1
|
Chen A, Zhang T, Cheng F, Yang H, Guo Z, Zhao S, Zhang YN, Qu J. Comprehensive analysis and risk assessment of Antibiotic contaminants, antibiotic-resistant bacteria, and resistance genes: Patterns, drivers, and implications in the Songliao Basin. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 361:124852. [PMID: 39216670 DOI: 10.1016/j.envpol.2024.124852] [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: 06/03/2024] [Revised: 07/28/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
The pervasive use of antibiotics has raised substantial environmental concerns, especially regarding their temporal and spatial distribution across diverse water systems. This study addressed the gap in comprehensive research on antibiotic contamination during different hydrological periods, focusing on the Jilin section of the Songliao Basin in Northeast China, an area with severe winter ice cover. The study examined the occurrence, distribution, influencing factors, and potential ecological risks of prevalent antibiotic contaminants. Findings revealed antibiotic concentrations ranging from 239.64 to 965.81 ng/L, with antibiotic resistance genes (ARGs) at 5.22 × 10-2 16S rRNA-1 and antibiotic-resistant bacteria (ARB) up to 5.76 log10 CFU/mL. Ecological risk assessments identified significant risks to algae from oxytetracycline, erythromycin, and amoxicillin. Redundancy analysis and co-occurrence networks with ordinary least squares (OLS) demonstrated that the dispersion of ARGs and ARB is significantly influenced by environmental factors such as total organic carbon (TOC), total phosphorus (TP), total nitrogen (TN), fluoride (F⁻), and nitrate (NO₃⁻). These elements, along with mobile genetic elements (MGEs), play crucial roles in ARG patterns (R2 = 0.94, p ≤ 0.01). This investigation offers foundational insights into antibiotic pollution dynamics in cold climates, supporting the development of targeted mitigation strategies for aquatic systems.
Collapse
Affiliation(s)
- Anjie Chen
- School of Environment, Northeast Normal University, Changchun, 130117, China
| | - Tingting Zhang
- School of Environment, Northeast Normal University, Changchun, 130117, China
| | - Fangyuan Cheng
- School of Environment, Northeast Normal University, Changchun, 130117, China
| | - Hao Yang
- School of Environment, Northeast Normal University, Changchun, 130117, China
| | - Zhengfeng Guo
- School of Environment, Northeast Normal University, Changchun, 130117, China
| | - Siyu Zhao
- School of Environment, Northeast Normal University, Changchun, 130117, China
| | - Ya-Nan Zhang
- School of Environment, Northeast Normal University, Changchun, 130117, China
| | - Jiao Qu
- School of Environment, Northeast Normal University, Changchun, 130117, China.
| |
Collapse
|
2
|
Mahmud HA, Wakeman CA. Navigating collateral sensitivity: insights into the mechanisms and applications of antibiotic resistance trade-offs. Front Microbiol 2024; 15:1478789. [PMID: 39512935 PMCID: PMC11540712 DOI: 10.3389/fmicb.2024.1478789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
The swift rise of antibiotic resistance, coupled with limited new antibiotic discovery, presents a significant hurdle to global public health, demanding innovative therapeutic solutions. Recently, collateral sensitivity (CS), the phenomenon in which resistance to one antibiotic increases vulnerability to another, has come to light as a potential path forward in this attempt. Targeting either unidirectional or reciprocal CS holds promise for constraining the emergence of drug resistance and notably enhancing treatment outcomes. Typically, the alteration of bacterial physiology, such as bacterial membrane potential, expression of efflux pumps, cell wall structures, and endogenous enzymatic actions, are involved in evolved collateral sensitivity. In this review, we present a thorough overview of CS in antibiotic therapy, including its definition, importance, and underlying mechanisms. We describe how CS can be exploited to prevent the emergence of resistance and enhance the results of treatment, but we also discuss the challenges and restrictions that come with implementing this practice. Our review underscores the importance of continued exploration of CS mechanisms in the broad spectrum and clinical validation of therapeutic approaches, offering insights into its role as a valuable tool in combating antibiotic resistance.
Collapse
Affiliation(s)
- Hafij Al Mahmud
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Catherine A. Wakeman
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
3
|
Zhao W, Ye C, Li J, Yu X. Increased risk of antibiotic resistance in surface water due to global warming. ENVIRONMENTAL RESEARCH 2024; 263:120149. [PMID: 39414103 DOI: 10.1016/j.envres.2024.120149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
As the pace of global warming accelerates, so do the threats to human health, urgent priority among them being antibiotic-resistant infections. In the context of global warming, this review summarises the direct and indirect effects of rising surface water temperatures on the development of bacterial antibiotic resistance. First, the resistance of typical pathogens such as E. coli increased with average temperature. This is not only related to increased bacterial growth rate and horizontal gene transfer frequency at high temperatures but also heat shock responses and cumulative effects. Secondly, the acceleration of bacterial growth indirectly promotes antibiotic residues in surface water, which is conducive to the growth and spread of resistant bacteria. Furthermore, the cascading effects of global warming, including the release of nutrients into the water and the resulting increase of bacteria and algae, indirectly promote the improvement of resistance. Water treatment processes exposed to high temperatures also increase the risk of resistance in surface water. The fitness costs of antibiotic resistance under these dynamic conditions are also discussed, concluding the relationship between various factors and resistance persistence. It was expected to provide a comprehensive basis for mitigating antibiotic resistance in the face of global warming.
Collapse
Affiliation(s)
- Wenya Zhao
- College of the Environment & Ecology, Xiamen University, Xiamen. 361102, China
| | - Chengsong Ye
- College of the Environment & Ecology, Xiamen University, Xiamen. 361102, China
| | - Jianguo Li
- College of the Environment & Ecology, Xiamen University, Xiamen. 361102, China
| | - Xin Yu
- College of the Environment & Ecology, Xiamen University, Xiamen. 361102, China.
| |
Collapse
|
4
|
Schmidlin K, Ogbunugafor CB, Alexander S, Geiler-Samerotte K. Environment by environment interactions (ExE) differ across genetic backgrounds (ExExG). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593194. [PMID: 38766025 PMCID: PMC11100745 DOI: 10.1101/2024.05.08.593194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
While the terms "gene-by-gene interaction" (GxG) and "gene-by-environment interaction" (GxE) are widely recognized in the fields of quantitative and evolutionary genetics, "environment-byenvironment interaction" (ExE) is a term used less often. In this study, we find that environmentby-environment interactions are a meaningful driver of phenotypes, and moreover, that they differ across different genotypes (suggestive of ExExG). To support this conclusion, we analyzed a large dataset of roughly 1,000 mutant yeast strains with varying degrees of resistance to different antifungal drugs. Our findings reveal that the effectiveness of a drug combination, relative to single drugs, often differs across drug resistant mutants. Remarkably, even mutants that differ by only a single nucleotide change can have dramatically different drug × drug (ExE) interactions. We also introduce a new framework that more accurately predicts the direction and magnitude of ExE interactions for some mutants. Understanding how ExE interactions change across genotypes (ExExG) is crucial not only for modeling the evolution of pathogenic microbes, but also for enhancing our knowledge of the underlying cell biology and the sources of phenotypic variance within populations. While the significance of ExExG interactions has been overlooked in evolutionary and population genetics, these fields and others stand to benefit from understanding how these interactions shape the complex behavior of living systems.
Collapse
Affiliation(s)
- Kara Schmidlin
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ, 85287
- School of Life Sciences, Arizona State University, Tempe AZ, 85287
| | - C. Brandon Ogbunugafor
- Department of Ecology & Evolutionary Biology, Yale University, New Haven, CT,06511
- Santa Fe Institute, Santa Fe, NM, 87501
| | - Sastokas Alexander
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ, 85287
- School of Life Sciences, Arizona State University, Tempe AZ, 85287
| | - Kerry Geiler-Samerotte
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ, 85287
- School of Life Sciences, Arizona State University, Tempe AZ, 85287
| |
Collapse
|
5
|
Okombo J, Fidock DA. Towards next-generation treatment options to combat Plasmodium falciparum malaria. Nat Rev Microbiol 2024:10.1038/s41579-024-01099-x. [PMID: 39367132 DOI: 10.1038/s41579-024-01099-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 10/06/2024]
Abstract
Malaria, which is caused by infection of red blood cells with Plasmodium parasites, can be fatal in non-immune individuals if left untreated. The recent approval of the pre-erythrocytic vaccines RTS, S/AS01 and R21/Matrix-M has ushered in hope of substantial reductions in mortality rates, especially when combined with other existing interventions. However, the efficacy of these vaccines is partial, and chemotherapy remains central to malaria treatment and control. For many antimalarial drugs, clinical efficacy has been compromised by the emergence of drug-resistant Plasmodium falciparum strains. Therefore, there is an urgent need for new antimalarial medicines to complement the existing first-line artemisinin-based combination therapies. In this Review, we discuss various opportunities to expand the present malaria treatment space, appraise the current antimalarial drug development pipeline and highlight examples of promising targets. We also discuss other approaches to circumvent antimalarial resistance and how potency against drug-resistant parasites could be retained.
Collapse
Affiliation(s)
- John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
6
|
Wang Y, Wang X, Zhang C, Li R, Li J, Shi H, Zhang C, Feng L. Customized A-D-A type molecule to construct a nitric oxide nanogenerator with enhanced antibacterial activity for infected wound healing. J Mater Chem B 2024; 12:9675-9685. [PMID: 39193614 DOI: 10.1039/d4tb01201a] [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: 08/29/2024]
Abstract
Bacterial infections pose an increasingly serious threat to global health due to the development of drug-resistant strains. Developing a method to efficiently kill bacteria and promote tissue repair is imperative to decrease the damage from bacterial infection, especially infected wounds. Herein, a biofriendly and light-controlled nitric oxide (NO) generator HFB with simultaneous bacterial killing and wound repair properties is reported based on a tailored light-responsive molecule F(EIBC)2. HFB demonstrates an appropriate photothermal conversion efficiency of 33.4% and type I reactive oxygen species (˙OH and H2O2) generation capability to simultaneously trigger NO generation and potently kill bacteria. Furthermore, HFB can effectively eradicate mature bacterial biofilms with the aid of favorable permeability of NO. Additionally, HFB effectively eradicates Staphylococcus aureus in infected wounds of living mice and accelerates healing via NO-induced angiogenesis and collagen deposition. Owing to the encapsulated human serum albumin (HSA), heavy metal-free feature, and synergistic killing mechanism, HFB exhibits good biosafety to surrounding tissue and major organs. This work provides a novel dual-functional photo-responsive molecule and a potential light-controlled release platform for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Yunxia Wang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
| | - Xiaohuan Wang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
| | - Chuangxin Zhang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
| | - Ruipeng Li
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
| | - Jing Li
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
| | - Hu Shi
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
| | - Caihong Zhang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
| | - Liheng Feng
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
- Institute for Carbon-Based Thin Film Electronics, Peking University, Shanxi (ICTFE-PKU), Taiyuan 030012, China
| |
Collapse
|
7
|
Yu W, Guo X, Li X, Wei Y, Lyu Y, Zhang L, Wang J, Shan A. Novel multidomain peptide self-assembly biomaterials based on bola structure and terminal anchoring: Nanotechnology meets antimicrobial therapy. Mater Today Bio 2024; 28:101183. [PMID: 39221200 PMCID: PMC11363844 DOI: 10.1016/j.mtbio.2024.101183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/28/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
To ameliorate the diminished antimicrobial efficiency and physiological stability associated with monomeric antimicrobial peptides (AMPs) molecules, future research will focus on the artificial design of self-assembling peptides to replace monomeric entities, aiming to combat the antibiotic resistance crisis caused by microbial infections. In this study, the "bola" structure was used as the foundational architecture driving molecular self-assembly, with hydrophobic amino acids at the termini to anchor and finely adjust the sequence, thereby organizing a range of novel multidomain peptides (MDPs) templates into an ABA block motif. The results indicate that FW2 (GMSI = 53.94) exhibits the highest selectivity index among all MDPs and can form spherical micelles in an aqueous medium without the addition of any exogenous additives. FW2 exhibited high stability in vitro in the presence of physiological salt ions, serum, and various pH conditions. It exhibited excellent biocompatibility and efficacy both in vivo and in vitro. Furthermore, FW2 strongly interacts with the lipid membrane and employs various synergistic mechanisms, such as reactive oxygen species (ROS) accumulation, collectively driving cellular apoptosis. This study demonstrates a straightforward strategy for designing self-assembling peptides and promotes the advancement of peptide-based biomaterials integration progress with nanotechnology.
Collapse
Affiliation(s)
- Weikang Yu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xu Guo
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xuefeng Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yingxin Wei
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yinfeng Lyu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Licong Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jiajun Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| |
Collapse
|
8
|
Rong L, Wu L, Zong L, Wang W, Xiao Y, Yang C, Pan H, Zou X. Evolution of the Black solider fly larvae gut antibiotic resistome during kitchen waste disposal. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:135878. [PMID: 39321479 DOI: 10.1016/j.jhazmat.2024.135878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/21/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Kitchen waste (KW) is an important reservoir of antibiotic resistance genes (ARGs). Black solider fly larvae (BSFL) are extensively employed in KW disposal, closely linking to their robust gut microbes. However, antibiotic resistome in BSFL gut during the KW disposal processes and the mechanism remain unclear. In the present study, the antibiotic resistome in BSFL gut within the 12 days KW disposal processes were investigated. Results showed that, ARGs abundance initially increased and subsequently decreased, the five most prevalent core ARG classes were tetracycline, aminoglycoside, cephalosporin, lincosamide and multidrug. A total of 7 MGE types were observed and the horizontal gene transfer (HGT) of ARGs was predominantly mediated by plasmids. Host microbes were mainly categorized into Proteobacteria (98.12 %) and their assemblies were mainly classified into the deterministic processes. To elucidate the driving mechanisms, the mantel test and the structural equation model (SEM) were developed. Results indicated that microbial functions (0.912, p < 0.0001) and microbial community (1.014, p = 0.036), consistently showed very significant relationships with the patterns of ARGs, which presented higher direct effects than indirect effects. Overall, this study makes an initial contribution to a more deepgoing comprehension of the gut antibiotic resistome of BSFL during KW disposal.
Collapse
Affiliation(s)
- Lingling Rong
- School of Life Science, Jinggangshan University, Ji'an 343009, China; College of Environmental Science and Engineering, Guilin University of Technology, Guilin 541004, China
| | - Ligui Wu
- School of Life Science, Jinggangshan University, Ji'an 343009, China
| | - Lihui Zong
- School of Life Science, Jinggangshan University, Ji'an 343009, China
| | - Wei Wang
- School of Life Science, Jinggangshan University, Ji'an 343009, China
| | - Yi Xiao
- School of Life Science, Jinggangshan University, Ji'an 343009, China
| | - Chunyan Yang
- School of Life Science, Jinggangshan University, Ji'an 343009, China
| | - Hongcheng Pan
- College of Environmental Science and Engineering, Guilin University of Technology, Guilin 541004, China.
| | - Xiaoming Zou
- School of Life Science, Jinggangshan University, Ji'an 343009, China.
| |
Collapse
|
9
|
Schmidlin K, Apodaca S, Newell D, Sastokas A, Kinsler G, Geiler-Samerotte K. Distinguishing mutants that resist drugs via different mechanisms by examining fitness tradeoffs. eLife 2024; 13:RP94144. [PMID: 39255191 PMCID: PMC11386965 DOI: 10.7554/elife.94144] [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] [Indexed: 09/12/2024] Open
Abstract
There is growing interest in designing multidrug therapies that leverage tradeoffs to combat resistance. Tradeoffs are common in evolution and occur when, for example, resistance to one drug results in sensitivity to another. Major questions remain about the extent to which tradeoffs are reliable, specifically, whether the mutants that provide resistance to a given drug all suffer similar tradeoffs. This question is difficult because the drug-resistant mutants observed in the clinic, and even those evolved in controlled laboratory settings, are often biased towards those that provide large fitness benefits. Thus, the mutations (and mechanisms) that provide drug resistance may be more diverse than current data suggests. Here, we perform evolution experiments utilizing lineage-tracking to capture a fuller spectrum of mutations that give yeast cells a fitness advantage in fluconazole, a common antifungal drug. We then quantify fitness tradeoffs for each of 774 evolved mutants across 12 environments, finding these mutants group into classes with characteristically different tradeoffs. Their unique tradeoffs may imply that each group of mutants affects fitness through different underlying mechanisms. Some of the groupings we find are surprising. For example, we find some mutants that resist single drugs do not resist their combination, while others do. And some mutants to the same gene have different tradeoffs than others. These findings, on one hand, demonstrate the difficulty in relying on consistent or intuitive tradeoffs when designing multidrug treatments. On the other hand, by demonstrating that hundreds of adaptive mutations can be reduced to a few groups with characteristic tradeoffs, our findings may yet empower multidrug strategies that leverage tradeoffs to combat resistance. More generally speaking, by grouping mutants that likely affect fitness through similar underlying mechanisms, our work guides efforts to map the phenotypic effects of mutation.
Collapse
Affiliation(s)
- Kara Schmidlin
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Sam Apodaca
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Daphne Newell
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Alexander Sastokas
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Grant Kinsler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, United States
| | - Kerry Geiler-Samerotte
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| |
Collapse
|
10
|
Ziller L, Blum PC, Buhl EM, Krüttgen A, Horz HP, Tagliaferri TL. Newly isolated Drexlerviridae phage LAPAZ is physically robust and fosters eradication of Klebsiella pneumoniae in combination with meropenem. Virus Res 2024; 347:199417. [PMID: 38880333 PMCID: PMC11245953 DOI: 10.1016/j.virusres.2024.199417] [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: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Due to the spread of multidrug resistance there is a renewed interest in using bacteriophages (briefly: phages) for controlling bacterial pathogens. The objective of this study was the characterization of a newly isolated phage (i.e. phage LAPAZ, vB_KpnD-LAPAZ), its antimicrobial activity against multidrug resistant Klebsiella pneumoniae and potential synergistic interactions with antibiotics. LAPAZ belongs to the family Drexlerviridae (genus: Webervirus) and lysed 30 % of tested strains, whereby four distinct capsular types can be infected. The genome consists of 51,689 bp and encodes 84 ORFs. The latent period is 30 min with an average burst size of 27 PFU/cell. Long-term storage experiments show that LAPAZ is significantly more stable in wastewater compared to laboratory media. A phage titre of 90 % persists up to 30 min at 50 ˚C and entire phage loss was seen only at temperatures > 66 ˚C. Besides stability against UV-C, antibacterial activity in liquid culture medium was consistent at pH values ranging from 4 to 10. Unlike exposure to phage or antibiotic alone, synergistic interactions and a complete bacterial eradication was achieved when combining LAPAZ with meropenem. In addition, synergism with the co-presence of ciprofloxacin was observed and phage resistance emergence could be delayed. Without co-addition of the antibiotic, phage resistant mutants readily emerged and showed a mixed pattern of drug sensitivity alterations. Around 88 % became less sensitive towards ceftazidime, meropenem and gentamicin. Conversely, around 44 % showed decreased resistance levels against ciprofloxacin. Whole genome analysis of a phage-resistant mutant with a 16-fold increased sensitivity towards ciprofloxacin revealed one de novo frameshift mutation leading to a gene fusion affecting two transport proteins belonging to the major facilitator-superfamily (MFS). Apparently, this mutation compromises ciprofloxacin efflux efficiency and further studies are warranted to understand how the non-mutated protein might be involved in phage-host adsorption.
Collapse
Affiliation(s)
- Leonie Ziller
- Institute of Medical Microbiology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | | | - Eva Miriam Buhl
- Electron Microscopy Facility, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Alex Krüttgen
- Laboratory Diagnostic Center, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Hans-Peter Horz
- Institute of Medical Microbiology, RWTH Aachen University Hospital, 52074 Aachen, Germany.
| | | |
Collapse
|
11
|
Yang A, Song J, Li J, Li Y, Bai S, Zhou C, Wang M, Zhou Y, Wen K, Luo M, Chen P, Liu B, Yang H, Bai Y, Wong WL, Cai Q, Pu H, Qian Y, Hu W, Huang W, Wan M, Zhang C, Feng X. Ligand-Receptor Interaction-Induced Intracellular Phase Separation: A Global Disruption Strategy for Resistance-Free Lethality of Pathogenic Bacteria. J Am Chem Soc 2024; 146:23121-23137. [PMID: 38980064 DOI: 10.1021/jacs.4c04749] [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: 07/10/2024]
Abstract
Addressing the global challenge of bacterial resistance demands innovative approaches, among which multitargeting is a widely used strategy. Current strategies of multitargeting, typically achieved through drug combinations or single agents inherently aiming at multiple targets, face challenges such as stringent pharmacokinetic and pharmacodynamic requirements and cytotoxicity concerns. In this report, we propose a bacterial-specific global disruption approach as a vastly expanded multitargeting strategy that effectively disrupts bacterial subcellular organization. This effect is achieved through a pioneering chemical design of ligand-receptor interaction-induced aggregation of small molecules, i.e., DNA-induced aggregation of a diarginine peptidomimetic within bacterial cells. These intracellular aggregates display affinity toward various proteins and thus substantially interfere with essential bacterial functions and rupture bacterial cell membranes in an "inside-out" manner, leading to robust antibacterial activities and suppression of drug resistance. Additionally, biochemical analysis of macromolecule binding affinity, cytoplasmic localization patterns, and bacterial stress responses suggests that this bacterial-specific intracellular aggregation mechanism is fundamentally different from nonselective classic DNA or membrane binding mechanisms. These mechanistic distinctions, along with the peptidomimetic's selective permeation of bacterial membranes, contribute to its favorable biocompatibility and pharmacokinetic properties, enabling its in vivo antimicrobial efficacy in several animal models, including mice-based superficial wound models, subcutaneous abscess models, and septicemia infection models. These results highlight the great promise of ligand-receptor interaction-induced intracellular aggregation in achieving a globally disruptive multitargeting effect, thereby offering potential applications in the treatment of malignant cells, including pathogens, tumor cells, and infected tissues.
Collapse
Affiliation(s)
- Anming Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Junfeng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Jiaqi Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Youzhi Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Silei Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Cailing Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Min Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yu Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Kang Wen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Miaomiao Luo
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Peiren Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Bo Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Huan Yang
- School of Medical Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Yugang Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR 999077, China
| | - Qingyun Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Huangsheng Pu
- College of Advanced Interdisciplinary Studies & Hunan Provincial Key Laboratory of Novel NanoOptoelectronic Information Materials and Devices, National University of Defense Technology, Changsha, Hunan 410073, China
- Nanhu Laser Laboratory, National University of Defense Technology, Changsha 410073, China
| | - Yu Qian
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenhao Hu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wei Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Muyang Wan
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Chunhui Zhang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Xinxin Feng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
12
|
Nyhoegen C, Bonhoeffer S, Uecker H. The many dimensions of combination therapy: How to combine antibiotics to limit resistance evolution. Evol Appl 2024; 17:e13764. [PMID: 39100751 PMCID: PMC11297101 DOI: 10.1111/eva.13764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/30/2024] [Accepted: 07/14/2024] [Indexed: 08/06/2024] Open
Abstract
In combination therapy, bacteria are challenged with two or more antibiotics simultaneously. Ideally, separate mutations are required to adapt to each of them, which is a priori expected to hinder the evolution of full resistance. Yet, the success of this strategy ultimately depends on how well the combination controls the growth of bacteria with and without resistance mutations. To design a combination treatment, we need to choose drugs and their doses and decide how many drugs get mixed. Which combinations are good? To answer this question, we set up a stochastic pharmacodynamic model and determine the probability to successfully eradicate a bacterial population. We consider bacteriostatic and two types of bactericidal drugs-those that kill independent of replication and those that kill during replication. To establish results for a null model, we consider non-interacting drugs and implement the two most common models for drug independence-Loewe additivity and Bliss independence. Our results show that combination therapy is almost always better in limiting the evolution of resistance than administering just one drug, even though we keep the total drug dose constant for a 'fair' comparison. Yet, exceptions exist for drugs with steep dose-response curves. Combining a bacteriostatic and a bactericidal drug which can kill non-replicating cells is particularly beneficial. Our results suggest that a 50:50 drug ratio-even if not always optimal-is usually a good and safe choice. Applying three or four drugs is beneficial for treatment of strains with large mutation rates but adding more drugs otherwise only provides a marginal benefit or even a disadvantage. By systematically addressing key elements of treatment design, our study provides a basis for future models which take further factors into account. It also highlights conceptual challenges with translating the traditional concepts of drug independence to the single-cell level.
Collapse
Affiliation(s)
- Christin Nyhoegen
- Research Group Stochastic Evolutionary Dynamics, Department of Theoretical BiologyMax Planck Institute for Evolutionary BiologyPlonGermany
| | - Sebastian Bonhoeffer
- Department of Environmental Systems Science, Institute of Integrative BiologyETH ZurichZurichSwitzerland
| | - Hildegard Uecker
- Research Group Stochastic Evolutionary Dynamics, Department of Theoretical BiologyMax Planck Institute for Evolutionary BiologyPlonGermany
| |
Collapse
|
13
|
James B, Reesaul H, Kashif S, Behruznia M, Meehan CJ, Domingo-Sananes MR, Hubbard ATM. The effect of antibiotic selection on collateral effects and evolvability of uropathogenic Escherichia coli. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:19. [PMID: 39036800 PMCID: PMC11254750 DOI: 10.1038/s44259-024-00037-4] [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: 12/07/2023] [Accepted: 06/28/2024] [Indexed: 07/23/2024]
Abstract
Trimethoprim is recommended as a first-line treatment of urinary tract infections (UTIs) in the UK. In 2018, 31.4% of Escherichia coli isolated from UTIs in England were trimethoprim-resistant, leading to overreliance on other first and second-line antibiotics. Here, we assessed whether, in principle, prior selection with trimethoprim results in collateral effects to other antibiotics recommended for the treatment of UTIs. As collateral effects, we considered changes in susceptibility, mutation-selection window and population establishment probability. We selected 10 trimethoprim-resistant derivatives from three clinical isolates of uropathogenic Escherichia coli. We found that mutations conferring trimethoprim resistance did not have any collateral effects on fosfomycin. In contrast, resistance to trimethoprim resulted in decreased susceptibility (collateral resistance) to nitrofurantoin, below the clinical breakpoint and narrowed the mutation-selection window, thereby reducing the maximum concentration for selection of nitrofurantoin resistance mutations. Our analyses demonstrate that multiple collateral responses should be accounted for when predicting and optimising antibiotic use, limiting future antimicrobial resistance emergence.
Collapse
Affiliation(s)
- Beth James
- Department of Biosciences, Nottingham Trent University, Clifton Campus, College Drive, Clifton, Nottingham NG11 8NS UK
| | - Hishikha Reesaul
- Department of Biosciences, Nottingham Trent University, Clifton Campus, College Drive, Clifton, Nottingham NG11 8NS UK
| | - Sidra Kashif
- Department of Biosciences, Nottingham Trent University, Clifton Campus, College Drive, Clifton, Nottingham NG11 8NS UK
| | - Mahboobeh Behruznia
- Department of Biosciences, Nottingham Trent University, Clifton Campus, College Drive, Clifton, Nottingham NG11 8NS UK
| | - Conor J. Meehan
- Department of Biosciences, Nottingham Trent University, Clifton Campus, College Drive, Clifton, Nottingham NG11 8NS UK
| | - Maria Rosa Domingo-Sananes
- Department of Biosciences, Nottingham Trent University, Clifton Campus, College Drive, Clifton, Nottingham NG11 8NS UK
| | - Alasdair T. M. Hubbard
- Department of Biosciences, Nottingham Trent University, Clifton Campus, College Drive, Clifton, Nottingham NG11 8NS UK
| |
Collapse
|
14
|
Jung HS, Park YJ, Gu BH, Han G, Ji W, Hwang SM, Kim M. Coumarin derivatives ameliorate the intestinal inflammation and pathogenic gut microbiome changes in the model of infectious colitis through antibacterial activity. Front Cell Infect Microbiol 2024; 14:1362773. [PMID: 39081865 PMCID: PMC11287663 DOI: 10.3389/fcimb.2024.1362773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Coumarin, a phenolic compound, is a secondary metabolite produced by plants such as Tanga and Lime. Coumarin derivatives were prepared via Pechmann condensation. In this study, we performed in vitro and in vivo experiments to determine the antimicrobial and gut immune-regulatory functions of coumarin derivatives. For the in vitro antimicrobial activity assay, coumarin derivatives C1 and C2 were selected based on their pathogen-killing activity against various pathogenic microbes. We further demonstrated that the selected coumarin derivatives disrupted bacterial cell membranes. Next, we examined the regulatory function of the coumarin derivatives in gut inflammation using an infectious colitis model. In an in vivo infectious colitis model, administration of selected C1 coumarin derivatives reduced pathogen loads, the number of inflammatory immune cells (Th1 cells and Th17 cells), and inflammatory cytokine levels (IL-6 and IL-1b) in the intestinal tissue after pathogen infection. In addition, we found that the administration of C1 coumarin derivatives minimized abnormal gut microbiome shift-driven pathogen infection. Potential pathogenic gut microbes, such as Enterobacteriaceae and Staphylococcaceae, were increased by pathogen infection. However, this pathogenic microbial expansion was minimized and beneficial bacteria, such as Ligilactobacillus and Limosilactobacillus, increased with C1 coumarin derivative treatment. Functional gene enrichment assessment revealed that the relative abundance of genes associated with lipid and nucleotide metabolism was reduced by pathogen infection; however, this phenomenon was not observed in C1 coumarin derivative-treated animals. Collectively, our data suggest that C1 coumarin derivative is effective antibacterial agents that minimize pathogen-induced gut inflammation and abnormal gut microbiome modulation through their antibacterial activity.
Collapse
Affiliation(s)
- Hui-su Jung
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea
| | - Yei Ju Park
- R & D Center, EyeGene, Goyang, Republic of Korea
| | - Bon-Hee Gu
- Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| | - Goeun Han
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea
- Future Earth Research Institute, PNU JYS Science Academy, Pusan National University, Busan, Republic of Korea
| | - Woonhak Ji
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea
| | - Su mi Hwang
- Department of Biomedical Laboratory Science, College of Health and Medical Science, Sangji University, Wonju, Republic of Korea
| | - Myunghoo Kim
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea
- Future Earth Research Institute, PNU JYS Science Academy, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
15
|
Li Z, Ouyang L, Lu Y, Peng Q, Qiao X, Wu Q, Zhang B, Liu B, Wan F, Qian W. Antibiotics suppress the expression of antimicrobial peptides and increase sensitivity of Cydia pomonella to granulosis virus. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 947:174612. [PMID: 38992382 DOI: 10.1016/j.scitotenv.2024.174612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/13/2024]
Abstract
Cydia pomonella granulovirus (CpGV) is a highly specific and environmentally friendly pathogenic virus successfully used as a biological insecticide against codling moth larvae. Continuous application of CpGV has led to high levels of resistance in codling moth, Cydia pomonella (C. pomonella). Nevertheless, the specific molecular mechanisms underlying the development of resistance in codling moths to CpGV have been rarely investigated. This study explored the potential antiviral immune roles of codling moth antimicrobial peptides (AMPs) against CpGV. A total of 11 AMP genes classified in cecropin, defensin, gloverin, and attacin subfamilies, were identified in the codling moth genome. The cecropin and gloverin subfamilies were found to be the ancestral genes of the AMP gene family. The expression of two AMP genes (CmGlo1 and CmAtt1) significantly increased following CpGV challenge, and CmGlo1 and CmAtt1 gene silencing resulted in a significant increase in CpGV replication in codling moth larvae. The hemolymph and fat body serve as major viral immune functional tissues in codling moth larvae. Moreover, zhongshengmycin significantly reduced the diversity and abundance of codling moth larvae gut microbiota, thereby suppressing the expression of CmAtt1 AMP gene. We also found that the combination of the virus with zhongshengmycin would enhance the insecticidal effects of CpGV. This study provides the first explanation of the molecular mechanisms driving CpGV immune function development in codling moths, approached from the perspective of the codling moth itself. Additionally, we introduced an alternative approach to combat codling moth in the field by combining antibiotics with biopesticides to amplify the insecticidal effects of the latter.
Collapse
Affiliation(s)
- Zaiyuan Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China.
| | - Lan Ouyang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China; College of Plant Health & Medicine, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Yin Lu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China; School of Life Sciences, Henan University, Kaifeng 475004, China; Shenzhen Research Institute, Henan University, Shenzhen 518000, China.
| | - Qi Peng
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China; College of Plant Health & Medicine, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Xi Qiao
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China.
| | - Qiang Wu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China.
| | - Bin Zhang
- College of Plant Health & Medicine, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Bo Liu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China; College of Plant Health & Medicine, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Fanghao Wan
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China; College of Plant Health & Medicine, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Wanqiang Qian
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China.
| |
Collapse
|
16
|
Antunes B, Zanchi C, Johnston PR, Maron B, Witzany C, Regoes RR, Hayouka Z, Rolff J. The evolution of antimicrobial peptide resistance in Pseudomonas aeruginosa is severely constrained by random peptide mixtures. PLoS Biol 2024; 22:e3002692. [PMID: 38954678 PMCID: PMC11218975 DOI: 10.1371/journal.pbio.3002692] [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: 02/26/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024] Open
Abstract
The prevalence of antibiotic-resistant pathogens has become a major threat to public health, requiring swift initiatives for discovering new strategies to control bacterial infections. Hence, antibiotic stewardship and rapid diagnostics, but also the development, and prudent use, of novel effective antimicrobial agents are paramount. Ideally, these agents should be less likely to select for resistance in pathogens than currently available conventional antimicrobials. The usage of antimicrobial peptides (AMPs), key components of the innate immune response, and combination therapies, have been proposed as strategies to diminish the emergence of resistance. Herein, we investigated whether newly developed random antimicrobial peptide mixtures (RPMs) can significantly reduce the risk of resistance evolution in vitro to that of single sequence AMPs, using the ESKAPE pathogen Pseudomonas aeruginosa (P. aeruginosa) as a model gram-negative bacterium. Infections of this pathogen are difficult to treat due the inherent resistance to many drug classes, enhanced by the capacity to form biofilms. P. aeruginosa was experimentally evolved in the presence of AMPs or RPMs, subsequentially assessing the extent of resistance evolution and cross-resistance/collateral sensitivity between treatments. Furthermore, the fitness costs of resistance on bacterial growth were studied and whole-genome sequencing used to investigate which mutations could be candidates for causing resistant phenotypes. Lastly, changes in the pharmacodynamics of the evolved bacterial strains were examined. Our findings suggest that using RPMs bears a much lower risk of resistance evolution compared to AMPs and mostly prevents cross-resistance development to other treatments, while maintaining (or even improving) drug sensitivity. This strengthens the case for using random cocktails of AMPs in favour of single AMPs, against which resistance evolved in vitro, providing an alternative to classic antibiotics worth pursuing.
Collapse
Affiliation(s)
- Bernardo Antunes
- Freie Universität Berlin, Evolutionary Biology, Berlin, Germany
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Caroline Zanchi
- Freie Universität Berlin, Evolutionary Biology, Berlin, Germany
| | - Paul R. Johnston
- Freie Universität Berlin, Evolutionary Biology, Berlin, Germany
- Berlin Centre for Genomics in Biodiversity Research, Berlin, Germany
- University of St. Andrews, School of Medicine, North Haugh, St Andrews, Fife, United Kingdom
| | - Bar Maron
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot, Israel
| | | | - Roland R. Regoes
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Jens Rolff
- Freie Universität Berlin, Evolutionary Biology, Berlin, Germany
- Berlin Centre for Genomics in Biodiversity Research, Berlin, Germany
| |
Collapse
|
17
|
Liang Y, Zhao H, Li Y, Gao F, Qiu J, Liu Z, Li Q. Joint effects about antibiotics combined using with antibiotics or phytochemicals on Aeromonas hydrophila. MARINE ENVIRONMENTAL RESEARCH 2024; 199:106594. [PMID: 38908112 DOI: 10.1016/j.marenvres.2024.106594] [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: 02/08/2024] [Revised: 05/01/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024]
Abstract
Aeromonas hydrophila is highly prevalent in aquaculture animals and aquaculture environment. Due to the abuse of antibiotics, A. hydrophila can change the antibiotic resistance spectrum directly and affect human health indirectly. The use of combined drugs replacement therapy and the long-term coexistence with drug-resistant bacteria are the reality that human beings have to face in dealing with the problem of antibiotic resistance in the future. This study showed the characteristics and trends through abundant results of combined effects related with the combinations of antibiotic and the combinations of antibiotic and phytochemical on A. hydrophila, and revealed the antagonism probability of combinations of antibiotic and phytochemical is significantly higher than that of the combinations of antibiotic. Meanwhile, the combinations of antibiotic and phytochemical could protect the host cells which also achieved the same effectiveness as combination of antibiotics, and the enrichment pathway was proved to be relatively discrete. In addition, the possible mechanism about the reverse "U" shape of the combined effect curve on wild/antibiotic-resistant bacteria was clarified, and it was confirmed that the antagonism for the combinations of antibiotic and phytochemical might has the significance in inhibiting the evolution of bacterial resistance mutations. This study was aims to provide theoretical basis and some clues for the antibiotic resistance control associated with A. hydrophila.
Collapse
Affiliation(s)
- Yannei Liang
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Haiqing Zhao
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Yun Li
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China.
| | - Fuqing Gao
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Jing Qiu
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Zhe Liu
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Qiongyan Li
- Key Laboratory of Agro-products Safety & Quality of Ministry of Agriculture, Beijing, PR China; Institute of Quality Standards & Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, PR China
| |
Collapse
|
18
|
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
|
19
|
Taha BA, Ahmed NM, Talreja RK, Haider AJ, Al Mashhadany Y, Al-Jubouri Q, Huddin AB, Mokhtar MHH, Rustagi S, Kaushik A, Chaudhary V, Arsad N. Synergizing Nanomaterials and Artificial Intelligence in Advanced Optical Biosensors for Precision Antimicrobial Resistance Diagnosis. ACS Synth Biol 2024; 13:1600-1620. [PMID: 38842483 DOI: 10.1021/acssynbio.4c00070] [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] [Indexed: 06/07/2024]
Abstract
Antimicrobial resistance (AMR) poses a critical global One Health concern, ensuing from unintentional and continuous exposure to antibiotics, as well as challenges in accurate contagion diagnostics. Addressing AMR requires a strategic approach that emphasizes early stage prevention through screening in clinical, environmental, farming, and livestock settings to identify nonvulnerable antimicrobial agents and the associated genes. Conventional AMR diagnostics, like antibiotic susceptibility testing, possess drawbacks, including high costs, time-consuming processes, and significant manpower requirements, underscoring the need for intelligent, prompt, and on-site diagnostic techniques. Nanoenabled artificial intelligence (AI)-supported smart optical biosensors present a potential solution by facilitating rapid point-of-care AMR detection with real-time, sensitive, and portable capabilities. This Review comprehensively explores various types of optical nanobiosensors, such as surface plasmon resonance sensors, whispering-gallery mode sensors, optical coherence tomography, interference reflection imaging sensors, surface-enhanced Raman spectroscopy, fluorescence spectroscopy, microring resonance sensors, and optical tweezer biosensors, for AMR diagnostics. By harnessing the unique advantages of these nanoenabled smart biosensors, a revolutionary paradigm shift in AMR diagnostics can be achieved, characterized by rapid results, high sensitivity, portability, and integration with Internet-of-Things (IoT) technologies. Moreover, nanoenabled optical biosensors enable personalized monitoring and on-site detection, significantly reducing turnaround time and eliminating the human resources needed for sample preservation and transportation. Their potential for holistic environmental surveillance further enhances monitoring capabilities in diverse settings, leading to improved modern-age healthcare practices and more effective management of antimicrobial treatments. Embracing these advanced diagnostic tools promises to bolster global healthcare capacity to combat AMR and safeguard One Health.
Collapse
Affiliation(s)
- Bakr Ahmed Taha
- Department of Electrical, Electronic and Systems Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia UKM, 43600 Bangi, Malaysia
| | - Naser M Ahmed
- Department of Laser and Optoelectronics Engineering, Dijlah University College, 00964 Baghdad, Iraq
| | - Rishi Kumar Talreja
- Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi 110029, India
| | - Adawiya J Haider
- Applied Sciences Department/Laser Science and Technology Branch, University of Technology, 00964 Baghdad, Iraq
| | - Yousif Al Mashhadany
- Department of Electrical Engineering, College of Engineering, University of Anbar, Anbar 00964, Iraq
| | - Qussay Al-Jubouri
- Department of Communication Engineering, University of Technology, 00964 Baghdad, Iraq
| | - Aqilah Baseri Huddin
- Department of Electrical, Electronic and Systems Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia UKM, 43600 Bangi, Malaysia
| | - Mohd Hadri Hafiz Mokhtar
- Department of Electrical, Electronic and Systems Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia UKM, 43600 Bangi, Malaysia
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttrakhand 248007, India
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, Florida 33805, United States
| | - Vishal Chaudhary
- Physics Department, Bhagini Nivedita College, University of Delhi, New Delhi 110045, India
| | - Norhana Arsad
- Department of Electrical, Electronic and Systems Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia UKM, 43600 Bangi, Malaysia
| |
Collapse
|
20
|
Schmidlin, Apodaca, Newell, Sastokas, Kinsler, Geiler-Samerotte. Distinguishing mutants that resist drugs via different mechanisms by examining fitness tradeoffs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.17.562616. [PMID: 37905147 PMCID: PMC10614906 DOI: 10.1101/2023.10.17.562616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
There is growing interest in designing multidrug therapies that leverage tradeoffs to combat resistance. Tradeoffs are common in evolution and occur when, for example, resistance to one drug results in sensitivity to another. Major questions remain about the extent to which tradeoffs are reliable, specifically, whether the mutants that provide resistance to a given drug all suffer similar tradeoffs. This question is difficult because the drug-resistant mutants observed in the clinic, and even those evolved in controlled laboratory settings, are often biased towards those that provide large fitness benefits. Thus, the mutations (and mechanisms) that provide drug resistance may be more diverse than current data suggests. Here, we perform evolution experiments utilizing lineage-tracking to capture a fuller spectrum of mutations that give yeast cells a fitness advantage in fluconazole, a common antifungal drug. We then quantify fitness tradeoffs for each of 774 evolved mutants across 12 environments, finding these mutants group into 6 classes with characteristically different tradeoffs. Their unique tradeoffs may imply that each group of mutants affects fitness through different underlying mechanisms. Some of the groupings we find are surprising. For example, we find some mutants that resist single drugs do not resist their combination, while others do. And some mutants to the same gene have different tradeoffs than others. These findings, on one hand, demonstrate the difficulty in relying on consistent or intuitive tradeoffs when designing multidrug treatments. On the other hand, by demonstrating that hundreds of adaptive mutations can be reduced to a few groups with characteristic tradeoffs, our findings may yet empower multidrug strategies that leverage tradeoffs to combat resistance. More generally speaking, by grouping mutants that likely affect fitness through similar underlying mechanisms, our work guides efforts to map the phenotypic effects of mutation.
Collapse
Affiliation(s)
- Schmidlin
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Apodaca
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Newell
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Sastokas
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Kinsler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Geiler-Samerotte
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| |
Collapse
|
21
|
Guerrero RF, Dorji T, Harris RM, Shoulders MD, Ogbunugafor CB. Evolutionary druggability for low-dimensional fitness landscapes toward new metrics for antimicrobial applications. eLife 2024; 12:RP88480. [PMID: 38833384 PMCID: PMC11149929 DOI: 10.7554/elife.88480] [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] [Indexed: 06/06/2024] Open
Abstract
The term 'druggability' describes the molecular properties of drugs or targets in pharmacological interventions and is commonly used in work involving drug development for clinical applications. There are no current analogues for this notion that quantify the drug-target interaction with respect to a given target variant's sensitivity across a breadth of drugs in a panel, or a given drug's range of effectiveness across alleles of a target protein. Using data from low-dimensional empirical fitness landscapes composed of 16 β-lactamase alleles and 7 β-lactam drugs, we introduce two metrics that capture (i) the average susceptibility of an allelic variant of a drug target to any available drug in a given panel ('variant vulnerability'), and (ii) the average applicability of a drug (or mixture) across allelic variants of a drug target ('drug applicability'). Finally, we (iii) disentangle the quality and magnitude of interactions between loci in the drug target and the seven drug environments in terms of their mutation by mutation by environment (G x G x E) interactions, offering mechanistic insight into the variant variability and drug applicability metrics. Summarizing, we propose that our framework can be applied to other datasets and pathogen-drug systems to understand which pathogen variants in a clinical setting are the most concerning (low variant vulnerability), and which drugs in a panel are most likely to be effective in an infection defined by standing genetic variation in the pathogen drug target (high drug applicability).
Collapse
Affiliation(s)
- Rafael F Guerrero
- Department of Biological Sciences, North Carolina State UniversityRaleighUnited States
| | - Tandin Dorji
- Department of Mathematics and Statistics, University of VermontBurlingtonUnited States
| | - Ra'Mal M Harris
- Department of Chemistry, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Matthew D Shoulders
- Department of Chemistry, Massachusetts Institute of TechnologyCambridgeUnited States
| | - C Brandon Ogbunugafor
- Department of Chemistry, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Ecology and Evolutionary Biology, Yale UniversityNew HavenUnited States
- Santa Fe InstituteSanta FeUnited States
- Public Health Modeling Unit, Yale School of Public HealthNew HavenUnited States
| |
Collapse
|
22
|
De Meester L, Vázquez-Domínguez E, Kassen R, Forest F, Bellon MR, Koskella B, Scherson RA, Colli L, Hendry AP, Crandall KA, Faith DP, Starger CJ, Geeta R, Araki H, Dulloo EM, Souffreau C, Schroer S, Johnson MTJ. A link between evolution and society fostering the UN sustainable development goals. Evol Appl 2024; 17:e13728. [PMID: 38884021 PMCID: PMC11178947 DOI: 10.1111/eva.13728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Given the multitude of challenges Earth is facing, sustainability science is of key importance to our continued existence. Evolution is the fundamental biological process underlying the origin of all biodiversity. This phylogenetic diversity fosters the resilience of ecosystems to environmental change, and provides numerous resources to society, and options for the future. Genetic diversity within species is also key to the ability of populations to evolve and adapt to environmental change. Yet, the value of evolutionary processes and the consequences of their impairment have not generally been considered in sustainability research. We argue that biological evolution is important for sustainability and that the concepts, theory, data, and methodological approaches used in evolutionary biology can, in crucial ways, contribute to achieving the UN Sustainable Development Goals (SDGs). We discuss how evolutionary principles are relevant to understanding, maintaining, and improving Nature Contributions to People (NCP) and how they contribute to the SDGs. We highlight specific applications of evolution, evolutionary theory, and evolutionary biology's diverse toolbox, grouped into four major routes through which evolution and evolutionary insights can impact sustainability. We argue that information on both within-species evolutionary potential and among-species phylogenetic diversity is necessary to predict population, community, and ecosystem responses to global change and to make informed decisions on sustainable production, health, and well-being. We provide examples of how evolutionary insights and the tools developed by evolutionary biology can not only inspire and enhance progress on the trajectory to sustainability, but also highlight some obstacles that hitherto seem to have impeded an efficient uptake of evolutionary insights in sustainability research and actions to sustain SDGs. We call for enhanced collaboration between sustainability science and evolutionary biology to understand how integrating these disciplines can help achieve the sustainable future envisioned by the UN SDGs.
Collapse
Affiliation(s)
- Luc De Meester
- Leibniz Institute of Freshwater Ecology and Inland Fisheries (IGB) Berlin Germany
- Laboratory of Aquatic Ecology, Evolution and Conservation KU Leuven Leuven Belgium
- Institute of Biology Freie University Berlin Berlin Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research (BBIB) Berlin Germany
| | - Ella Vázquez-Domínguez
- Departamento de Ecología de la Biodiversidad, Instituto de Ecología, Universidad Nacional Autónoma de México Ciudad Universitaria Ciudad de México Mexico
- Conservation and Evolutionary Genetics Group Estación Biológica de Doñana (EBD-CSIC) Sevilla Spain
| | - Rees Kassen
- Department of Biology McGill University Montreal Quebec Canada
| | | | - Mauricio R Bellon
- Comisión Nacional Para el Conocimiento y Uso de la Biodiversidad (CONABIO) México City Mexico
- Swette Center for Sustainable Food Systems Arizona State University Tempe Arizona USA
| | - Britt Koskella
- Department of Integrative Biology University of California Berkeley California USA
| | - Rosa A Scherson
- Laboratorio Evolución y Sistemática, Departamento de Silvicultura y Conservación de la Naturaleza Universidad de Chile Santiago Chile
| | - Licia Colli
- Dipartimento di Scienze Animali, Della Nutrizione e Degli Alimenti, BioDNA Centro di Ricerca Sulla Biodiversità e Sul DNA Antico, Facoltà di Scienze Agrarie, Alimentari e Ambientali Università Cattolica del Sacro Cuore Piacenza Italy
| | - Andrew P Hendry
- Redpath Museum & Department of Biology McGill University Montreal Quebec Canada
| | - Keith A Crandall
- Department of Biostatistics and Bioinformatics George Washington University Washington DC USA
- Department of Invertebrate Zoology, US National Museum of Natural History Smithsonian Institution Washington DC USA
| | | | - Craig J Starger
- School of Global Environmental Sustainability Colorado State University Fort Collins Colorado USA
| | - R Geeta
- Department of Botany University of Delhi New Delhi India
| | - Hitoshi Araki
- Research Faculty of Agriculture Hokkaido University Sapporo Japan
| | - Ehsan M Dulloo
- Effective Genetic Resources Conservation and Use Alliance of Bioversity International and CIAT Rome Italy
| | - Caroline Souffreau
- Laboratory of Aquatic Ecology, Evolution and Conservation KU Leuven Leuven Belgium
| | - Sibylle Schroer
- Leibniz Institute of Freshwater Ecology and Inland Fisheries (IGB) Berlin Germany
| | - Marc T J Johnson
- Department of Biology & Centre for Urban Environments University of Toronto Mississauga Mississauga Ontario Canada
| |
Collapse
|
23
|
Almowallad SJ, Alqahtani LS. Synergistic antimicrobial action of chitosan-neem extracts nanoformulation as a promising strategy for overcoming multi-drug resistant bacteria. Int J Biol Macromol 2024; 272:132337. [PMID: 38797302 DOI: 10.1016/j.ijbiomac.2024.132337] [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: 01/13/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/29/2024]
Abstract
The objective of the present study was to analyze and identify the phytochemical components found in neem leaf extracts using gas chromatography-mass spectrometry (GC-MS) and Fourier-transform infrared spectroscopy (FTIR) methods. The extract samples were acquired using ethyl acetate (EA) and petroleum ether (PE) solvents. Moreover, the extracts were assessed for their antibacterial and antioxidant features. In addition, chitosan nanoparticles (Cs NPs) containing neem extracts were synthesized and evaluated for their potential antibacterial properties, explicitly targeting multi-drug resistant (MDR) bacteria. The neem extracts were analyzed using GC-MS, which identified components such as hydrocarbons, phenolic compounds, terpenoids, alkaloids, and glycosides. Results revealed that the PE extract showed significant antibacterial activity against a range of bacteria. In addition, the PE extract exhibited significant antioxidant activity, exceeding both the EA extract and vitamin C. In addition, both extracts exhibited notable antibiofilm activity, significantly inhibiting the production of biofilm. The Cs NPs, loaded with neem extracts, exhibited significant antibacterial action against multidrug-resistant (MDR) microorganisms. The Cs NPs/EA materials had the greatest zone of inhibition values of 24 ± 2.95 mm against Pseudomonas aeruginosa. Similarly, the Cs NPs/PE materials exhibited a zone of inhibition measurement of 22 ± 3.14 mm against P. aeruginosa. This work highlights the various biochemical components of neem extracts, their strong abilities to combat bacteria and oxidative stress, and the possibility of Cs NPs containing neem extracts as effective treatments for antibiotic-resistant bacterial strains.
Collapse
Affiliation(s)
- Sanaa J Almowallad
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71421, Saudi Arabia.
| | - Leena S Alqahtani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 23445, Saudi Arabia
| |
Collapse
|
24
|
Fik-Jaskółka M, Mittova V, Motsonelidze C, Vakhania M, Vicidomini C, Roviello GN. Antimicrobial Metabolites of Caucasian Medicinal Plants as Alternatives to Antibiotics. Antibiotics (Basel) 2024; 13:487. [PMID: 38927153 PMCID: PMC11200912 DOI: 10.3390/antibiotics13060487] [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: 05/09/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
This review explores the potential of antimicrobial metabolites derived from Caucasian medicinal plants as alternatives to conventional antibiotics. With the rise of antibiotic resistance posing a global health threat, there is a pressing need to investigate alternative sources of antimicrobial agents. Caucasian medicinal plants have traditionally been used for their therapeutic properties, and recent research has highlighted their potential as sources of antimicrobial compounds. Representatives of 15 families of Caucasian medicinal plant extracts (24 species) have been explored for their efficacy against these pathogens. The effect of these plants on Gram-positive and Gram-negative bacteria and fungi is discussed in this paper. By harnessing the bioactive metabolites present in these plants, this study aims to contribute to the development of new antimicrobial treatments that can effectively combat bacterial infections while minimizing the risk of resistance emergence. Herein we discuss the following classes of bioactive compounds exhibiting antimicrobial activity: phenolic compounds, flavonoids, tannins, terpenes, saponins, alkaloids, and sulfur-containing compounds of Allium species. The review discusses the pharmacological properties of selected Caucasian medicinal plants, the extraction and characterization of these antimicrobial metabolites, the mechanisms of action of antibacterial and antifungal plant compounds, and their potential applications in clinical settings. Additionally, challenges and future directions in the research of antimicrobial metabolites from Caucasian medicinal plants are addressed.
Collapse
Affiliation(s)
- Marta Fik-Jaskółka
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Valentina Mittova
- Teaching University Geomedi, 4 King Solomon II Str., Tbilisi 0114, Georgia; (V.M.)
| | | | - Malkhaz Vakhania
- Teaching University Geomedi, 4 King Solomon II Str., Tbilisi 0114, Georgia; (V.M.)
| | - Caterina Vicidomini
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Giovanni N. Roviello
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| |
Collapse
|
25
|
Chen H, Gao J, Wang Q, Liu Y, Wu L, Fu X, Guo Y, Wang H, Wang Y. The synergistic effect of periodate/ferrate (VI) system on disinfection of antibiotic resistant bacteria and removal of antibiotic resistant genes: The dominance of Fe (IV)/Fe (V). JOURNAL OF HAZARDOUS MATERIALS 2024; 470:134132. [PMID: 38554510 DOI: 10.1016/j.jhazmat.2024.134132] [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: 02/11/2024] [Revised: 03/12/2024] [Accepted: 03/24/2024] [Indexed: 04/01/2024]
Abstract
The proliferation of antibiotic resistant genes (ARGs) and antibiotic resistant bacteria (ARB) caused by antibiotic abuse has raised concerns about the global infectious-disease crisis. This study employed periodate (PI)/ferrate (VI) (Fe (VI)) system to disinfect Gram-negative ARB (Escherichia coli DH5α) and Gram-positive bacteria (Bacillus subtilis ATCC6633). The PI/Fe (VI) system could inactivate 1 × 108 CFU/mL of Gram-negative ARB and Gram-positive bacteria by 4.0 and 2.8 log in 30 min. Neutral and acidic pH, increase of PI dosage and Fe (VI) dosage had positive impacts on the inactivation efficiency of ARB, while alkaline solution and the coexistence of 10 mM Cl-, NO3-, SO42- and 20 mg/L humic acid had slightly negative impacts. The reactive species generated by PI/Fe (VI) system could disrupt the integrity of cell membrane and wall, leading to oxidative stress and lipid peroxidation. Intracellular hereditary substance, including DNA and ARGs (tetA), would leak into the external environment through damaged cells and be degraded. The electron spin resonance analysis and quenching experiments indicated that Fe (IV)/Fe (V) played a leading role in disinfection. Meanwhile, PI/Fe (VI) system also had an efficient removal effect on sulfadiazine, which was expected to inhibit the ARGs transmission from the source.
Collapse
Affiliation(s)
- Hao Chen
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, Beijing 100124, China
| | - Jingfeng Gao
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, Beijing 100124, China.
| | - Qian Wang
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, Beijing 100124, China
| | - Ying Liu
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, Beijing 100124, China
| | - Lei Wu
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, Beijing 100124, China
| | - Xiaoyu Fu
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yi Guo
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, Beijing 100124, China
| | - Hanyi Wang
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yuxuan Wang
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
26
|
Simoni A, Schwartz L, Junquera GY, Ching CB, Spencer JD. Current and emerging strategies to curb antibiotic-resistant urinary tract infections. Nat Rev Urol 2024:10.1038/s41585-024-00877-9. [PMID: 38714857 PMCID: PMC11540872 DOI: 10.1038/s41585-024-00877-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/23/2024]
Abstract
Rising rates of antibiotic resistance in uropathogenic bacteria compromise patient outcomes and prolong hospital stays. Consequently, new strategies are needed to prevent and control the spread of antibiotic resistance in uropathogenic bacteria. Over the past two decades, sizeable clinical efforts and research advances have changed urinary tract infection (UTI) treatment and prevention strategies to conserve antibiotic use. The emergence of antimicrobial stewardship, policies from national societies, and the development of new antimicrobials have shaped modern UTI practices. Future UTI management practices could be driven by the evolution of antimicrobial stewardship, improved and readily available diagnostics, and an improved understanding of how the microbiome affects UTI. Forthcoming UTI treatment and prevention strategies could employ novel bactericidal compounds, combinations of new and classic antimicrobials that enhance bacterial killing, medications that prevent bacterial attachment to uroepithelial cells, repurposing drugs, and vaccines to curtail the rising rates of antibiotic resistance in uropathogenic bacteria and improve outcomes in people with UTI.
Collapse
Affiliation(s)
- Aaron Simoni
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
| | - Laura Schwartz
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- Department of Pediatrics, Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Guillermo Yepes Junquera
- Department of Pediatrics, Division of Infectious Diseases, Nationwide Children's, Columbus, OH, USA
| | - Christina B Ching
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- Department of Urology, Nationwide Children's, Columbus, OH, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- Department of Pediatrics, Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
27
|
Xia L, Li Y, Wang Y, Zhou H, Dandekar AA, Wang M, Xu F. Quorum-sensing regulation of phenazine production heightens Pseudomonas aeruginosa resistance to ciprofloxacin. Antimicrob Agents Chemother 2024; 68:e0011824. [PMID: 38526048 PMCID: PMC11064481 DOI: 10.1128/aac.00118-24] [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: 01/21/2024] [Accepted: 03/07/2024] [Indexed: 03/26/2024] Open
Abstract
Quorum sensing is a type of cell-cell communication that modulates various biological activities of bacteria. Previous studies indicate that quorum sensing contributes to the evolution of bacterial resistance to antibiotics, but the underlying mechanisms are not fully understood. In this study, we grew Pseudomonas aeruginosa in the presence of sub-lethal concentrations of ciprofloxacin, resulting in a large increase in ciprofloxacin minimal inhibitory concentration. We discovered that quorum sensing-mediated phenazine biosynthesis was significantly enhanced in the resistant isolates, where the quinolone circuit was the predominant contributor to this phenomenon. We found that production of pyocyanin changed carbon flux and showed that the effect can be partially inhibited by the addition of pyruvate to cultures. This study illustrates the role of quorum sensing-mediated phenotypic resistance and suggests a strategy for its prevention.
Collapse
Affiliation(s)
- Lexin Xia
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Li
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Yufan Wang
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Hui Zhou
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ajai A. Dandekar
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Meizhen Wang
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
28
|
Wang H, Yang Y, Wang S, Badawy S, Ares I, Martínez M, Lopez-Torres B, Martínez-Larrañaga MR, Wang X, Anadón A, Martínez MA. Antimicrobial sensitisers: Gatekeepers to avoid the development of multidrug-resistant bacteria. J Control Release 2024; 369:25-38. [PMID: 38508527 DOI: 10.1016/j.jconrel.2024.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/23/2024] [Accepted: 03/16/2024] [Indexed: 03/22/2024]
Abstract
The resistance of multidrug-resistant bacteria to existing antibiotics forces the continued development of new antibiotics and antibacterial agents, but the high costs and long timeframe involved in the development of new agents renders the hope that existing antibiotics may again play a part. The "antibiotic adjuvant" is an indirect antibacterial strategy, but its vague concept has, in the past, limited the development speed of related drugs. In this review article, we put forward an accurate concept of a "non-self-antimicrobial sensitisers (NSAS)", to distinguish it from an "antibiotic adjuvant", and then discuss several scientific methods to restore bacterial sensitivity to antibiotics, and the sources and action mechanism of existing NSAS, in order to guide the development and further research of NSAS.
Collapse
Affiliation(s)
- Hanfei Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yingying Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Simeng Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Sara Badawy
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Pathology Department of Animal Medicine, Faculty of Veterinary Medicine, Benha University, Egypt
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital, 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Marta Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital, 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital, 12 de Octubre (i+12), 28040 Madrid, Spain
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital, 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital, 12 de Octubre (i+12), 28040 Madrid, Spain.
| | - María-Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital, 12 de Octubre (i+12), 28040 Madrid, Spain
| |
Collapse
|
29
|
Chen C, Shi J, Wang D, Kong P, Wang Z, Liu Y. Antimicrobial peptides as promising antibiotic adjuvants to combat drug-resistant pathogens. Crit Rev Microbiol 2024; 50:267-284. [PMID: 36890767 DOI: 10.1080/1040841x.2023.2186215] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/19/2022] [Accepted: 10/26/2022] [Indexed: 03/10/2023]
Abstract
The widespread antimicrobial resistance (AMR) calls for the development of new antimicrobial strategies. Antibiotic adjuvant rescues antibiotic activity and increases the life span of the antibiotics, representing a more productive, timely, and cost-effective strategy in fighting drug-resistant pathogens. Antimicrobial peptides (AMPs) from synthetic and natural sources are considered new-generation antibacterial agents. Besides their direct antimicrobial activity, growing evidence shows that some AMPs effectively enhance the activity of conventional antibiotics. The combinations of AMPs and antibiotics display an improved therapeutic effect on antibiotic-resistant bacterial infections and minimize the emergence of resistance. In this review, we discuss the value of AMPs in the age of resistance, including modes of action, limiting evolutionary resistance, and their designing strategies. We summarise the recent advances in combining AMPs and antibiotics against antibiotic-resistant pathogens, as well as their synergistic mechanisms. Lastly, we highlight the challenges and opportunities associated with the use of AMPs as potential antibiotic adjuvants. This will shed new light on the deployment of synergistic combinations to address the AMR crisis.
Collapse
Affiliation(s)
- Chen Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jingru Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Dejuan Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Pan Kong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
30
|
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
|
31
|
Lyu Z, Ling Y, van Hoof A, Ling J. Inactivation of the ribosome assembly factor RimP causes streptomycin resistance and impairs motility in Salmonella. Antimicrob Agents Chemother 2024:e0000224. [PMID: 38629858 DOI: 10.1128/aac.00002-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/27/2024] [Indexed: 05/02/2024] Open
Abstract
The ribosome is the central hub for protein synthesis and the target of many antibiotics. Although the majority of ribosome-targeting antibiotics inhibit protein synthesis and are bacteriostatic, aminoglycosides promote protein mistranslation and are bactericidal. Understanding the resistance mechanisms of bacteria against aminoglycosides is not only vital for improving the efficacy of this critically important group of antibiotics but also crucial for studying the molecular basis of translational fidelity. In this work, we analyzed Salmonella mutants evolved in the presence of the aminoglycoside streptomycin (Str) and identified a novel gene rimP to be involved in Str resistance. RimP is a ribosome assembly factor critical for the maturation of the 30S small subunit that binds Str. Deficiency in RimP increases resistance against Str and facilitates the development of even higher resistance. Deleting rimP decreases mistranslation and cellular uptake of Str and further impairs flagellar motility. Our work thus highlights a previously unknown mechanism of aminoglycoside resistance via defective ribosome assembly.
Collapse
Affiliation(s)
- Zhihui Lyu
- Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, Maryland, USA
| | - Yunyi Ling
- Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, Maryland, USA
| | - Ambro van Hoof
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jiqiang Ling
- Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, Maryland, USA
| |
Collapse
|
32
|
Weaver DT, King ES, Maltas J, Scott JG. Reinforcement learning informs optimal treatment strategies to limit antibiotic resistance. Proc Natl Acad Sci U S A 2024; 121:e2303165121. [PMID: 38607932 PMCID: PMC11032439 DOI: 10.1073/pnas.2303165121] [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: 02/24/2023] [Accepted: 02/23/2024] [Indexed: 04/14/2024] Open
Abstract
Antimicrobial resistance was estimated to be associated with 4.95 million deaths worldwide in 2019. It is possible to frame the antimicrobial resistance problem as a feedback-control problem. If we could optimize this feedback-control problem and translate our findings to the clinic, we could slow, prevent, or reverse the development of high-level drug resistance. Prior work on this topic has relied on systems where the exact dynamics and parameters were known a priori. In this study, we extend this work using a reinforcement learning (RL) approach capable of learning effective drug cycling policies in a system defined by empirically measured fitness landscapes. Crucially, we show that it is possible to learn effective drug cycling policies despite the problems of noisy, limited, or delayed measurement. Given access to a panel of 15 [Formula: see text]-lactam antibiotics with which to treat the simulated Escherichia coli population, we demonstrate that RL agents outperform two naive treatment paradigms at minimizing the population fitness over time. We also show that RL agents approach the performance of the optimal drug cycling policy. Even when stochastic noise is introduced to the measurements of population fitness, we show that RL agents are capable of maintaining evolving populations at lower growth rates compared to controls. We further tested our approach in arbitrary fitness landscapes of up to 1,024 genotypes. We show that minimization of population fitness using drug cycles is not limited by increasing genome size. Our work represents a proof-of-concept for using AI to control complex evolutionary processes.
Collapse
Affiliation(s)
- Davis T. Weaver
- Case Western Reserve University School of Medicine, Cleveland, OH44106
- Translational Hematology Oncology Research, Cleveland Clinic, Cleveland, OH44106
| | - Eshan S. King
- Case Western Reserve University School of Medicine, Cleveland, OH44106
- Translational Hematology Oncology Research, Cleveland Clinic, Cleveland, OH44106
| | - Jeff Maltas
- Translational Hematology Oncology Research, Cleveland Clinic, Cleveland, OH44106
| | - Jacob G. Scott
- Case Western Reserve University School of Medicine, Cleveland, OH44106
- Translational Hematology Oncology Research, Cleveland Clinic, Cleveland, OH44106
- Department of Physics, Case Western Reserve University, Cleveland, OH44106
| |
Collapse
|
33
|
Chen J, Ren J, Wu Y, Hu N, Zhao F, Zhang L. Wet adhesive hydrogels based on niobium carbide for experimental research of oral mucosal impairment. RSC Adv 2024; 14:12935-12946. [PMID: 38650683 PMCID: PMC11033722 DOI: 10.1039/d4ra01352b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/13/2024] [Indexed: 04/25/2024] Open
Abstract
Oral mucosal impairment is a prevalent oral disease that frequently causes pain for patients. Conventional treatments have limited effectiveness and can cause adverse reactions. Furthermore, the moist and dynamic nature of the oral mucosal environment makes persistent adherence of conventional materials challenging, which can affect treatment efficacy. In this study, we investigated the potential of a NbC/TA-GelMA hydrogel system, where niobium carbide (NbC) and tannic acid (TA) were added to gelatin methacryloyl (GelMA), for repairing oral mucosal impairment. The wet adhesion properties of NbC/TA-GelMA hydrogels were confirmed by the inclusion of TA with a catechol-rich group. In addition, the photothermal effect of NbC/TA-GelMA hydrogel under near-infrared light, synergizing with TA, provided sustained antibacterial action. Furthermore, the NbC/TA-GelMA hydrogel effectively healed damaged oral mucosa of rats.
Collapse
Affiliation(s)
- Jiayuan Chen
- First Affiliated Hospital of Harbin Medical University, College of Stomatology, Harbin Medical University No. 143 Yiman Street, Nangang District Harbin 150001 China
| | - Junyu Ren
- Oral Implant Center, Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Harbin Heilongjiang China
| | - Yingjie Wu
- Key Laboratory of Microsystems and Microstructures Manufacturing (Ministry of Education), School of Medicine and Health, Harbin Institute of Technology No. 92 XiDaZhi Street Harbin 150001 China
| | - Narisu Hu
- Oral Implant Center, Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Harbin Heilongjiang China
| | - Fang Zhao
- Department of Dentistry, Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Harbin Heilongjiang China
| | - Lin Zhang
- First Affiliated Hospital of Harbin Medical University, College of Stomatology, Harbin Medical University No. 143 Yiman Street, Nangang District Harbin 150001 China
| |
Collapse
|
34
|
Eckartt KA, Delbeau M, Munsamy-Govender V, DeJesus MA, Azadian ZA, Reddy AK, Chandanani J, Poulton NC, Quiñones-Garcia S, Bosch B, Landick R, Campbell EA, Rock JM. Compensatory evolution in NusG improves fitness of drug-resistant M. tuberculosis. Nature 2024; 628:186-194. [PMID: 38509362 PMCID: PMC10990936 DOI: 10.1038/s41586-024-07206-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024]
Abstract
Drug-resistant bacteria are emerging as a global threat, despite frequently being less fit than their drug-susceptible ancestors1-8. Here we sought to define the mechanisms that drive or buffer the fitness cost of rifampicin resistance (RifR) in the bacterial pathogen Mycobacterium tuberculosis (Mtb). Rifampicin inhibits RNA polymerase (RNAP) and is a cornerstone of modern short-course tuberculosis therapy9,10. However, RifR Mtb accounts for one-quarter of all deaths due to drug-resistant bacteria11,12. We took a comparative functional genomics approach to define processes that are differentially vulnerable to CRISPR interference (CRISPRi) inhibition in RifR Mtb. Among other hits, we found that the universally conserved transcription factor NusG is crucial for the fitness of RifR Mtb. In contrast to its role in Escherichia coli, Mtb NusG has an essential RNAP pro-pausing function mediated by distinct contacts with RNAP and the DNA13. We find this pro-pausing NusG-RNAP interface to be under positive selection in clinical RifR Mtb isolates. Mutations in the NusG-RNAP interface reduce pro-pausing activity and increase fitness of RifR Mtb. Collectively, these results define excessive RNAP pausing as a molecular mechanism that drives the fitness cost of RifR in Mtb, identify a new mechanism of compensation to overcome this cost, suggest rational approaches to exacerbate the fitness cost, and, more broadly, could inform new therapeutic approaches to develop drug combinations to slow the evolution of RifR in Mtb.
Collapse
Affiliation(s)
- Kathryn A Eckartt
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, NY, USA
| | - Madeleine Delbeau
- Laboratory of Molecular Biophysics, The Rockefeller University, New York, NY, USA
| | | | - Michael A DeJesus
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, NY, USA
| | - Zachary A Azadian
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, NY, USA
| | - Abhijna K Reddy
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, NY, USA
| | - Joshua Chandanani
- Laboratory of Molecular Biophysics, The Rockefeller University, New York, NY, USA
| | - Nicholas C Poulton
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, NY, USA
| | | | - Barbara Bosch
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, NY, USA
| | - Robert Landick
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth A Campbell
- Laboratory of Molecular Biophysics, The Rockefeller University, New York, NY, USA.
| | - Jeremy M Rock
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
35
|
Exciting times for evolutionary biology. Nat Ecol Evol 2024; 8:593-594. [PMID: 38605230 DOI: 10.1038/s41559-024-02402-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
|
36
|
Jiang J, Lv X, Cheng H, Yang D, Xu W, Hu Y, Song Y, Zeng G. Type I photodynamic antimicrobial therapy: Principles, progress, and future perspectives. Acta Biomater 2024; 177:1-19. [PMID: 38336269 DOI: 10.1016/j.actbio.2024.02.005] [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: 12/21/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
The emergence of drug-resistant bacteria has significantly diminished the efficacy of existing antibiotics in the treatment of bacterial infections. Consequently, the need for finding a strategy capable of effectively combating bacterial infections has become increasingly urgent. Photodynamic therapy (PDT) is considered one of the most promising emerging antibacterial strategies due to its non-invasiveness, low adverse effect, and the fact that it does not lead to the development of drug resistance. However, bacteria at the infection sites often exist in the form of biofilm instead of the planktonic form, resulting in a hypoxic microenvironment. This phenomenon compromises the treatment outcome of oxygen-dependent type-II PDT. Compared to type-II PDT, type-I PDT is not constrained by the oxygen concentration in the infected tissues. Therefore, in the treatment of bacterial infections, type-I PDT exhibits significant advantages over type-II PDT. In this review, we first introduce the fundamental principles of type-I PDT in details, including its physicochemical properties and how it generates reactive oxygen species (ROS). Next, we explore several specific antimicrobial mechanisms utilized by type-I PDT and summarize the recent applications of type-I PDT in antimicrobial treatment. Finally, the limitations and future development directions of type-I photosensitizers are discussed. STATEMENT OF SIGNIFICANCE: The misuse and overuse of antibiotics have accelerated the development of bacterial resistance. To achieve the effective eradication of resistant bacteria, pathfinders have devised various treatment strategies. Among these strategies, type I photodynamic therapy has garnered considerable attention owing to its non-oxygen dependence. The utilization of non-oxygen-dependent photodynamic therapy not only enables the effective elimination of drug-resistant bacteria but also facilitates the successful eradication of hypoxic biofilms, which exhibits promising prospects for treating biofilm-associated infections. Based on the current research status, we anticipate that the novel type I photodynamic therapy agent can surmount the biofilm barrier, enabling efficient treatment of hypoxic biofilm infections.
Collapse
Affiliation(s)
- Jingai Jiang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Xinyi Lv
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Huijuan Cheng
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China.
| | - Wenjia Xu
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing 211200, China.
| | - Yanling Hu
- Nanjing Polytechnic Institute, Nanjing 210048, China.
| | - Yanni Song
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Guisheng Zeng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648.
| |
Collapse
|
37
|
Wang M, Zhang J, Wei J, Jiang L, Jiang L, Sun Y, Zeng Z, Wang Z. Phage-inspired strategies to combat antibacterial resistance. Crit Rev Microbiol 2024; 50:196-211. [PMID: 38400715 DOI: 10.1080/1040841x.2023.2181056] [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: 12/09/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023]
Abstract
Antimicrobial resistance (AMR) in clinically priority pathogensis now a major threat to public health worldwide. Phages are bacterial parasites that efficiently infect or kill specific strains and represent the most abundant biological entities on earth, showing great attraction as potential antibacterial therapeutics in combating AMR. This review provides a summary of phage-inspired strategies to combat AMR. We firstly cover the phage diversity, and then explain the biological principles of phage therapy that support the use of phages in the post-antimicrobial era. Furthermore, we state the versatility methods of phage therapy both from direct access as well as collateral access. Among the direct access approaches, we discuss the use of phage cocktail therapy, phage-encoded endolysins and the bioengineering for function improvement of used phages or endolysins. On the other hand, we introduce the collateral access, including the phages antimicrobial immunity combined therapy and phage-based novel antibacterial mimic molecules. Nowadays, more and more talented and enthusiastic scientist, doctors, pharmacists, media, authorities, and industry are promoting the progress of phage therapy, and proposed more phages-inspired strategy to make them more tractable to combat AMR and benefit more people, more animal and diverse environment in "one health" framework.
Collapse
Affiliation(s)
- Mianzhi Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Junxuan Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Jingyi Wei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Lei Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Li Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yongxue Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhenling Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, China
| |
Collapse
|
38
|
Zhou Y, Xu L, Sun X, Zhan W, Liang G. In situ peptide assemblies for bacterial infection imaging and treatment. NANOSCALE 2024; 16:3211-3225. [PMID: 38288668 DOI: 10.1039/d3nr05557d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Bacterial infections, especially antibiotic-resistant ones, remain a major threat to human health. Advances in nanotechnology have led to the development of numerous antimicrobial nanomaterials. Among them, in situ peptide assemblies, formed by biomarker-triggered self-assembly of peptide-based building blocks, have received increasing attention due to their unique merits of good spatiotemporal controllability and excellent disease accumulation and retention. In recent years, a variety of "turn on" imaging probes and activatable antibacterial agents based on in situ peptide assemblies have been developed, providing promising alternatives for the treatment and diagnosis of bacterial infections. In this review, we introduce representative design strategies for in situ peptide assemblies and highlight the bacterial infection imaging and treatment applications of these supramolecular materials. Besides, current challenges in this field are proposed.
Collapse
Affiliation(s)
- Yanyan Zhou
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing 210093, China
- Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Lingling Xu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, Jiangsu 210096, China.
| | - Xianbao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, Jiangsu 210096, China.
| | - Wenjun Zhan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, Jiangsu 210096, China.
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, Jiangsu 210096, China.
| |
Collapse
|
39
|
Guo W, Yang Z, Wang K, Li W, Zhao Y, Yang Y, Chang W, Gong Z, Liu Z, Chen Y, Li Q. Discovery of Unique Bis-Substituted Aromatic Amide Derivatives as Novel Highly Potent Antibiotics for Combating Methicillin-Resistant Staphylococcus aureus (MRSA). J Med Chem 2024; 67:2129-2151. [PMID: 38289145 DOI: 10.1021/acs.jmedchem.3c02064] [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: 02/09/2024]
Abstract
Due to the increasing antibiotic resistance, developing novel antimicrobials to fight infections caused by resistant bacteria is imperative. Herein, a series of novel bis-substituted aromatic amides were designed and synthesized through modifying the hit compound 1, and their antimicrobial activities were evaluated. Among them, compound 4t, as the most potent lead, exhibited excellent antimicrobial activities against Gram-positive bacteria, including clinical methicillin-resistant Staphylococcus aureus (MRSA) isolates, while keeping weak hemolytic and mammalian cytotoxic activities. Furthermore, compound 4t displayed rapid bactericidal capabilities, low tendency to produce resistance, and favorable capacities to destroy bacterial biofilms. Further explorations indicated that compound 4t induces bacterial death by binding to cardiolipin (CL) on the bacterial membrane, disrupting the cell membrane, and facilitating the accumulation of reactive oxygen species (ROS). Additionally, compound 4t showed remarkable anti-MRSA activity in vivo, demonstrating compound 4t could be developed as a potential candidate to combat MRSA infections.
Collapse
Affiliation(s)
- Weikai Guo
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Zhengfan Yang
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Kexiao Wang
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Wenyu Li
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Yanyang Zhao
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Yuqing Yang
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Wenjing Chang
- Henan Provincial Center for Disease Control and Prevention, Zhengzhou 450003, China
| | - Zhen Gong
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhou Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yihua Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qiming Li
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| |
Collapse
|
40
|
Allemailem KS. Recent Advances in Understanding the Molecular Mechanisms of Multidrug Resistance and Novel Approaches of CRISPR/Cas9-Based Genome-Editing to Combat This Health Emergency. Int J Nanomedicine 2024; 19:1125-1143. [PMID: 38344439 PMCID: PMC10859101 DOI: 10.2147/ijn.s453566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/26/2024] [Indexed: 02/15/2024] Open
Abstract
The rapid spread of multidrug resistance (MDR), due to abusive use of antibiotics has led to global health emergency, causing substantial morbidity and mortality. Bacteria attain MDR by different means such as antibiotic modification/degradation, target protection/modification/bypass, and enhanced efflux mechanisms. The classical approaches of counteracting MDR bacteria are expensive and time-consuming, thus, it is highly significant to understand the molecular mechanisms of this resistance to curb the problem from core level. The revolutionary approach of clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated sequence 9 (CRISPR/Cas9), considered as a next-generation genome-editing tool presents an innovative opportunity to precisely target and edit bacterial genome to alter their MDR strategy. Different bacteria possessing antibiotic resistance genes such as mecA, ermB, ramR, tetA, mqrB and blaKPC that have been targeted by CRISPR/Cas9 to re-sensitize these pathogens against antibiotics, such as methicillin, erythromycin, tigecycline, colistin and carbapenem, respectively. The CRISPR/Cas9 from S. pyogenes is the most widely studied genome-editing tool, consisting of a Cas9 DNA endonuclease associated with tracrRNA and crRNA, which can be systematically coupled as sgRNA. The targeting strategies of CRISPR/Cas9 to bacterial cells is mediated through phage, plasmids, vesicles and nanoparticles. However, the targeting approaches of this genome-editing tool to specific bacteria is a challenging task and still remains at a very preliminary stage due to numerous obstacles awaiting to be solved. This review elaborates some recent updates about the molecular mechanisms of antibiotic resistance and the innovative role of CRISPR/Cas9 system in modulating these resistance mechanisms. Furthermore, the delivery approaches of this genome-editing system in bacterial cells are discussed. In addition, some challenges and future prospects are also described.
Collapse
Affiliation(s)
- Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah51452, Saudi Arabia
| |
Collapse
|
41
|
Lyu Z, Ling Y, van Hoof A, Ling J. Deficiency in ribosome biogenesis causes streptomycin resistance and impairs motility in Salmonella. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574728. [PMID: 38260697 PMCID: PMC10802465 DOI: 10.1101/2024.01.08.574728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The ribosome is the central hub for protein synthesis and the target of many antibiotics. Whereas the majority of ribosome-targeting antibiotics inhibit protein synthesis and are bacteriostatic, aminoglycosides promote protein mistranslation and are bactericidal. Understanding the resistance mechanisms of bacteria against aminoglycosides is not only vital for improving the efficacy of this critically important group of antibiotics but also crucial for studying the molecular basis of translational fidelity. In this work, we analyzed Salmonella mutants evolved in the presence of the aminoglycoside streptomycin (Str) and identified a novel gene rimP to be involved in Str resistance. RimP is a ribosome assembly factor critical for the maturation of the 30S small subunit that binds Str. Deficiency in RimP increases resistance against Str and facilitates the development of even higher resistance. Deleting rimP decreases mistranslation and cellular uptake of Str, and further impairs flagellar motility. Our work thus highlights a previously unknown mechanism of aminoglycoside resistance via defective ribosome assembly.
Collapse
Affiliation(s)
- Zhihui Lyu
- Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, MD 20742, USA
| | - Yunyi Ling
- Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, MD 20742, USA
| | - Ambro van Hoof
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jiqiang Ling
- Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
42
|
Huang Q, Yang Z, Tao X, Ma C, Cao P, Wei P, Jiang C, Ren H, Li X. Sprayable chitosan nanogel with nitric oxide to accelerate diabetic wound healing through bacteria inhibition, biofilm eradication and macrophage polarization. Int J Biol Macromol 2024; 254:127806. [PMID: 37918593 DOI: 10.1016/j.ijbiomac.2023.127806] [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: 09/19/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/04/2023]
Abstract
Bacterial infection and chronic inflammation are two major risks in diabetic wound healing, which increase patient mortality. In this study, a multifunctional sprayable nanogel (Ag-G@CS) based on chitosan has been developed to synergistically inhibit bacterial infection, eradicate biofilm, and relieve inflammation of diabetic wounds. The nanogel is successfully crafted by encapsulating with a nitric oxide (NO) donor and performing in-situ reduction of silver nanoparticles (Ag). The released NO enhances the antibacterial efficacy of Ag, nearly achieving complete eradication of biofilms in vitro. Upon application on both normal or diabetic chronic wounds, the combination effects of released NO and Ag offer a notable antibacterial effect. Furthermore, after bacteria inhibition and biofilm eradication, the NO released by the nanogel orchestrates a transformation of M1 macrophages into M2 macrophages, significantly reducing tumor necrosis factor α (TNF-α) release and relieving inflammation. Remarkably, the released NO also promotes M2a to M2c macrophages, thereby facilitating tissue remodeling in chronic wounds. More importantly, it upregulates the expression of vascular endothelial growth factor (VEGF), further accelerating the wound healing process. Collectively, the formed sprayable nanogel exhibits excellent inhibition of bacterial infections and biofilms, and promotes chronic wound healing via inflammation resolution, which has excellent potential for clinical use in the future.
Collapse
Affiliation(s)
- Qinqin Huang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Zheng Yang
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China
| | - Xinyue Tao
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China
| | - Chenyu Ma
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China
| | - Peiyao Cao
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China
| | - Ping Wei
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Chenxiao Jiang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Hao Ren
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China.
| | - Xueming Li
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
43
|
Kaushik A, Kaushik M, Kaur G, Gupta V. Perspective of Secondary Metabolites in Respect of Multidrug Resistance (MDR): A Review. Infect Disord Drug Targets 2024; 24:40-52. [PMID: 38031773 DOI: 10.2174/0118715265210606231113105225] [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: 05/02/2023] [Revised: 10/07/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023]
Abstract
Aberrant and haphazard use of antibiotics has created the development of antimicrobial resistance which is a bizarre challenge for human civilization. This emerging crisis of antibiotic resistance for microbial pathogens is alarming all the nations posing a global threat to human health. It is difficult to treat bacterial infections as they develop resistance to all antimicrobial resistance. Currently used antibacterial agents inhibit a variety of essential metabolic pathways in bacteria, including macro-molecular synthesis (MMS) pathways (e.g. protein, DNA, RNA, cell wall) most often by targeting a specific enzyme or subcellular component e.g. DNA gyrase, RNA polymerase, ribosomes, transpeptidase. Despite the availability of diverse synthetic molecules, there are still many complications in managing progressive and severe antimicrobial resistance. Currently not even a single antimicrobial agent is available for which the microbes do not show resistance. Thus, the lack of efficient drug molecules for combating microbial resistance requires continuous research efforts to overcome the problem of multidrug-resistant bacteria. The phytochemicals from various plants have the potential to combat the microbial resistance produced by bacteria, fungi, protozoa and viruses without producing any side effects. This review is a concerted effort to identify some of the major active phytoconstituents from various medicinal plants which might have the potential to be used as an alternative and effective strategy to fight against microbial resistance and can promote research for the treatment of MDR.
Collapse
Affiliation(s)
- Aditi Kaushik
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar, Punjab, India
| | - Manish Kaushik
- KC Group of Institutions, UNA, H.P, MMDU, Mullana, Ambala, Haryana, India
| | - Gagandeep Kaur
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Vrinda Gupta
- Chitkara Group of Institutions, Chitkara University, Chandigarh, India
| |
Collapse
|
44
|
Liu F, Greenwood AI, Xiong Y, Miceli RT, Fu R, Anderson KW, McCallum SA, Mihailescu M, Gross R, Cotten ML. Host Defense Peptide Piscidin and Yeast-Derived Glycolipid Exhibit Synergistic Antimicrobial Action through Concerted Interactions with Membranes. JACS AU 2023; 3:3345-3365. [PMID: 38155643 PMCID: PMC10751773 DOI: 10.1021/jacsau.3c00506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/25/2023] [Accepted: 09/29/2023] [Indexed: 12/30/2023]
Abstract
Developing new antimicrobials as alternatives to conventional antibiotics has become an urgent race to eradicate drug-resistant bacteria and to save human lives. Conventionally, antimicrobial molecules are studied independently even though they can be cosecreted in vivo. In this research, we investigate two classes of naturally derived antimicrobials: sophorolipid (SL) esters as modified yeast-derived glycolipid biosurfactants that feature high biocompatibility and low production cost; piscidins, which are host defense peptides (HDPs) from fish. While HDPs such as piscidins target the membrane of pathogens, and thus result in low incidence of resistance, SLs are not well understood on a mechanistic level. Here, we demonstrate that combining SL-hexyl ester (SL-HE) with subinhibitory concentration of piscidins 1 (P1) and 3 (P3) stimulates strong antimicrobial synergy, potentiating a promising therapeutic window. Permeabilization assays and biophysical studies employing circular dichroism, NMR, mass spectrometry, and X-ray diffraction are performed to investigate the mechanism underlying this powerful synergy. We reveal four key mechanistic features underlying the synergistic action: (1) P1/3 binds to SL-HE aggregates, becoming α-helical; (2) piscidin-glycolipid assemblies synergistically accumulate on membranes; (3) SL-HE used alone or bound to P1/3 associates with phospholipid bilayers where it induces defects; (4) piscidin-glycolipid complexes disrupt the bilayer structure more dramatically and differently than either compound alone, with phase separation occurring when both agents are present. Overall, dramatic enhancement in antimicrobial activity is associated with the use of two membrane-active agents, with the glycolipid playing the roles of prefolding the peptide, coordinating the delivery of both agents to bacterial surfaces, recruiting the peptide to the pathogenic membranes, and supporting membrane disruption by the peptide. Given that SLs are ubiquitously and safely used in consumer products, the SL/peptide formulation engineered and mechanistically characterized in this study could represent fertile ground to develop novel synergistic agents against drug-resistant bacteria.
Collapse
Affiliation(s)
- Fei Liu
- Department
of Chemistry, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Alexander I. Greenwood
- Department
of Applied Science, William & Mary, Williamsburg, Virginia 23185, United States
| | - Yawei Xiong
- Department
of Applied Science, William & Mary, Williamsburg, Virginia 23185, United States
| | - Rebecca T. Miceli
- Department
of Chemistry, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Center
for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Riqiang Fu
- Center
of Interdisciplinary Magnetic Resonance, National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - Kyle W. Anderson
- National
Institute of Standards and Technology, Rockville, Maryland 20850, United States
| | - Scott A. McCallum
- Center
for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Mihaela Mihailescu
- Institute
for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland 20850, United States
| | - Richard Gross
- Department
of Chemistry, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Center
for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Myriam L. Cotten
- Department
of Applied Science, William & Mary, Williamsburg, Virginia 23185, United States
| |
Collapse
|
45
|
Zhukovets AA, Chernyshov VV, Al’mukhametov AZ, Seregina TA, Revtovich SV, Kasatkina MA, Isakova YE, Kulikova VV, Morozova EA, Cherkasova AI, Mannanov TA, Anashkina AA, Solyev PN, Mitkevich VA, Ivanov RA. Novel Hydroxamic Acids Containing Aryl-Substituted 1,2,4- or 1,3,4-Oxadiazole Backbones and an Investigation of Their Antibiotic Potentiation Activity. Int J Mol Sci 2023; 25:96. [PMID: 38203266 PMCID: PMC10779255 DOI: 10.3390/ijms25010096] [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: 11/02/2023] [Revised: 12/05/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC) is a zinc amidase that catalyzes the second step of the biosynthesis of lipid A, which is an outer membrane essential structural component of Gram-negative bacteria. Inhibitors of this enzyme can be attributed to two main categories, non-hydroxamate and hydroxamate inhibitors, with the latter being the most effective given the chelation of Zn2+ in the active site. Compounds containing diacetylene or acetylene tails and the sulfonic head, as well as oxazoline derivatives of hydroxamic acids, are among the LpxC inhibitors with the most profound antibacterial activity. The present article describes the synthesis of novel functional derivatives of hydroxamic acids-bioisosteric to oxazoline inhibitors-containing 1,2,4- and 1,3,4-oxadiazole cores and studies of their cytotoxicity, antibacterial activity, and antibiotic potentiation. Some of the hydroxamic acids we obtained (9c, 9d, 23a, 23c, 30b, 36) showed significant potentiation in nalidixic acid, rifampicin, and kanamycin against the growth of laboratory-strain Escherichia coli MG1655. Two lead compounds (9c, 9d) significantly reduced Pseudomonas aeruginosa ATCC 27853 growth in the presence of nalidixic acid and rifampicin.
Collapse
Affiliation(s)
- Anastasia A. Zhukovets
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sochi, Russia; (A.A.Z.); (A.Z.A.); (M.A.K.); (Y.E.I.); (A.I.C.); (T.A.M.); (R.A.I.)
| | - Vladimir V. Chernyshov
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sochi, Russia; (A.A.Z.); (A.Z.A.); (M.A.K.); (Y.E.I.); (A.I.C.); (T.A.M.); (R.A.I.)
| | - Aidar Z. Al’mukhametov
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sochi, Russia; (A.A.Z.); (A.Z.A.); (M.A.K.); (Y.E.I.); (A.I.C.); (T.A.M.); (R.A.I.)
| | - Tatiana A. Seregina
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., 119991 Moscow, Russia; (T.A.S.); (S.V.R.); (V.V.K.); (E.A.M.); (A.A.A.); (P.N.S.); (V.A.M.)
| | - Svetlana V. Revtovich
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., 119991 Moscow, Russia; (T.A.S.); (S.V.R.); (V.V.K.); (E.A.M.); (A.A.A.); (P.N.S.); (V.A.M.)
| | - Mariia A. Kasatkina
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sochi, Russia; (A.A.Z.); (A.Z.A.); (M.A.K.); (Y.E.I.); (A.I.C.); (T.A.M.); (R.A.I.)
| | - Yulia E. Isakova
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sochi, Russia; (A.A.Z.); (A.Z.A.); (M.A.K.); (Y.E.I.); (A.I.C.); (T.A.M.); (R.A.I.)
| | - Vitalia V. Kulikova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., 119991 Moscow, Russia; (T.A.S.); (S.V.R.); (V.V.K.); (E.A.M.); (A.A.A.); (P.N.S.); (V.A.M.)
| | - Elena A. Morozova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., 119991 Moscow, Russia; (T.A.S.); (S.V.R.); (V.V.K.); (E.A.M.); (A.A.A.); (P.N.S.); (V.A.M.)
| | - Anastasia I. Cherkasova
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sochi, Russia; (A.A.Z.); (A.Z.A.); (M.A.K.); (Y.E.I.); (A.I.C.); (T.A.M.); (R.A.I.)
| | - Timur A. Mannanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sochi, Russia; (A.A.Z.); (A.Z.A.); (M.A.K.); (Y.E.I.); (A.I.C.); (T.A.M.); (R.A.I.)
| | - Anastasia A. Anashkina
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., 119991 Moscow, Russia; (T.A.S.); (S.V.R.); (V.V.K.); (E.A.M.); (A.A.A.); (P.N.S.); (V.A.M.)
| | - Pavel N. Solyev
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., 119991 Moscow, Russia; (T.A.S.); (S.V.R.); (V.V.K.); (E.A.M.); (A.A.A.); (P.N.S.); (V.A.M.)
| | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., 119991 Moscow, Russia; (T.A.S.); (S.V.R.); (V.V.K.); (E.A.M.); (A.A.A.); (P.N.S.); (V.A.M.)
| | - Roman A. Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sochi, Russia; (A.A.Z.); (A.Z.A.); (M.A.K.); (Y.E.I.); (A.I.C.); (T.A.M.); (R.A.I.)
| |
Collapse
|
46
|
Lässig M, Mustonen V, Nourmohammad A. Steering and controlling evolution - from bioengineering to fighting pathogens. Nat Rev Genet 2023; 24:851-867. [PMID: 37400577 PMCID: PMC11137064 DOI: 10.1038/s41576-023-00623-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 07/05/2023]
Abstract
Control interventions steer the evolution of molecules, viruses, microorganisms or other cells towards a desired outcome. Applications range from engineering biomolecules and synthetic organisms to drug, therapy and vaccine design against pathogens and cancer. In all these instances, a control system alters the eco-evolutionary trajectory of a target system, inducing new functions or suppressing escape evolution. Here, we synthesize the objectives, mechanisms and dynamics of eco-evolutionary control in different biological systems. We discuss how the control system learns and processes information about the target system by sensing or measuring, through adaptive evolution or computational prediction of future trajectories. This information flow distinguishes pre-emptive control strategies by humans from feedback control in biotic systems. We establish a cost-benefit calculus to gauge and optimize control protocols, highlighting the fundamental link between predictability of evolution and efficacy of pre-emptive control.
Collapse
Affiliation(s)
- Michael Lässig
- Institute for Biological Physics, University of Cologne, Cologne, Germany.
| | - Ville Mustonen
- Organismal and Evolutionary Biology Research Programme, Department of Computer Science, Institute of Biotechnology, University of Helsinki, Helsinki, Finland.
| | - Armita Nourmohammad
- Department of Physics, University of Washington, Seattle, WA, USA.
- Department of Applied Mathematics, University of Washington, Seattle, WA, USA.
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA.
- Herbold Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
47
|
Sun C, Liu W, Wang L, Meng R, Deng J, Qing R, Wang B, Hao S. Photopolymerized keratin-PGLa hydrogels for antibiotic resistance reversal and enhancement of infectious wound healing. Mater Today Bio 2023; 23:100807. [PMID: 37810750 PMCID: PMC10558788 DOI: 10.1016/j.mtbio.2023.100807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/04/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023] Open
Abstract
Infectious wounds have become serious challenges for both treatment and management in clinical practice, so development of new antibiotics has been considered an increasingly difficult task. Here, we report the design and synthesis of keratin 31 (K31)-peptide glycine-leucine-amide (PGLa) photopolymerized hydrogels to rescue the antibiotic activity of antibiotics for infectious wound healing promotion. K31-PGLa displayed an outstanding synergistic effect with commercial antibiotics against drug-resistant bacteria by down-regulating the synthesis genes of efflux pump. Furthermore, the photopolymerized K31-PGLa/PEGDA hydrogels effectively suppressed drug-resistant bacteria growth and enhanced skin wound closure in murine. This study provided a promising alternative strategy for infectious wound treatment.
Collapse
Affiliation(s)
- Changfa Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Wenjie Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Lili Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Run Meng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Jia Deng
- College of Environment and Resources, Chongqing Technology and Business University, Chongqing, 400067, China
| | - Rui Qing
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| |
Collapse
|
48
|
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
|
49
|
Weaver DT, King ES, Maltas J, Scott JG. Reinforcement Learning informs optimal treatment strategies to limit antibiotic resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523765. [PMID: 36711676 PMCID: PMC9882109 DOI: 10.1101/2023.01.12.523765] [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: 01/15/2023]
Abstract
Antimicrobial resistance was estimated to be associated with 4.95 million deaths worldwide in 2019. It is possible to frame the antimicrobial resistance problem as a feedback-control problem. If we could optimize this feedback-control problem and translate our findings to the clinic, we could slow, prevent or reverse the development of high-level drug resistance. Prior work on this topic has relied on systems where the exact dynamics and parameters were known a priori. In this study, we extend this work using a reinforcement learning (RL) approach capable of learning effective drug cycling policies in a system defined by empirically measured fitness landscapes. Crucially, we show that is possible to learn effective drug cycling policies despite the problems of noisy, limited, or delayed measurement. Given access to a panel of 15 β-lactam antibiotics with which to treat the simulated E. coli population, we demonstrate that RL agents outperform two naive treatment paradigms at minimizing the population fitness over time. We also show that RL agents approach the performance of the optimal drug cycling policy. Even when stochastic noise is introduced to the measurements of population fitness, we show that RL agents are capable of maintaining evolving populations at lower growth rates compared to controls. We further tested our approach in arbitrary fitness landscapes of up to 1024 genotypes. We show that minimization of population fitness using drug cycles is not limited by increasing genome size. Our work represents a proof-of-concept for using AI to control complex evolutionary processes.
Collapse
Affiliation(s)
- Davis T. Weaver
- Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Translational Hematology Oncology Research, Cleveland Clinic, Cleveland OH, 44106, USA
| | - Eshan S. King
- Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Translational Hematology Oncology Research, Cleveland Clinic, Cleveland OH, 44106, USA
| | - Jeff Maltas
- Translational Hematology Oncology Research, Cleveland Clinic, Cleveland OH, 44106, USA
| | - Jacob G. Scott
- Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Translational Hematology Oncology Research, Cleveland Clinic, Cleveland OH, 44106, USA
- Department of Physics, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
50
|
Chowdhury F, Findlay BL. Fitness Costs of Antibiotic Resistance Impede the Evolution of Resistance to Other Antibiotics. ACS Infect Dis 2023; 9:1834-1845. [PMID: 37726252 PMCID: PMC10581211 DOI: 10.1021/acsinfecdis.3c00156] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Indexed: 09/21/2023]
Abstract
Antibiotic resistance is a major threat to global health, claiming the lives of millions every year. With a nearly dry antibiotic development pipeline, novel strategies are urgently needed to combat resistant pathogens. One emerging strategy is the use of sequential antibiotic therapy, postulated to reduce the rate at which antibiotic resistance evolves. Here, we use the soft agar gradient evolution (SAGE) system to carry out high-throughput in vitro bacterial evolution against antibiotic pressure. We find that evolution of resistance to the antibiotic chloramphenicol (CHL) severely affects bacterial fitness, slowing the rate at which resistance to the antibiotics nitrofurantoin and streptomycin emerges. In vitro acquisition of compensatory mutations in the CHL-resistant cells markedly improves fitness and nitrofurantoin adaptation rates but fails to restore rates to wild-type levels against streptomycin. Genome sequencing reveals distinct evolutionary paths to resistance in fitness-impaired populations, suggesting resistance trade-offs in favor of mitigation of fitness costs. We show that the speed of bacterial fronts in SAGE plates is a reliable indicator of adaptation rates and evolutionary trajectories to resistance. Identification of antibiotics whose mutational resistance mechanisms confer stable impairments may help clinicians prescribe sequential antibiotic therapies that are less prone to resistance evolution.
Collapse
Affiliation(s)
- Farhan
R. Chowdhury
- Department
of Biology, Concordia University, Montréal, Québec H4B 1R6, Canada
| | - Brandon L. Findlay
- Department
of Biology, Concordia University, Montréal, Québec H4B 1R6, Canada
- Department
of Chemistry and Biochemistry, Concordia
University, Montréal, Québec H4B 1R6, Canada
| |
Collapse
|