1
|
Ding L, Zhang CM, Jiang HY. Lipid-lowering drug clofibric acid promotes conjugative transfer of RP4 plasmid carrying antibiotic resistance genes by multiple mechanisms. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137560. [PMID: 39952130 DOI: 10.1016/j.jhazmat.2025.137560] [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: 10/06/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Antibiotic resistance represents a growing global health crisis, predominantly driven by the selective pressure imposed by antibiotics, which facilitates horizontal gene transfer. However, the potential role of non-antibiotic pharmaceuticals in promoting or enabling the spread of antibiotic resistance genes (ARGs) remains poorly understood. This study provided novel insights into the capacity of clofibric acid, a lipid-lowering drug, to enhance the conjugative transfer of ARGs, and deeply explored the underlying multiple mechanisms. The findings revealed that clofibric acid, at concentrations ranging from 0.01 to 1000 μg/L, significantly promoted the transfer efficiency of the RP4 plasmid carrying multiple ARGs. This enhancement was accompanied by a cascade of stress responses in bacterial cells, including elevated production of reactive oxygen species, increased secretion of extracellular polymeric substances, reduced bacterial surface zeta potential, and heightened cell membrane permeability. The physiological alterations were closely linked to significant changes in the expression of genes associated with these processes. Our results highlighted the potential of non-antibiotic pharmaceuticals to contribute to the dissemination of antibiotic resistance, offering a critical foundation for further research into the environmental and public health implications of such compounds.
Collapse
Affiliation(s)
- Lin Ding
- School of Environmental and Municipal Engineering, Xi'an University of Architecture and Technology, Xi'an 710055, China; Key Laboratory of Northwest Water Resource, Environment and Ecology, Ministry of Education, Xi'an University of Architecture and Technology, Xi'an 710055, China; Shaanxi Key Laboratory of Environmental Engineering, Xi'an University of Architecture and Technology, Xi'an 710055, China
| | - Chong-Miao Zhang
- School of Environmental and Municipal Engineering, Xi'an University of Architecture and Technology, Xi'an 710055, China; Key Laboratory of Northwest Water Resource, Environment and Ecology, Ministry of Education, Xi'an University of Architecture and Technology, Xi'an 710055, China; Shaanxi Key Laboratory of Environmental Engineering, Xi'an University of Architecture and Technology, Xi'an 710055, China; International Science and Technology Cooperation Center for Urban Alternative Water Resources Development, Xi'an University of Architecture and Technology, Xi'an 710055, China.
| | - Hong-Yu Jiang
- School of Environmental and Municipal Engineering, Xi'an University of Architecture and Technology, Xi'an 710055, China; Key Laboratory of Northwest Water Resource, Environment and Ecology, Ministry of Education, Xi'an University of Architecture and Technology, Xi'an 710055, China; Shaanxi Key Laboratory of Environmental Engineering, Xi'an University of Architecture and Technology, Xi'an 710055, China
| |
Collapse
|
2
|
Zhou T, Liu G, Jin R, Zhou J. Altered Cell Viability, Morphology, and Motility under Ciprofloxacin Stress Influence the Transport and Resistance of Bacteria in Saturated Porous Media. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:8746-8755. [PMID: 40265891 DOI: 10.1021/acs.est.5c00322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The ubiquitous occurrence of antibiotics in the environment induces various stress responses of microbes and increases the risk of the emergence and spread of antimicrobial resistance (AMR). In this study, the transport and retention of Shewanella oneidensis cells in saturated porous media was investigated under different levels of ciprofloxacin (CIP) stress. Exposing to lethal CIP stress caused significant viability loss and stimulated cell transport due to increasing hydrophilicity and decreasing surface roughness. While exposure to sublethal CIP stress did not affect MR-1's viability, elongation of cells promoted their retention in sand columns via straining and orientation effects. The elongated cells likely adopted an end-on configuration to minimize repulsive interaction energy when approaching sand surfaces and deposited in a side-on position due to local surface roughness and charge heterogeneity of sands. The more diminished breakthrough of MR-1 cells in redox-active media was ascribed to their improving extracellular electron transfer and energy taxis activities under sublethal CIP stress. Moreover, the retention of elongated cells in porous media facilitated the de novo emergence of a resistant gyrase mutant, whose remobilization might exacerbate the AMR dissemination.
Collapse
Affiliation(s)
- Tianao Zhou
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Guangfei Liu
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Ruofei Jin
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Jiti Zhou
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Ali S, Mirza R, Shah KU, Javed A, Dilawar N. "Harnessing green synthesized zinc oxide nanoparticles for dual action in wound management: Antibiotic delivery and healing Promotion". Microb Pathog 2025; 200:107314. [PMID: 39848301 DOI: 10.1016/j.micpath.2025.107314] [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: 07/06/2024] [Revised: 01/02/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
Wound infections are characterized by the invasion of microorganisms into bodily tissues, leading to inflammation and potentially affecting any type of wound, including surgical incisions and chronic ulcers. If left untreated, they can delay recovery and cause tissue damage. Healthcare providers face challenges in treating these infections, which necessitate efficient treatment plans involving microbiological testing and clinical evaluation. The effectiveness of conventional treatments like antibiotics is limited by resistance. Various forms of nanotechnology have been developed, each exhibiting unique properties that address particular issues with conventional therapies. Among all the Nanocarriers, zinc oxide nanoparticles (ZnO NPs), offer promising treatments for persistent wound infections. ZnO NPs possess strong antibacterial, antioxidant, anti-inflammatory, and anti-diabetic properties, making them suitable for wound care applications. These nanoparticles can be produced economically and environmentally using green synthesis techniques that minimize toxicity and are biocompatible. While chemical and physical techniques offer precise control over nanoparticle characteristics, they often involve hazardous substances and energy-intensive procedures. The antibacterial qualities, low toxicity, and biological compatibility of green-synthesized ZnO NPs make them a promising treatment for wound infections. Their use in scaffolds, drug delivery systems, and wound dressings provides a viable approach to combat antibiotic resistance and enhance wound treatment outcomes. Furthermore research is necessary to fully realize the benefits of ZnO NPs in clinical practice.
Collapse
Affiliation(s)
- Sajid Ali
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Rashna Mirza
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Kifayat Ullah Shah
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Aqeedat Javed
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Naz Dilawar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
6
|
Tan L, Liang J, Qin Z, Ning T, Wei X, Yang B, Wang Q, Xu Y, Shen F. Unveiling the sustained effects of plant root exudates on soil microbiome and resistome and the related functional traits. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 376:124485. [PMID: 39938296 DOI: 10.1016/j.jenvman.2025.124485] [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: 11/05/2024] [Revised: 01/15/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Investigating the transmission mechanisms and influencing factors of antibiotic resistance genes (ARGs) in the soil-plant continuum is vital for mitigating ARG contamination and safeguarding plant and human health. Rhizosphere soil serves as a crucial interface for ARG propagation and transmission; however, it is unclear whether and how plant involved in regulating ARGs in their rhizosphere environment. Root exudates acting as vital links in the plant-soil-microbe interaction. Here, we examined the fluctuating patterns of the resistome and mobile genetic elements (MGEs) following exposure to four types of common root exudates: amino acids (AAs), sugars, long-chain organic acids (LCOAs), and short-chain organic acids (SCOAs). AAs exerted a rapid and pronounced effect, leading to a significant elevation in total ARG and MGE abundance by 3.18-fold and 21.06-fold, respectively, compared to the control group by day 7. Conversely, the impact of sugars manifested gradually over time. The influence of AAs and sugars persisted beyond 240 days post-treatment cessation. Importantly, the proliferation of ARGs was closely linked to the enrichment of plant growth-promoting bacteria (PGPBs) such as Pseudomonas, Cupriavidus, Azospirillum, Variovorax, and Ensifer. Functional analysis revealed that the potential features of ARGs and MGEs were associated with cell wall/membrane/envelope biogenesis, cell motility, and inorganic ion transport. This study offers novel insights into the factors influencing the dynamics of ARGs in the plant rhizosphere and may contribute to ecologically sustainable agricultural practices.
Collapse
Affiliation(s)
- Lu Tan
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Jiayin Liang
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Ziyi Qin
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Tianyang Ning
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Xiaocheng Wei
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Bo Yang
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Qiang Wang
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Yan Xu
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China.
| | - Feng Shen
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China; Key Laboratory of Rural Toilet and Sewage Treatment Technology, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China.
| |
Collapse
|
7
|
Zhu Y, Yao S, Wang X, Wang J, Cao H, Tao Y. Variable cyanobacterial death modes caused by ciprofloxacin in the aquatic environment: Prioritizing antibiotic-photosynthetic protein interactions for risk assessment. WATER RESEARCH 2025; 271:122885. [PMID: 39642793 DOI: 10.1016/j.watres.2024.122885] [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: 10/18/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Antibiotics continuously discharged into the aquatic environment pose threats to phototrophs via high-affinity binding to photosynthetic apparatuses and interfering with their energy metabolism and growth. However, studies attributed the sublethal effects of antibiotics on phototrophs to damaging photosystem (PS) II (PSII) proteins while neglecting PSI proteins as potential targets. Herein, we report that frequently detected ciprofloxacin (CIP) with concentrations of 3-8 μg/L was lethal to Microcystis aeruginosa, the widely distributed phytoplankton in freshwater, via damaging DNA. Besides, CIP damages on different photosynthetic proteins at different exposure levels were evidenced to influence the cyanobacterial death phenotypes. In detail, CIP at 3 μg/L bound to PSII D1 protein exclusively, activating the tricarboxylic acid cycle for energy and proline catabolism. This favored the execution of apoptosis-like regulated cell death (RCD). However, CIP at 8 μg/L exhibited additional binding to the PSI iron-sulfur reaction center, apart from PSII, inducing carbon and arginine starvation. This shifted the RCD from apoptosis-like RCD to mazEF-mediated RCD. Furthermore, microcystin-LR risks were elevated after CIP exposure with enhanced microcystin-LR release and biosynthesis for apoptosis-like and mazEF-mediated RCD, respectively. Thus, the present study underscores the intricate interactions between antibiotics and different photosynthetic apparatuses, which alter antibiotic lethal effects at different exposure levels. This could provide new perspectives on the risk assessment and prediction of antibiotics from the standpoint of chemical-photosynthesis interactions.
Collapse
Affiliation(s)
- Yinjie Zhu
- Guangdong Provincial Engineering Research Center for Urban Water Recycling and Environmental Safety, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Key Laboratory of Microorganism Application and Risk Control, Ministry of Ecology and Environment, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Shenzhen Key Laboratory of Ecological Remediation and Carbon Sequestration, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Tsinghua University-Kunming Joint Research Center for Dianchi Plateau Lake, Tsinghua University, Beijing 100084, PR China
| | - Shishi Yao
- Guangdong Provincial Engineering Research Center for Urban Water Recycling and Environmental Safety, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Key Laboratory of Microorganism Application and Risk Control, Ministry of Ecology and Environment, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China
| | - Xiaoxiong Wang
- Institute for Ocean Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China
| | - Jian Wang
- Guangdong Provincial Engineering Research Center for Urban Water Recycling and Environmental Safety, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Key Laboratory of Microorganism Application and Risk Control, Ministry of Ecology and Environment, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Shenzhen Key Laboratory of Ecological Remediation and Carbon Sequestration, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Tsinghua University-Kunming Joint Research Center for Dianchi Plateau Lake, Tsinghua University, Beijing 100084, PR China
| | - Huansheng Cao
- Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan, Jiangsu 215300, PR China
| | - Yi Tao
- Guangdong Provincial Engineering Research Center for Urban Water Recycling and Environmental Safety, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Key Laboratory of Microorganism Application and Risk Control, Ministry of Ecology and Environment, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Shenzhen Key Laboratory of Ecological Remediation and Carbon Sequestration, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518055, PR China; Tsinghua University-Kunming Joint Research Center for Dianchi Plateau Lake, Tsinghua University, Beijing 100084, PR China.
| |
Collapse
|
8
|
Kaur N, Sahoo J, De M. Development of Nanomaterials-Based Agents for Selective Antibacterial Activity. Chembiochem 2025; 26:e202400693. [PMID: 39632741 DOI: 10.1002/cbic.202400693] [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: 08/20/2024] [Revised: 10/26/2024] [Indexed: 12/07/2024]
Abstract
Bacterial infections continue to threaten public health due to limitations in rapid and accurate diagnostic techniques. While broad-spectrum antibiotics offer empirical treatment, their overuse has fuelled the emergence of antimicrobial resistance (AMR) pathogens, posing a critical global public health challenge. In this critical scenario, nanomaterial-based antibacterial agents emerge as a promising solution to combat bacteria and inhibit their proliferation. However, selective elimination of pathogenic bacteria is paramount. This review highlights recent advancements in developing nanomaterials for selective antibacterial activity. We categorize these agents based on their mode of action, exploring how they selectively interact with bacteria and their potential antibacterial mechanisms. This review offers crucial insights for researchers exploring the potential of nanotechnology to address the growing threat of AMR.
Collapse
Affiliation(s)
- Navjot Kaur
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Jagabandhu Sahoo
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Mrinmoy De
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
9
|
Maltas J, Huynh A, Wood KB. Dynamic collateral sensitivity profiles highlight opportunities and challenges for optimizing antibiotic treatments. PLoS Biol 2025; 23:e3002970. [PMID: 39774800 PMCID: PMC11709278 DOI: 10.1371/journal.pbio.3002970] [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: 08/06/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
As failure rates for traditional antimicrobial therapies escalate, recent focus has shifted to evolution-based therapies to slow resistance. Collateral sensitivity-the increased susceptibility to one drug associated with evolved resistance to a different drug-offers a potentially exploitable evolutionary constraint, but the manner in which collateral effects emerge over time is not well understood. Here, we use laboratory evolution in the opportunistic pathogen Enterococcus faecalis to phenotypically characterize collateral profiles through evolutionary time. Specifically, we measure collateral profiles for 400 strain-antibiotic combinations over the course of 4 evolutionary time points as strains are selected in increasing concentrations of antibiotic. We find that at a global level-when results from all drugs are combined-collateral resistance dominates during early phases of adaptation, when resistance to the selecting drug is lower, while collateral sensitivity becomes increasingly likely with further selection. At the level of individual populations; however, the trends are idiosyncratic; for example, the frequency of collateral sensitivity to ceftriaxone increases over time in isolates selected by linezolid but decreases in isolates selected by ciprofloxacin. We then show experimentally how dynamic collateral sensitivity relationships can lead to time-dependent dosing windows that depend on finely timed switching between drugs. Finally, we develop a stochastic mathematical model based on a Markov decision process consistent with observed dynamic collateral profiles to show measurements across time are required to optimally constrain antibiotic resistance.
Collapse
Affiliation(s)
- Jeff Maltas
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Anh Huynh
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kevin B. Wood
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Physics, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
10
|
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
|
11
|
Xu Y, Li H, Ding Y, Zhang D, Liu W. How nanoscale plastics facilitate the evolution of antibiotic resistance? JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136157. [PMID: 39423639 DOI: 10.1016/j.jhazmat.2024.136157] [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: 06/18/2024] [Revised: 08/29/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
The plastic can enhance the proliferation of antibiotic resistance genes (ARGs), however, the effect of nanoplastics (NPLs) on bacterial antibiotic resistance has not been clearly explained. Herein, we explored the effects and mechanisms of NPLs of different sizes (200 and 600 nm) on the evolution of antibiotic resistance in Serratia marcescens. The results indicated that the evolution of bacterial antibiotic resistance could be promoted under NPLs exposure, which the median of relative abundance of ARGs was 1.11-1.46 times compared to the treatment without NPLs. Transcriptomic analysis showed that the larger size of NPLs mainly increased the permeability of bacterial cell membranes to efflux antibiotics, thus potentiating antibiotic resistance. While, the smaller NPLs is more than that, its enhanced the expression of antibiotic resistance by modulating bacterial metabolic processes. The genome SNP analysis found that the NPLs could cause the genetic mutation occurrence to alter the membrane transport and metabolism processes, and it increased at a size of 200 nm more than at 600 nm NPLs. Importantly, we demonstrated that the horizontal transfer of ARGs was augmented due to the NPLs could dock to bacterial surface proteins and pull their movement to contact with other bacteria (binding energy of membrane proteins: -8.54 kcal/mol), especially the smaller size. It suggests that NPLs will also contribute to the proliferation of ARGs in the environment. This study provides data for understanding the risk of bacterial resistance.
Collapse
Affiliation(s)
- Yan Xu
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Houyu Li
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Yinuo Ding
- Jilin Agriculture University, College of Life Science, Jilin 130118, China
| | - Dandan Zhang
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Wei Liu
- Department F.A. Forel for Environmental and Aquatic Sciences, Section of Earth and Environmental Sciences and Institute for Environmental Sciences, University of Geneva, Switzerland.
| |
Collapse
|
12
|
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
|
13
|
Xu T, Dai Y, Ge A, Chen X, Gong Y, Lam TH, Lee K, Han X, Ji Y, Shen W, Liu J, Sun L, Xu J, Ma B. Ultrafast Evolution of Bacterial Antimicrobial Resistance by Picoliter-Scale Centrifugal Microfluidics. Anal Chem 2024; 96:18842-18851. [PMID: 39531253 DOI: 10.1021/acs.analchem.4c04482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Experimental evolution is a powerful approach for scrutinizing and dissecting the development of antimicrobial resistance; nevertheless, it typically demands an extended duration to detect evolutionary changes. Here, a centrifugal microfluidics system is designed to accelerate the process. Through a simple step of on-chip centrifugation, a highly condensed bacterial matrix of ∼1012 cells/mL at the enrichment tip of the chip channel is derived, enabling bacteria encapsulated to survive in antimicrobial concentrations several times higher than the minimum inhibitory concentration (MIC) and rapidly develop resistance in the first 10 h. After 48 h of on-chip evolution, the E. coli strain demonstrated a 64 to 128-fold reduction in sensitivity to disinfectants (triclosan) as well as antibiotics (ciprofloxacin and amikacin), a rate substantially swifter compared to conventional continuous inoculation-based experimental evolution. The speed and simplicity of this microfluidic system suggest its broad application for uncovering resistance mechanisms and identifying targets of biocides and antibiotics.
Collapse
Affiliation(s)
- Teng Xu
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
| | - Yajie Dai
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
| | - Anle Ge
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
| | - Xueqian Chen
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
| | - Yanhai Gong
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
| | - Tze Hau Lam
- Procter & Gamble Singapore Innovation Center, Singapore 138668, Singapore
| | - Kelvin Lee
- Procter & Gamble Singapore Innovation Center, Singapore 138668, Singapore
| | - Xiao Han
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
| | - Yuetong Ji
- Qingdao Single-Cell Biotech. Ltd., Qingdao, Shandong 266100, China
| | - Wei Shen
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
| | - Jiquan Liu
- Procter & Gamble Singapore Innovation Center, Singapore 138668, Singapore
| | - Luyang Sun
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
- Laboratory of Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266101, China
| | - Jian Xu
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
- Laboratory of Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266101, China
| | - Bo Ma
- Single-Cell Center, Key Laboratory of Photoelectric Conversion and Utilization of Solar Energy, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong 266101, China
- Shandong Energy Institute, Qingdao, Shandong 266101,China
- Laboratory of Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266101, China
| |
Collapse
|
14
|
Vergalli J, Réfrégiers M, Ruggerone P, Winterhalter M, Pagès JM. Advances in methods and concepts provide new insight into antibiotic fluxes across the bacterial membrane. Commun Biol 2024; 7:1508. [PMID: 39543341 PMCID: PMC11564671 DOI: 10.1038/s42003-024-07168-4] [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: 05/16/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
The sophisticated envelope of Gram-negative bacteria modulates the uptake of small molecules in a side-chain-sensitive manner. Despite intensive theoretical and experimental investigations, a general set of pathways underpinning antibiotic uptake has not been identified. This manuscript discusses the passive influx versus active efflux of antibiotics, considering the responsible membrane proteins and the transported molecules. Recent methods have analyzed drug transport across the bacterial membrane in order to understand their activity. The combination of in vitro, in cellulo and in silico methods shed light on the key, mainly electrostatic, interactions between the molecule surface, porins and transporters during permeation. A key factor is the relationship between the dose of an active compound near its target and its antibacterial activity during the critical early window. Today, methodology breakthroughs provide fruitful tools to precisely dissect drug transport, identify key steps in drug resistance associated with membrane impermeability and efflux, and highlight key parameters to generate more effective drugs.
Collapse
Affiliation(s)
| | | | - Paolo Ruggerone
- Department of Physics, University of Cagliari, 09042, Monserrato, CA, Italy
| | - Mathias Winterhalter
- Department of Life Sciences and Chemistry, Constructor University, 28719, Bremen, Germany
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
| | | |
Collapse
|
15
|
Dou L, Wang X, Bai Y, Li Q, Luo L, Yu W, Wang Z, Wen K, Shen J. Mussel-like polydopamine-assisted aggregation-induced emission nanodot for enhanced broad-spectrum antimicrobial activity: In vitro and in vivo validation. Int J Biol Macromol 2024; 282:136762. [PMID: 39486741 DOI: 10.1016/j.ijbiomac.2024.136762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/22/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
Emerging luminogens with aggregation-induced emission properties, namely AIEgens, demonstrated excellent anti-bacteria activity potential. However, their application still limited by the low antibacterial activity caused by the poor binding with bacteria. Polydopamine (PDA), an important biological macromolecule, possesses superior adhesion ability toward various material surface, including bacteria. In this study, the novel mussel-like PDA-assisted AIE Nanodot was proposed, achieving with robust bacterial binding ability and enhanced broad-spectrum antibacterial activity. Binding ability inherited from the PDA enables the constructed PDA-assisted AIE Nanodot to adhere efficiently to the bacterial membrane surface. Meanwhile, the AIE properties endowed them with monitoring capability, allowing for tracking their interaction with bacteria through facile fluorescence imaging in real time. More importantly, excellent killing of both Gram-positive and Gram-negative bacteria were successfully achieved in vitro antibacterial tests with excellent biocompatibility. Furthermore, in the treatment of Methicillin-resistant S. aureus (MRSA)-infected mouse-wound model, the mice exhibited accelerated wound healing with low bacterial load. This novel integrated strategy introduced a simple but effectively design to enhance the binding and antibacterial ability of AIEgens and would diversify the existing pool of antibacterial agents.
Collapse
Affiliation(s)
- Leina Dou
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China; College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shanxi, China
| | - Xiaonan Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Yuchen Bai
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Liang Luo
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Wenbo Yu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Kai Wen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China.
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China.
| |
Collapse
|
16
|
Wen T, Xiong S, Zhao H, Wang J, Wang C, Long Z, Xiong L, Qian G. Polylactic acid-based dressing with oxygen generation and enzyme-like activity for accelerating both light-driven biofilm elimination and wound healing. BURNS & TRAUMA 2024; 12:tkae041. [PMID: 39464502 PMCID: PMC11510456 DOI: 10.1093/burnst/tkae041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 10/29/2024]
Abstract
Background Photodynamic therapy (PDT) is a widely used therapeutic approach for eradicating bacterial biofilms in infected wound, but its effectiveness is limited by the hypoxic environment within the biofilm. This study aimed to investigate whether the efficiency of photodynamic removing biofilm is improving by providing oxygen (O2), as well as the expression of cytokines involved in infected wound healing. Methods Manganese dioxide (MnO2) nanoparticles with catalase-like activity were grown in situ on graphitic phase carbon nitride (g-C3N4, CN) nanosheets to construct an all-in-one CN-MnO2 nanozyme, which was then incorporated into poly-L-lactic acid (PLLA) to prepare CN-MnO2/PLLA wound dressing by electrospinning. Subsequently, the in vitro antibacterial biofilm ratio and antibacterial ratio of CN-MnO2/PLLA wound dressing were examined by spread plate and crystal violet staining under irradiation with 808 nm near-infrared light and 660 nm visible light. Meanwhile, the rat skin injury model was established, and hematoxylin and eosin (H&E), Masson's, tumor necrosis factor-α (TNF-α), Arginase 1 (Arg-1), vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (BFGF) were evaluated in vivo to assess the effect of CN-MnO2/PLLA wound dressing on wound healing. Results Biofilm density caused by Staphylococcus aureus and Pseudomonas aeruginosa had elimination rates of 83 and 62%, respectively, when treated with CN-MnO2/PLLA dressing. Additionally, the dressing exhibited high antibacterial efficacy against both bacteria, achieving 99 and 98.7% elimination of Staphylococcus aureus and Pseudomonas aeruginosa, respectively. Furthermore, in vivo experiments showed that the CN-MnO2/PLLA wound dressing achieved complete healing of infected wounds on Day 14, with a wound healing rate of >99% by increasing collagen deposition, expression of anti-inflammatory cytokine Arg-1, vascularization cytokine VEGF, and epithelial cell BFGF, and inhibiting the expression of inflammatory cytokine TNF-α. Conclusions The CN-MnO2/PLLA wound dressing exhibited excellent antibacterial properties in vitro and in vivo. In addition, CN-MnO2/PLLA wound dressing accelerated rapid wound healing through an anti-inflammatory, pro-vascular regeneration and skin tissue remodeling mechanism.
Collapse
Affiliation(s)
- Tianci Wen
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, No. 1180, Shuanggang East Street, Qingshanhu District, Nanchang City, Jiangxi Province 330013, P.R. China
| | - Shilang Xiong
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang 330006, P.R. China
| | - Huihui Zhao
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, No. 1180, Shuanggang East Street, Qingshanhu District, Nanchang City, Jiangxi Province 330013, P.R. China
| | - Junzhe Wang
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, No. 1180, Shuanggang East Street, Qingshanhu District, Nanchang City, Jiangxi Province 330013, P.R. China
| | - Chunming Wang
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, No. 1180, Shuanggang East Street, Qingshanhu District, Nanchang City, Jiangxi Province 330013, P.R. China
| | - Zhisheng Long
- Department of Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No. 152, Aiguo Road, Donghu District, Nanchang City, Jiangxi Province 330006, P.R. China
| | - Long Xiong
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang City, Jiangxi Province 330008, P.R. China
| | - Guowen Qian
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, No. 1180, Shuanggang East Street, Qingshanhu District, Nanchang City, Jiangxi Province 330013, P.R. China
| |
Collapse
|
17
|
Abdollahi M, Baharian A, Mohamadhoseini M, Hassanpour M, Makvandi P, Habibizadeh M, Jafari B, Nouri R, Mohamadnia Z, Nikfarjam N. Advances in ionic liquid-based antimicrobial wound healing platforms. J Mater Chem B 2024; 12:9478-9507. [PMID: 39206539 DOI: 10.1039/d4tb00841c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Wound infections, marked by the proliferation of microorganisms at surgical sites, necessitate the development of innovative wound dressings with potent bactericidal properties to curb microbial growth and prevent bacterial infiltration. This study explores the recent strides in utilizing ionic liquid-based polymers as highly promising antimicrobial agents for advanced wound healing applications. Specifically, cationic polymers containing quaternary ammonium, imidazolium, guanidinium, pyridinium, triazolium, or phosphonium groups have emerged as exceptionally effective antimicrobial compounds. Their mechanism of action involves disrupting bacterial membranes, thereby preventing the development of resistance and minimizing toxicity to mammalian cells. This comprehensive review not only elucidates the intricate dynamics of the skin's immune response and the various stages of wound healing but also delves into the synthesis methodologies of ionic liquid-based polymers. By spotlighting the practical applications of antimicrobial wound dressings, particularly those incorporating ionic liquid-based materials, this review aims to lay the groundwork for future research endeavors in this burgeoning field. Through a nuanced examination of these advancements, this article seeks to contribute to the ongoing progress in developing cutting-edge wound healing platforms that can effectively address the challenges posed by microbial infections in surgical wounds.
Collapse
Affiliation(s)
- Mahin Abdollahi
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Aysan Baharian
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Masoumeh Mohamadhoseini
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Mahnaz Hassanpour
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Pooyan Makvandi
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3JL, UK
| | - Mina Habibizadeh
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Bahman Jafari
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Roya Nouri
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Zahra Mohamadnia
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Nasser Nikfarjam
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
- Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina, Columbia 29208, SC, USA
| |
Collapse
|
18
|
Biswas T, Ahmed M, Mondal S. Mixed species biofilm: Structure, challenge and its intricate involvement in hospital associated infections. Microb Pathog 2024; 195:106866. [PMID: 39159773 DOI: 10.1016/j.micpath.2024.106866] [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: 05/07/2024] [Revised: 08/06/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Hospital associated infections or healthcare associated infections (HAIs) are a major threat to healthcare and medical management, mostly because of their recalcitrant nature. The primary cause of these HAIs is bacterial associations, especially the interspecies interactions. In interspecies interactions, more than one species co-exists in a common platform of extracellular polymeric substances (EPS), establishing a strong interspecies crosstalk and thereby lead to the formation of mixed species biofilms. In this process, the internal microenvironment and the surrounding EPS matrix of the biofilms ensure the protection of the microorganisms and allow them to survive under antagonistic conditions. The communications between the biofilm members as well as the interactions between the bacterial cells and the matrix polymers, also aid in the rigidity of the biofilm structure and allow the microorganisms to evade both the host immune response and a wide range of anti-microbials. Therefore, to design a treatment protocol for HAIs is difficult and it has become a growing point of concern. This review therefore first aims to discuss the role of microenvironment, molecular structure, cell-cell communication, and metabolism of mixed species biofilms in manifestation of HAIs. In addition, we discuss the electrochemical properties of mixed-species biofilms and their mechanism in developing drug resistance. Then we focus on the most dreaded bacterial HAI including oral and gut multi-species infections, catheter-associated urinary tract infections, surgical site infections, and ventilator-associated pneumonia. Further, we highlight the challenges to eradication of the mixed species biofilms and the current and prospective future strategies for the treatment of mixed species-associated HAI. Together, the review presents a comprehensive understanding of mixed species biofilm-mediated infections in clinical scenario, and summarizes the current challenge and prospect of therapeutic strategies against HAI.
Collapse
Affiliation(s)
| | - Mehnaz Ahmed
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Susmita Mondal
- Department of Life Sciences, Presidency University, Kolkata, India.
| |
Collapse
|
19
|
Abbara A, Pagani L, García-Pareja C, Bitbol AF. Mutant fate in spatially structured populations on graphs: Connecting models to experiments. PLoS Comput Biol 2024; 20:e1012424. [PMID: 39241045 PMCID: PMC11410244 DOI: 10.1371/journal.pcbi.1012424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 09/18/2024] [Accepted: 08/15/2024] [Indexed: 09/08/2024] Open
Abstract
In nature, most microbial populations have complex spatial structures that can affect their evolution. Evolutionary graph theory predicts that some spatial structures modelled by placing individuals on the nodes of a graph affect the probability that a mutant will fix. Evolution experiments are beginning to explicitly address the impact of graph structures on mutant fixation. However, the assumptions of evolutionary graph theory differ from the conditions of modern evolution experiments, making the comparison between theory and experiment challenging. Here, we aim to bridge this gap by using our new model of spatially structured populations. This model considers connected subpopulations that lie on the nodes of a graph, and allows asymmetric migrations. It can handle large populations, and explicitly models serial passage events with migrations, thus closely mimicking experimental conditions. We analyze recent experiments in light of this model. We suggest useful parameter regimes for future experiments, and we make quantitative predictions for these experiments. In particular, we propose experiments to directly test our recent prediction that the star graph with asymmetric migrations suppresses natural selection and can accelerate mutant fixation or extinction, compared to a well-mixed population.
Collapse
Affiliation(s)
- Alia Abbara
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Lisa Pagani
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Celia García-Pareja
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Anne-Florence Bitbol
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
20
|
Krajewska J, Tyski S, Laudy AE. In Vitro Resistance-Predicting Studies and In Vitro Resistance-Related Parameters-A Hit-to-Lead Perspective. Pharmaceuticals (Basel) 2024; 17:1068. [PMID: 39204172 PMCID: PMC11357384 DOI: 10.3390/ph17081068] [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: 07/26/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Despite the urgent need for new antibiotics, very few innovative antibiotics have recently entered clinics or clinical trials. To provide a constant supply of new drug candidates optimized in terms of their potential to select for resistance in natural settings, in vitro resistance-predicting studies need to be improved and scaled up. In this review, the following in vitro parameters are presented: frequency of spontaneous mutant selection (FSMS), mutant prevention concentration (MPC), dominant mutant prevention concentration (MPC-D), inferior-mutant prevention concentration (MPC-F), and minimal selective concentration (MSC). The utility of various adaptive laboratory evolution (ALE) approaches (serial transfer, continuous culture, and evolution in spatiotemporal microenvironments) for comparing hits in terms of the level and time required for multistep resistance to emerge is discussed. We also consider how the hit-to-lead stage can benefit from high-throughput ALE setups based on robotic workstations, do-it-yourself (DIY) continuous cultivation systems, microbial evolution and growth arena (MEGA) plates, soft agar gradient evolution (SAGE) plates, microfluidic chips, or microdroplet technology. Finally, approaches for evaluating the fitness of in vitro-generated resistant mutants are presented. This review aims to draw attention to newly emerged ideas on how to improve the in vitro forecasting of the potential of compounds to select for resistance in natural settings.
Collapse
Affiliation(s)
- Joanna Krajewska
- Department of Environmental Health and Safety, National Institute of Public Health NIH—National Research Institute, 00-791 Warsaw, Poland;
| | - Stefan Tyski
- Department of Pharmaceutical Microbiology and Laboratory Diagnostic, National Medicines Institute, 00-725 Warsaw, Poland;
| | - Agnieszka E. Laudy
- Department of Pharmaceutical Microbiology and Bioanalysis, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
21
|
Rolff J, Bonhoeffer S, Kloft C, Leistner R, Regoes R, Hochberg ME. Forecasting antimicrobial resistance evolution. Trends Microbiol 2024; 32:736-745. [PMID: 38238231 DOI: 10.1016/j.tim.2023.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 08/09/2024]
Abstract
Antimicrobial resistance (AMR) is a major global health issue. Current measures for tackling it comprise mainly the prudent use of drugs, the development of new drugs, and rapid diagnostics. Relatively little attention has been given to forecasting the evolution of resistance. Here, we argue that forecasting has the potential to be a great asset in our arsenal of measures to tackle AMR. We argue that, if successfully implemented, forecasting resistance will help to resolve the antibiotic crisis in three ways: it will (i) guide a more sustainable use (and therefore lifespan) of antibiotics and incentivize investment in drug development, (ii) reduce the spread of AMR genes and pathogenic microbes in the environment and between patients, and (iii) allow more efficient treatment of persistent infections, reducing the continued evolution of resistance. We identify two important challenges that need to be addressed for the successful establishment of forecasting: (i) the development of bespoke technology that allows stakeholders to empirically assess the risks of resistance evolving during the process of drug development and therapeutic/preventive use, and (ii) the transformative shift in mindset from the current praxis of mostly addressing the problem of antibiotic resistance a posteriori to a concept of a priori estimating, and acting on, the risks of resistance.
Collapse
Affiliation(s)
- Jens Rolff
- Evolutionary Biology, Institute of Biology, Freie Universität Berlin, Berlin, Germany.
| | | | - Charlotte Kloft
- Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Rasmus Leistner
- Charité-Universitätsmedizin Berlin Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Berlin, Germany
| | - Roland Regoes
- Institute of Integrative Biology, ETH Zurich, 8092 Zurich, Switzerland
| | - Michael E Hochberg
- ISEM, Université de Montpellier, CNRS, IRD, EPHE, 34095 Montpellier, France; Santa Fe Institute, Santa Fe, NM 87501, USA
| |
Collapse
|
22
|
Abou-Jaoudeh C, Khalil J, El-Hayek E, Abou-Khalil R. Food safety control in poultry industry: prevalence and antimicrobial susceptibility of Escherichia coli isolated from raw chicken and the potential use of Origanum essential oils as alternative to antibiotics. Br Poult Sci 2024; 65:494-501. [PMID: 38994872 DOI: 10.1080/00071668.2024.2346307] [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: 09/13/2023] [Accepted: 03/28/2024] [Indexed: 07/13/2024]
Abstract
1. The extensive use of antimicrobials in poultry production may contribute to the emergence of resistant bacteria. This study was conducted to determine the prevalence and resistance of different E. coli strains isolated from raw chicken meat and to investigate the possibility to use Lebanese native oregano essential oils as alternatives.2. In total, 250 chickens from Lebanese markets were examined for the presence of E. coli. Isolates were then screened for susceptibility using 19 antibiotics and two essential oils extracted from oregano plants.3. Of the 250 chickens tested, 80% were contaminated with E. coli. Main resistance was seen against amoxycillin, ampicillin, penicillin, tetracycline, tylosin, streptomycin and erythromycin. The highest rate of sensitivity was found in 86.1% of strains to Amoxycillin/Clavulanic acid, 80.09% to Tilmicosin. Both essential oils from Origanum syriacum (98%) and O. ehrenbergii (97.3%) showed promising potential in inhibiting the growth of the tested bacteria. Oil from O. syriacum exhibited superior efficacy against 200 E. coli strains, inhibiting 46.1% at 200 mg/l and all at 400 mg/l, while O. ehrenbergii oil showed slightly lower inhibition, affecting 41.6% at 200 mg/l and all at 400 mg/l.
Collapse
Affiliation(s)
- C Abou-Jaoudeh
- Faculty of Arts and Sciences, Biology Department, Holy Spirit University of Kaslik, Jounieh, Lebanon
| | - J Khalil
- Faculty of Arts and Sciences, Biology Department, Holy Spirit University of Kaslik, Jounieh, Lebanon
| | - E El-Hayek
- Faculty of Arts and Sciences, Biology Department, Holy Spirit University of Kaslik, Jounieh, Lebanon
| | - R Abou-Khalil
- Faculty of Arts and Sciences, Biology Department, Holy Spirit University of Kaslik, Jounieh, Lebanon
| |
Collapse
|
23
|
Zhu L, Xu Y, Li J, Lin G, Han X, Yi J, Jayaprada T, Zhou Z, Ying Y, Wang M. Environmentally persistent microbial contamination in agricultural soils: High risk of pathogenicity and antibiotic resistance. ENVIRONMENT INTERNATIONAL 2024; 190:108902. [PMID: 39059024 DOI: 10.1016/j.envint.2024.108902] [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: 05/29/2024] [Revised: 07/20/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024]
Abstract
Persistent microbial contamination commonly occurs in the environment. However, the characteristics and associated risks remain largely unknown. The coexistence of virulence factor genes (VFGs) and "last-resort" antibiotic resistance genes (LARGs) on human bacterial pathogens (HBPs) are notorious, creating ecological concerns and health risks. Herein, we explored the pathogenicity and antibiotic resistance levels of LARG-harboring HBPs in agricultural soils. Our findings revealed a high distribution level of VFGs and LARGs in soils (an absolute abundance up to 4.7 × 107 gene copies/g soil) by quantitative PCR (qPCR). Furthermore, most isolated LARG-harboring HBPs exhibited a 100 % lethality rate to Galleria mellonella. LARG-carrying plasmids had a low fitness cost to their host bacteria, implying the high adaptation of these plasmids within the HBPs. Most importantly, multiple LARG and VFG plasmid fusion and core genetic arrangements suggested that these LARG/VFG-linked plasmids endowed the stable and persistent horizontal spread of these genes in and/or cross the species and environments. This study not only unveiled high risk, multisource, compliance and stability aspects of environmentally persistent microbial contamination but also illuminated the importance of linking the phenotype-genotype-niche colonization of environmental microbial contamination within "One Health" framework.
Collapse
Affiliation(s)
- Lin Zhu
- International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development & Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Yongchang Xu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Jingpeng Li
- International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development & Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Guoping Lin
- International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development & Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Xuezhu Han
- International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development & Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Jiaming Yi
- International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development & Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Thilini Jayaprada
- International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development & Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China
| | - Zhenchao Zhou
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yiqian Ying
- Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Meizhen Wang
- International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development & Zhejiang Provincial Key Laboratory of Solid Waste Treatment and Recycling, School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310012, China.
| |
Collapse
|
24
|
Mohamed DS, Abd El-Baky RM, El-Mokhtar MA, Ghanem SK, Yahia R, Alqahtani AM, Abourehab MAS, Ahmed EF. Influence of selected non-antibiotic pharmaceuticals on antibiotic resistance gene transfer in Escherichia coli. PLoS One 2024; 19:e0304980. [PMID: 38905247 PMCID: PMC11192386 DOI: 10.1371/journal.pone.0304980] [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: 04/06/2023] [Accepted: 05/20/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Antibiotic resistance genes (ARGs) transfer rapidly among bacterial species all over the world contributing to the aggravation of antibiotic resistance crisis. Antibiotics at sub-inhibitory concentration induce horizontal gene transfer (HRT) between bacteria, especially through conjugation. The role of common non-antibiotic pharmaceuticals in the market in disseminating antibiotic resistance is not well studied. OBJECTIVES In this work, we indicated the effect of some commonly used non-antibiotic pharmaceuticals including antiemetic (metoclopramide HCl) and antispasmodics (hyoscine butyl bromide and tiemonium methyl sulfate) on the plasmid-mediated conjugal transfer of antibiotic resistance genes between pathogenic E. coli in the gastric intestinal tract (GIT). METHODS Broth microdilution assay was used to test the antibacterial activity of the tested non-antibiotic pharmaceuticals. A conjugation mating system was applied in presence of the studied non-antibiotic pharmaceuticals to test their effect on conjugal transfer frequency. Plasmid extraction and PCR were performed to confirm the conjugation process. Transmission electron microscopy (TEM) was used for imaging the effect of non-antibiotic pharmaceuticals on bacterial cells. RESULTS No antibacterial activity was reported for the used non-antibiotic pharmaceuticals. Plasmid-mediated conjugal transfer between isolates was induced by metoclopramide HCl but suppressed by hyoscine butyl bromide. Tiemonium methylsulfate slightly promoted conjugal transfer. Aggregation between cells and periplasmic bridges was clear in the case of metoclopramide HCl while in presence of hyoscine butyl bromide little affinity was observed. CONCLUSION This study indicates the contribution of non-antibiotic pharmaceuticals to the dissemination and evolution of antibiotic resistance at the community level. Metoclopramide HCl showed an important role in the spread of antibiotic resistance.
Collapse
Affiliation(s)
- Doaa Safwat Mohamed
- Microbiology & Immunology Department, Faculty of Pharmacy, Sohag University, Sohag Al Gadida City, Egypt
| | - Rehab Mahmoud Abd El-Baky
- Microbiology & Immunology Department, Faculty of Pharmacy, Minia University, Minia, Egypt
- Microbiology and Immunology Department, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Mohamed Ahmed El-Mokhtar
- Medical Microbiology & Immunology Department, Faculty of Medicine, Assiut University, El Fateh, Egypt
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Sahar K. Ghanem
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Sohag University, Sohag Al Gadida City, Egypt
| | - Ramadan Yahia
- Microbiology and Immunology Department, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Alaa M. Alqahtani
- Pharmaceutical Chemistry Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Eman Farouk Ahmed
- Microbiology & Immunology Department, Faculty of Pharmacy, Sohag University, Sohag Al Gadida City, Egypt
| |
Collapse
|
25
|
Nguyen AV, Yaghoobi M, Zhang S, Li P, Li Q, Dogan B, Ahnrud GP, Flock G, Marek P, Simpson KW, Abbaspourrad A. Adaptive Laboratory Evolution of Probiotics toward Oxidative Stress Using a Microfluidic-Based Platform. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306974. [PMID: 38247174 DOI: 10.1002/smll.202306974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/28/2023] [Indexed: 01/23/2024]
Abstract
Adaptive laboratory evolution (ALE) can be used to make bacteria less susceptible to oxidative stress. An alternative to large batch scale ALE cultures is to use microfluidic platforms, which are often more economical and more efficient. Microfluidic ALE platforms have shown promise, but many have suffered from subpar cell passaging mechanisms and poor spatial definition. A new approach is presented using a microfluidic Evolution on a Chip (EVoc) design which progressively drives microbial cells from areas of lower H2O2 concentration to areas of higher concentration. Prolonged exposure, up to 72 h, revealed the survival of adaptive strains of Lacticaseibacillus rhamnosus GG, a beneficial probiotic often included in food products. After performing ALE on this microfluidic platform, the bacteria persisted under high H2O2 concentrations in repeated trials. After two progressive exposures, the ability of L. rhamnosus to grow in the presence of H2O2 increased from 1 mm H2O2 after a lag time of 31 h to 1 mm after 21 h, 2 mm after 28 h, and 3 mm after 42 h. The adaptive strains have different morphology, and gene expression compared to wild type, and genome sequencing revealed a potentially meaningful single nucleotide mutation in the protein omega-amidase.
Collapse
Affiliation(s)
- Ann V Nguyen
- Department of Food Science, College of Agricultural and Life Sciences, Cornell University, Stocking Hall, Ithaca, NY, 14853, USA
| | - Mohammad Yaghoobi
- Department of Food Science, College of Agricultural and Life Sciences, Cornell University, Stocking Hall, Ithaca, NY, 14853, USA
| | - Shiying Zhang
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, 602 Tower Rd., Ithaca, NY, 14853, USA
| | - Peilong Li
- Department of Food Science, College of Agricultural and Life Sciences, Cornell University, Stocking Hall, Ithaca, NY, 14853, USA
| | - Qike Li
- Department of Food Science, College of Agricultural and Life Sciences, Cornell University, Stocking Hall, Ithaca, NY, 14853, USA
| | - Belgin Dogan
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, 602 Tower Rd., Ithaca, NY, 14853, USA
| | - Gianna P Ahnrud
- DEVCOM Soldier Center, Soldier Sustainment Directorate, Combat Feeding Division, Food Protection & Innovative Packaging Team, Natick, MA, 01760, USA
| | - Genevieve Flock
- DEVCOM Soldier Center, Soldier Sustainment Directorate, Combat Feeding Division, Food Protection & Innovative Packaging Team, Natick, MA, 01760, USA
| | - Patrick Marek
- DEVCOM Soldier Center, Soldier Sustainment Directorate, Combat Feeding Division, Food Protection & Innovative Packaging Team, Natick, MA, 01760, USA
| | - Kenneth W Simpson
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, 602 Tower Rd., Ithaca, NY, 14853, USA
| | - Alireza Abbaspourrad
- Department of Food Science, College of Agricultural and Life Sciences, Cornell University, Stocking Hall, Ithaca, NY, 14853, USA
| |
Collapse
|
26
|
A Malik A, Dangroo NA, Kaur P, Attery S, A Rather M, Khan A, Ara T, Nandanwar H. Discovery of novel dihydronaphthalene-imidazole ligands as potential inhibitors of Staphylococcus aureus multidrug resistant NorA efflux pump: A combination of experimental and in silico molecular docking studies. Microb Pathog 2024; 190:106627. [PMID: 38521473 DOI: 10.1016/j.micpath.2024.106627] [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: 12/19/2023] [Revised: 02/22/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024]
Abstract
Overexpression of the efflux pump is a predominant mechanism by which bacteria show antimicrobial resistance (AMR) and leads to the global emergence of multidrug resistance (MDR). In this work, the inhibitory potential of library of dihydronapthyl scaffold-based imidazole derivatives having structural resemblances with some known efflux pump inhibitors (EPI) were designed, synthesized and evaluated against efflux pump inhibitor against overexpressing bacterial strains to study the synergistic effect of compounds and antibiotics. Out of 15 compounds, four compounds (Dz-1, Dz-3, Dz-7, and Dz-8) were found to be highly active. DZ-3 modulated the MIC of ciprofloxacin, erythromycin, and tetracycline by 128-fold each against 1199B, XU212 and RN4220 strains of S. aureus respectively. DZ-3 also potentiated tetracycline by 64-fold in E. coli AG100 strain. DZ-7 modulated the MIC of both tetracycline and erythromycin 128-fold each in S. aureus XU212 and S. aureus RN4220 strains. DZ-1 and DZ-8 showed the moderate reduction in MIC of tetracycline in E. coli AG100 only by 16-fold and 8-fold, respectively. DZ-3 was found to be the potential inhibitor of NorA as determined by ethidium bromide efflux inhibition and accumulation studies employing NorA overexpressing strain SA-1199B. DZ-3 displayed EPI activity at non-cytotoxic concentration to human cells and did not possess any antibacterial activity. Furthermore, molecular docking studies of DZ-3 was carried out in order to understand the possible binding sites of DZ-3 with the active site of the protein. These studies indicate that dihydronaphthalene scaffolds could serve as valuable cores for the development of promising EPIs.
Collapse
Affiliation(s)
- Asif A Malik
- Department of Chemistry, National Institute of Technology, Srinagar, J&K, 190006, India
| | - Nisar A Dangroo
- Department of Chemistry, Islamic University of Science and Technology, Awantipora, J &K, 192122, India.
| | - Parminder Kaur
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Shobit Attery
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Manzoor A Rather
- Department of Chemistry, Islamic University of Science and Technology, Awantipora, J &K, 192122, India.
| | - Abrar Khan
- Department of Chemistry, National Institute of Technology, Srinagar, J&K, 190006, India
| | - Tabassum Ara
- Department of Chemistry, National Institute of Technology, Srinagar, J&K, 190006, India.
| | - Hemraj Nandanwar
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India.
| |
Collapse
|
27
|
Freire TFA, Hu Z, Wood KB, Gjini E. Modeling spatial evolution of multi-drug resistance under drug environmental gradients. PLoS Comput Biol 2024; 20:e1012098. [PMID: 38820350 PMCID: PMC11142541 DOI: 10.1371/journal.pcbi.1012098] [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: 11/17/2023] [Accepted: 04/23/2024] [Indexed: 06/02/2024] Open
Abstract
Multi-drug combinations to treat bacterial populations are at the forefront of approaches for infection control and prevention of antibiotic resistance. Although the evolution of antibiotic resistance has been theoretically studied with mathematical population dynamics models, extensions to spatial dynamics remain rare in the literature, including in particular spatial evolution of multi-drug resistance. In this study, we propose a reaction-diffusion system that describes the multi-drug evolution of bacteria based on a drug-concentration rescaling approach. We show how the resistance to drugs in space, and the consequent adaptation of growth rate, is governed by a Price equation with diffusion, integrating features of drug interactions and collateral resistances or sensitivities to the drugs. We study spatial versions of the model where the distribution of drugs is homogeneous across space, and where the drugs vary environmentally in a piecewise-constant, linear and nonlinear manner. Although in many evolution models, per capita growth rate is a natural surrogate for fitness, in spatially-extended, potentially heterogeneous habitats, fitness is an emergent property that potentially reflects additional complexities, from boundary conditions to the specific spatial variation of growth rates. Applying concepts from perturbation theory and reaction-diffusion equations, we propose an analytical metric for characterization of average mutant fitness in the spatial system based on the principal eigenvalue of our linear problem, λ1. This enables an accurate translation from drug spatial gradients and mutant antibiotic susceptibility traits to the relative advantage of each mutant across the environment. Our approach allows one to predict the precise outcomes of selection among mutants over space, ultimately from comparing their λ1 values, which encode a critical interplay between growth functions, movement traits, habitat size and boundary conditions. Such mathematical understanding opens new avenues for multi-drug therapeutic optimization.
Collapse
Affiliation(s)
- Tomas Ferreira Amaro Freire
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Zhijian Hu
- Departments of Biophysics and Physics, University of Michigan, United States of America
| | - Kevin B. Wood
- Departments of Biophysics and Physics, University of Michigan, United States of America
| | - Erida Gjini
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
28
|
Hancock AM, Datta SS. Interplay between environmental yielding and dynamic forcing modulates bacterial growth. Biophys J 2024; 123:957-967. [PMID: 38454600 PMCID: PMC11052696 DOI: 10.1016/j.bpj.2024.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024] Open
Abstract
Many bacterial habitats-ranging from gels and tissues in the body to cell-secreted exopolysaccharides in biofilms-are rheologically complex, undergo dynamic external forcing, and have unevenly distributed nutrients. How do these features jointly influence how the resident cells grow and proliferate? Here, we address this question by studying the growth of Escherichia coli dispersed in granular hydrogel matrices with defined and highly tunable structural and rheological properties, under different amounts of external forcing imposed by mechanical shaking, and in both aerobic and anaerobic conditions. Our experiments establish a general principle: that the balance between the yield stress of the environment that the cells inhabit, σy, and the external stress imposed on the environment, σ, modulates bacterial growth by altering transport of essential nutrients to the cells. In particular, when σy<σ, the environment is easily fluidized and mixed over large scales, providing nutrients to the cells and sustaining complete cellular growth. By contrast, when σy>σ, the elasticity of the environment suppresses large-scale fluid mixing, limiting nutrient availability and arresting cellular growth. Our work thus reveals a new mechanism, beyond effects that change cellular behavior via local forcing, by which the rheology of the environment may modulate microbial physiology in diverse natural and industrial settings.
Collapse
Affiliation(s)
- Anna M Hancock
- Chemical and Biological Engineering, Princeton University, Princeton, New Jersey
| | - Sujit S Datta
- Chemical and Biological Engineering, Princeton University, Princeton, New Jersey.
| |
Collapse
|
29
|
Liu X, Li J, Zhang Z, He Y, Wang M, Zhao Y, Lin S, Liu T, Liao Y, Zhang N, Yuan K, Ling Y, Liu Z, Chen X, Chen Z, Chen R, Wang X, Gu B. Acetylation of xenogeneic silencer H-NS regulates biofilm development through the nitrogen homeostasis regulator in Shewanella. Nucleic Acids Res 2024; 52:2886-2903. [PMID: 38142446 PMCID: PMC11014242 DOI: 10.1093/nar/gkad1219] [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: 02/14/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/26/2023] Open
Abstract
Adjusting intracellular metabolic pathways and adopting suitable live state such as biofilms, are crucial for bacteria to survive environmental changes. Although substantial progress has been made in understanding how the histone-like nucleoid-structuring (H-NS) protein modulates the expression of the genes involved in biofilm formation, the precise modification that the H-NS protein undergoes to alter its DNA binding activity is still largely uncharacterized. This study revealed that acetylation of H-NS at Lys19 inhibits biofilm development in Shewanella oneidensis MR-1 by downregulating the expression of glutamine synthetase, a critical enzyme in glutamine synthesis. We further found that nitrogen starvation, a likely condition in biofilm development, induces deacetylation of H-NS and the trimerization of nitrogen assimilation regulator GlnB. The acetylated H-NS strain exhibits significantly lower cellular glutamine concentration, emphasizing the requirement of H-NS deacetylation in Shewanella biofilm development. Moreover, we discovered in vivo that the activation of glutamine biosynthesis pathway and the concurrent suppression of the arginine synthesis pathway during both pellicle and attached biofilms development, further suggesting the importance of fine tune nitrogen assimilation by H-NS acetylation in Shewanella. In summary, posttranslational modification of H-NS endows Shewanella with the ability to respond to environmental needs by adjusting the intracellular metabolism pathways.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
| | - Jun Li
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Zhixuan Zhang
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510080, China
| | - Yizhou He
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
| | - Mingfang Wang
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Yunhu Zhao
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Shituan Lin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianlang Liu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiwen Liao
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Ni Zhang
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Kaixuan Yuan
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Yong Ling
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Ziyao Liu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaozhong Chen
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Zhe Chen
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ran Chen
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
| | - Xiaoxue Wang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Gu
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| |
Collapse
|
30
|
Yoon Y, Song S. Structural Insights into the Lipopolysaccharide Transport (Lpt) System as a Novel Antibiotic Target. J Microbiol 2024; 62:261-275. [PMID: 38816673 DOI: 10.1007/s12275-024-00137-w] [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: 03/19/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 06/01/2024]
Abstract
Lipopolysaccharide (LPS) is a critical component of the extracellular leaflet within the bacterial outer membrane, forming an effective physical barrier against environmental threats in Gram-negative bacteria. After LPS is synthesized and matured in the bacterial cytoplasm and the inner membrane (IM), LPS is inserted into the outer membrane (OM) through the ATP-driven LPS transport (Lpt) pathway, which is an energy-intensive process. A trans-envelope complex that contains seven Lpt proteins (LptA-LptG) is crucial for extracting LPS from the IM and transporting it across the periplasm to the OM. The last step in LPS transport involves the mediation of the LptDE complex, facilitating the insertion of LPS into the outer leaflet of the OM. As the Lpt system plays an essential role in maintaining the impermeability of the OM via LPS decoration, the interactions between these interconnected subunits, which are meticulously regulated, may be potential targets for the development of new antibiotics to combat multidrug-resistant Gram-negative bacteria. In this review, we aimed to provide an overview of current research concerning the structural interactions within the Lpt system and their implications to clarify the function and regulation of LPS transport in the overall process of OM biogenesis. Additionally, we explored studies on the development of therapeutic inhibitors of LPS transport, the factors that limit success, and future prospects.
Collapse
Affiliation(s)
- Yurim Yoon
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Saemee Song
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea.
| |
Collapse
|
31
|
Rodrigues M, Sabaeifard P, Yildiz MS, Lyon A, Coughlin L, Ahmed S, Poulides N, Toprak AC, Behrendt C, Wang X, Monogue M, Kim J, Gan S, Zhan X, Filkins L, Williams NS, Hooper LV, Koh AY, Toprak E. Susceptible bacteria can survive antibiotic treatment in the mammalian gastrointestinal tract without evolving resistance. Cell Host Microbe 2024; 32:396-410.e6. [PMID: 38359828 PMCID: PMC10942764 DOI: 10.1016/j.chom.2024.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 12/13/2023] [Accepted: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Antibiotic resistance and evasion are incompletely understood and complicated by the fact that murine interval dosing models do not fully recapitulate antibiotic pharmacokinetics in humans. To better understand how gastrointestinal bacteria respond to antibiotics, we colonized germ-free mice with a pan-susceptible genetically barcoded Escherichia coli clinical isolate and administered the antibiotic cefepime via programmable subcutaneous pumps, allowing closer emulation of human parenteral antibiotic dynamics. E. coli was only recovered from intestinal tissue, where cefepime concentrations were still inhibitory. Strikingly, "some" E. coli isolates were not cefepime resistant but acquired mutations in genes involved in polysaccharide capsular synthesis increasing their invasion and survival within human intestinal cells. Deleting wbaP involved in capsular polysaccharide synthesis mimicked this phenotype, allowing increased invasion of colonocytes where cefepime concentrations were reduced. Additionally, "some" mutant strains exhibited a persister phenotype upon further cefepime exposure. This work uncovers a mechanism allowing "select" gastrointestinal bacteria to evade antibiotic treatment.
Collapse
Affiliation(s)
- Marinelle Rodrigues
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Parastoo Sabaeifard
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Muhammed Sadik Yildiz
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adam Lyon
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laura Coughlin
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sara Ahmed
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nicole Poulides
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ahmet C Toprak
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Cassie Behrendt
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaoyu Wang
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Marguerite Monogue
- Department of Pharmacy, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jiwoong Kim
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shuheng Gan
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaowei Zhan
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laura Filkins
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Noelle S Williams
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lora V Hooper
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; The Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrew Y Koh
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Erdal Toprak
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Lyda Hill Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
32
|
Maltas J, Tadele DS, Durmaz A, McFarland CD, Hinczewski M, Scott JG. Frequency-dependent ecological interactions increase the prevalence, and shape the distribution, of pre-existing drug resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.16.533001. [PMID: 36993678 PMCID: PMC10055114 DOI: 10.1101/2023.03.16.533001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The evolution of resistance remains one of the primary challenges for modern medicine from infectious diseases to cancers. Many of these resistance-conferring mutations often carry a substantial fitness cost in the absence of treatment. As a result, we would expect these mutants to undergo purifying selection and be rapidly driven to extinction. Nevertheless, pre-existing resistance is frequently observed from drug-resistant malaria to targeted cancer therapies in non-small cell lung cancer (NSCLC) and melanoma. Solutions to this apparent paradox have taken several forms from spatial rescue to simple mutation supply arguments. Recently, in an evolved resistant NSCLC cell line, we found that frequency-dependent ecological interactions between ancestor and resistant mutant ameliorate the cost of resistance in the absence of treatment. Here, we hypothesize that frequency-dependent ecological interactions in general play a major role in the prevalence of pre-existing resistance. We combine numerical simulations with robust analytical approximations to provide a rigorous mathematical framework for studying the effects of frequency-dependent ecological interactions on the evolutionary dynamics of pre-existing resistance. First, we find that ecological interactions significantly expand the parameter regime under which we expect to observe pre-existing resistance. Next, even when positive ecological interactions between mutants and ancestors are rare, these resistant clones provide the primary mode of evolved resistance because even weak positive interaction leads to significantly longer extinction times. We then find that even in the case where mutation supply alone is sufficient to predict pre-existing resistance, frequency-dependent ecological forces still contribute a strong evolutionary pressure that selects for increasingly positive ecological effects (negative frequency-dependent selection). Finally, we genetically engineer several of the most common clinically observed resistance mechanisms to targeted therapies in NSCLC, a treatment notorious for pre-existing resistance. We find that each engineered mutant displays a positive ecological interaction with their ancestor. As a whole, these results suggest that frequency-dependent ecological effects can play a crucial role in shaping the evolutionary dynamics of pre-existing resistance.
Collapse
Affiliation(s)
- Jeff Maltas
- Cleveland Clinic, Translational Hematology Oncology Research, Cleveland, OH
- Case Western Reserve University, School of Medicine, Cleveland, OH
| | - Dagim Shiferaw Tadele
- Cleveland Clinic, Translational Hematology Oncology Research, Cleveland, OH
- Oslo University Hospital, Ullevål, Department of Medical Genetics, Oslo, Norway
| | - Arda Durmaz
- Cleveland Clinic, Translational Hematology Oncology Research, Cleveland, OH
| | - Christopher D. McFarland
- Case Western Reserve University, School of Medicine, Cleveland, OH
- Case Comprehensive Cancer Center, Cleveland, OH
| | | | - Jacob G. Scott
- Cleveland Clinic, Translational Hematology Oncology Research, Cleveland, OH
- Case Western Reserve University, School of Medicine, Cleveland, OH
- Case Western Reserve University, Department of Physics, Cleveland, OH
- Case Comprehensive Cancer Center, Cleveland, OH
| |
Collapse
|
33
|
Zoheir AE, Stolle C, Rabe KS. Microfluidics for adaptation of microorganisms to stress: design and application. Appl Microbiol Biotechnol 2024; 108:162. [PMID: 38252163 PMCID: PMC10803453 DOI: 10.1007/s00253-024-13011-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: 10/31/2023] [Revised: 12/22/2023] [Accepted: 01/11/2024] [Indexed: 01/23/2024]
Abstract
Microfluidic systems have fundamentally transformed the realm of adaptive laboratory evolution (ALE) for microorganisms by offering unparalleled control over environmental conditions, thereby optimizing mutant generation and desired trait selection. This review summarizes the substantial influence of microfluidic technologies and their design paradigms on microbial adaptation, with a primary focus on leveraging spatial stressor concentration gradients to enhance microbial growth in challenging environments. Specifically, microfluidic platforms tailored for scaled-down ALE processes not only enable highly autonomous and precise setups but also incorporate novel functionalities. These capabilities encompass fostering the growth of biofilms alongside planktonic cells, refining selection gradient profiles, and simulating adaptation dynamics akin to natural habitats. The integration of these aspects enables shaping phenotypes under pressure, presenting an unprecedented avenue for developing robust, stress-resistant strains, a feat not easily attainable using conventional ALE setups. The versatility of these microfluidic systems is not limited to fundamental research but also offers promising applications in various areas of stress resistance. As microfluidic technologies continue to evolve and merge with cutting-edge methodologies, they possess the potential not only to redefine the landscape of microbial adaptation studies but also to expedite advancements in various biotechnological areas. KEY POINTS: • Microfluidics enable precise microbial adaptation in controlled gradients. • Microfluidic ALE offers insights into stress resistance and distinguishes between resistance and persistence. • Integration of adaptation-influencing factors in microfluidic setups facilitates efficient generation of stress-resistant strains.
Collapse
Affiliation(s)
- Ahmed E Zoheir
- Department of Genetics and Cytology, Biotechnology Research Institute, National Research Centre (NRC), 33 El Buhouth St., Dokki, Cairo, 12622, Egypt
| | - Camilla Stolle
- Institute for Biological Interfaces 1 (IBG-1), Biomolecular Micro- and Nanostructures, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Kersten S Rabe
- Institute for Biological Interfaces 1 (IBG-1), Biomolecular Micro- and Nanostructures, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany.
| |
Collapse
|
34
|
Chang B, Chen J, Bao J, Sun T, Cheng Z. Molecularly Engineered Room-Temperature Phosphorescence for Biomedical Application: From the Visible toward Second Near-Infrared Window. Chem Rev 2023; 123:13966-14037. [PMID: 37991875 DOI: 10.1021/acs.chemrev.3c00401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Phosphorescence, characterized by luminescent lifetimes significantly longer than that of biological autofluorescence under ambient environment, is of great value for biomedical applications. Academic evidence of fluorescence imaging indicates that virtually all imaging metrics (sensitivity, resolution, and penetration depths) are improved when progressing into longer wavelength regions, especially the recently reported second near-infrared (NIR-II, 1000-1700 nm) window. Although the emission wavelength of probes does matter, it is not clear whether the guideline of "the longer the wavelength, the better the imaging effect" is still suitable for developing phosphorescent probes. For tissue-specific bioimaging, long-lived probes, even if they emit visible phosphorescence, enable accurate visualization of large deep tissues. For studies dealing with bioimaging of tiny biological architectures or dynamic physiopathological activities, the prerequisite is rigorous planning of long-wavelength phosphorescence, being aware of the cooperative contribution of long wavelengths and long lifetimes for improving the spatiotemporal resolution, penetration depth, and sensitivity of bioimaging. In this Review, emerging molecular engineering methods of room-temperature phosphorescence are discussed through the lens of photophysical mechanisms. We highlight the roles of phosphorescence with emission from visible to NIR-II windows toward bioapplications. To appreciate such advances, challenges and prospects in rapidly growing studies of room-temperature phosphorescence are described.
Collapse
Affiliation(s)
- Baisong Chang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Jie Chen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Jiasheng Bao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Taolei Sun
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264000, China
| |
Collapse
|
35
|
Kerek Á, Török B, Laczkó L, Kardos G, Bányai K, Somogyi Z, Kaszab E, Bali K, Jerzsele Á. In Vitro Microevolution and Co-Selection Assessment of Florfenicol Impact on Escherichia coli Resistance Development. Antibiotics (Basel) 2023; 12:1728. [PMID: 38136762 PMCID: PMC10740648 DOI: 10.3390/antibiotics12121728] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
The issue of antimicrobial resistance is becoming an increasingly serious challenge in both human and veterinary medicine. Prudent antimicrobial use in veterinary medicine is warranted and supported by international guidelines, with the Antimicrobial Advice Ad Hoc Expert Group (AMEG) placing particular emphasis on the critically important group B antimicrobials. These antimicrobials are commonly employed, especially in the poultry and swine industry. The impact of florfenicol, a veterinary antibiotic, was studied on the resistance development of Escherichia coli. The aim of the study was to investigate the effect of the use of florfenicol on the development of phenotypic and genomic resistances, not only to the drug itself but also to other drugs. The minimum inhibitory concentrations (MICs) of the antibiotics were investigated at 1×, 10×, 100× and 1000× concentrations using the adapted Microbial Evolution and Growth Arena (MEGA-plate) method. The results demonstrate that florfenicol can select for resistance to fluoroquinolone antibiotics (167× MIC value increase) and cephalosporins (67× MIC value increase). A total of 44 antimicrobial resistance genes were identified, the majority of which were consistent across the samples. Chromosomal point mutations, including alterations in resistance-associated and regulatory genes (acrB, acrR, emrR and robA), are thought to trigger multiple drug efflux pump activations, leading to phenotypically increased resistance. The study underscores the impact of florfenicol and its role in the development of antimicrobial resistance, particularly concerning fluoroquinolone antibiotics and cephalosporins. This study is the first to report florfenicol's dose-dependent enhancement of other antibiotics' MICs, linked to mutations in SOS-box genes (mdtABC-tolC, emrAB-tolC and acrAB-tolC) and increased multidrug efflux pump genes. Mutations in the regulatory genes acrR, emrR and rpbA support the possibility of increased gene expression. The results are crucial for understanding antimicrobial resistance and its development, highlighting the promising potential of in vitro evolutionary and coselection studies for future research.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
| | - Bence Török
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
| | - Levente Laczkó
- Institute of Metagenomics, University of Debrecen, 4032 Debrecen, Hungary;
| | - Gábor Kardos
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
- Institute of Metagenomics, University of Debrecen, 4032 Debrecen, Hungary;
| | - Krisztián Bányai
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
- Veterinary Medical Research Institute, 1143 Budapest, Hungary;
| | - Zoltán Somogyi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
| | - Eszter Kaszab
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
- Veterinary Medical Research Institute, 1143 Budapest, Hungary;
| | - Krisztina Bali
- Veterinary Medical Research Institute, 1143 Budapest, Hungary;
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (B.T.); (K.B.); (Z.S.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary; (G.K.); (E.K.)
| |
Collapse
|
36
|
Freire T, Hu Z, Wood KB, Gjini E. Modeling spatial evolution of multi-drug resistance under drug environmental gradients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567447. [PMID: 38014279 PMCID: PMC10680811 DOI: 10.1101/2023.11.16.567447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Multi-drug combinations to treat bacterial populations are at the forefront of approaches for infection control and prevention of antibiotic resistance. Although the evolution of antibiotic resistance has been theoretically studied with mathematical population dynamics models, extensions to spatial dynamics remain rare in the literature, including in particular spatial evolution of multi-drug resistance. In this study, we propose a reaction-diffusion system that describes the multi-drug evolution of bacteria, based on a rescaling approach (Gjini and Wood, 2021). We show how the resistance to drugs in space, and the consequent adaptation of growth rate is governed by a Price equation with diffusion. The covariance terms in this equation integrate features of drug interactions and collateral resistances or sensitivities to the drugs. We study spatial versions of the model where the distribution of drugs is homogeneous across space, and where the drugs vary environmentally in a piecewise-constant, linear and nonlinear manner. Applying concepts from perturbation theory and reaction-diffusion equations, we propose an analytical characterization of average mutant fitness in the spatial system based on the principal eigenvalue of our linear problem. This enables an accurate translation from drug spatial gradients and mutant antibiotic susceptibility traits, to the relative advantage of each mutant across the environment. Such a mathematical understanding allows to predict the precise outcomes of selection over space, ultimately from the fundamental balance between growth and movement traits, and their diversity in a population.
Collapse
Affiliation(s)
- Tomas Freire
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Zhijian Hu
- Departments of Biophysics and Physics, University of Michigan, USA
| | - Kevin B. Wood
- Departments of Biophysics and Physics, University of Michigan, USA
| | - Erida Gjini
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
37
|
Svet L, Parijs I, Isphording S, Lories B, Marchal K, Steenackers HP. Competitive interactions facilitate resistance development against antimicrobials. Appl Environ Microbiol 2023; 89:e0115523. [PMID: 37819078 PMCID: PMC10617502 DOI: 10.1128/aem.01155-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 10/13/2023] Open
Abstract
While the evolution of antimicrobial resistance is well studied in free-living bacteria, information on resistance development in dense and diverse biofilm communities is largely lacking. Therefore, we explored how the social interactions in a duo-species biofilm composed of the brewery isolates Pseudomonas rhodesiae and Raoultella terrigena influence the adaptation to the broad-spectrum antimicrobial sulfathiazole. Previously, we showed that the competition between these brewery isolates enhances the antimicrobial tolerance of P. rhodesiae. Here, we found that this enhanced tolerance in duo-species biofilms is associated with a strongly increased antimicrobial resistance development in P. rhodesiae. Whereas P. rhodesiae was not able to evolve resistance against sulfathiazole in monospecies conditions, it rapidly evolved resistance in the majority of the duo-species communities. Although the initial presence of R. terrigena was thus required for P. rhodesiae to acquire resistance, the resistance mechanisms did not depend on the presence of R. terrigena. Whole genome sequencing of resistant P. rhodesiae clones showed no clear mutational hot spots. This indicates that the acquired resistance phenotype depends on complex interactions between low-frequency mutations in the genetic background of the strains. We hypothesize that the increased tolerance in duo-species conditions promotes resistance by enhancing the selection of partially resistant mutants and opening up novel evolutionary trajectories that enable such genetic interactions. This hypothesis is reinforced by experimentally excluding potential effects of increased initial population size, enhanced mutation rate, and horizontal gene transfer. Altogether, our observations suggest that the community mode of life and the social interactions therein strongly affect the accessible evolutionary pathways toward antimicrobial resistance.IMPORTANCEAntimicrobial resistance is one of the most studied bacterial properties due to its enormous clinical and industrial relevance; however, most research focuses on resistance development of a single species in isolation. In the present study, we showed that resistance evolution of brewery isolates can differ greatly between single- and mixed-species conditions. Specifically, we observed that the development of antimicrobial resistance in certain species can be significantly enhanced in co-culture as compared to the single-species conditions. Overall, the current study emphasizes the need of considering the within bacterial interactions in microbial communities when evaluating antimicrobial treatments and resistance evolution.
Collapse
Affiliation(s)
- Luka Svet
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), Leuven, Belgium
| | - Ilse Parijs
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), Leuven, Belgium
| | - Simon Isphording
- Department of Plant Biotechnology and Bioinformatics, Data Integration and Biological Networks, UGent, Technologiepark 15, Gent, Belgium
| | - Bram Lories
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), Leuven, Belgium
| | - Kathleen Marchal
- Department of Plant Biotechnology and Bioinformatics, Data Integration and Biological Networks, UGent, Technologiepark 15, Gent, Belgium
| | - Hans P. Steenackers
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), Leuven, Belgium
| |
Collapse
|
38
|
Hu G, Wang Y, Liu X, Strube ML, Wang B, Kovács ÁT. Species and condition shape the mutational spectrum in experimentally evolved biofilms. mSystems 2023; 8:e0054823. [PMID: 37768063 PMCID: PMC10654089 DOI: 10.1128/msystems.00548-23] [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: 05/27/2023] [Accepted: 08/11/2023] [Indexed: 09/29/2023] Open
Abstract
IMPORTANCE Biofilm formation is a vital factor for the survival and adaptation of bacteria in diverse environmental niches. Experimental evolution combined with the advancement of whole-population genome sequencing provides us a powerful tool to understand the genomic dynamic of evolutionary adaptation to different environments, such as during biofilm development. Previous studies described the genetic and phenotypic changes of selected clones from experimentally evolved Bacillus thuringiensis and Bacillus subtilis that were adapted under abiotic and biotic biofilm conditions. However, the full understanding of the dynamic evolutionary landscapes was lacking. Furthermore, the differences and similarities of adaptive mechanisms in B. thuringiensis and B. subtilis were not identified. To overcome these limitations, we performed longitudinal whole-population genome sequencing to study the underlying genetic dynamics at high resolution. Our study provides the first comprehensive mutational landscape of two bacterial species' biofilms that is adapted to an abiotic and biotic surface.
Collapse
Affiliation(s)
- Guohai Hu
- China National GeneBank, BGI, Shenzhen, China
- BGI Research, Shenzhen, China
- Bacterial Interactions and Evolution Group, DTU Bioengineering, Technical University of Denmark, Lyngby, Denmark
| | - Yue Wang
- China National GeneBank, BGI, Shenzhen, China
- BGI Research, Shenzhen, China
- BGI Research, Beijing, China
| | - Xin Liu
- China National GeneBank, BGI, Shenzhen, China
- BGI Research, Shenzhen, China
- BGI Research, Beijing, China
| | - Mikael Lenz Strube
- Bacterial Ecophysiology and Biotechnology Group, DTU Bioengineering, Technical University of Denmark, Lyngby, Denmark
| | - Bo Wang
- China National GeneBank, BGI, Shenzhen, China
- BGI Research, Shenzhen, China
- Shenzhen Key Laboratory of Environmental Microbial Genomics and Application, BGI Research, Shenzhen, China
| | - Ákos T. Kovács
- Bacterial Interactions and Evolution Group, DTU Bioengineering, Technical University of Denmark, Lyngby, Denmark
- Institute of Biology, Leiden University, Leiden, The Netherlands
| |
Collapse
|
39
|
Raza S, Wdowiak M, Grotek M, Adamkiewicz W, Nikiforow K, Mente P, Paczesny J. Enhancing the antimicrobial activity of silver nanoparticles against ESKAPE bacteria and emerging fungal pathogens by using tea extracts. NANOSCALE ADVANCES 2023; 5:5786-5798. [PMID: 37881701 PMCID: PMC10597549 DOI: 10.1039/d3na00220a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/12/2023] [Indexed: 10/27/2023]
Abstract
The sale of antibiotics and antifungals has skyrocketed since 2020. The increasing threat of pathogens like ESKAPE bacteria (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.), which are effective in evading existing antibiotics, and yeasts like Candida auris or Cryptococcus neoformans is pressing to develop efficient antimicrobial alternatives. Nanoparticles, especially silver nanoparticles (AgNPs), are believed to be promising candidates to supplement or even replace antibiotics in some applications. Here, we propose a way to increase the antimicrobial efficiency of silver nanoparticles by using tea extracts (black, green, or red) for their synthesis. This allows for using lower concentrations of nanoparticles and obtaining the antimicrobial effect in a short time. We found that AgNPs synthesized using green tea extract (G-TeaNPs) are the most effective, causing approximately 80% bacterial cell death in Gram-negative bacteria within only 3 hours at a concentration of 0.1 mg mL-1, which is better than antibiotics. Ampicillin at the same concentration (0.1 mg mL-1) and within the same duration (3 h) causes only up to 40% decrease in the number of S. aureus and E. cloacae cells (non-resistant strains). The tested silver nanoparticles also have antifungal properties and are effective against C. auris and C. neoformans, which are difficult to eradicate using other means. We established that silver nanoparticles synthesized with tea extracts have higher antibacterial properties than silver nanoparticles alone. Such formulations using inexpensive tea extracts and lower concentrations of silver nanoparticles show a promising solution to fight various pathogens.
Collapse
Affiliation(s)
- Sada Raza
- Institute of Physical Chemistry, Polish Academy of Sciences Kasprzaka 44/52 01-224 Warsaw Poland +48 22 343 2071
| | - Mateusz Wdowiak
- Institute of Physical Chemistry, Polish Academy of Sciences Kasprzaka 44/52 01-224 Warsaw Poland +48 22 343 2071
| | - Mateusz Grotek
- Institute of Physical Chemistry, Polish Academy of Sciences Kasprzaka 44/52 01-224 Warsaw Poland +48 22 343 2071
- Military University of Technology gen. Sylwestra Kaliskiego 2 00-908 Warsaw Poland
| | - Witold Adamkiewicz
- Institute of Physical Chemistry, Polish Academy of Sciences Kasprzaka 44/52 01-224 Warsaw Poland +48 22 343 2071
| | - Kostiantyn Nikiforow
- Institute of Physical Chemistry, Polish Academy of Sciences Kasprzaka 44/52 01-224 Warsaw Poland +48 22 343 2071
| | - Pumza Mente
- Institute of Physical Chemistry, Polish Academy of Sciences Kasprzaka 44/52 01-224 Warsaw Poland +48 22 343 2071
| | - Jan Paczesny
- Institute of Physical Chemistry, Polish Academy of Sciences Kasprzaka 44/52 01-224 Warsaw Poland +48 22 343 2071
| |
Collapse
|
40
|
Piskovsky V, Oliveira NM. Bacterial motility can govern the dynamics of antibiotic resistance evolution. Nat Commun 2023; 14:5584. [PMID: 37696800 PMCID: PMC10495427 DOI: 10.1038/s41467-023-41196-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 08/24/2023] [Indexed: 09/13/2023] Open
Abstract
Spatial heterogeneity in antibiotic concentrations is thought to accelerate the evolution of antibiotic resistance, but current theory and experiments have overlooked the effect of cell motility on bacterial adaptation. Here, we study bacterial evolution in antibiotic landscapes with a quantitative model where bacteria evolve under the stochastic processes of proliferation, death, mutation and migration. Numerical and analytical results show that cell motility can both accelerate and decelerate bacterial adaptation by affecting the degree of genotypic mixing and ecological competition. Moreover, we find that for sufficiently high rates, cell motility can limit bacterial survival, and we derive conditions for all these regimes. Similar patterns are observed in more complex scenarios, namely where bacteria can bias their motion in chemical gradients (chemotaxis) or switch between motility phenotypes either stochastically or in a density-dependent manner. Overall, our work reveals limits to bacterial adaptation in antibiotic landscapes that are set by cell motility.
Collapse
Affiliation(s)
- Vit Piskovsky
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Wilberforce Road, Cambridge, CB3 0WA, UK
- Mathematical Institute, University of Oxford, Woodstock Road, Oxford, OX2 6GG, UK
| | - Nuno M Oliveira
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Wilberforce Road, Cambridge, CB3 0WA, UK.
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK.
| |
Collapse
|
41
|
Zhai Y, Pribis JP, Dooling SW, Garcia-Villada L, Minnick P, Xia J, Liu J, Mei Q, Fitzgerald DM, Herman C, Hastings P, Costa-Mattioli M, Rosenberg SM. Drugging evolution of antibiotic resistance at a regulatory network hub. SCIENCE ADVANCES 2023; 9:eadg0188. [PMID: 37352342 PMCID: PMC10289659 DOI: 10.1126/sciadv.adg0188] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 05/22/2023] [Indexed: 06/25/2023]
Abstract
Evolution of antibiotic resistance is a world health crisis, fueled by new mutations. Drugs to slow mutagenesis could, as cotherapies, prolong the shelf-life of antibiotics, yet evolution-slowing drugs and drug targets have been underexplored and ineffective. Here, we used a network-based strategy to identify drugs that block hubs of fluoroquinolone antibiotic-induced mutagenesis. We identify a U.S. Food and Drug Administration- and European Medicines Agency-approved drug, dequalinium chloride (DEQ), that inhibits activation of the Escherichia coli general stress response, which promotes ciprofloxacin-induced (stress-induced) mutagenic DNA break repair. We uncover the step in the pathway inhibited: activation of the upstream "stringent" starvation stress response, and find that DEQ slows evolution without favoring proliferation of DEQ-resistant mutants. Furthermore, we demonstrate stress-induced mutagenesis during mouse infections and its inhibition by DEQ. Our work provides a proof-of-concept strategy for drugs to slow evolution in bacteria and generally.
Collapse
Affiliation(s)
- Yin Zhai
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - John P. Pribis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sean W. Dooling
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Libertad Garcia-Villada
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - P.J. Minnick
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jun Xia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jingjing Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qian Mei
- The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX 77030, USA
| | - Devon M. Fitzgerald
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christophe Herman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
- The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - P.J. Hastings
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mauro Costa-Mattioli
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Susan M. Rosenberg
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
- The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
42
|
Saeed A, Ali H, Yasmin A, Baig M, Ullah A, Kazmi A, Ahmed MA, Albadrani GM, El-Demerdash FM, Bibi M, Abdel-Daim MM, Ali I, Hussain S. Unveiling the Antibiotic Susceptibility and Antimicrobial Potential of Bacteria from Human Breast Milk of Pakistani Women: An Exploratory Study. BIOMED RESEARCH INTERNATIONAL 2023; 2023:6399699. [PMID: 37377461 PMCID: PMC10292949 DOI: 10.1155/2023/6399699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND Human life quality and expectancy have increased dramatically over the past 5 decades because of improvements in nutrition and antibiotic's usage fighting against infectious diseases. Yet, it was soon revealed that the microbes adapted to develop resistance to any of the drugs that were used. Recently, there is great concern that commensal bacteria from food and the gastrointestinal tract of humans and animals could act as a reservoir for antibiotic resistance genes. Methodology. This study was intended for evaluating the phenotypic antibiotic resistance/sensitivity profiles of probiotic bacteria from human breast milk and evaluating the inhibitory effect of the probiotic bacteria against both Gram-negative and Gram-positive bacteria. RESULTS The results point out that some of the isolated bacteria were resistant to diverse antibiotics including gentamycin, imipenem, trimethoprim sulfamethoxazole, and nalidixic acid. Susceptibility profile to certain antibiotics like vancomycin, tetracycline, ofloxacin, chloramphenicol, streptomycin, rifampicin, and bacitracin was also observed. The antimicrobial qualities of cell-free supernatants of some probiotic bacteria inhibited the growth of indicator bacteria. Also, antimicrobial properties of the probiotic bacteria from the present study attributed to the production of organic acid, bacterial adhesion to hydrocarbons (BATH), salt aggregation, coaggregation with pathogens, and bacteriocin production. Some isolated bacteria from human milk displayed higher hydrophobicity in addition to intrinsic probiotic properties like Gram-positive classification, catalase-negative activity, resistance to gastric juice (pH 2), and bile salt (0.3%) concentration. CONCLUSION This study has added to the data of the antibiotic and antimicrobial activity of some probiotic bacteria from some samples of Pakistani women breast milk. Probiotic bacteria are usually considered to decrease gastrointestinal tract diseases by adhering to the gut epithelial and reducing population of pathogens and in the case of Streptococcus lactarius MB622 and Streptococcus salivarius MB620 in terms of hydrophobicity and exclusion of indicator pathogenic strains.
Collapse
Affiliation(s)
- Ayesha Saeed
- Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University, Rawalpindi, Pakistan
| | - Hina Ali
- Quaid-e-Azam Medical College, Bahawalpur, Punjab, Pakistan
| | - Azra Yasmin
- Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University, Rawalpindi, Pakistan
| | - Mehreen Baig
- Surgical Unit II, Foundation University, Islamabad, Pakistan
| | - Abd Ullah
- Xinjiang Key Laboratory of Desert Plant Root Ecology and Vegetation Restoration, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi, China
- Cele National Station of Observation and Research for Desert-Grassland Ecosystems, Cele, China
| | - Abeer Kazmi
- Institute of Hydrobiology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (UCAS), Wuhan, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, 84428, Riyadh 11671, Saudi Arabia
| | - Fatma M. El-Demerdash
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Monaza Bibi
- Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University, Rawalpindi, Pakistan
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Iftikhar Ali
- Centre for Plant Sciences and Biodiversity, University of Swat, Charbagh 19120, Pakistan
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sadam Hussain
- University of Health Sciences, Lahore, Punjab, Pakistan
| |
Collapse
|
43
|
Fang P, Elena AX, Kunath MA, Berendonk TU, Klümper U. Reduced selection for antibiotic resistance in community context is maintained despite pressure by additional antibiotics. ISME COMMUNICATIONS 2023; 3:52. [PMID: 37258727 PMCID: PMC10232432 DOI: 10.1038/s43705-023-00262-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
Selection for antibiotic resistance at very low antibiotic concentrations has been demonstrated for individual antibiotics in single species experiments. Furthermore, selection in these focal strains is reduced when taking place in complex microbial community context. However, in the environment, bacteria are rarely exposed to single, but rather complex mixtures of selective agents. Here, we explored how the presence of a second selective agent affects selection dynamics between isogenic pairs of focal E. coli strains, differing exclusively in a single resistance determinant, in the absence and presence of a model wastewater community across a gradient of antibiotics. An additional antibiotic that exclusively affects the model wastewater community, but to which the focal strains are resistant to, was chosen as the second selective agent. This allowed exploring how inhibition alters the community's ability to reduce selection. In the presence of the community, the selection coefficient at specific antibiotic concentrations was consistently decreased compared to the absence of the community. While pressure through the second antibiotic significantly decreased the activity and diversity of the community, its ability to reduce selection was consistently maintained at levels comparable to those recorded in absence of the second antibiotic. This indicates that the observed effects of community context on selection dynamics are rather based on competitive or protective effects between the focal strains and a small proportion of bacteria within the community, than on general competition for nutrients. These findings have implications for our understanding of the evolution and selection for multi-drug resistant strains.
Collapse
Affiliation(s)
- Peiju Fang
- Technische Universität Dresden, Institute of Hydrobiology, Zellescher Weg 40, Dresden, Germany
| | - Alan Xavier Elena
- Technische Universität Dresden, Institute of Hydrobiology, Zellescher Weg 40, Dresden, Germany
| | - Maxi Antonia Kunath
- Technische Universität Dresden, Institute of Hydrobiology, Zellescher Weg 40, Dresden, Germany
| | - Thomas U Berendonk
- Technische Universität Dresden, Institute of Hydrobiology, Zellescher Weg 40, Dresden, Germany
| | - Uli Klümper
- Technische Universität Dresden, Institute of Hydrobiology, Zellescher Weg 40, Dresden, Germany.
| |
Collapse
|
44
|
Liu C, Chen J, Yang Y, Teng Y, Chen H. Biogeography and diversity patterns of antibiotic resistome in the sediments of global lakes. J Environ Sci (China) 2023; 127:421-430. [PMID: 36522074 DOI: 10.1016/j.jes.2022.06.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/11/2022] [Accepted: 06/15/2022] [Indexed: 06/17/2023]
Abstract
Lakes act as one of the reservoirs and dispersal routes of antibiotic resistance genes (ARGs) and pathogenic resistant bacteria in aquatic environments. Previous studies reported the occurrence and distribution of ARGs in lakes worldwide; however, few investigated the biogeography and diversity patterns of antibiotic resistome in the environment. To fill this gap, a large-scale data set of sediment metagenomes was collected from globally distributed lakes and characterized comprehensively using metagenomic assembly-based analysis, aiming to shed light on the biogeography and diversity patterns of ARGs in lake ecosystems from a global perspective. Our analyses showed that abundant and diverse ARGs were found in the global lake sediments, including a set of emerging ARGs such as mcr-type and carbapenem-resistant Enterobacteriaceae related genes. Most of the identified ARGs were generally associated with the commonly used antibiotics, suggesting the role of increasing antibiotic consumptions on the resistome prevalence. Spatially, the composition and diversity of ARGs varied across geographical distances and exhibited a scale-dependent distance-decay relationship. Notably, the composition of ARGs was largely shaped by bacterial community structure, and their diversities were co-governed by stochastic process (∼48%) and deterministic process (∼52%). Findings provide a valuable insight to better understand ecological mechanisms of ARGs in lake ecosystems and have important implication for the prevention and control of resistome risk.
Collapse
Affiliation(s)
- Chang Liu
- College of Water Sciences, Beijing Normal University, Beijing 100875, China
| | - Jinping Chen
- College of Water Sciences, Beijing Normal University, Beijing 100875, China
| | - Yuyi Yang
- Key Laboratory of Aquatic Botany and Watershed Ecology, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| | - Yanguo Teng
- College of Water Sciences, Beijing Normal University, Beijing 100875, China; Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, Beijing 100875, China
| | - Haiyang Chen
- College of Water Sciences, Beijing Normal University, Beijing 100875, China; Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, Beijing 100875, China.
| |
Collapse
|
45
|
Zhai Y, Minnick PJ, Pribis JP, Garcia-Villada L, Hastings PJ, Herman C, Rosenberg SM. ppGpp and RNA-polymerase backtracking guide antibiotic-induced mutable gambler cells. Mol Cell 2023; 83:1298-1310.e4. [PMID: 36965481 PMCID: PMC10317147 DOI: 10.1016/j.molcel.2023.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/14/2022] [Accepted: 03/02/2023] [Indexed: 03/27/2023]
Abstract
Antibiotic resistance is a global health threat and often results from new mutations. Antibiotics can induce mutations via mechanisms activated by stress responses, which both reveal environmental cues of mutagenesis and are weak links in mutagenesis networks. Network inhibition could slow the evolution of resistance during antibiotic therapies. Despite its pivotal importance, few identities and fewer functions of stress responses in mutagenesis are clear. Here, we identify the Escherichia coli stringent starvation response in fluoroquinolone-antibiotic ciprofloxacin-induced mutagenesis. Binding of response-activator ppGpp to RNA polymerase (RNAP) at two sites leads to an antibiotic-induced mutable gambler-cell subpopulation. Each activates a stress response required for mutagenic DNA-break repair: surprisingly, ppGpp-site-1-RNAP triggers the DNA-damage response, and ppGpp-site-2-RNAP induces σS-response activity. We propose that RNAP regulates DNA-damage processing in transcribed regions. The data demonstrate a critical node in ciprofloxacin-induced mutagenesis, imply RNAP-regulation of DNA-break repair, and identify promising targets for resistance-resisting drugs.
Collapse
Affiliation(s)
- Yin Zhai
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - P J Minnick
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - John P Pribis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Libertad Garcia-Villada
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - P J Hastings
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christophe Herman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA; The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Susan M Rosenberg
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA; The Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
46
|
Li C, McCrone S, Warrick JW, Andes DR, Hite Z, Volk CF, Rose WE, Beebe DJ. Under-oil open microfluidic systems for rapid phenotypic antimicrobial susceptibility testing. LAB ON A CHIP 2023; 23:2005-2015. [PMID: 36883560 PMCID: PMC10581760 DOI: 10.1039/d3lc00066d] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Antimicrobial susceptibility testing (AST) remains the cornerstone of effective antimicrobial selection and optimization in patients. Despite recent advances in rapid pathogen identification and resistance marker detection with molecular diagnostics (e.g., qPCR, MALDI-TOF MS), phenotypic (i.e., microbial culture-based) AST methods - the gold standard in hospitals/clinics - remain relatively unchanged over the last few decades. Microfluidics-based phenotypic AST has been growing fast in recent years, aiming for rapid (i.e., turnaround time <8 h), high-throughput, and automated species identification, resistance detection, and antibiotics screening. In this pilot study, we describe the application of a multi-liquid-phase open microfluidic system, named under-oil open microfluidic systems (UOMS), to achieve a rapid phenotypic AST. UOMS provides an open microfluidics-based solution for rapid phenotypic AST (UOMS-AST) by implementing and recording a pathogen's antimicrobial activity in micro-volume testing units under an oil overlay. UOMS-AST allows free physical access (e.g., by standard pipetting) to the system and label-free, single-cell resolution optical access. UOMS-AST can accurately and rapidly determine antimicrobial activities [including susceptibility/resistance breakpoint and minimum inhibitory concentration (MIC)] from nominal sample/bacterial cells in a system aligned with clinical laboratory standards where open systems and optical microscopy are predominantly adopted. Further, we combine UOMS-AST with a cloud lab data analytic technique for real-time image analysis and report generation to provide a rapid (<4 h) sample-to-report turnaround time, shedding light on its utility as a versatile (e.g., low-resource setting and manual laboratory operation, or high-throughput automated system) phenotypic AST platform for hospital/clinic use.
Collapse
Affiliation(s)
- Chao Li
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sue McCrone
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jay W. Warrick
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David R. Andes
- Department of Medicine, Division of Infectious Diseases, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachary Hite
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cecilia F. Volk
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Warren E. Rose
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, Division of Infectious Diseases, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David J. Beebe
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
47
|
Barkan CO, Wang S. Multiple phase transitions shape biodiversity of a migrating population. Phys Rev E 2023; 107:034405. [PMID: 37072956 DOI: 10.1103/physreve.107.034405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/28/2023] [Indexed: 04/20/2023]
Abstract
In a wide variety of natural systems, closely related microbial strains coexist stably, resulting in high levels of fine-scale biodiversity. However, the mechanisms that stabilize this coexistence are not fully understood. Spatial heterogeneity is one common stabilizing mechanism, but the rate at which organisms disperse throughout the heterogeneous environment may strongly impact the stabilizing effect that heterogeneity can provide. An intriguing example is the gut microbiome, where active mechanisms affect the movement of microbes and potentially maintain diversity. We investigate how biodiversity is affected by migration rate using a simple evolutionary model with heterogeneous selection pressure. We find that the biodiversity-migration rate relationship is shaped by multiple phase transitions, including a reentrant phase transition to coexistence. At each transition, an ecotype goes extinct and dynamics exhibit critical slowing down (CSD). CSD is encoded in the statistics of fluctuations due to demographic noise-this may provide an experimental means for detecting and altering impending extinction.
Collapse
Affiliation(s)
- Casey O Barkan
- Department of Physics and Astronomy, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Shenshen Wang
- Department of Physics and Astronomy, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
48
|
First Synthesis of DBU-Conjugated Cationic Carbohydrate Derivatives and Investigation of Their Antibacterial and Antifungal Activity. Int J Mol Sci 2023; 24:ijms24043550. [PMID: 36834964 PMCID: PMC9968064 DOI: 10.3390/ijms24043550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The emergence of drug-resistant bacteria and fungi represents a serious health problem worldwide. It has long been known that cationic compounds can inhibit the growth of bacteria and fungi by disrupting the cell membrane. The advantage of using such cationic compounds is that the microorganisms would not become resistant to cationic agents, since this type of adaptation would mean significantly altering the structure of their cell walls. We designed novel, DBU (1,8-diazabicyclo[5.4.0]undec-7-ene)-derived amidinium salts of carbohydrates, which may be suitable for disturbing the cell walls of bacteria and fungi due to their quaternary ammonium moiety. A series of saccharide-DBU conjugates were prepared from 6-iodo derivatives of d-glucose, d-mannose, d-altrose and d-allose by nucleophilic substitution reactions. We optimized the synthesis of a d-glucose derivative, and studied the protecting group free synthesis of the glucose-DBU conjugates. The effect of the obtained quaternary amidinium salts against Escherichia coli and Staphylococcus aureus bacterial strains and Candida albicans yeast was investigated, and the impact of the used protecting groups and the sugar configuration on the antimicrobial activity was analyzed. Some of the novel sugar quaternary ammonium compounds with lipophilic aromatic groups (benzyl and 2-napthylmethyl) showed particularly good antifungal and antibacterial activity.
Collapse
|
49
|
Chodkowski JL, Shade A. A coevolution experiment between Flavobacterium johnsoniae and Burkholderia thailandensis reveals parallel mutations that reduce antibiotic susceptibility. MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 36724091 DOI: 10.1099/mic.0.001267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
One interference mechanism of bacterial competition is the production of antibiotics. Bacteria exposed to antibiotics can resist antibiotic inhibition through intrinsic or acquired mechanisms. Here, we performed a coevolution experiment to understand the long-term consequences of antibiotic production and antibiotic susceptibility for two environmental bacterial strains. We grew five independent lines of the antibiotic-producing environmental strain, Burkholderia thailandensis E264, and the antibiotic-inhibited environmental strain, Flavobacterium johnsoniae UW101, together and separately on agar plates for 7.5 months (1.5 month incubations), transferring each line five times to new agar plates. We observed that the F. johnsoniae ancestor could tolerate the B. thailandensis-produced antibiotic through efflux mechanisms, but that the coevolved lines had reduced susceptibility. We then sequenced genomes from the coevolved and monoculture F. johnsoniae lines, and uncovered mutational ramifications for the long-term antibiotic exposure. The coevolved genomes from F. johnsoniae revealed four potential mutational signatures of reduced antibiotic susceptibility that were not observed in the evolved monoculture lines. Two mutations were found in tolC: one corresponding to a 33 bp deletion and the other corresponding to a nonsynonymous mutation. A third mutation was observed as a 1 bp insertion coding for a RagB/SusD nutrient uptake protein. The last mutation was a G83R nonsynonymous mutation in acetyl-coA carboxylayse carboxyltransferase subunit alpha (AccA). Deleting the 33 bp from tolC in the F. johnsoniae ancestor reduced antibiotic susceptibility, but not to the degree observed in coevolved lines. Furthermore, the accA mutation matched a previously described mutation conferring resistance to B. thailandensis-produced thailandamide. Analysis of B. thailandensis transposon mutants for thailandamide production revealed that thailandamide was bioactive against F. johnsoniae, but also suggested that additional B. thailandensis-produced antibiotics were involved in the inhibition of F. johnsoniae. This study reveals how multi-generational interspecies interactions, mediated through chemical exchange, can result in novel interaction-specific mutations, some of which may contribute to reductions in antibiotic susceptibility.
Collapse
Affiliation(s)
- John L Chodkowski
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Ashley Shade
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.,Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, MI 48824, USA.,Program in Ecology, Evolution and Behavior, Michigan State University, East Lansing, MI 48824, USA.,Univ Lyon, CNRS, INSA Lyon, Université Claude Bernard Lyon 1, Ecole Centrale de Lyon, Ampère, UMR5005, 69134, Ecully cedex, France
| |
Collapse
|
50
|
Rodrigues M, Sabaeifard P, Yildiz MS, Coughlin L, Ahmed S, Behrendt C, Wang X, Monogue M, Kim J, Gan S, Zhan X, Filkins L, Williams NS, Hooper LV, Koh AY, Toprak E. Susceptible bacteria survive antibiotic treatment in the mammalian gastrointestinal tract without evolving resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523617. [PMID: 36711614 PMCID: PMC9882032 DOI: 10.1101/2023.01.11.523617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In vitro systems have provided great insight into the mechanisms of antibiotic resistance. Yet, in vitro approaches cannot reflect the full complexity of what transpires within a host. As the mammalian gut is host to trillions of resident bacteria and thus a potential breeding ground for antibiotic resistance, we sought to better understand how gut bacteria respond to antibiotic treatment in vivo . Here, we colonized germ-free mice with a genetically barcoded antibiotic pan-susceptible Escherichia coli clinical isolate and then administered the antibiotic cefepime via programmable subcutaneous pumps which allowed for closer emulation of human parenteral antibiotic pharmacokinetics/dynamics. After seven days of antibiotics, we were unable to culture E. coli from feces. We were, however, able to recover barcoded E. coli from harvested gastrointestinal (GI) tissue, despite high GI tract and plasma cefepime concentrations. Strikingly, these E. coli isolates were not resistant to cefepime but had acquired mutations â€" most notably in the wbaP gene, which encodes an enzyme required for the initiation of the synthesis of the polysaccharide capsule and lipopolysaccharide O antigen - that increased their ability to invade and survive within intestinal cells, including cultured human colonocytes. Further, these E. coli mutants exhibited a persister phenotype when exposed to cefepime, allowing for greater survival to pulses of cefepime treatment when compared to the wildtype strain. Our findings highlight a mechanism by which bacteria in the gastrointestinal tract can adapt to antibiotic treatment by increasing their ability to persist during antibiotic treatment and invade intestinal epithelial cells where antibiotic concentrations are substantially reduced.
Collapse
|