCID: PMC11500821 DOI: 10.1103/prxlife.2.013010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
To optimize their fitness, cells face the crucial task of efficiently responding to various stresses. This necessitates striking a balance between conserving resources for survival and allocating resources for growth and division. The fundamental principles governing these tradeoffs is an outstanding challenge in the physics of living systems. In this study, we introduce a coarse-grained theoretical framework for bacterial physiology that establishes a connection between the physiological state of cells and their survival outcomes in dynamic environments, particularly in the context of antibiotic exposure. Predicting bacterial survival responses to varying antibiotic doses proves challenging due to the profound influence of the physiological state on critical parameters, such as the minimum inhibitory concentration (MIC) and killing rates, even within an isogenic cell population. Our proposed theoretical model bridges the gap by linking extracellular antibiotic concentration and nutrient quality to intracellular damage accumulation and gene expression. This framework allows us to predict and explain the control of cellular growth rate, death rate, MIC, and survival fraction in a wide range of time-varying environments. Surprisingly, our model reveals that cell death is rarely due to antibiotic levels being above the maximum physiological limit, but instead survival is limited by the inability to alter gene expression sufficiently quickly to transition to a less susceptible physiological state. Moreover, bacteria tend to overexpress stress response genes at the expense of reduced growth, conferring greater protection against further antibiotic exposure. This strategy is in contrast to those employed in different nutrient environments, in which bacteria allocate resources to maximize growth rate. This highlights an important tradeoff between the cellular capacity for growth and the ability to survive antibiotic exposure.
Collapse
Affiliation(s)
- Josiah C. Kratz
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| | - Shiladitya Banerjee
- Department of Physics, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| |
Collapse
|
16
|
Bren A, Glass DS, Kohanim YK, Mayo A, Alon U. Tradeoffs in bacterial physiology determine the efficiency of antibiotic killing. Proc Natl Acad Sci U S A 2023; 120:e2312651120. [PMID: 38096408 PMCID: PMC10742385 DOI: 10.1073/pnas.2312651120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Antibiotic effectiveness depends on a variety of factors. While many mechanistic details of antibiotic action are known, the connection between death rate and bacterial physiology is poorly understood. A common observation is that death rate in antibiotics rises linearly with growth rate; however, it remains unclear how other factors, such as environmental conditions and whole-cell physiological properties, affect bactericidal activity. To address this, we developed a high-throughput assay to precisely measure antibiotic-mediated death. We found that death rate is linear in growth rate, but the slope depends on environmental conditions. Growth under stress lowers death rate compared to nonstressed environments with similar growth rate. To understand stress's role, we developed a mathematical model of bacterial death based on resource allocation that includes a stress-response sector; we identify this sector using RNA-seq. Our model accurately predicts the minimal inhibitory concentration (MIC) with zero free parameters across a wide range of growth conditions. The model also quantitatively predicts death and MIC when sectors are experimentally modulated using cyclic adenosine monophosphate (cAMP), including protection from death at very low cAMP levels. The present study shows that different conditions with equal growth rate can have different death rates and establishes a quantitative relation between growth, death, and MIC that suggests approaches to improve antibiotic efficacy.
Collapse
Affiliation(s)
- Anat Bren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - David S. Glass
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Yael Korem Kohanim
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06520
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| |
Collapse
|
17
|
Lee S, Schefter BR, Taheri-Araghi S, Ha BY. Modeling selectivity of antimicrobial peptides: how it depends on the presence of host cells and cell density. RSC Adv 2023; 13:34167-34182. [PMID: 38020026 PMCID: PMC10663724 DOI: 10.1039/d3ra06030f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
Antimicrobial peptides (AMPs), naturally-occurring peptide antibiotics, are known to attack bacteria selectively over the host cells. The emergence of drug-resistant bacteria has spurred much effort in utilizing optimized (more selective) AMPs as new peptide antibiotics. Cell selectivity of these peptides depends on various factors or parameters such as their binding affinity for cell membranes, peptide trapping in cells, peptide coverages on cell membranes required for membrane rupture, and cell densities. In this work, using a biophysical model of peptide selectivity, we show this dependence quantitatively especially for a mixture of bacteria and host cells. The model suggests a rather nontrivial dependence of the selectivity on the presence of host cells, cell density, and peptide trapping. In a typical biological setting, peptide trapping works in favor of host cells; the selectivity increases with increasing host-cell density but decreases with bacterial cell density. Because of the cell-density dependence of peptide activity, the selectivity can be overestimated by two or three orders of magnitude. The model also clarifies how the cell selectivity of AMPs differs from their membrane selectivity.
Collapse
Affiliation(s)
- Suemin Lee
- Department of Physics and Astronomy, University of Waterloo Waterloo Ontario N2L 3G1 Canada
| | - Bethany R Schefter
- Department of Physics and Astronomy, University of Western Ontario London Ontario N6A 3K7 Canada
| | - Sattar Taheri-Araghi
- Department of Physics and Astronomy, California State University Northridge CA 91330 USA
| | - Bae-Yeun Ha
- Department of Physics and Astronomy, University of Waterloo Waterloo Ontario N2L 3G1 Canada
| |
Collapse
|
18
|
Hernández-Navarro L, Asker M, Rucklidge AM, Mobilia M. Coupled environmental and demographic fluctuations shape the evolution of cooperative antimicrobial resistance. J R Soc Interface 2023; 20:20230393. [PMID: 37907094 PMCID: PMC10618063 DOI: 10.1098/rsif.2023.0393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/06/2023] [Indexed: 11/02/2023] Open
Abstract
There is a pressing need to better understand how microbial populations respond to antimicrobial drugs, and to find mechanisms to possibly eradicate antimicrobial-resistant cells. The inactivation of antimicrobials by resistant microbes can often be viewed as a cooperative behaviour leading to the coexistence of resistant and sensitive cells in large populations and static environments. This picture is, however, greatly altered by the fluctuations arising in volatile environments, in which microbial communities commonly evolve. Here, we study the eco-evolutionary dynamics of a population consisting of an antimicrobial-resistant strain and microbes sensitive to antimicrobial drugs in a time-fluctuating environment, modelled by a carrying capacity randomly switching between states of abundance and scarcity. We assume that antimicrobial resistance (AMR) is a shared public good when the number of resistant cells exceeds a certain threshold. Eco-evolutionary dynamics is thus characterised by demographic noise (birth and death events) coupled to environmental fluctuations which can cause population bottlenecks. By combining analytical and computational means, we determine the environmental conditions for the long-lived coexistence and fixation of both strains, and characterise a fluctuation-driven AMR eradication mechanism, where resistant microbes experience bottlenecks leading to extinction. We also discuss the possible applications of our findings to laboratory-controlled experiments.
Collapse
Affiliation(s)
- Lluís Hernández-Navarro
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, UK
| | - Matthew Asker
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, UK
| | - Alastair M. Rucklidge
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, UK
| | - Mauro Mobilia
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
19
|
Alexander HK. Quantifying stochastic establishment of mutants in microbial adaptation. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001365. [PMID: 37561015 PMCID: PMC10482372 DOI: 10.1099/mic.0.001365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023]
Abstract
Studies of microbial evolution, especially in applied contexts, have focused on the role of selection in shaping predictable, adaptive responses to the environment. However, chance events - the appearance of novel genetic variants and their establishment, i.e. outgrowth from a single cell to a sizeable population - also play critical initiating roles in adaptation. Stochasticity in establishment has received little attention in microbiology, potentially due to lack of awareness as well as practical challenges in quantification. However, methods for high-replicate culturing, mutant labelling and detection, and statistical inference now make it feasible to experimentally quantify the establishment probability of specific adaptive genotypes. I review methods that have emerged over the past decade, including experimental design and mathematical formulas to estimate establishment probability from data. Quantifying establishment in further biological settings and comparing empirical estimates to theoretical predictions represent exciting future directions. More broadly, recognition that adaptive genotypes may be stochastically lost while rare is significant both for interpreting common lab assays and for designing interventions to promote or inhibit microbial evolution.
Collapse
Affiliation(s)
- Helen K. Alexander
- Institute of Ecology & Evolution, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
20
|
Czuppon P, Day T, Débarre F, Blanquart F. A stochastic analysis of the interplay between antibiotic dose, mode of action, and bacterial competition in the evolution of antibiotic resistance. PLoS Comput Biol 2023; 19:e1011364. [PMID: 37578976 PMCID: PMC10449190 DOI: 10.1371/journal.pcbi.1011364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 08/24/2023] [Accepted: 07/17/2023] [Indexed: 08/16/2023] Open
Abstract
The use of an antibiotic may lead to the emergence and spread of bacterial strains resistant to this antibiotic. Experimental and theoretical studies have investigated the drug dose that minimizes the risk of resistance evolution over the course of treatment of an individual, showing that the optimal dose will either be the highest or the lowest drug concentration possible to administer; however, no analytical results exist that help decide between these two extremes. To address this gap, we develop a stochastic mathematical model of bacterial dynamics under antibiotic treatment. We explore various scenarios of density regulation (bacterial density affects cell birth or death rates), and antibiotic modes of action (biostatic or biocidal). We derive analytical results for the survival probability of the resistant subpopulation until the end of treatment, the size of the resistant subpopulation at the end of treatment, the carriage time of the resistant subpopulation until it is replaced by a sensitive one after treatment, and we verify these results with stochastic simulations. We find that the scenario of density regulation and the drug mode of action are important determinants of the survival of a resistant subpopulation. Resistant cells survive best when bacterial competition reduces cell birth and under biocidal antibiotics. Compared to an analogous deterministic model, the population size reached by the resistant type is larger and carriage time is slightly reduced by stochastic loss of resistant cells. Moreover, we obtain an analytical prediction of the antibiotic concentration that maximizes the survival of resistant cells, which may help to decide which drug dosage (not) to administer. Our results are amenable to experimental tests and help link the within and between host scales in epidemiological models.
Collapse
Affiliation(s)
- Peter Czuppon
- Institute for Evolution and Biodiversity, University of Münster, Münster, Germany
- Institute of Ecology and Environmental Sciences of Paris, Sorbonne Université, UPEC, CNRS, IRD, INRA, Paris, France
- Center for Interdisciplinary Research in Biology, CNRS, Collège de France, PSL Research University, Paris, France
| | - Troy Day
- Department of Mathematics and Statistics, Department of Biology, Queen’s University, Kingston, Canada
| | - Florence Débarre
- Institute of Ecology and Environmental Sciences of Paris, Sorbonne Université, UPEC, CNRS, IRD, INRA, Paris, France
| | - François Blanquart
- Center for Interdisciplinary Research in Biology, CNRS, Collège de France, PSL Research University, Paris, France
| |
Collapse
|
21
|
Marrec L, Bank C. Evolutionary rescue in a fluctuating environment: periodic versus quasi-periodic environmental changes. Proc Biol Sci 2023; 290:20230770. [PMID: 37253425 PMCID: PMC10229231 DOI: 10.1098/rspb.2023.0770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/02/2023] [Indexed: 06/01/2023] Open
Abstract
No environment is constant over time, and environmental fluctuations impact the outcome of evolutionary dynamics. Survival of a population not adapted to some environmental conditions is threatened unless, for example, a mutation rescues it, an eco-evolutionary process termed evolutionary rescue. We here investigate evolutionary rescue in an environment that fluctuates between a favourable state, in which the population grows, and a harsh state, in which the population declines. We develop a stochastic model that includes both population dynamics and genetics. We derive analytical predictions for the mean extinction time of a non-adapted population given that it is not rescued, the probability of rescue by a mutation, and the mean appearance time of a rescue mutant, which we validate using numerical simulations. We find that stochastic environmental fluctuations, resulting in quasi-periodic environmental changes, accelerate extinction and hinder evolutionary rescue compared with deterministic environmental fluctuations, resulting in periodic environmental changes. We demonstrate that high equilibrium population sizes and per capita growth rates maximize the chances of evolutionary rescue. We show that an imperfectly harsh environment, which does not fully prevent births but makes the death rate to birth rate ratio much greater than unity, has almost the same rescue probability as a perfectly harsh environment, which fully prevents births. Finally, we put our results in the context of antimicrobial resistance and conservation biology.
Collapse
Affiliation(s)
- Loïc Marrec
- Institut für Ökologie und Evolution, Universität Bern, Baltzerstrasse 6, 3012 Bern, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Claudia Bank
- Institut für Ökologie und Evolution, Universität Bern, Baltzerstrasse 6, 3012 Bern, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| |
Collapse
|
22
|
Akshay SD, Nayak S, Deekshit VK, Rohit A, Maiti B. Differential expression of outer membrane proteins and quinolone resistance determining region mutations can lead to ciprofloxacin resistance in Salmonella Typhi. Arch Microbiol 2023; 205:136. [PMID: 36961627 DOI: 10.1007/s00203-023-03485-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/17/2023] [Accepted: 03/13/2023] [Indexed: 03/25/2023]
Abstract
Multi-drug resistance in Salmonella Typhi remains a public health concern globally. This study aimed to investigate the function of quinolone resistance determining region (QRDR) of gyrA and parC in ciprofloxacin (CIP) resistant isolates and examine the differential expression of outer membrane proteins (OMPs) on exposure to sub-lethal concentrations of CIP in S. Typhi. The CIP-resistant isolates were screened for mutations in the QRDR and analyzed for bacterial growth. Furthermore, major OMPs encoding genes such as ompF, lamB, yaeT, tolC, ompS1, and phoE were examined for differential expression under the sub-lethal concentrations of CIP by real-time PCR and SDS-PAGE. Notably, our study has shown a single-point mutation in gyrA at codon 83 (Ser83-tyrosine and Ser83-phenylalanine), also the rare amino acid substitution in parC gene at codon 80 (Glu80-glycine) in CIP-resistant isolates. Additionally, CIP-resistant isolates showed moderate growth compared to susceptible isolates. Although most of the OMP-encoding genes (tolC, ompS1, and phoE) showed some degree of upregulation, a significant level of upregulation (p < 0.05) was observed only for yaeT. However, ompF and lamB genes were down-regulated compared to CIP-susceptible isolates. Whereas OMPs profiling using SDS-PAGE did not show any changes in the banding pattern. These results provide valuable information on the QRDR mutation, and the difference in the growth, and expression of OMP-encoding genes in resistant and susceptible isolates of S. Typhi. This further provides insight into the involvement of QRDR mutation and OMPs associated with CIP resistance in S. Typhi.
Collapse
Affiliation(s)
- Sadanand Dangari Akshay
- Division of Infectious Diseases, Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Paneer Campus, Deralakatte, Mangalore, 575018, India
| | - Srajana Nayak
- Division of Infectious Diseases, Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Paneer Campus, Deralakatte, Mangalore, 575018, India
| | - Vijaya Kumar Deekshit
- Division of Infectious Diseases, Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Paneer Campus, Deralakatte, Mangalore, 575018, India
| | - Anusha Rohit
- Division of Infectious Diseases, Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Paneer Campus, Deralakatte, Mangalore, 575018, India
- Department of Microbiology, The Madras Medical Mission, 4-A, Dr, Mogappair, Chennai, Tamil Nadu, 600037, India
| | - Biswajit Maiti
- Division of Infectious Diseases, Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Paneer Campus, Deralakatte, Mangalore, 575018, India.
| |
Collapse
|
23
|
Bogri A, Otani S, Aarestrup FM, Brinch C. Interplay between strain fitness and transmission frequency determines prevalence of antimicrobial resistance. Front Ecol Evol 2023. [DOI: 10.3389/fevo.2023.981377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023] Open
Abstract
The steep rise of infections caused by bacteria that are resistant to antimicrobial agents threatens global health. However, the association between antimicrobial use and the prevalence of resistance is not straightforward. Therefore, it is necessary to quantify the importance of additional factors that affect this relationship. We theoretically explore how the prevalence of resistance is affected by the combination of three factors: antimicrobial use, bacterial transmission, and fitness cost of resistance. We present a model that combines within-host, between-hosts and between-populations dynamics, built upon the competitive Lotka-Volterra equations. We developed the model in a manner that allows future experimental validation of the findings with single isolates in the laboratory. Each host may carry two strains (susceptible and resistant) that represent the host’s commensal microbiome and are not the target of the antimicrobial treatment. The model simulates a population of hosts who are treated periodically with antibiotics and transmit bacteria to each other. We show that bacterial transmission results in strain co-existence. Transmission disseminates resistant bacteria in the population, increasing the levels of resistance. Counterintuitively, when the cost of resistance is low, high transmission frequencies reduce resistance prevalence. Transmission between host populations leads to more similar resistance levels, increasing the susceptibility of the population with higher antimicrobial use. Overall, our results indicate that the interplay between bacterial transmission and strain fitness affects the prevalence of resistance in a non-linear way. We then place our results within the context of ecological theory, particularly on temporal niche partitioning and metapopulation rescue, and we formulate testable experimental predictions for future research.
Collapse
|
24
|
Genome Analysis of Pseudomonas aeruginosa Strains from Chronically Infected Patients with High Levels of Persister Formation. Pathogens 2023; 12:pathogens12030426. [PMID: 36986348 PMCID: PMC10051920 DOI: 10.3390/pathogens12030426] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
The appearance of persister cells with low metabolic rates are key factors leading to antibiotic treatment failure. Such persisters are multidrug tolerant and play a key role in the recalcitrance of biofilm-based chronic infections. Here, we present the genomic analyses of three distinct Pseudomonas aeruginosa Egyptian persister-isolates recovered from chronic human infections. To calculate the persister frequencies, viable counts were determined before and after treatment with levofloxacin. The susceptibilities of isolates to different antibiotics were determined using the agar-dilution method. To determine their recalcitrance, the levofloxacin persisters were further challenged with lethal concentrations of meropenem, tobramycin, or colistin. Furthermore, the biofilm formation of the persister strains was estimated phenotypically, and they were reported to be strong biofilm-forming strains. The genotypic characterization of the persisters was performed using whole genome sequencing (WGS) followed by phylogenetic analysis and resistome profiling. Interestingly, out of the thirty-eight clinical isolates, three isolates (8%) demonstrated a persister phenotype. The three levofloxacin-persister isolates were tested for their susceptibility to selected antibiotics; all of the tested isolates were multidrug resistant (MDR). Additionally, the P. aeruginosa persisters were capable of surviving over 24 h and were not eradicated after exposure to 100X-MIC of levofloxacin. WGS for the three persisters revealed a smaller genome size compared to PAO1-genome. Resistome profiling indicated the presence of a broad collection of antibiotic-resistance genes, including genes encoding for antibiotic-modifying enzymes and efflux pump. Phylogenetic analysis indicated that the persister isolates belong to a distinct clade rather than the deposited P. aeruginosa strains in the GenBank. Conclusively, the persister isolates in our study are MDR and form a highly strong biofilm. WGS revealed a smaller genome that belongs to a distinct clade.
Collapse
|
25
|
Le D, Akiyama T, Weiss D, Kim M. Dissociation kinetics of small-molecule inhibitors in Escherichia coli is coupled to physiological state of cells. Commun Biol 2023; 6:223. [PMID: 36841892 PMCID: PMC9968327 DOI: 10.1038/s42003-023-04604-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/16/2023] [Indexed: 02/27/2023] Open
Abstract
Bioactive small-molecule inhibitors represent a treasure chest for future drugs. In vitro high-throughput screening is a common approach to identify the small-molecule inhibitors that bind tightly to purified targets. Here, we investigate the inhibitor-target binding/unbinding kinetics in E. coli cells using a benzimidazole-derivative DNA inhibitor as a model system. We find that its unbinding rate is not constant but depends on cell growth rate. This dependence is mediated by the cellular activity, forming a feedback loop with the inhibitor's activity. In accordance with this feedback, we find cell-to-cell heterogeneity in inhibitor-target interaction, leading to co-existence of two distinct subpopulations: actively growing cells that dissociate the inhibitors from the targets and non-growing cells that do not. We find similar heterogeneity for other clinical DNA inhibitors. Our studies reveal a mechanism that couples inhibitor-target kinetics to cell physiology and demonstrate the significant effect of this coupling on drug efficacy.
Collapse
Affiliation(s)
- Dai Le
- Department of Physics, Emory University, Atlanta, GA, 30322, USA
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Tatsuya Akiyama
- Department of Physics, Emory University, Atlanta, GA, 30322, USA
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA
| | - David Weiss
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Antibiotic Research Center, Emory University, Atlanta, GA, 30322, USA
| | - Minsu Kim
- Department of Physics, Emory University, Atlanta, GA, 30322, USA.
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA.
- Antibiotic Research Center, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
26
|
The Assessment of Antimicrobial Resistance in Gram-Negative and Gram-Positive Infective Endocarditis: A Multicentric Retrospective Analysis. Medicina (B Aires) 2023; 59:medicina59030457. [PMID: 36984458 PMCID: PMC10054718 DOI: 10.3390/medicina59030457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/08/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Background and Objectives: Multidrug-resistant microorganisms have made treating bacterial infections challenging. Resistance to antibiotics is expected to overcome efforts to produce new, effective antibacterial medication that is lifesaving in many situations. Infective endocarditis (IE) is a life-threatening infection that affects 5–15 per 100,000 patients annually and requires rapid antibiotic therapy to prevent morbidity and mortality. Materials and Methods: The present research assessed IE cases over five years, from a multicentric database, with the main objective of determining the degree of antibiotic resistance in these patients, stratified by Gram-positive and Gram-negative bacteria. Results: Bad oral hygiene was present in 58.6% of patients from the Gram-negative group (vs. 38.7% in the Gram-positive group). Non-valvular heart disease was identified in approximately 40% of all patients, and valvopathies in approximately 20%. It was observed that 37.9% of Gram-negative IE bacteria were resistant to three or more antibiotics, whereas 20.7% were susceptible. Among Gram-positive infections, S. aureus was the most commonly involved pathogen, with a multidrug-resistant pattern in 11.2% of patients, while Acinetobacter baumannii had the highest resistance pattern of all Gram-negative pathogens, with 27.4% of all samples resistant to three or more antibiotics. Patients with Gram-negative IE were 4.2 times more likely to die. The mortality risk was 4 times higher when bacteria resistant to two or more antibiotics was involved and 5.7 times higher with resistance patterns to three or more antibiotics than the reference group with no antibiotic resistance. Peripheral catheters were the most common cause of multi-resistant IE, followed by heart surgery, dental procedures, and ENT interventions. Conclusions: Even though Gram-positive infections were the most frequent (83.0% of all cases), Gram-negative IE infections are substantially more deadly than Gram-positive IE infections. However, it was also observed that patients with Gram-negative infections were more likely to have underlying comorbidities, be institutionalized, and be underweight. Although the Gram-negative infections were more severe, their resistance patterns were similar to Gram-positive bacteria. As resistance patterns increase, more efforts should be made to prevent a healthcare catastrophe. At the same time, careful prophylaxis should be considered in patients at risk, including those with central catheters, undergoing dental procedures, and with poor oral hygiene.
Collapse
|
27
|
Antibiotics that affect translation can antagonize phage infectivity by interfering with the deployment of counter-defenses. Proc Natl Acad Sci U S A 2023; 120:e2216084120. [PMID: 36669116 PMCID: PMC9942909 DOI: 10.1073/pnas.2216084120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
It is becoming increasingly clear that antibiotics can both positively and negatively impact the infectivity of bacteriophages (phage), but the underlying mechanisms often remain unclear. Here we demonstrate that antibiotics that target the protein translation machinery can fundamentally alter the outcome of bacteria-phage interactions by interfering with the production of phage-encoded counter-defense proteins. Specifically, using Pseudomonas aeruginosa PA14 and phage DMS3vir as a model, we show that bacteria with Clustered Regularly Interspaced Short Palindromic Repeat, CRISPR associated (CRISPR-Cas) immune systems have elevated levels of immunity against phage that encode anti-CRISPR (acr) genes when translation inhibitors are present in the environment. CRISPR-Cas are highly prevalent defense systems that enable bacteria to detect and destroy phage genomes in a sequence-specific manner. In response, many phages encode acr genes that are expressed immediately following the infection to inhibit key steps of the CRISPR-Cas immune response. Our data show that while phage-carrying acr genes can amplify efficiently on bacteria with CRISPR-Cas immune systems in the absence of antibiotics, the presence of antibiotics that act on protein translation prevents phage amplification, while protecting bacteria from lysis.
Collapse
|
28
|
Diaz-Tang G, Meneses EM, Patel K, Mirkin S, García-Diéguez L, Pajon C, Barraza I, Patel V, Ghali H, Tracey AP, Blanar CA, Lopatkin AJ, Smith RP. Growth productivity as a determinant of the inoculum effect for bactericidal antibiotics. SCIENCE ADVANCES 2022; 8:eadd0924. [PMID: 36516248 PMCID: PMC9750144 DOI: 10.1126/sciadv.add0924] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 11/11/2022] [Indexed: 06/10/2023]
Abstract
Understanding the mechanisms by which populations of bacteria resist antibiotics has implications in evolution, microbial ecology, and public health. The inoculum effect (IE), where antibiotic efficacy declines as the density of a bacterial population increases, has been observed for multiple bacterial species and antibiotics. Several mechanisms to account for IE have been proposed, but most lack experimental evidence or cannot explain IE for multiple antibiotics. We show that growth productivity, the combined effect of growth and metabolism, can account for IE for multiple bactericidal antibiotics and bacterial species. Guided by flux balance analysis and whole-genome modeling, we show that the carbon source supplied in the growth medium determines growth productivity. If growth productivity is sufficiently high, IE is eliminated. Our results may lead to approaches to reduce IE in the clinic, help standardize the analysis of antibiotics, and further our understanding of how bacteria evolve resistance.
Collapse
Affiliation(s)
- Gabriela Diaz-Tang
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Estefania Marin Meneses
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Kavish Patel
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Sophia Mirkin
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Laura García-Diéguez
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Camryn Pajon
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Ivana Barraza
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Vijay Patel
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Helana Ghali
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Angelica P. Tracey
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Christopher A. Blanar
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Allison J. Lopatkin
- Department of Biology, Barnard College, Columbia University, New York, NY10025, USA
- Data Science Institute, Columbia University, New York, NY10025, USA
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY10025, USA
| | - Robert P. Smith
- Department of Biological Sciences, Halmos College of Arts and Science, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
- Cell Therapy Institute, Kiran Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| |
Collapse
|
29
|
Moffett AS, Thomas PJ, Hinczewski M, Eckford AW. Cheater suppression and stochastic clearance through quorum sensing. PLoS Comput Biol 2022; 18:e1010292. [PMID: 35901008 PMCID: PMC9333318 DOI: 10.1371/journal.pcbi.1010292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 06/09/2022] [Indexed: 11/24/2022] Open
Abstract
The evolutionary consequences of quorum sensing in regulating bacterial cooperation are not fully understood. In this study, we reveal unexpected effects of regulating public good production through quorum sensing on bacterial population dynamics, showing that quorum sensing can be a collectively harmful alternative to unregulated production. We analyze a birth-death model of bacterial population dynamics accounting for public good production and the presence of non-producing cheaters. Our model demonstrates that when demographic noise is a factor, the consequences of controlling public good production according to quorum sensing depend on the cost of public good production and the growth rate of populations in the absence of public goods. When public good production is inexpensive, quorum sensing is a destructive alternative to unconditional production, in terms of the mean population extinction time. When costs are higher, quorum sensing becomes a constructive strategy for the producing strain, both stabilizing cooperation and decreasing the risk of population extinction. Quorum sensing is a process through which bacteria can regulate gene expression according to their population density. The reasons for why bacteria use quorum sensing to regulate production of “public goods”, biochemical products that benefit nearby bacteria, are not entirely clear. We use mathematical modeling to explore how quorum sensing compares to other strategies for controlling production of public goods, namely unconditional production independent on population density, in small populations of bacteria where the random nature of growth is significant. Our model captures both how likely “cheater” strains, which do not produce public goods but benefit from them, are to take over a population and how long on average the population will last before going extinct. We find that depending on how expensive public good production is and how critical public goods are for growth, quorum sensing can decrease or increase the mean time to extinction compared with unconditional production, while always reducing the likelihood of cheaters taking over. Our results could have important implications for the growth of bacterial infections, for example Pseudomonas aeruginosa infections of the lungs of cystic fibrosis patients.
Collapse
Affiliation(s)
- Alexander S. Moffett
- Department of Electrical Engineering and Computer Science, York University, Toronto, Ontario, Canada
| | - Peter J. Thomas
- Department of Mathematics, Applied Mathematics, and Statistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Michael Hinczewski
- Department of Physics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Andrew W. Eckford
- Department of Electrical Engineering and Computer Science, York University, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
30
|
Garzon M, Sosik P, Drastík J, Skalli O. A Self-Controlled and Self-Healing Model of Bacterial Cells. MEMBRANES 2022; 12:678. [PMID: 35877878 PMCID: PMC9324567 DOI: 10.3390/membranes12070678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022]
Abstract
A new kind of self-assembly model, morphogenetic (M) systems, assembles spatial units into larger structures through local interactions of simpler components and enables discovery of new principles for cellular membrane assembly, development, and its interface function. The model is based on interactions among three kinds of constitutive objects such as tiles and protein-like elements in discrete time and continuous 3D space. It was motivated by achieving a balance between three conflicting goals: biological, physical-chemical, and computational realism. A recent example is a unified model of morphogenesis of a single biological cell, its membrane and cytoskeleton formation, and finally, its self-reproduction. Here, a family of dynamic M systems (Mbac) is described with similar characteristics, modeling the process of bacterial cell formation and division that exhibits bacterial behaviors of living cells at the macro-level (including cell growth that is self-controlled and sensitive to the presence/absence of nutrients transported through membranes), as well as self-healing properties. Remarkably, it consists of only 20 or so developmental rules. Furthermore, since the model exhibits membrane formation and septic mitosis, it affords more rigorous definitions of concepts such as injury and self-healing that enable quantitative analyses of these kinds of properties. Mbac shows that self-assembly and interactions of living organisms with their environments and membrane interfaces are critical for self-healing, and that these properties can be defined and quantified more rigorously and precisely, despite their complexity.
Collapse
Affiliation(s)
- Max Garzon
- Department of Computer Science, The University of Memphis, Memphis, TN 38152, USA;
| | - Petr Sosik
- Research Institute of the IT4Innovations Centre of Excellence, Silesian University in Opava, 74601 Opava, Czech Republic;
| | - Jan Drastík
- Research Institute of the IT4Innovations Centre of Excellence, Silesian University in Opava, 74601 Opava, Czech Republic;
| | - Omar Skalli
- Department of Biology, The University of Memphis, Memphis, TN 38152, USA;
| |
Collapse
|
31
|
Saebelfeld M, Das SG, Hagenbeek A, Krug J, de Visser JAGM. Stochastic establishment of β-lactam-resistant Escherichia coli mutants reveals conditions for collective resistance. Proc Biol Sci 2022; 289:20212486. [PMID: 35506221 PMCID: PMC9065960 DOI: 10.1098/rspb.2021.2486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
For antibiotic resistance to arise, new resistant mutants must establish in a bacterial population before they can spread via natural selection. Comprehending the stochastic factors that influence mutant establishment is crucial for a quantitative understanding of antibiotic resistance emergence. Here, we quantify the single-cell establishment probability of four Escherichia coli strains expressing β-lactamase alleles with different activity against the antibiotic cefotaxime, as a function of antibiotic concentration in both unstructured (liquid) and structured (agar) environments. We show that concentrations well below the minimum inhibitory concentration (MIC) can substantially hamper establishment, particularly for highly resistant mutants. While the pattern of establishment suppression is comparable in both tested environments, we find greater variability in establishment probability on agar. Using a simple branching model, we investigate possible sources of this stochasticity, including environment-dependent lineage variability, but cannot reject other possible causes. Lastly, we use the single-cell establishment probability to predict each strain's MIC in the absence of social interactions. We observe substantially higher measured than predicted MIC values, particularly for highly resistant strains, which indicates cooperative effects among resistant cells at large cell numbers, such as in standard MIC assays.
Collapse
Affiliation(s)
- Manja Saebelfeld
- Institute for Biological Physics, University of Cologne, Cologne, Germany,Laboratory of Genetics, Wageningen University, Wageningen, The Netherlands
| | - Suman G. Das
- Institute for Biological Physics, University of Cologne, Cologne, Germany
| | - Arno Hagenbeek
- Laboratory of Genetics, Wageningen University, Wageningen, The Netherlands
| | - Joachim Krug
- Institute for Biological Physics, University of Cologne, Cologne, Germany
| | | |
Collapse
|
32
|
Pedreira A, Vázquez JA, García MR. Kinetics of Bacterial Adaptation, Growth, and Death at Didecyldimethylammonium Chloride sub-MIC Concentrations. Front Microbiol 2022; 13:758237. [PMID: 35464917 PMCID: PMC9023358 DOI: 10.3389/fmicb.2022.758237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/09/2022] [Indexed: 11/24/2022] Open
Abstract
Minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) are standard indexes for determining disinfection effectiveness. Nevertheless, they are static values disregarding the kinetics at sub-MIC concentrations where adaptation, growth, stationary, and death phases can be observed. The understanding of these dynamic mechanisms is crucial to designing effective disinfection strategies. In this study, we studied the 48 h kinetics of Bacillus cereus and Escherichia coli cells exposed to sub-MIC concentrations of didecyldimethylammonium chloride (DDAC). Two mathematical models were employed to reproduce the experiments: the only-growth classical logistic model and a mechanistic model including growth and death dynamics. Although both models reproduce the lag, exponential and stationary phases, only the mechanistic model is able to reproduce the death phase and reveals the concentration dependence of the bactericidal/bacteriostatic activity of DDAC. This model could potentially be extended to study other antimicrobials and reproduce changes in optical density (OD) and colony-forming units (CFUs) with the same parameters and mechanisms of action.
Collapse
Affiliation(s)
- Adrián Pedreira
- Biosystems and Bioprocess Engineering (Bio2Eng), Marine Research Institute-Spanish National Research Council (IIM-CSIC), Eduardo Cabello, Vigo, Spain
- Group of Recycling and Valorization of Waste Materials (REVAL), Marine Research Institute-Spanish National Research Council (IIM-CSIC), Eduardo Cabello, Vigo, Spain
| | - José A. Vázquez
- Group of Recycling and Valorization of Waste Materials (REVAL), Marine Research Institute-Spanish National Research Council (IIM-CSIC), Eduardo Cabello, Vigo, Spain
- *Correspondence: José A. Vázquez
| | - Míriam R. García
- Biosystems and Bioprocess Engineering (Bio2Eng), Marine Research Institute-Spanish National Research Council (IIM-CSIC), Eduardo Cabello, Vigo, Spain
- Míriam R. García
| |
Collapse
|
33
|
Taylor D, Verdon N, Lomax P, Allen RJ, Titmuss S. Tracking the stochastic growth of bacterial populations in microfluidic droplets. Phys Biol 2022; 19:026003. [PMID: 35042205 PMCID: PMC7613235 DOI: 10.1088/1478-3975/ac4c9b] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 01/18/2022] [Indexed: 11/11/2022]
Abstract
Bacterial growth in microfluidic droplets is relevant in biotechnology, in microbial ecology, and in understanding stochastic population dynamics in small populations. However, it has proved challenging to automate measurement of absolute bacterial numbers within droplets, forcing the use of proxy measures for population size. Here we present a microfluidic device and imaging protocol that allows high-resolution imaging of thousands of droplets, such that individual bacteria stay in the focal plane and can be counted automatically. Using this approach, we track the stochastic growth of hundreds of replicateEscherichia colipopulations within droplets. We find that, for early times, the statistics of the growth trajectories obey the predictions of the Bellman-Harris model, in which there is no inheritance of division time. Our approach should allow further testing of models for stochastic growth dynamics, as well as contributing to broader applications of droplet-based bacterial culture.
Collapse
Affiliation(s)
- Daniel Taylor
- School of Physics and Astronomy, University of Edinburgh, James Clerk Maxwell Building, Peter Guthrie Tait Road, Edinburgh EH9 3FD, United Kingdom
| | - Nia Verdon
- School of Physics and Astronomy, University of Edinburgh, James Clerk Maxwell Building, Peter Guthrie Tait Road, Edinburgh EH9 3FD, United Kingdom
| | - Peter Lomax
- Scottish Microelectronics Centre, Alexander Crum Brown Road, King's Buildings, Edinburgh, EH9 3FF, United Kingdom
| | - Rosalind J Allen
- School of Physics and Astronomy, University of Edinburgh, James Clerk Maxwell Building, Peter Guthrie Tait Road, Edinburgh EH9 3FD, United Kingdom
| | - Simon Titmuss
- School of Physics and Astronomy, University of Edinburgh, James Clerk Maxwell Building, Peter Guthrie Tait Road, Edinburgh EH9 3FD, United Kingdom
| |
Collapse
|
34
|
Morsky B, Vural DC. Suppressing evolution of antibiotic resistance through environmental switching. THEOR ECOL-NETH 2022. [DOI: 10.1007/s12080-022-00530-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
35
|
Martini CL, Coronado AZ, Melo MCN, Gobbi CN, Lopez ÚS, de Mattos MC, Amorim TT, Botelho AMN, Vasconcelos ATR, Almeida LGP, Planet PJ, Zingali RB, Figueiredo AMS, Ferreira-Carvalho BT. Cellular Growth Arrest and Efflux Pumps Are Associated With Antibiotic Persisters in Streptococcus pyogenes Induced in Biofilm-Like Environments. Front Microbiol 2021; 12:716628. [PMID: 34621249 PMCID: PMC8490960 DOI: 10.3389/fmicb.2021.716628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Streptococcus pyogenes (group A Streptococcus-GAS) is an important pathogen for humans. GAS has been associated with severe and invasive diseases. Despite the fact that these bacteria remain universally susceptible to penicillin, therapeutic failures have been reported in some GAS infections. Many hypotheses have been proposed to explain these antibiotic-unresponsive infections; however, none of them have fully elucidated this phenomenon. In this study, we show that GAS strains have the ability to form antimicrobial persisters when inoculated on abiotic surfaces to form a film of bacterial agglomerates (biofilm-like environment). Our data suggest that efflux pumps were possibly involved in this phenomenon. In fact, gene expression assays by real-time qRT-PCR showed upregulation of some genes associated with efflux pumps in persisters arising in the presence of penicillin. Phenotypic reversion assay and whole-genome sequencing indicated that this event was due to non-inherited resistance mechanisms. The persister cells showed downregulation of genes associated with protein biosynthesis and cell growth, as demonstrated by gene expression assays. Moreover, the proteomic analysis revealed that susceptible cells express higher levels of ribosome proteins. It is remarkable that previous studies have reported the recovery of S. pyogenes viable cells from tissue biopsies of patients presented with GAS invasive infections and submitted to therapy with antibiotics. The persistence phenomenon described herein brings new insights into the origin of therapeutic failures in S. pyogenes infections. Multifactorial mechanisms involving protein synthesis inhibition, cell growth impairment and efflux pumps seem to play roles in the formation of antimicrobial persisters in S. pyogenes.
Collapse
Affiliation(s)
- Caroline Lopes Martini
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amada Zambrana Coronado
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Celeste Nunes Melo
- Departamento de Microbiologia e Parasitologia, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Clarice Neffa Gobbi
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Úrsula Santos Lopez
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Correa de Mattos
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thais Tavares Amorim
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Maria Nunes Botelho
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Paul J Planet
- Department of Pediatrics, Perelman College of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, United States.,Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Russolina Benedeta Zingali
- Unidade de Espectrometria de Massas e Proteomica - UEMP, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Agnes Marie Sá Figueiredo
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
36
|
Teimouri H, Nguyen TN, Kolomeisky AB. Single-cell stochastic modelling of the action of antimicrobial peptides on bacteria. J R Soc Interface 2021; 18:20210392. [PMID: 34520689 PMCID: PMC8440028 DOI: 10.1098/rsif.2021.0392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/18/2021] [Indexed: 11/12/2022] Open
Abstract
Antimicrobial peptides (AMPs) produced by multi-cellular organisms as their immune system's defence against microbes are actively considered as natural alternatives to conventional antibiotics. Although substantial progress has been achieved in studying the AMPs, the microscopic mechanisms of their functioning remain not well understood. Here, we develop a new theoretical framework to investigate how the AMPs are able to efficiently neutralize bacteria. In our minimal theoretical model, the most relevant processes, AMPs entering into and the following inhibition of the single bacterial cell, are described stochastically. Using complementary master equations approaches, all relevant features of bacteria clearance dynamics by AMPs, such as the probability of inhibition and the mean times before the clearance, are explicitly evaluated. It is found that both processes, entering and inhibition, are equally important for the efficient functioning of AMPs. Our theoretical method naturally explains a wide spectrum of efficiencies of existing AMPs and their heterogeneity at the single-cell level. Theoretical calculations are also consistent with existing single-cell measurements. Thus, the presented theoretical approach clarifies some microscopic aspects of the action of AMPs on bacteria.
Collapse
Affiliation(s)
- Hamid Teimouri
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
| | - Thao N. Nguyen
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
| | - Anatoly B. Kolomeisky
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX 77005, USA
- Department of Physics and Astronomy, Rice University, Houston, TX 77005, USA
| |
Collapse
|
37
|
Saebelfeld M, Das SG, Brink J, Hagenbeek A, Krug J, de Visser JAGM. Antibiotic Breakdown by Susceptible Bacteria Enhances the Establishment of β-Lactam Resistant Mutants. Front Microbiol 2021; 12:698970. [PMID: 34489889 PMCID: PMC8417073 DOI: 10.3389/fmicb.2021.698970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022] Open
Abstract
For a better understanding of the evolution of antibiotic resistance, it is imperative to study the factors that determine the initial establishment of mutant resistance alleles. In addition to the antibiotic concentration, the establishment of resistance alleles may be affected by interactions with the surrounding susceptible cells from which they derive, for instance via the release of nutrients or removal of the antibiotic. Here, we investigate the effects of social interactions with surrounding susceptible cells on the establishment of Escherichia coli mutants with increasing β-lactamase activity (i.e., the capacity to hydrolyze β-lactam antibiotics) from single cells under the exposure of the antibiotic cefotaxime (CTX) on agar plates. We find that relatively susceptible cells, expressing a β-lactamase with very low antibiotic-hydrolyzing activity, increase the probability of mutant cells to survive and outgrow into colonies due to the active breakdown of the antibiotic. However, the rate of breakdown by the susceptible strain is much higher than expected based on its low enzymatic activity. A detailed theoretical model suggests that this observation may be explained by cell filamentation causing delayed lysis. While susceptible cells may hamper the spread of higher-resistant β-lactamase mutants at relatively high frequencies, our findings show that they promote their initial establishment.
Collapse
Affiliation(s)
- Manja Saebelfeld
- Institute for Biological Physics, University of Cologne, Cologne, Germany
- Laboratory of Genetics, Department of the Plant Sciences Group, Wageningen University and Research, Wageningen, Netherlands
| | - Suman G. Das
- Institute for Biological Physics, University of Cologne, Cologne, Germany
| | - Jorn Brink
- Laboratory of Genetics, Department of the Plant Sciences Group, Wageningen University and Research, Wageningen, Netherlands
| | - Arno Hagenbeek
- Laboratory of Genetics, Department of the Plant Sciences Group, Wageningen University and Research, Wageningen, Netherlands
| | - Joachim Krug
- Institute for Biological Physics, University of Cologne, Cologne, Germany
| | - J. Arjan G. M. de Visser
- Laboratory of Genetics, Department of the Plant Sciences Group, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
38
|
Akiyama T, Kim M. Stochastic response of bacterial cells to antibiotics: its mechanisms and implications for population and evolutionary dynamics. Curr Opin Microbiol 2021; 63:104-108. [PMID: 34325154 DOI: 10.1016/j.mib.2021.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/22/2021] [Accepted: 07/01/2021] [Indexed: 11/20/2022]
Abstract
The effectiveness of antibiotics against bacterial infections has been declining due to the emergence of resistance. Precisely understanding the response of bacteria to antibiotics is critical to maximizing antibiotic-induced bacterial eradication while minimizing the emergence of antibiotic resistance. Cell-to-cell heterogeneity in antibiotic susceptibility is observed across various bacterial species for a wide range of antibiotics. Heterogeneity in antibiotic susceptibility is not always due to the genetic differences. Rather, it can be caused by non-genetic mechanisms such as stochastic gene expression and biased partitioning upon cell division. Heterogeneous susceptibility leads to the stochastic growth and death of individual cells and stochastic fluctuations in population size. These fluctuations have important implications for the eradication of bacterial populations and the emergence of genotypic resistance.
Collapse
Affiliation(s)
- Tatsuya Akiyama
- Department of Physics, Emory University, Atlanta, GA, 30322, USA; Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Minsu Kim
- Department of Physics, Emory University, Atlanta, GA, 30322, USA; Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA; Emory Antibiotic Resistance Center, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
39
|
Gjini E, Wood KB. Price equation captures the role of drug interactions and collateral effects in the evolution of multidrug resistance. eLife 2021; 10:e64851. [PMID: 34289932 PMCID: PMC8331190 DOI: 10.7554/elife.64851] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 07/08/2021] [Indexed: 01/03/2023] Open
Abstract
Bacterial adaptation to antibiotic combinations depends on the joint inhibitory effects of the two drugs (drug interaction [DI]) and how resistance to one drug impacts resistance to the other (collateral effects [CE]). Here we model these evolutionary dynamics on two-dimensional phenotype spaces that leverage scaling relations between the drug-response surfaces of drug-sensitive (ancestral) and drug-resistant (mutant) populations. We show that evolved resistance to the component drugs - and in turn, the adaptation of growth rate - is governed by a Price equation whose covariance terms encode geometric features of both the two-drug-response surface (DI) in ancestral cells and the correlations between resistance levels to those drugs (CE). Within this framework, mean evolutionary trajectories reduce to a type of weighted gradient dynamics, with the drug interaction dictating the shape of the underlying landscape and the collateral effects constraining the motion on those landscapes. We also demonstrate how constraints on available mutational pathways can be incorporated into the framework, adding a third key driver of evolution. Our results clarify the complex relationship between drug interactions and collateral effects in multidrug environments and illustrate how specific dosage combinations can shift the weighting of these two effects, leading to different and temporally explicit selective outcomes.
Collapse
Affiliation(s)
- Erida Gjini
- Center for Computational and Stochastic Mathematics, Instituto Superior Tecnico, University of Lisbon, PortugalLisbonPortugal
| | - Kevin B Wood
- Departments of Biophysics and Physics, University of MichiganAnn ArborUnited States
| |
Collapse
|
40
|
Bistable Bacterial Growth Dynamics in the Presence of Antimicrobial Agents. Antibiotics (Basel) 2021; 10:antibiotics10010087. [PMID: 33477524 PMCID: PMC7831100 DOI: 10.3390/antibiotics10010087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 11/22/2022] Open
Abstract
The outcome of an antibiotic treatment on the growth capacity of bacteria is largely dependent on the initial population size (Inoculum Effect). We characterized and built a model of this effect in E. coli cultures using a large variety of antimicrobials, including conventional antibiotics, and for the first time, cationic antimicrobial peptides (CAMPs). Our results show that all classes of antimicrobial drugs induce an inoculum effect, which, as we explain, implies that the dynamic is bistable: For a range of anti-microbial densities, a very small inoculum decays whereas a larger inoculum grows, and the threshold inoculum depends on the drug concentration. We characterized three distinct classes of drug-induced bistable growth dynamics and demonstrate that in rich medium, CAMPs correspond to the simplest class, bacteriostatic antibiotics to the second class, and all other traditional antibiotics to the third, more complex class. These findings provide a unifying universal framework for describing the dynamics of the inoculum effect induced by antimicrobials with inherently different killing mechanisms.
Collapse
|
41
|
Revitt-Mills SA, Robinson A. Antibiotic-Induced Mutagenesis: Under the Microscope. Front Microbiol 2020; 11:585175. [PMID: 33193230 PMCID: PMC7642495 DOI: 10.3389/fmicb.2020.585175] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/28/2020] [Indexed: 11/24/2022] Open
Abstract
The development of antibiotic resistance poses an increasing threat to global health. Understanding how resistance develops in bacteria is critical for the advancement of new strategies to combat antibiotic resistance. In the 1980s, it was discovered that certain antibiotics induce elevated rates of mutation in bacteria. From this, an “increased evolvability” hypothesis was proposed: antibiotic-induced mutagenesis increases the genetic diversity of bacterial populations, thereby increasing the rate at which bacteria develop antibiotic resistance. However, antibiotic-induced mutagenesis is one of multiple competing factors that act on bacterial populations exposed to antibiotics. Its relative importance in shaping evolutionary outcomes, including the development of antibiotic resistance, is likely to depend strongly on the conditions. Presently, there is no quantitative model that describes the relative contribution of antibiotic-induced mutagenesis to bacterial evolution. A far more complete understanding could be reached if we had access to technology that enabled us to study antibiotic-induced mutagenesis at the molecular-, cellular-, and population-levels simultaneously. Direct observations would, in principle, allow us to directly link molecular-level events with outcomes in individual cells and cell populations. In this review, we highlight microscopy studies which have allowed various aspects of antibiotic-induced mutagenesis to be directly visualized in individual cells for the first time. These studies have revealed new links between error-prone DNA polymerases and recombinational DNA repair, evidence of spatial regulation occurring during the SOS response, and enabled real-time readouts of mismatch and mutation rates. Further, we summarize the recent discovery of stochastic population fluctuations in cultures exposed to sub-inhibitory concentrations of bactericidal antibiotics and discuss the implications of this finding for the study of antibiotic-induced mutagenesis. The studies featured here demonstrate the potential of microscopy to provide direct observation of phenomena relevant to evolution under antibiotic-induced mutagenesis.
Collapse
Affiliation(s)
- Sarah A Revitt-Mills
- Molecular Horizons Institute and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| | - Andrew Robinson
- Molecular Horizons Institute and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| |
Collapse
|
42
|
Morphogenetic systems: Models and experiments. Biosystems 2020; 198:104270. [PMID: 33038464 DOI: 10.1016/j.biosystems.2020.104270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/14/2020] [Accepted: 10/03/2020] [Indexed: 11/21/2022]
Abstract
M systems are mathematical models of morphogenesis developed to gain insights into its relations to phenomena such as self-assembly, self-controlled growth, homeostasis, self-healing and self-reproduction, in both natural and artificial systems. M systems rely on basic principles of membrane computing and self-assembly, as well as explicit emphasis on geometrical structures (location and shape) in 2D, 3D or higher dimensional Euclidean spaces. They can be used for principled studies of these phenomena, both theoretically and experimentally, at a computational level abstracted from their detailed implementation. In particular, they afford 2D and 3D models to explore biological morphogenetic processes. Theoretical studies have shown that M systems are powerful tools (e.g., computational universal, i.e. can become as complex as any computer program) and their parallelism allows for trading space for time in solving efficiently problems considered infeasible on conventional computers (NP-hard problems). In addition, they can also exhibit properties such as robustness to injuries and degrees of self-healing. This paper focuses on the experimental side of M systems. To this end, we have developed a high-level morphogenetic simulator, Cytos, to implement and visualize M systems in silico in order to verify theoretical results and facilitate research in M systems. We summarize the software package and make a brief comparison with some other simulators of membrane systems. The core of the article is a description of a range of experiments inspired by aspects of morphogenesis in both prokaryotic and eukaryotic cells. The experiments explore the regulatory role of the septum and of the cytoskeleton in cell fission, the robustness of cell models against injuries, and, finally, the impact of changing nutrient concentration on population growth.
Collapse
|
43
|
Marrec L, Bitbol AF. Adapt or Perish: Evolutionary Rescue in a Gradually Deteriorating Environment. Genetics 2020; 216:573-583. [PMID: 32855198 PMCID: PMC7536851 DOI: 10.1534/genetics.120.303624] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/24/2020] [Indexed: 12/31/2022] Open
Abstract
We investigate the evolutionary rescue of a microbial population in a gradually deteriorating environment, through a combination of analytical calculations and stochastic simulations. We consider a population destined for extinction in the absence of mutants, which can survive only if mutants sufficiently adapted to the new environment arise and fix. We show that mutants that appear later during the environment deterioration have a higher probability to fix. The rescue probability of the population increases with a sigmoidal shape when the product of the carrying capacity and of the mutation probability increases. Furthermore, we find that rescue becomes more likely for smaller population sizes and/or mutation probabilities if the environment degradation is slower, which illustrates the key impact of the rapidity of environment degradation on the fate of a population. We also show that our main conclusions are robust across various types of adaptive mutants, including specialist and generalist ones, as well as mutants modeling antimicrobial resistance evolution. We further express the average time of appearance of the mutants that do rescue the population and the average extinction time of those that do not. Our methods can be applied to other situations with continuously variable fitnesses and population sizes, and our analytical predictions are valid in the weak-to-moderate mutation regime.
Collapse
Affiliation(s)
- Loïc Marrec
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratoire Jean Perrin (UMR 8237), 75005 Paris, France
| | - Anne-Florence Bitbol
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratoire Jean Perrin (UMR 8237), 75005 Paris, France
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
44
|
Ojkic N, Lilja E, Direito S, Dawson A, Allen RJ, Waclaw B. A Roadblock-and-Kill Mechanism of Action Model for the DNA-Targeting Antibiotic Ciprofloxacin. Antimicrob Agents Chemother 2020; 64:e02487-19. [PMID: 32601161 PMCID: PMC7449190 DOI: 10.1128/aac.02487-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 06/19/2020] [Indexed: 12/19/2022] Open
Abstract
Fluoroquinolones, antibiotics that cause DNA damage by inhibiting DNA topoisomerases, are clinically important, but their mechanism of action is not yet fully understood. In particular, the dynamical response of bacterial cells to fluoroquinolone exposure has hardly been investigated, although the SOS response, triggered by DNA damage, is often thought to play a key role. Here, we investigated the growth inhibition of the bacterium Escherichia coli by the fluoroquinolone ciprofloxacin at low concentrations. We measured the long-term and short-term dynamical response of the growth rate and DNA production rate to ciprofloxacin at both the population and single-cell levels. We show that, despite the molecular complexity of DNA metabolism, a simple roadblock-and-kill model focusing on replication fork blockage and DNA damage by ciprofloxacin-poisoned DNA topoisomerase II (gyrase) quantitatively reproduces long-term growth rates in the presence of ciprofloxacin. The model also predicts dynamical changes in the DNA production rate in wild-type E. coli and in a recombination-deficient mutant following a step-up of ciprofloxacin. Our work highlights that bacterial cells show a delayed growth rate response following fluoroquinolone exposure. Most importantly, our model explains why the response is delayed: it takes many doubling times to fragment the DNA sufficiently to inhibit gene expression. We also show that the dynamical response is controlled by the timescale of DNA replication and gyrase binding/unbinding to the DNA rather than by the SOS response, challenging the accepted view. Our work highlights the importance of including detailed biophysical processes in biochemical-systems models to quantitatively predict the bacterial response to antibiotics.
Collapse
Affiliation(s)
- Nikola Ojkic
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, United Kingdom
| | - Elin Lilja
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, United Kingdom
| | - Susana Direito
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, United Kingdom
| | - Angela Dawson
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, United Kingdom
| | - Rosalind J Allen
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Synthetic and Systems Biology, Edinburgh, United Kingdom
| | - Bartlomiej Waclaw
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Synthetic and Systems Biology, Edinburgh, United Kingdom
| |
Collapse
|
45
|
Alexander HK, MacLean RC. Stochastic bacterial population dynamics restrict the establishment of antibiotic resistance from single cells. Proc Natl Acad Sci U S A 2020; 117:19455-19464. [PMID: 32703812 PMCID: PMC7431077 DOI: 10.1073/pnas.1919672117] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A better understanding of how antibiotic exposure impacts the evolution of resistance in bacterial populations is crucial for designing more sustainable treatment strategies. The conventional approach to this question is to measure the range of concentrations over which resistant strain(s) are selectively favored over a sensitive strain. Here, we instead investigate how antibiotic concentration impacts the initial establishment of resistance from single cells, mimicking the clonal expansion of a resistant lineage following mutation or horizontal gene transfer. Using two Pseudomonas aeruginosa strains carrying resistance plasmids, we show that single resistant cells have <5% probability of detectable outgrowth at antibiotic concentrations as low as one-eighth of the resistant strain's minimum inhibitory concentration (MIC). This low probability of establishment is due to detrimental effects of antibiotics on resistant cells, coupled with the inherently stochastic nature of cell division and death on the single-cell level, which leads to loss of many nascent resistant lineages. Our findings suggest that moderate doses of antibiotics, well below the MIC of resistant strains, may effectively restrict de novo emergence of resistance even though they cannot clear already-large resistant populations.
Collapse
Affiliation(s)
- Helen K Alexander
- Department of Zoology, University of Oxford, Oxford OX1 3PS, United Kingdom;
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh EH9 3FL, United Kingdom
| | - R Craig MacLean
- Department of Zoology, University of Oxford, Oxford OX1 3PS, United Kingdom
| |
Collapse
|
46
|
Taitelbaum A, West R, Assaf M, Mobilia M. Population Dynamics in a Changing Environment: Random versus Periodic Switching. PHYSICAL REVIEW LETTERS 2020; 125:048105. [PMID: 32794803 DOI: 10.1103/physrevlett.125.048105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/13/2020] [Accepted: 06/23/2020] [Indexed: 06/11/2023]
Abstract
Environmental changes greatly influence the evolution of populations. Here, we study the dynamics of a population of two strains, one growing slightly faster than the other, competing for resources in a time-varying binary environment modeled by a carrying capacity switching either randomly or periodically between states of abundance and scarcity. The population dynamics is characterized by demographic noise (birth and death events) coupled to a varying environment. We elucidate the similarities and differences of the evolution subject to a stochastically and periodically varying environment. Importantly, the population size distribution is generally found to be broader under intermediate and fast random switching than under periodic variations, which results in markedly different asymptotic behaviors between the fixation probability of random and periodic switching. We also determine the detailed conditions under which the fixation probability of the slow strain is maximal.
Collapse
Affiliation(s)
- Ami Taitelbaum
- Racah Institute of Physics, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Robert West
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Michael Assaf
- Racah Institute of Physics, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Mauro Mobilia
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
47
|
Vlazaki M, Rossi O, Price DJ, McLean C, Grant AJ, Mastroeni P, Restif O. A data-based mathematical modelling study to quantify the effects of ciprofloxacin and ampicillin on the within-host dynamics of Salmonella enterica during treatment and relapse. J R Soc Interface 2020; 17:20200299. [PMID: 32634369 DOI: 10.1098/rsif.2020.0299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Antibiotic therapy has drastically reduced the mortality and sequelae of bacterial infections. From naturally occurring to chemically synthesized, different classes of antibiotics have been successfully used without detailed knowledge of how they affect bacterial dynamics in vivo. However, a proportion of patients receiving antimicrobial therapy develop recrudescent infections post-treatment. Relapsing infections are attributable to incomplete clearance of bacterial populations following antibiotic administration; the metabolic profile of this antibiotic-recalcitrant bacterial subpopulation, the spatio-temporal context of its emergence and the variance of antibiotic-bacterial interactions in vivo remain unclear. Here, we develop and apply a mechanistic mathematical model to data from a study comparing the effects of ciprofloxacin and ampicillin on the within-host dynamics of Salmonella enterica serovar Typhimurium in murine infections. Using the inferential capacity of our model, we show that the antibiotic-recalcitrant bacteria following ampicillin, but not ciprofloxacin, treatment belong to a non-replicating phenotype. Aligning with previous studies, we independently estimate that the lymphoid tissues and spleen are important reservoirs of non-replicating bacteria. Finally, we predict that post-treatment, the progenitors of the non-growing and growing bacterial populations replicate and die at different rates. Ultimately, the liver, spleen and mesenteric lymph nodes are all repopulated by progenitors of the previously non-growing phenotype in ampicillin-treated mice.
Collapse
Affiliation(s)
- Myrto Vlazaki
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Omar Rossi
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | - David J Price
- Centre of Epidemiology and Biostatistics, University of Melbourne, Grattan Street, Parkville, Victoria 3010, Australia.,The Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, Victoria 3000, Australia
| | - Callum McLean
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Andrew J Grant
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Pietro Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Olivier Restif
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| |
Collapse
|
48
|
Fan Y, Lim Y, Wyss LS, Park S, Xu C, Fu H, Fei J, Hong Y, Wang B. Mechanical expansion microscopy. Methods Cell Biol 2020; 161:125-146. [PMID: 33478686 DOI: 10.1016/bs.mcb.2020.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
This chapter describes two mechanical expansion microscopy methods with accompanying step-by-step protocols. The first method, mechanically resolved expansion microscopy, uses non-uniform expansion of partially digested samples to provide the imaging contrast that resolves local mechanical properties. Examining bacterial cell wall with this method, we are able to distinguish bacterial species in mixed populations based on their distinct cell wall rigidity and detect cell wall damage caused by various physiological and chemical perturbations. The second method is mechanically locked expansion microscopy, in which we use a mechanically stable gel network to prevent the original polyacrylate network from shrinking in ionic buffers. This method allows us to use anti-photobleaching buffers in expansion microscopy, enabling detection of novel ultra-structures under the optical diffraction limit through super-resolution single molecule localization microscopy on bacterial cells and whole-mount immunofluorescence imaging in thick animal tissues. We also discuss potential applications and assess future directions.
Collapse
Affiliation(s)
- Yuhang Fan
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Youngbin Lim
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Livia S Wyss
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Seongjin Park
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, United States
| | - Cancan Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Huikang Fu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Jingyi Fei
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, United States; Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, CA, United States; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
49
|
Teimouri H, Kolomeisky AB. Theoretical investigation of stochastic clearance of bacteria: first-passage analysis. J R Soc Interface 2020; 16:20180765. [PMID: 30890051 DOI: 10.1098/rsif.2018.0765] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Understanding mechanisms of bacterial eradication is critically important for overcoming failures of antibiotic treatments. Current studies suggest that the clearance of large bacterial populations proceeds deterministically, while for smaller populations, the stochastic effects become more relevant. Here, we develop a theoretical approach to investigate the bacterial population dynamics under the effect of antibiotic drugs using a method of first-passage processes. It allows us to explicitly evaluate the most important characteristics of bacterial clearance dynamics such as extinction probabilities and extinction times. The new meaning of minimal inhibitory concentrations for stochastic clearance of bacterial populations is also discussed. In addition, we investigate the effect of fluctuations in population growth rates on the dynamics of bacterial eradication. It is found that extinction probabilities and extinction times generally do not correlate with each other when random fluctuations in the growth rates are taking place. Unexpectedly, for a significant range of parameters, the extinction times increase due to these fluctuations, indicating a slowing in the bacterial clearance dynamics. It is argued that this might be one of the initial steps in the pathway for the development of antibiotic resistance. Furthermore, it is suggested that extinction times is a convenient measure of bacterial tolerance.
Collapse
Affiliation(s)
- Hamid Teimouri
- 1 Department of Chemistry, Rice University , Houston, TX , USA.,3 Center for Theoretical Biological Physics, Rice University , Houston, TX , USA
| | - Anatoly B Kolomeisky
- 1 Department of Chemistry, Rice University , Houston, TX , USA.,2 Department of Chemical and Biomolecular Engineering, Rice University , Houston, TX , USA.,3 Center for Theoretical Biological Physics, Rice University , Houston, TX , USA
| |
Collapse
|
50
|
Marrec L, Bitbol AF. Resist or perish: Fate of a microbial population subjected to a periodic presence of antimicrobial. PLoS Comput Biol 2020; 16:e1007798. [PMID: 32275712 PMCID: PMC7176291 DOI: 10.1371/journal.pcbi.1007798] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/22/2020] [Accepted: 03/19/2020] [Indexed: 12/22/2022] Open
Abstract
The evolution of antimicrobial resistance can be strongly affected by variations of antimicrobial concentration. Here, we study the impact of periodic alternations of absence and presence of antimicrobial on resistance evolution in a microbial population, using a stochastic model that includes variations of both population composition and size, and fully incorporates stochastic population extinctions. We show that fast alternations of presence and absence of antimicrobial are inefficient to eradicate the microbial population and strongly favor the establishment of resistance, unless the antimicrobial increases enough the death rate. We further demonstrate that if the period of alternations is longer than a threshold value, the microbial population goes extinct upon the first addition of antimicrobial, if it is not rescued by resistance. We express the probability that the population is eradicated upon the first addition of antimicrobial, assuming rare mutations. Rescue by resistance can happen either if resistant mutants preexist, or if they appear after antimicrobial is added to the environment. Importantly, the latter case is fully prevented by perfect biostatic antimicrobials that completely stop division of sensitive microorganisms. By contrast, we show that the parameter regime where treatment is efficient is larger for biocidal drugs than for biostatic drugs. This sheds light on the respective merits of different antimicrobial modes of action.
Collapse
Affiliation(s)
- Loïc Marrec
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratoire Jean Perrin (UMR 8237), F-75005 Paris, France
| | - Anne-Florence Bitbol
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratoire Jean Perrin (UMR 8237), F-75005 Paris, France
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|