Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Evol Biol ; 37(4): 371-382, 2024 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-38386697

RESUMEN

Viruses that infect bacteria, known as bacteriophages or phages, are the most prevalent entities on Earth. Their genetic diversity in nature is well documented, and members of divergent lineages can be found sharing the same ecological niche. This viral diversity can be influenced by a number of factors, including productivity, spatial structuring of the environment, and host-range trade-offs. Rapid evolution is also known to promote diversity by buffering ecological systems from extinction. There is, however, little known about the impact of coevolution on the maintenance of viral diversity within a microbial community. To address this, we developed a 4 species experimental system where two bacterial hosts, a generalist and a specialist phage, coevolved in a spatially homogenous environment over time. We observed the persistence of both viruses if the resource availability was sufficiently high. This coexistence occurred in the absence of any detectable host-range trade-offs that are costly for generalists and thus known to promote viral diversity. However, the coexistence was lost if two bacteria were not permitted to evolve alongside the phages or if two phages coevolved with a single bacterial host. Our findings indicate that a host's resistance response in mixed-species communities plays a significant role in maintaining viral diversity in the environment.


Asunto(s)
Bacteriófagos , Bacteriófagos/genética , Especificidad del Huésped , Bacterias/genética
2.
Nat Commun ; 13(1): 2917, 2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35614098

RESUMEN

Antibiotic resistance represents a growing medical concern where raw, clinical datasets are under-exploited as a means to track the scale of the problem. We therefore sought patterns of antibiotic resistance in the Antimicrobial Testing Leadership and Surveillance (ATLAS) database. ATLAS holds 6.5M minimal inhibitory concentrations (MICs) for 3,919 pathogen-antibiotic pairs isolated from 633k patients in 70 countries between 2004 and 2017. We show most pairs form coherent, although not stationary, timeseries whose frequencies of resistance are higher than other databases, although we identified no systematic bias towards including more resistant strains in ATLAS. We sought data anomalies whereby MICs could shift for methodological and not clinical or microbiological reasons and found artefacts in over 100 pathogen-antibiotic pairs. Using an information-optimal clustering methodology to classify pathogens into low and high antibiotic susceptibilities, we used ATLAS to predict changes in resistance. Dynamics of the latter exhibit complex patterns with MIC increases, and some decreases, whereby subpopulations' MICs can diverge. We also identify pathogens at risk of developing clinical resistance in the near future.


Asunto(s)
Antiinfecciosos , Metadatos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/farmacología , Farmacorresistencia Microbiana , Humanos , Pruebas de Sensibilidad Microbiana
3.
PLoS Comput Biol ; 17(3): e1008817, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33735173

RESUMEN

Developing mathematical models to accurately predict microbial growth dynamics remains a key challenge in ecology, evolution, biotechnology, and public health. To reproduce and grow, microbes need to take up essential nutrients from the environment, and mathematical models classically assume that the nutrient uptake rate is a saturating function of the nutrient concentration. In nature, microbes experience different levels of nutrient availability at all environmental scales, yet parameters shaping the nutrient uptake function are commonly estimated for a single initial nutrient concentration. This hampers the models from accurately capturing microbial dynamics when the environmental conditions change. To address this problem, we conduct growth experiments for a range of micro-organisms, including human fungal pathogens, baker's yeast, and common coliform bacteria, and uncover the following patterns. We observed that the maximal nutrient uptake rate and biomass yield were both decreasing functions of initial nutrient concentration. While a functional form for the relationship between biomass yield and initial nutrient concentration has been previously derived from first metabolic principles, here we also derive the form of the relationship between maximal nutrient uptake rate and initial nutrient concentration. Incorporating these two functions into a model of microbial growth allows for variable growth parameters and enables us to substantially improve predictions for microbial dynamics in a range of initial nutrient concentrations, compared to keeping growth parameters fixed.


Asunto(s)
Candida , Enterobacteriaceae , Modelos Biológicos , Saccharomyces cerevisiae , Biotecnología , Candida/citología , Candida/crecimiento & desarrollo , Candida/fisiología , Proliferación Celular/fisiología , Biología Computacional , Ecología , Enterobacteriaceae/citología , Enterobacteriaceae/crecimiento & desarrollo , Enterobacteriaceae/fisiología , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/fisiología
4.
Proc Biol Sci ; 287(1931): 20200761, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32673559

RESUMEN

Antimicrobial resistance frequently carries a fitness cost to a pathogen, measured as a reduction in growth rate compared to the sensitive wild-type, in the absence of antibiotics. Existing empirical evidence points to the following relationship between cost of resistance and virulence. If a resistant pathogen suffers a fitness cost in terms of reduced growth rate it commonly has lower virulence compared to the sensitive wild-type. If this cost is absent so is the reduction in virulence. Here we show, using experimental evolution of drug resistance in the fungal human pathogen Candida glabrata, that reduced growth rate of resistant strains need not result in reduced virulence. Phenotypically heterogeneous populations were evolved in parallel containing highly resistant sub-population small colony variants (SCVs) alongside sensitive sub-populations. Despite their low growth rate in the absence of an antifungal drug, the SCVs did not suffer a marked alteration in virulence compared with the wild-type ancestral strain, or their co-isolated sensitive strains. This contrasts with classical theory that assumes growth rate to positively correlate with virulence. Our work thus highlights the complexity of the relationship between resistance, basic life-history traits and virulence.


Asunto(s)
Candida glabrata , Farmacorresistencia Fúngica , Antifúngicos , Proteínas Fúngicas , Humanos , Pruebas de Sensibilidad Microbiana , Fenotipo
5.
J R Soc Interface ; 17(166): 20190776, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32453982

RESUMEN

Microbes occupy almost every niche within and on their human hosts. Whether colonizing the gut, mouth or bloodstream, microorganisms face temporal fluctuations in resources and stressors within their niche but we still know little of how environmental fluctuations mediate certain microbial phenotypes, notably antimicrobial-resistant ones. For instance, do rapid or slow fluctuations in nutrient and antimicrobial concentrations select for, or against, resistance? We tackle this question using an ecological approach by studying the dynamics of a synthetic and pathogenic microbial community containing two species, one sensitive and the other resistant to an antibiotic drug where the community is exposed to different rates of environmental fluctuation. We provide mathematical models, supported by experimental data, to demonstrate that simple community outcomes, such as competitive exclusion, can shift to coexistence and ecosystem bistability as fluctuation rates vary. Theory gives mechanistic insight into how these dynamical regimes are related. Importantly, our approach highlights a fundamental difference between resistance in single-species populations, the context in which it is usually assayed, and that in communities. While fast environmental changes are known to select against resistance in single-species populations, here we show that they can promote the resistant species in mixed-species communities. Our theoretical observations are verified empirically using a two-species Candida community.


Asunto(s)
Antibacterianos , Ecosistema , Antibacterianos/farmacología , Humanos , Modelos Teóricos , Dinámica Poblacional
6.
Nat Ecol Evol ; 2(11): 1824, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30237543

RESUMEN

In the version of this Article originally published, the following sentence was missing from the Acknowledgements: "R.E.B. is an EPSRC Healthcare Technologies Impact Fellow EP/N033671/1; I.G. is funded by ERC Consolidator grant 647292 MathModExp; A.J.P.B., N.A.R.G. and A.T. were funded by BBSRC grant BB/F00513X/1; K.H., I.G., S.N. and E.C. were funded by BBSRC grant BB/F005210/2." This text has now been added.

7.
Nat Ecol Evol ; 2(8): 1312-1320, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29988162

RESUMEN

Microbes rarely exist in isolation, rather, they form intricate multi-species communities that colonize our bodies and inserted medical devices. However, the efficacy of antimicrobials is measured in clinical laboratories exclusively using microbial monocultures. Here, to determine how multi-species interactions mediate selection for resistance during antibiotic treatment, particularly following drug withdrawal, we study a laboratory community consisting of two microbial pathogens. Single-species dose responses are a poor predictor of community dynamics during treatment so, to better understand those dynamics, we introduce the concept of a dose-response mosaic, a multi-dimensional map that indicates how species' abundance is affected by changes in abiotic conditions. We study the dose-response mosaic of a two-species community with a 'Gene × Gene × Environment × Environment' ecological interaction whereby Candida glabrata, which is resistant to the antifungal drug fluconazole, competes for survival with Candida albicans, which is susceptible to fluconazole. The mosaic comprises several zones that delineate abiotic conditions where each species dominates. Zones are separated by loci of bifurcations and tipping points that identify what environmental changes can trigger the loss of either species. Observations of the laboratory communities corroborated theory, showing that changes in both antibiotic concentration and nutrient availability can push populations beyond tipping points, thus creating irreversible shifts in community composition from drug-sensitive to drug-resistant species. This has an important consequence: resistant species can increase in frequency even if an antibiotic is withdrawn because, unwittingly, a tipping point was passed during treatment.


Asunto(s)
Antifúngicos/farmacología , Candida albicans/efectos de los fármacos , Candida glabrata/efectos de los fármacos , Farmacorresistencia Fúngica , Fluconazol/farmacología , Interacciones Microbianas , Candida albicans/crecimiento & desarrollo , Candida glabrata/crecimiento & desarrollo , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga
8.
PLoS Comput Biol ; 12(11): e1005216, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27898662

RESUMEN

Many antimicrobial and anti-tumour drugs elicit hormetic responses characterised by low-dose stimulation and high-dose inhibition. While this can have profound consequences for human health, with low drug concentrations actually stimulating pathogen or tumour growth, the mechanistic understanding behind such responses is still lacking. We propose a novel, simple but general mechanism that could give rise to hormesis in systems where an inhibitor acts on an enzyme. At its core is one of the basic building blocks in intracellular signalling, the dual phosphorylation-dephosphorylation motif, found in diverse regulatory processes including control of cell proliferation and programmed cell death. Our analytically-derived conditions for observing hormesis provide clues as to why this mechanism has not been previously identified. Current mathematical models regularly make simplifying assumptions that lack empirical support but inadvertently preclude the observation of hormesis. In addition, due to the inherent population heterogeneities, the presence of hormesis is likely to be masked in empirical population-level studies. Therefore, examining hormetic responses at single-cell level coupled with improved mathematical models could substantially enhance detection and mechanistic understanding of hormesis.


Asunto(s)
Fenómenos Fisiológicos Celulares/efectos de los fármacos , Hormesis/fisiología , Modelos Biológicos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Quinasas/metabolismo , Animales , Simulación por Computador , Humanos , Modelos Químicos , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas/química , Proteínas Quinasas/efectos de los fármacos
9.
Evol Appl ; 8(10): 945-55, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26640520

RESUMEN

Antibiotic resistance is a growing concern to public health. New treatment strategies may alleviate the situation by slowing down the evolution of resistance. Here, we evaluated sequential treatment protocols using two fully independent laboratory-controlled evolution experiments with the human pathogen Pseudomonas aeruginosa PA14 and two pairs of clinically relevant antibiotics (doripenem/ciprofloxacin and cefsulodin/gentamicin). Our results consistently show that the sequential application of two antibiotics decelerates resistance evolution relative to monotherapy. Sequential treatment enhanced population extinction although we applied antibiotics at sublethal dosage. In both experiments, we identified an order effect of the antibiotics used in the sequential protocol, leading to significant variation in the long-term efficacy of the tested protocols. These variations appear to be caused by asymmetric evolutionary constraints, whereby adaptation to one drug slowed down adaptation to the other drug, but not vice versa. An understanding of such asymmetric constraints may help future development of evolutionary robust treatments against infectious disease.

10.
PLoS Biol ; 12(8): e1001928, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25136970

RESUMEN

Phenotypic heterogeneity can confer clonal groups of organisms with new functionality. A paradigmatic example is the bistable expression of virulence genes in Salmonella typhimurium, which leads to phenotypically virulent and phenotypically avirulent subpopulations. The two subpopulations have been shown to divide labor during S. typhimurium infections. Here, we show that heterogeneous virulence gene expression in this organism also promotes survival against exposure to antibiotics through a bet-hedging mechanism. Using microfluidic devices in combination with fluorescence time-lapse microscopy and quantitative image analysis, we analyzed the expression of virulence genes at the single cell level and related it to survival when exposed to antibiotics. We found that, across different types of antibiotics and under concentrations that are clinically relevant, the subpopulation of bacterial cells that express virulence genes shows increased survival after exposure to antibiotics. Intriguingly, there is an interplay between the two consequences of phenotypic heterogeneity. The bet-hedging effect that arises through heterogeneity in virulence gene expression can protect clonal populations against avirulent mutants that exploit and subvert the division of labor within these populations. We conclude that bet-hedging and the division of labor can arise through variation in a single trait and interact with each other. This reveals a new degree of functional complexity of phenotypic heterogeneity. In addition, our results suggest a general principle of how pathogens can evade antibiotics: Expression of virulence factors often entails metabolic costs and the resulting growth retardation could generally increase tolerance against antibiotics and thus compromise treatment.


Asunto(s)
Adaptación Fisiológica/genética , Antibacterianos/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidad , Adaptación Fisiológica/efectos de los fármacos , Genes Bacterianos , Mutación/genética , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/fisiología , Selección Genética/efectos de los fármacos , Virulencia/efectos de los fármacos , Virulencia/genética
11.
Ecol Lett ; 16(10): 1267-76, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23902419

RESUMEN

Understanding how populations and communities respond to competition is a central concern of ecology. A seminal theoretical solution first formalised by Levins (and re-derived in multiple fields) showed that, in theory, the form of a trade-off should determine the outcome of competition. While this has become a central postulate in ecology it has evaded experimental verification, not least because of substantial technical obstacles. We here solve the experimental problems by employing synthetic ecology. We engineer strains of Escherichia coli with fixed resource allocations enabling accurate measurement of trade-off shapes between bacterial survival and multiplication in multiple environments. A mathematical chemostat model predicts different, and experimentally verified, trajectories of gene frequency changes as a function of condition-specific trade-offs. The results support Levins' postulate and demonstrates that otherwise paradoxical alternative outcomes witnessed in subtly different conditions are predictable.


Asunto(s)
Ecosistema , Escherichia coli/fisiología , Modelos Biológicos , Proteínas Bacterianas/metabolismo , Escherichia coli/crecimiento & desarrollo , Viabilidad Microbiana , Factor sigma/metabolismo
12.
Nature ; 472(7343): 342-6, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21441905

RESUMEN

How is diversity maintained? Environmental heterogeneity is considered to be important, yet diversity in seemingly homogeneous environments is nonetheless observed. This, it is assumed, must either be owing to weak selection, mutational input or a fitness advantage to genotypes when rare. Here we demonstrate the possibility of a new general mechanism of stable diversity maintenance, one that stems from metabolic and physiological trade-offs. The model requires that such trade-offs translate into a fitness landscape in which the most fit has unfit near-mutational neighbours, and a lower fitness peak also exists that is more mutationally robust. The 'survival of the fittest' applies at low mutation rates, giving way to 'survival of the flattest' at high mutation rates. However, as a consequence of quasispecies-level negative frequency-dependent selection and differences in mutational robustness we observe a transition zone in which both fittest and flattest coexist. Although diversity maintenance is possible for simple organisms in simple environments, the more trade-offs there are, the wider the maintenance zone becomes. The principle may be applied to lineages within a species or species within a community, potentially explaining why competitive exclusion need not be observed in homogeneous environments. This principle predicts the enigmatic richness of metabolic strategies in clonal bacteria and questions the safety of lethal mutagenesis as an antimicrobial treatment.


Asunto(s)
Biodiversidad , Evolución Biológica , Aptitud Genética , Metabolismo/genética , Modelos Biológicos , Selección Genética , Adaptación Biológica/genética , Aptitud Genética/genética , Genotipo , Modelos Genéticos , Mutagénesis/genética , Saccharomyces cerevisiae , Selección Genética/genética , Procesos Estocásticos
13.
Math Biosci Eng ; 7(4): 923-33, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21077716

RESUMEN

We ask the question Which antibiotic deployment protocols select best against drug-resistant microbes: mixing or periodic cycling? and demonstrate that the statistical distribution of the performances of both sets of protocols, mixing and periodic cycling, must have overlapping supports. In other words, it is a general, mathematical result that there must be mixing policies that outperform cycling policies and vice versa. As a result, we agree with the tenet of Bonhoefer et al. [1] that one should not apply the results of [2] to conclude that an antibiotic cycling policy that implements cycles of drug restriction and prioritisation on an ad-hoc basis can select against drug-resistant microbial pathogens in a clinical setting any better than random drug use. However, nor should we conclude that a random, per-patient drug-assignment protocol is the de facto optimal method for allocating antibiotics to patients in any general sense.


Asunto(s)
Antibacterianos/uso terapéutico , Farmacorresistencia Microbiana , Modelos Biológicos , Humanos
14.
Math Biosci Eng ; 7(3): 527-52, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20578784

RESUMEN

The purpose of this paper is to use mathematical models to investigate the claim made in the medical literature over a decade ago that the routine rotation of antibiotics in an intensive care unit (ICU) will select against the evolution and spread of antibiotic-resistant pathogens. In contrast, previous theoretical studies addressing this question have demonstrated that routinely changing the drug of choice for a given pathogenic infection may in fact lead to a greater incidence of drug resistance in comparison to the random deployment of different drugs. Using mathematical models that do not explicitly incorporate the spatial dynamics of pathogen transmission within the ICU or hospital and assuming the antibiotics are from distinct functional groups, we use a control theoretic-approach to prove that one can relax the medical notion of what constitutes an antibiotic rotation and so obtain protocols that are arbitrarily close to the optimum. Finally, we show that theoretical feedback control measures that rotate between different antibiotics motivated directly by the outcome of clinical studies can be deployed to good effect to reduce the prevalence of antibiotic resistance below what can be achieved with random antibiotic use.


Asunto(s)
Antibacterianos/administración & dosificación , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/tratamiento farmacológico , Modelos Biológicos , Animales , Bacterias/efectos de los fármacos , Infecciones Bacterianas/microbiología , Esquema de Medicación , Farmacorresistencia Bacteriana , Unidades de Cuidados Intensivos
15.
Nature ; 455(7210): 220-3, 2008 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-18784724

RESUMEN

Given the difficulty of testing evolutionary and ecological theory in situ, in vitro model systems are attractive alternatives; however, can we appraise whether an experimental result is particular to the in vitro model, and, if so, characterize the systems likely to behave differently and understand why? Here we examine these issues using the relationship between phenotypic diversity and resource input in the T7-Escherichia coli co-evolving system as a case history. We establish a mathematical model of this interaction, framed as one instance of a super-class of host-parasite co-evolutionary models, and show that it captures experimental results. By tuning this model, we then ask how diversity as a function of resource input could behave for alternative co-evolving partners (for example, E. coli with lambda bacteriophages). In contrast to populations lacking bacteriophages, variation in diversity with differences in resources is always found for co-evolving populations, supporting the geographic mosaic theory of co-evolution. The form of this variation is not, however, universal. Details of infectivity are pivotal: in T7-E. coli with a modified gene-for-gene interaction, diversity is low at high resource input, whereas, for matching-allele interactions, maximal diversity is found at high resource input. A combination of in vitro systems and appropriately configured mathematical models is an effective means to isolate results particular to the in vitro system, to characterize systems likely to behave differently and to understand the biology underpinning those alternatives.


Asunto(s)
Bacteriófago T7/fisiología , Evolución Biológica , Escherichia coli/virología , Modelos Biológicos , Bacteriófago T7/genética , Bacteriófago T7/patogenicidad , Ecología , Escherichia coli/genética , Variación Genética , Interacciones Huésped-Patógeno , Fenotipo , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...