1
|
Hall KM, Williams LG, Smith RD, Kuang EA, Ernst RK, Bojanowski CM, Wimley WC, Morici LA, Pursell ZF. Mutational signature analysis predicts bacterial hypermutation and multidrug resistance. Nat Commun 2025; 16:19. [PMID: 39746975 PMCID: PMC11695600 DOI: 10.1038/s41467-024-55206-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Bacteria of clinical importance, such as Pseudomonas aeruginosa, can become hypermutators upon loss of DNA mismatch repair (MMR) and are clinically correlated with high rates of multidrug resistance (MDR). Here, we demonstrate that hypermutated MMR-deficient P. aeruginosa has a unique mutational signature and rapidly acquires MDR upon repeated exposure to first-line or last-resort antibiotics. MDR acquisition was irrespective of drug class and instead arose through common resistance mechanisms shared between the initial and secondary drugs. Rational combinations of drugs having distinct resistance mechanisms prevented MDR acquisition in hypermutated MMR-deficient P. aeruginosa. Mutational signature analysis of P. aeruginosa across different human disease contexts identified appreciable quantities of MMR-deficient clinical isolates that were already MDR or prone to future MDR acquisition. Mutational signature analysis of patient samples is a promising diagnostic tool that may predict MDR and guide precision-based medical care.
Collapse
Affiliation(s)
- Kalen M Hall
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
- Informuta, Inc., San Diego, CA, USA
| | - Leonard G Williams
- Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
- Informuta, Inc., San Diego, CA, USA
- Bioinnovation Program, Tulane University, New Orleans, LA, USA
| | - Richard D Smith
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Erin A Kuang
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Robert K Ernst
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | | | - William C Wimley
- Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Lisa A Morici
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Zachary F Pursell
- Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA.
- Tulane Cancer Center, School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
2
|
Martian PC, Tertis M, Leonte D, Hadade N, Cristea C, Crisan O. Cyclic peptides: A powerful instrument for advancing biomedical nanotechnologies and drug development. J Pharm Biomed Anal 2025; 252:116488. [PMID: 39388867 DOI: 10.1016/j.jpba.2024.116488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/05/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
Cyclic peptides have emerged as an essential tool in the advancement of biomedical nanotechnologies, offering unique structural and functional advantages over linear peptides. This review article aims to highlight the roles of cyclic peptides in the development of biomedical fields, with a particular focus on their application in drug discovery and delivery. Cyclic peptides exhibit exceptional stability, bioavailability, and binding specificity, making them ideal candidates for therapeutic and diagnostic applications. We explore the synthesis and design strategies that enable the precise control of cyclic peptide structures, leading to enhanced performance in targeting specific cellular pathways. The article also highlights recent breakthroughs in the use of cyclic peptides for creating innovative drug delivery systems, including nanoparticle conjugates and peptide-drug conjugates, which have shown promise in improving the efficacy and safety profiles of existing traditional treatments. The integration of cyclic peptides into nanotechnological frameworks holds significant promise for addressing unmet medical needs, providing a foundation for future advancements in personalized medicine and targeted drug delivery.
Collapse
Affiliation(s)
- Paul Cristian Martian
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 4 Pasteur Street, Cluj-Napoca 400021, Romania
| | - Mihaela Tertis
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 4 Pasteur Street, Cluj-Napoca 400021, Romania
| | - Denisa Leonte
- Department of Organic Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 28 Victor Babes Street, Cluj-Napoca 400023, Romania
| | - Niculina Hadade
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babes Bolyai University, 11 Arany Janos Street, Cluj-Napoca 400028, Romania
| | - Cecilia Cristea
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 4 Pasteur Street, Cluj-Napoca 400021, Romania.
| | - Ovidiu Crisan
- Department of Organic Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 28 Victor Babes Street, Cluj-Napoca 400023, Romania
| |
Collapse
|
3
|
Ma B, Zhou S, Fang C, Wang M, Xue X, Xie J, Liu J, Hou Z. Membrane anchoring of New Delhi metallo-β-lactamase-1 alters the fitness of Escherichia coli and increases its susceptibility to colistin by inducing outer membrane destabilization. FEBS J 2024. [PMID: 39658304 DOI: 10.1111/febs.17351] [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: 04/01/2024] [Revised: 08/02/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
New Delhi metallo-β-lactamase-1 (NDM-1)-producing bacteria are resistant to nearly all available β-lactam antibiotics and have become a public health threat. Antibiotic resistance often carries fitness costs, which typically manifest as a reduced bacterial growth rate. Here, we investigated the mechanism of fitness cost in NDM-1-producing bacteria. Our findings revealed that strains expressing blaNDM-1 exhibited a significant growth defect under high osmotic stress. This fitness cost was attributed to the anchoring of NDM-1 to the bacterial outer membrane via its leader peptide, which destabilized the outer membrane. Replacing the membrane-anchoring residue Cys26 in the leader peptide with alanine not only restored outer membrane stability but also ameliorated the bacterial fitness cost. Furthermore, the anchoring of NDM-1 to the membrane increased bacterial susceptibility to the membrane-disrupting antibiotic colistin, both in vitro and in vivo, as confirmed in engineered and clinically isolated strains. In conclusion, membrane anchoring of NDM-1 increased the permeability of the bacterial outer membrane, thereby reducing the fitness of NDM-1-producing bacteria and enhancing their susceptibility to colistin. These results not only elucidate the mechanism of fitness cost associated with NDM-1 but also provide new insights into the rational use of colistin to combat infections caused by NDM-1-producing bacteria.
Collapse
Affiliation(s)
- Bo Ma
- State Key Laboratory of Toxicology and Medical Countermeasures, Academy of Military Medical Sciences, Beijing, China
- Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, China
| | - Shan Zhou
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Chao Fang
- Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, China
| | - Mingzhi Wang
- Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, China
| | - Xiaoyan Xue
- Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, China
| | - Jianwei Xie
- State Key Laboratory of Toxicology and Medical Countermeasures, Academy of Military Medical Sciences, Beijing, China
| | - Jiayun Liu
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Zheng Hou
- Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
4
|
Suprenant MP, Ching C, Gross N, Sutradhar I, Anderson JE, El Sherif N, Zaman MH. The impact of zinc pre-exposure on ciprofloxacin resistance development in E. coli. Front Microbiol 2024; 15:1491532. [PMID: 39717277 PMCID: PMC11663908 DOI: 10.3389/fmicb.2024.1491532] [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: 09/05/2024] [Accepted: 11/01/2024] [Indexed: 12/25/2024] Open
Abstract
Introduction Antimicrobial resistance (AMR) is a global health crisis that is predicted to worsen in the coming years. While improper antibiotic usage is an established driver, less is known about the impact of other endogenous and exogeneous environmental factors, such as metals, on AMR. One metal of interest is zinc as it is often used as a supplement for diarrhea treatment prior to antibiotics. Materials and methods Here, we probed the impact of zinc on ciprofloxacin resistance in E. coli via altering zinc exposure time and order. We found that the order of exposure to zinc impacted resistance development. These impacted samples then underwent whole genome and RNA sequencing analysis. Results Zinc pre-exposure led to a subsequent acceleration of ciprofloxacin resistance. Specifically, we saw that 5 days of zinc pre-exposure led samples to have nearly a 4× and 3× higher MIC after 2 and 3 days of subinhibitory antibiotics, respectively, compared to samples not pre-exposed to zinc, but only if ciprofloxacin exposure happened in the absence of zinc. Additionally, for samples that underwent the same pre-exposure treatment, those exposed to a combination of zinc and ciprofloxacin saw delayed ciprofloxacin resistance compared to those exposed to only ciprofloxacin resulting in up to a 5× lower MIC within the first 2 days of antibiotic exposure. We did not observe any genetic changes or changes in antibiotic tolerance in cells after zinc pre-exposure, suggesting changes in gene expression may underlie these phenotypes. Discussion These results highlight the need to reexamine the role of zinc, and supplements more broadly, on antibiotic resistance evolution.
Collapse
Affiliation(s)
- Mark P. Suprenant
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Carly Ching
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Neila Gross
- Department of Materials Science and Engineering, Boston University, Boston, MA, United States
| | - Indorica Sutradhar
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Jessica E. Anderson
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Nourhan El Sherif
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Muhammad H. Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
- Department of Materials Science and Engineering, Boston University, Boston, MA, United States
- Howard Hughes Medical Institute, Boston University, Boston, MA, United States
- Center on Forced Displacement, Boston University, Boston, MA, United States
| |
Collapse
|
5
|
Zhang S, Wang J, Yu R, Liu H, Liu S, Luo K, Lei J, Han B, Chen Y, Han S, Yang E, Xun M, Han L. The role of universal stress protein Usp1413 in meropenem adaptive resistance and environmental stress responses in Acinetobacter baumannii. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 8:100332. [PMID: 39758053 PMCID: PMC11699434 DOI: 10.1016/j.crmicr.2024.100332] [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] [Indexed: 01/07/2025] Open
Abstract
Although various mechanisms of carbapenem-resistance have been identified in the nosocomial pathogen Acinetobacter baumannii, the critical process of resistance evolution and the factors involved in are not well understood. Herein, we identified a universal stress protein Usp1413 which played an important role in adaptive resistance of A. baumannii to meropenem (MEM). Based on RNA-Seq and genome sequencing, Usp1413 was not only one of the most downregulated USPs, but also the bare one having mutation of tyrosine and glycine inserted at the site of 229-230 (YG229-230) under the stimulation of MEM. Deletion of Usp1413 resulted in increased MEM resistance. In addition, Usp1413 affected the bacterial abilities of biofilm formation and swarm motility, as well as helped A. baumannii response to various environmental stresses. These effects of Usp1413 were achieved by regulating its interaction proteins, within the functions of YigZ family protein, acetyltransferase, and SulP family inorganic anion transporter. The insertion mutation of YG229-230 influenced both the expression of interaction proteins and the phenotypes of bacteria. Finally, the promotor region of Usp1413 was convinced by point mutations. Overall, our findings identified the universal stress protein Usp1413 as a contributor involved in MEM adaptive resistance and responded to numerous environmental stresses. This study provides novel insights into the mechanism of universal stress proteins in participating antibiotic resistance, and affords a potential target for controlling drug resistance development in A. baumannii.
Collapse
Affiliation(s)
- Sirui Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jingdan Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Rong Yu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Haiping Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Xi'an Daxing Hospital, Xi'an, China
| | - Shuyan Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kai Luo
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jin'e Lei
- Department of Laboratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bei Han
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yanjiong Chen
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Shaoshan Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - E Yang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Meng Xun
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Lei Han
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
6
|
Abdelmassih MM, Ismail MM, Kashef MT, Essam T. Repurposing fusidic acid as an antimicrobial against enterococci with a low probability of resistance development. Int Microbiol 2024; 27:1807-1819. [PMID: 38532184 PMCID: PMC11611940 DOI: 10.1007/s10123-024-00506-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/26/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024]
Abstract
Drug repurposing constitutes a strategy to combat antimicrobial resistance, by using agents with known safety, pharmacokinetics, and pharmacodynamics. Previous studies have implemented new fusidic acid (FA) front-loading-dose regimens, allowing higher serum levels than those achievable with ordinary doses. As susceptibility breakpoints are affected by serum level, we evaluated the repurposing of FA as an antimicrobial product against enterococci. FA minimum inhibitory concentrations (MICs) against standard enterococci strains; Enterococcus faecalis ATCC 29212 and Enterococcus faecium ATCC 27270 were 2 and 4 µg/mL, respectively. The MIC against 98 enterococcal clinical isolates was ≤ 8 µg/mL; all would be susceptible if categorized according to recalculated breakpoints (≥ 16 µg/mL), based on the serum level achieved using the front-loading regimen. FA administration in vivo, using the BALB/c mouse infection model, significantly reduced bacterial burden by two to three log10 units in the liver and spleen of mice infected with vancomycin-susceptible and -resistant strains. Exposure of the standard enterococcal strains to increasing, but not fixed, FA concentrations resulted in resistant strains (MIC = 128 µg/mL), with thicker cell walls and slower growth rates. Only one mutation (M651I) was detected in the fusA gene of the resistant strain derived from serial passage of E. faecium ATCC 27270, which was retained in the revertant strain after passage in the FA-free medium. In conclusion, FA can be repurposed as an antimicrobial drug against enterococci with a low probability of mutational resistance development, and can be employed for treatment of infections attributable to vancomycin-resistant enterococci.
Collapse
Affiliation(s)
- Mark M Abdelmassih
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Maha M Ismail
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Mona T Kashef
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Tamer Essam
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
7
|
Yang J, Xu JF, Liang S. Antibiotic resistance in Pseudomonas aeruginosa: mechanisms and emerging treatment. Crit Rev Microbiol 2024:1-19. [PMID: 39556143 DOI: 10.1080/1040841x.2024.2429599] [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: 02/02/2024] [Revised: 08/22/2024] [Accepted: 11/10/2024] [Indexed: 11/19/2024]
Abstract
Pseudomonas aeruginosa, able to survive on the surfaces of medical devices, is a life-threatening pathogen that mainly leads to nosocomial infection especially in immunodeficient and cystic fibrosis (CF) patients. The antibiotic resistance in P. aeruginosa has become a world-concerning problem, which results in reduced and ineffective therapy efficacy. Besides intrinsic properties to decrease the intracellular content and activity of antibiotics, P. aeruginosa develops acquired resistance by gene mutation and acquisition, as well as adaptive resistance under specific situations. With in-depth research on drug resistance mechanisms and the development of biotechnology, innovative strategies have emerged and yielded benefits such as screening for new antibiotics based on artificial intelligence technology, utilizing drugs synergistically, optimizing administration, and developing biological therapy. This review summarizes the recent advances in the mechanisms of antibiotic resistance and emerging treatments for combating resistance, aiming to provide a reference for the development of therapy against drug-resistant P. aeruginosa.
Collapse
Affiliation(s)
- Jian Yang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuo Liang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Ma R, Peng L, Tang R, Jiang T, Chang J, Li G, Wang J, Yang Y, Yuan J. Bioaerosol emission characteristics and potential risks during composting: Focus on pathogens and antimicrobial resistance. JOURNAL OF HAZARDOUS MATERIALS 2024; 481:136466. [PMID: 39549575 DOI: 10.1016/j.jhazmat.2024.136466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/19/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024]
Abstract
In this study, we analyzed bioaerosol emission characteristics and potential risks of antimicrobial resistance (AMR) during composting using the impaction culture method and metagenomic sequencing. The results showed that the highly saturated water vapor in the emission gas mitigated particulate matter emission during the thermophilic period. About the bioaerosols, the airborne aerobic bacterial emissions were suppressed as composting enters the mature period, and the airborne fungi are usually present as single-cell or small-cell aggregates (< 3.3 µm). In addition, the microbial community structure in bioaerosols was stable and independent of composting time. Most importantly, the PM2.5 in bioaerosols contained large amounts of antibiotic resistance genes (ARGs), potential pathogens, and multidrug resistant pathogens, which were diverse and present in high concentrations. Among them, ARGs concentrations encoding 21 antibiotics ranged from - 4.50 to 0.70 ppm/m3 (Log10 ARGs). Among the 89 potential human pathogens detected, Escherichia coli, Salmonella enterica, Klebsiella pneumoniae, and Staphylococcus aureus were the only culturable potentially multidrug resistant pathogens carrying multiple ARGs encoding resistance at high concentrations (- 0.57 to 1.15 ppm/m3 (Log10 ARGs)), and were more likely to persist and multiply in oligotrophic environments. Our findings indicate that composting technology can transfer AMR from solid compost to gas phase and increase the risk of AMR transmission.
Collapse
Affiliation(s)
- Ruonan Ma
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Science, China Agricultural University, Beijing 100193, China
| | - Lijuan Peng
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Science, China Agricultural University, Beijing 100193, China
| | - Ruolan Tang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Science, China Agricultural University, Beijing 100193, China
| | - Tao Jiang
- School of New Energy Materials and Chemistry, Leshan Normal University, Sichuan 614000, China
| | - Jiali Chang
- School of New Energy Materials and Chemistry, Leshan Normal University, Sichuan 614000, China
| | - Guoxue Li
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Science, China Agricultural University, Beijing 100193, China
| | - Jiani Wang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Science, China Agricultural University, Beijing 100193, China
| | - Yan Yang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Science, China Agricultural University, Beijing 100193, China
| | - Jing Yuan
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Science, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
9
|
Dong L, Sun L, Yang Y, Yuan L, Gao W, Yu D, Meng Q, Shi W, Wang Q, Li Y, Zhang Y, You X, Yao K. Non-antibiotic pharmaceutical phenylbutazone binding to MexR reduces the antibiotic susceptibility of Pseudomonas aeruginosa. Microbiol Res 2024; 288:127872. [PMID: 39146705 DOI: 10.1016/j.micres.2024.127872] [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: 05/12/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
Antimicrobial resistance has been an increasingly serious threat to global public health. The contribution of non-antibiotic pharmaceuticals to the development of antibiotic resistance has been overlooked. Our study found that the anti-inflammatory drug phenylbutazone could protect P. aeruginosa against antibiotic mediated killing by binding to the efflux pump regulator MexR. In this study, antibiotic activity against P. aeruginosa alone or in combination with phenylbutazone was evaluated in vitro and in vivo. Resazurin accumulation assay, transcriptomic sequencing, and PISA assay were conducted to explore the underlying mechanism for the reduced antibiotic susceptibility caused by phenylbutazone. Then EMSA, ITC, molecular dynamic simulations, and amino acid substitutions were used to investigate the interactions between phenylbutazone and MexR. We found that phenylbutazone could reduce the susceptibility of P. aeruginosa to multiple antibiotics, including parts of β-lactams, fluoroquinolones, tetracyclines, and macrolides. Phenylbutazone could directly bind to MexR, then promote MexR dissociating from the mexA-mexR intergenic region and de-repress the expression of MexAB-OprM efflux pump. The overexpressed MexAB-OprM pump resulted in the reduced antibiotic susceptibility. And the His41 and Arg21 residues of MexR were involved in the phenylbutazone-MexR interaction. We hope this study would imply the potential risk of antibiotic resistance caused by non-antibiotic pharmaceuticals.
Collapse
Affiliation(s)
- Limin Dong
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Lang Sun
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yan Yang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lin Yuan
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wei Gao
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Dan Yu
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Qinghong Meng
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wei Shi
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Qing Wang
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Yue Li
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Youwen Zhang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Xuefu You
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Kaihu Yao
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| |
Collapse
|
10
|
Lei TY, Liao BB, Yang LR, Wang Y, Chen XB. Hypervirulent and carbapenem-resistant Klebsiella pneumoniae: A global public health threat. Microbiol Res 2024; 288:127839. [PMID: 39141971 DOI: 10.1016/j.micres.2024.127839] [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: 06/06/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 08/16/2024]
Abstract
The evolution of hypervirulent and carbapenem-resistant Klebsiella pneumoniae can be categorized into three main patterns: the evolution of KL1/KL2-hvKp strains into CR-hvKp, the evolution of carbapenem-resistant K. pneumoniae (CRKp) strains into hv-CRKp, and the acquisition of hybrid plasmids carrying carbapenem resistance and virulence genes by classical K. pneumoniae (cKp). These strains are characterized by multi-drug resistance, high virulence, and high infectivity. Currently, there are no effective methods for treating and surveillance this pathogen. In addition, the continuous horizontal transfer and clonal spread of these bacteria under the pressure of hospital antibiotics have led to the emergence of more drug-resistant strains. This review discusses the evolution and distribution characteristics of hypervirulent and carbapenem-resistant K. pneumoniae, the mechanisms of carbapenem resistance and hypervirulence, risk factors for susceptibility, infection syndromes, treatment regimens, real-time surveillance and preventive control measures. It also outlines the resistance mechanisms of antimicrobial drugs used to treat this pathogen, providing insights for developing new drugs, combination therapies, and a "One Health" approach. Narrowing the scope of surveillance but intensifying implementation efforts is a viable solution. Monitoring of strains can be focused primarily on hospitals and urban wastewater treatment plants.
Collapse
Affiliation(s)
- Ting-Yu Lei
- College of Pharmaceutical Science, Dali University, Dali 671000, China.
| | - Bin-Bin Liao
- College of Pharmaceutical Science, Dali University, Dali 671000, China.
| | - Liang-Rui Yang
- First Affiliated Hospital of Dali University, Yunnan 671000, China.
| | - Ying Wang
- College of Pharmaceutical Science, Dali University, Dali 671000, China.
| | - Xu-Bing Chen
- College of Pharmaceutical Science, Dali University, Dali 671000, China.
| |
Collapse
|
11
|
Aseem A, Sagar P, Reddy NS, Veleri S. The antimicrobial resistance profile in poultry of Central and Southern India is evolving with distinct features. Comp Immunol Microbiol Infect Dis 2024; 114:102255. [PMID: 39432940 DOI: 10.1016/j.cimid.2024.102255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024]
Abstract
Antimicrobial resistance (AMR) is fast emerging and is depleting antibiotics repertoire. Poultry is a major source for AMR because focus to enhance its production by modern practices widely uses antibiotics. India and China are major producers of meat and have hotspots of AMR. The Central and Southern India were predicted as emerging hotspots for AMR in poultry but no data available to substantiate it. To this end, we collected chicken feces from poultry farms in these regions and isolated genomic DNA. Further, shotgun whole genome sequencing was performed for metagenomics analysis. For the first time, we report the AMR gene profiles in poultry from Kerala and Telangana. The samples exhibited a higher prevalence of gram-negative and anaerobic species. The high priority pathogens in India were detected, like E.coli, Clostridium perfringens, Klebsiella pneumonia Staphylococcus aureus, Enterococcous faecalis, Pseudomonas aeruginosa, Bacteriodes fragiles. Conspicuously, the Southern India had the highest abundance of AMR genes than the Central India. E.coli was significantly more prevalent in the southernmost zone of India than in other sites. Our data had many common AMR profile features of the European Union (EU) poultry farms but lacked mcr-1, which was a lately emerged AMR gene in E.coli. Our data revealed the extent of AMR gene evolved in the Central and Southern India. It is comparable to the EU data but severity is lesser than in the EU.
Collapse
Affiliation(s)
- Ajmal Aseem
- Drug Safety Division, ICMR-National Institute of Nutrition, Hyderabad 500007, India
| | - Prarthi Sagar
- Drug Safety Division, ICMR-National Institute of Nutrition, Hyderabad 500007, India
| | | | - Shobi Veleri
- Drug Safety Division, ICMR-National Institute of Nutrition, Hyderabad 500007, India.
| |
Collapse
|
12
|
Wang X, Chen Q, Pang R, Zhang C, Huang G, Han Z, Su Y. Exposure modes determined the effects of nanomaterials on antibiotic resistance genes: The different roles of oxidative stress and quorum sensing. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 360:124772. [PMID: 39168438 DOI: 10.1016/j.envpol.2024.124772] [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: 07/01/2024] [Revised: 08/09/2024] [Accepted: 08/17/2024] [Indexed: 08/23/2024]
Abstract
The effects of co-occurrent pollutants on antibiotic resistance genes (ARGs) in wastewater treatment plants (WWTPs) have raised attentions. However, how the different realistic exposure scenarios determining the effects of nanomaterials (NMs) on ARGs, was still unknown. Herein, the effects of NMs on ARGs under two realistic scenarios was investigated by short-term and long-term exposure modes. The presence of NMs with two different exposure modes could both promote the dissemination of ARGs, and the results were dose-, type- and duration-dependent. Compared to short-term exposure, the long-term exposure increased the abundances of ARGs with a greater extent except nano-ZnO. The long-term exposure increased the overall abundances of target ARGs by 2.9%-20.4%, while shot-term exposure caused the 3.4%-10.5% increment. The mechanisms of ARGs fates driven by NMs exposure were further investigated from the levels of microbial community shift, intracellular oxidative stress, and gene abundance. The variations of several potential bacterial hosts did not contribute to the difference in the ARGs transmission with different exposure modes because NMs types played more vital roles in the shift of microbial community compared to the exposure modes. For the short-term exposure, NMs were capable of triggering the QS by upregulating relevant genes, and further activated the production of surfactin and increased membrane permeability, resulting in the facilitation of ARGs transfer. However, NMs under long-term exposure scenario preferentially stimulated oxidative stress by generating more ROS, which then enhanced ARGs dissemination. Therefore, the exposure mode of NMs was one of the pivotal factors determining the ARGs fates by different triggering mechanisms. This study highlighted the importance of exposure scenario of co-occurrent pollutants on ARGs spread, which will benefit the comprehensive understanding of the actual environmental fates of ARGs.
Collapse
Affiliation(s)
- Xueting Wang
- Shanghai Engineering Research Center of Biotransformation on Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China
| | - Qirui Chen
- Shanghai Engineering Research Center of Biotransformation on Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China
| | - Ruirui Pang
- Shanghai Engineering Research Center of Biotransformation on Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China
| | - Congyan Zhang
- Shanghai Engineering Research Center of Biotransformation on Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China
| | - Guangchen Huang
- Shanghai Engineering Research Center of Biotransformation on Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhibang Han
- Shanghai Engineering Research Center of Biotransformation on Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China
| | - Yinglong Su
- Shanghai Engineering Research Center of Biotransformation on Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China; Engineering Research Center for Nanophotonics & Advanced Instrument, Ministry of Education, East China Normal University, Shanghai, 200241, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, China.
| |
Collapse
|
13
|
Sakagianni A, Koufopoulou C, Koufopoulos P, Feretzakis G, Kalles D, Paxinou E, Myrianthefs P, Verykios VS. The Synergy of Machine Learning and Epidemiology in Addressing Carbapenem Resistance: A Comprehensive Review. Antibiotics (Basel) 2024; 13:996. [PMID: 39452262 PMCID: PMC11505168 DOI: 10.3390/antibiotics13100996] [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: 09/19/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES Carbapenem resistance poses a significant threat to public health by undermining the efficacy of one of the last lines of antibiotic defense. Addressing this challenge requires innovative approaches that can enhance our understanding and ability to combat resistant pathogens. This review aims to explore the integration of machine learning (ML) and epidemiological approaches to understand, predict, and combat carbapenem-resistant pathogens. It examines how leveraging large datasets and advanced computational techniques can identify patterns, predict outbreaks, and inform targeted intervention strategies. METHODS The review synthesizes current knowledge on the mechanisms of carbapenem resistance, highlights the strengths and limitations of traditional epidemiological methods, and evaluates the transformative potential of ML. Real-world applications and case studies are used to demonstrate the practical benefits of combining ML and epidemiology. Technical and ethical challenges, such as data quality, model interpretability, and biases, are also addressed, with recommendations provided for overcoming these obstacles. RESULTS By integrating ML with epidemiological analysis, significant improvements can be made in predictive accuracy, identifying novel patterns in disease transmission, and designing effective public health interventions. Case studies illustrate the benefits of interdisciplinary collaboration in tackling carbapenem resistance, though challenges such as model interpretability and data biases must be managed. CONCLUSIONS The combination of ML and epidemiology holds great promise for enhancing our capacity to predict and prevent carbapenem-resistant infections. Future research should focus on overcoming technical and ethical challenges to fully realize the potential of these approaches. Interdisciplinary collaboration is key to developing sustainable strategies to combat antimicrobial resistance (AMR), ultimately improving patient outcomes and safeguarding public health.
Collapse
Affiliation(s)
| | - Christina Koufopoulou
- Anesthesiology Department, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Petros Koufopoulos
- Internal Medicine Department, Sismanogleio General Hospital, 15126 Marousi, Greece;
| | - Georgios Feretzakis
- School of Science and Technology, Hellenic Open University, 26335 Patras, Greece; (G.F.); (D.K.); (E.P.)
| | - Dimitris Kalles
- School of Science and Technology, Hellenic Open University, 26335 Patras, Greece; (G.F.); (D.K.); (E.P.)
| | - Evgenia Paxinou
- School of Science and Technology, Hellenic Open University, 26335 Patras, Greece; (G.F.); (D.K.); (E.P.)
| | - Pavlos Myrianthefs
- Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vassilios S. Verykios
- School of Science and Technology, Hellenic Open University, 26335 Patras, Greece; (G.F.); (D.K.); (E.P.)
| |
Collapse
|
14
|
Shafiq M, Obinwanne Okoye C, Nazar M, Ali Khattak W, Algammal AM. Ecological consequences of antimicrobial residues and bioactive chemicals on antimicrobial resistance in agroecosystems. J Adv Res 2024:S2090-1232(24)00467-3. [PMID: 39414225 DOI: 10.1016/j.jare.2024.10.013] [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: 08/11/2024] [Revised: 09/30/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND The widespread use of antimicrobials in agriculture, coupled with bioactive chemicals like pesticides and growth-promoting agents, has accelerated the global crisis of antimicrobial resistance (AMR). Agroecosystems provides a platform in the evolution and dissemination of antibiotic-resistant bacteria (ARB) and antibiotic resistance genes (ARGs), which pose significant threats to both environmental and public health. AIM OF REVIEW This review explores the ecological consequences of antimicrobial residues and bioactive chemicals in agroecosystems, with a focus on their role in shaping AMR. It delves into the mechanisms by which these substances enter agricultural environments, their interactions with soil microbiomes, and the subsequent impacts on microbial community structure. KEY SCIENTIFIC CONCEPTS OF REVIEW Evidence indicates that the accumulation of antimicrobials promotes resistance gene transfer among microorganisms, potentially compromising ecosystem health and agricultural productivity. By synthesizing current research, we identify critical gaps in knowledge and propose strategies for mitigating the ecological risks associated with antimicrobial residues. Moreover, this review highlights the urgent need for integrated management approaches to preserve ecosystem health and combat the spread of AMR in agricultural settings.
Collapse
Affiliation(s)
- Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Department of Pharmacology, Shantou University Medical College, Shantou 515041, China.
| | - Charles Obinwanne Okoye
- Biofuels Institute, School of Environment & Safety Engineering, Jiangsu University, Zhenjiang 212013, China; School of Life Sciences, Jiangsu University, Zhenjiang 212013, China; Department of Zoology & Environmental Biology, University of Nigeria, Nsukka 410001, Nigeria
| | - Mudasir Nazar
- Institute of Animal Science, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
| | - Wajid Ali Khattak
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Abdelazeem M Algammal
- Department of Bacteriology, Immunology, and Mycology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
15
|
Nasr J, Abdessamad H, Mina J, Haykal T, Jamil Y, Abboud E, Mahdi A, Asmar R, Abi Assaad R, Alameddine D, Bourji A, Mahdi M, Abdulaal R, Tomassian S, El Ahmadieh H, Azzam W, Mokhbat JE, Moghnieh R, Rodriguez-Morales AJ, Husni R. The epidemiology of gram-negative bacteremia in Lebanon: a study in four hospitals. Ann Clin Microbiol Antimicrob 2024; 23:90. [PMID: 39385237 PMCID: PMC11465513 DOI: 10.1186/s12941-024-00740-0] [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: 03/05/2024] [Accepted: 08/16/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Gram-negative bacteremia is a life-threatening infection with high morbidity and mortality. Its incidence is rising worldwide, and treatment has become more challenging due to emerging bacterial resistance. Little data is available on the burden and outcome of such infections in Lebanon. METHODS We conducted this retrospective study in four Lebanese hospitals. Data on medical conditions and demographics of 2400 patients diagnosed with a bloodstream infection based on a positive blood culture were collected between January 2014 and December 2020. RESULTS Most bacteremias were caused by Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter baumannii, with the more resistant organisms being hospital-acquired. Third-generation cephalosporin and quinolone resistance was steady throughout the study, but carbapenem resistance increased. Mortality with such infections is high, but carbapenem resistance or infection with Pseudomonas or Acinetobacter species were significant risk factors for poor outcomes. CONCLUSION This is the first multi-center study from Lebanon on gram-negative bacteremia, resistance patterns, and factors associated with a poor outcome. More surveillance is needed to provide data to guide empirical treatment for bacteremia in Lebanon.
Collapse
Affiliation(s)
- Janane Nasr
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Hilal Abdessamad
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Johnathan Mina
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Tony Haykal
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Yasser Jamil
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Emma Abboud
- Laboratory Director, Mount Lebanon Hospital University Medical Center, Beirut, 1102, Lebanon
| | - Ahmad Mahdi
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rana Asmar
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rawad Abi Assaad
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Dana Alameddine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Alaa Bourji
- Department of Surgery, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Mahmoud Mahdi
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Razan Abdulaal
- Department of Internal Medicine, University of Balamand, Balamand, Lebanon
| | - Serge Tomassian
- Department of Internal Medicine, University of Balamand, Balamand, Lebanon
| | - Hanane El Ahmadieh
- Infection Control Coordination, Mount Lebanon Hospital University Medical Center, Beirut, 1102, Lebanon
| | - Wael Azzam
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Jacques E Mokhbat
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rima Moghnieh
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Alfonso J Rodriguez-Morales
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
- Master of Clinical Epidemiology and Biostatistics, Universidad Cientifica del Sur, Lima, 15067, Peru
| | - Rola Husni
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon.
| |
Collapse
|
16
|
Freitas PR, de Araújo ACJ, Araújo IM, de Almeida RS, Borges JAO, Paulo CLR, Oliveira-Tintino CDM, Miranda GM, Araújo-Neto JB, Nascimento IJS, Araújo-Júnior JX, Silva JMA, Balbino TCL, Silva-Júnior EF, Aquino TM, Mendonca-Junior FJB, Marinho ES, Santos HS, Lima CMG, Obaidullah AJ, Bin Emran T, Cunha FAB, Menezes IRA, Tintino SR, Coutinho HDM. Thiazine-derived compounds in inhibiting efflux pump in Staphylococcus aureus K2068, mepA gene expression, and membrane permeability alteration. Biomed Pharmacother 2024; 179:117291. [PMID: 39146766 DOI: 10.1016/j.biopha.2024.117291] [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: 06/06/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
Staphylococcus aureus is a bacterium responsible for resistance to multiple drugs and the efflux system is widely studied among the resistance mechanisms developed by this species. The present study evaluates the inhibition of the MepA efflux pump by thiadiazine-derived compounds. For this purpose, thiadiazine-derived compounds (IJ-14 to IJ-20) were tested against S. aureus K2068 strains. Microdilution tests were initially conducted to assess the Minimum Inhibitory Concentration (MIC) of the compounds and their efflux pump inhibition activity. In addition, fluorimetry tests were performed using BrEt emission and tests were conducted to inhibit the expression of the mepA gene. This involved comparing the bacterial gene expression with the antibiotic alone to the gene expression after combining compounds (IJ-17 and IJ-20) with the antibiotic. Furthermore, membrane permeability assessment tests and in silico molecular docking tests were performed. It was observed that the IJ17 and IJ20 compounds exhibited direct activity against the tested strain. The IJ17 compound produced significant results in the gene inhibition tests, which was also evidenced through the membrane permeability alteration test. These findings suggest that thiadiazine-derived compounds have promising effects against one of the main resistance mechanisms, with the IJ17 compound presenting observable mechanisms of action.
Collapse
Affiliation(s)
- Priscilla R Freitas
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil
| | - Ana C J de Araújo
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil
| | - Isaac M Araújo
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil
| | - Ray S de Almeida
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil
| | - João A O Borges
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil
| | - Cícera L R Paulo
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil
| | | | | | - José B Araújo-Neto
- Postgraduate Program in Biological Sciences, Biosciences Center, Federal University of Pernambuco, Recife, PE 50740-570, Brazil
| | - Igor J S Nascimento
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió, AL 57072-900, Brazil
| | - João X Araújo-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió, AL 57072-900, Brazil
| | - Julia M A Silva
- Department of Microbiology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Tereza C L Balbino
- Department of Microbiology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Edeildo F Silva-Júnior
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL 57072-900, Brazil
| | - Thiago M Aquino
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL 57072-900, Brazil
| | | | - Emmanuel S Marinho
- Laboratory of Chemistry of Natural and Synthetic Product, State University of Ceará, UECE, Fortaleza, CE 60741-835, Brazil
| | - Hélcio S Santos
- Laboratory of Chemistry of Natural and Synthetic Product, State University of Ceará, UECE, Fortaleza, CE 60741-835, Brazil
| | | | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Talha Bin Emran
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Legorreta Cancer Center, Brown University, Providence, RI 02912, USA; Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh.
| | - Francisco A B Cunha
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil
| | - Irwin R A Menezes
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil
| | - Saulo R Tintino
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil
| | - Henrique D M Coutinho
- Department of Biological Chemistry, Universidade Regional do Cariri, Crato-CE, Brazil.
| |
Collapse
|
17
|
Pal A, Ghosh D, Thakur P, Nagpal P, Irulappan M, Maruthan K, Mukherjee S, Patil N, Dutta T, Veeraraghavan B, Vivekanandan P. Clinically relevant mutations in regulatory regions of metabolic genes facilitate early adaptation to ciprofloxacin in Escherichia coli. Nucleic Acids Res 2024; 52:10385-10399. [PMID: 39180403 PMCID: PMC11417348 DOI: 10.1093/nar/gkae719] [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: 12/04/2023] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
The genomic landscape associated with early adaptation to ciprofloxacin is poorly understood. Although the interplay between core metabolism and antimicrobial resistance is being increasingly recognized, mutations in metabolic genes and their biological role remain elusive. Here, we exposed Escherichia coli to increasing gradients of ciprofloxacin with intermittent transfer-bottlenecking and identified mutations in three non-canonical targets linked to metabolism including a deletion (tRNA-ArgΔ414-bp) and point mutations in the regulatory regions of argI (ARG box) and narU. Our findings suggest that these mutations modulate arginine and carbohydrate metabolism, facilitate anaerobiosis and increased ATP production during ciprofloxacin stress. Furthermore, mutations in the regulatory regions of argI and narU were detected in over 70% of sequences from clinical E. coli isolates and were overrepresented among ciprofloxacin-resistant isolates. In sum, we have identified clinically relevant mutations in the regulatory regions of metabolic genes as a central theme that drives physiological changes necessary for adaptation to ciprofloxacin stress.
Collapse
Affiliation(s)
- Arijit Pal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Department of Zoology, Raiganj Surendranath Mahavidyalaya, Sudarshanpur, Raiganj, Uttar Dinajpur, West Bengal733134, India
| | - Dipannita Ghosh
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Pratyusha Thakur
- RNA Biology Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Priya Nagpal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Madhumathi Irulappan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Karthik Maruthan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Sanket Mukherjee
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Nikita G Patil
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Amity Institute of Virology and Immunology, Amity University, Noida, Uttar Pradesh, India
| | - Tanmay Dutta
- RNA Biology Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Balaji Veeraraghavan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Perumal Vivekanandan
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
18
|
Crow JC, Geng H, Sullivan TJ, Soucy SM, Schultz D. Dynamics of drug delivery determines course of evolution of antibiotic responses in bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.29.569327. [PMID: 38076825 PMCID: PMC10705423 DOI: 10.1101/2023.11.29.569327] [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: 12/24/2023]
Abstract
To adjust to sudden shifts in conditions, microbes possess regulated genetic mechanisms that sense environmental challenges and induce the appropriate responses. The initial evolution of microbes in new environments is thought to be driven by regulatory mutations, but it is not clear how this evolution is affected by how quickly conditions change (i.e. dynamics). Here, we perform experimental evolution on continuous cultures of tetracycline resistant E. coli in different dynamical regimens of drug administration. We find that cultures evolved under gradually increasing drug concentrations acquire fine-tuning mutations adapting an alternative efflux pump to tetracycline. However, cultures that are instead periodically exposed to large drug doses evolve transposon insertions resulting in loss of regulation of the main mechanism of tetracycline resistance. A mathematical model shows that sudden drug exposures overwhelm regulated responses, which cannot induce resistance fast enough. These results help explain the frequent loss of regulation of resistance in clinical pathogens.
Collapse
Affiliation(s)
- John C. Crow
- Department of Microbiology & Immunology, Dartmouth – Geisel School of Medicine, Hanover, NH 03755, USA
| | - Hao Geng
- Department of Microbiology & Immunology, Dartmouth – Geisel School of Medicine, Hanover, NH 03755, USA
| | - Timothy J. Sullivan
- Department of Biomedical Data Science, Dartmouth – Geisel School of Medicine, Hanover, NH 03755, USA
| | - Shannon M. Soucy
- Department of Biomedical Data Science, Dartmouth – Geisel School of Medicine, Hanover, NH 03755, USA
| | - Daniel Schultz
- Department of Microbiology & Immunology, Dartmouth – Geisel School of Medicine, Hanover, NH 03755, USA
| |
Collapse
|
19
|
Shen J, Yu D, Liu Z, Di H, He JZ. Land use conversion to uplands significantly increased the risk of antibiotic resistance genes in estuary area. ENVIRONMENT INTERNATIONAL 2024; 191:108953. [PMID: 39153385 DOI: 10.1016/j.envint.2024.108953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/28/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Land use conversion in estuary wetlands may affect the transmission of antibiotic resistance genes (ARGs), while the risk rank of the ARGs and the change of clinically relevant ARGs under various land-use types are not well understood. This study used metagenomics to reveal the diversity and abundance of ARGs across five distinct land uses: reed wetland, tidal flat, grassland, agricultural land and fallow land, as well as their distribution and potential health risks. Results showed that high numbers of ARG subtypes and classes were detected irrespective of land-use types, notably higher in agricultural land (144 ARG subtypes). The most shared ARG subtypes were multidrug resistance genes across all the land uses (29 subtypes, 4.7 × 10-2-1.5 × 10-1 copies per 16S rRNA gene copy). Proteobacteria and Actinobacteria were primary ARG hosts, with 18 and 15 ARGs were found in both of them, respectively. The ARG subtype mdtB was the most dominant clinical ARG detected with 90 % amino acid identity. The change of ARGs exhibited a consistent trend across land uses in terms of health risk ranks, with the highest observed in fallow land and the lowest in reed wetland. This study reveals the distribution pattern of ARGs across various land-use types, and enhances our understanding of the potential health risks associated with ARGs in the context of coastal wetland conversion in estuary areas.
Collapse
Affiliation(s)
- Jupei Shen
- Key Laboratory of Humid Subtropical Eco-geographical Process of Ministry of Education, School of Geographical Sciences/School of Carbon Neutrality Future Technology, Fujian Normal University, Fuzhou 350007, China
| | - Danting Yu
- Key Laboratory of Humid Subtropical Eco-geographical Process of Ministry of Education, School of Geographical Sciences/School of Carbon Neutrality Future Technology, Fujian Normal University, Fuzhou 350007, China.
| | - Zikai Liu
- Key Laboratory of Humid Subtropical Eco-geographical Process of Ministry of Education, School of Geographical Sciences/School of Carbon Neutrality Future Technology, Fujian Normal University, Fuzhou 350007, China
| | - Hongjie Di
- Centre for Soil and Environmental Research, Lincoln University, Lincoln, 7674, Christchurch, New Zealand
| | - Ji-Zheng He
- Key Laboratory of Humid Subtropical Eco-geographical Process of Ministry of Education, School of Geographical Sciences/School of Carbon Neutrality Future Technology, Fujian Normal University, Fuzhou 350007, China; School of Agriculture, Food and Ecosystem Sciences, The University of Melbourne, Parkville 3010, Victoria, Australia.
| |
Collapse
|
20
|
Dulanto Chiang A, Dekker JP. Efflux pump-mediated resistance to new beta lactam antibiotics in multidrug-resistant gram-negative bacteria. COMMUNICATIONS MEDICINE 2024; 4:170. [PMID: 39210044 PMCID: PMC11362173 DOI: 10.1038/s43856-024-00591-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
The emergence and spread of bacteria resistant to commonly used antibiotics poses a critical threat to modern medical practice. Multiple classes of bacterial efflux pump systems play various roles in antibiotic resistance, and members of the resistance-nodulation-division (RND) transporter superfamily are among the most important determinants of efflux-mediated resistance in gram-negative bacteria. RND pumps demonstrate broad substrate specificities, facilitating extrusion of multiple chemical classes of antibiotics from the bacterial cell. Several newer beta-lactams and beta-lactam/beta-lactamase inhibitor combinations (BL/BLI) have been developed to treat infections caused by multidrug resistant bacteria. Here we review recent studies that suggest RND efflux pumps in clinically relevant gram-negative bacteria may play critical but underappreciated roles in the development of resistance to beta-lactams and novel BL/BLI combinations. Improved understanding of the genetic and structural basis of RND efflux pump-mediated resistance may identify new antibiotic targets as well as strategies to minimize the emergence of resistance.
Collapse
Affiliation(s)
- Augusto Dulanto Chiang
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
- Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN, 37232, USA
| | - John P Dekker
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA.
- National Institutes of Health Clinical Center, NIH, Bethesda, MD, USA.
| |
Collapse
|
21
|
Semeraro M, Boubaker G, Scaccaglia M, Müller J, Vigneswaran A, Hänggeli KPA, Amdouni Y, Kramer LH, Vismarra A, Genchi M, Pelosi G, Bisceglie F, Heller M, Uldry AC, Braga-Lagache S, Hemphill A. Transient Adaptation of Toxoplasma gondii to Exposure by Thiosemicarbazone Drugs That Target Ribosomal Proteins Is Associated with the Upregulated Expression of Tachyzoite Transmembrane Proteins and Transporters. Int J Mol Sci 2024; 25:9067. [PMID: 39201756 PMCID: PMC11354806 DOI: 10.3390/ijms25169067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Thiosemicarbazones and their metal complexes have been studied for their biological activities against bacteria, cancer cells and protozoa. Short-term in vitro treatment with one gold (III) complex (C3) and its salicyl-thiosemicarbazone ligand (C4) selectively inhibited proliferation of T. gondii. Transmission Electron Microscopy (TEM) detected transient structural alterations in the parasitophorous vacuole membrane and the tachyzoite cytoplasm, but the mitochondrial membrane potential appeared unaffected by these compounds. Proteins potentially interacting with C3 and C4 were identified using differential affinity chromatography coupled with mass spectrometry (DAC-MS). Moreover, long-term in vitro treatment was performed to investigate parasitostatic or parasiticidal activity of the compounds. DAC-MS identified 50 ribosomal proteins binding both compounds, and continuous drug treatments for up to 6 days caused the loss of efficacy. Parasite tolerance to both compounds was, however, rapidly lost in their absence and regained shortly after re-exposure. Proteome analyses of six T. gondii ME49 clones adapted to C3 and C4 compared to the non-adapted wildtype revealed overexpression of ribosomal proteins, of two transmembrane proteins involved in exocytosis and of an alpha/beta hydrolase fold domain-containing protein. Results suggest that C3 and C4 may interfere with protein biosynthesis and that adaptation may be associated with the upregulated expression of tachyzoite transmembrane proteins and transporters, suggesting that the in vitro drug tolerance in T. gondii might be due to reversible, non-drug specific stress-responses mediated by phenotypic plasticity.
Collapse
Affiliation(s)
- Manuela Semeraro
- Department of Veterinary Medicine Sciences, University of Parma, Strada del Taglio 10, 43126 Parma, Italy; (M.S.); (L.H.K.); (A.V.); (M.G.)
| | - Ghalia Boubaker
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (J.M.); (A.V.); (K.P.A.H.); (Y.A.)
| | - Mirco Scaccaglia
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/a, 43124 Parma, Italy; (M.S.); (G.P.); (F.B.)
| | - Joachim Müller
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (J.M.); (A.V.); (K.P.A.H.); (Y.A.)
| | - Anitha Vigneswaran
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (J.M.); (A.V.); (K.P.A.H.); (Y.A.)
| | - Kai Pascal Alexander Hänggeli
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (J.M.); (A.V.); (K.P.A.H.); (Y.A.)
| | - Yosra Amdouni
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (J.M.); (A.V.); (K.P.A.H.); (Y.A.)
| | - Laura Helen Kramer
- Department of Veterinary Medicine Sciences, University of Parma, Strada del Taglio 10, 43126 Parma, Italy; (M.S.); (L.H.K.); (A.V.); (M.G.)
| | - Alice Vismarra
- Department of Veterinary Medicine Sciences, University of Parma, Strada del Taglio 10, 43126 Parma, Italy; (M.S.); (L.H.K.); (A.V.); (M.G.)
| | - Marco Genchi
- Department of Veterinary Medicine Sciences, University of Parma, Strada del Taglio 10, 43126 Parma, Italy; (M.S.); (L.H.K.); (A.V.); (M.G.)
| | - Giorgio Pelosi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/a, 43124 Parma, Italy; (M.S.); (G.P.); (F.B.)
| | - Franco Bisceglie
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/a, 43124 Parma, Italy; (M.S.); (G.P.); (F.B.)
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (M.H.); (A.-C.U.); (S.B.-L.)
| | - Anne-Christine Uldry
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (M.H.); (A.-C.U.); (S.B.-L.)
| | - Sophie Braga-Lagache
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (M.H.); (A.-C.U.); (S.B.-L.)
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (J.M.); (A.V.); (K.P.A.H.); (Y.A.)
| |
Collapse
|
22
|
Belay WY, Getachew M, Tegegne BA, Teffera ZH, Dagne A, Zeleke TK, Abebe RB, Gedif AA, Fenta A, Yirdaw G, Tilahun A, Aschale Y. Mechanism of antibacterial resistance, strategies and next-generation antimicrobials to contain antimicrobial resistance: a review. Front Pharmacol 2024; 15:1444781. [PMID: 39221153 PMCID: PMC11362070 DOI: 10.3389/fphar.2024.1444781] [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: 06/06/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Antibacterial drug resistance poses a significant challenge to modern healthcare systems, threatening our ability to effectively treat bacterial infections. This review aims to provide a comprehensive overview of the types and mechanisms of antibacterial drug resistance. To achieve this aim, a thorough literature search was conducted to identify key studies and reviews on antibacterial resistance mechanisms, strategies and next-generation antimicrobials to contain antimicrobial resistance. In this review, types of resistance and major mechanisms of antibacterial resistance with examples including target site modifications, decreased influx, increased efflux pumps, and enzymatic inactivation of antibacterials has been discussed. Moreover, biofilm formation, and horizontal gene transfer methods has also been included. Furthermore, measures (interventions) taken to control antimicrobial resistance and next-generation antimicrobials have been discussed in detail. Overall, this review provides valuable insights into the diverse mechanisms employed by bacteria to resist the effects of antibacterial drugs, with the aim of informing future research and guiding antimicrobial stewardship efforts.
Collapse
Affiliation(s)
- Wubetu Yihunie Belay
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Melese Getachew
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu Teffera
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Dagne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Tirsit Ketsela Zeleke
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Rahel Belete Abebe
- Department of clinical pharmacy, College of medicine and health sciences, University of Gondar, Gondar, Ethiopia
| | - Abebaw Abie Gedif
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Fenta
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getasew Yirdaw
- Department of environmental health science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Tilahun
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
23
|
Kumari K, Aggarwal Y, Singh RP. Molecular characterization and in-depth genomic analysis to unravel the pathogenic features of an environmental isolate Enterobacter sp. S-33. Int Microbiol 2024; 27:1095-1110. [PMID: 38044418 DOI: 10.1007/s10123-023-00461-y] [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: 08/17/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Enterobacter species represent widely distributed opportunistic pathogens, commonly associated with plants and humans. In the present study, we performed a detailed molecular characterization as well as genomic study of a type VI secretion system (T6SS) bacterium belonging to member of the family Enterobacteriaceae and named Enterobacter sp. S-33. The comparative sequence analysis of the 16S rRNA gene showed that the strain was closely related to other Enterobacter species. The complete genome of the strain with a genome size of 4.6 Mbp and GC-content of 55.63% was obtained through high-quality sequencing. The genomic analysis with online tools unravelled the various genes belonging to the bacterial secretion system, antibiotic resistance, virulence, efflux pumps, etc. The isolate showed the motility behavior that contributes to Enterobacter persistence in a stressed environment and further supports infections. PCR amplification and further sequencing confirmed the presence of drug-efflux genes acrA, acrB, and outer membrane genes, viz. OmpA, OmpC, and OmpF. The cell surface hydrophobicity and co-aggregation assay against different bacterial strains illustrated its putative pathogenic nature. Genome mining identified various biosynthetic gene clusters (BGCs) corresponding to non-ribosomal proteins (NRPS), siderophore, and arylpolyene production. Briefly, genome sequencing and detailed characterization of environmental Enterobacter isolate will assist in understanding the epidemiology of Enterobacter species, and the further prevention and treatment of infectious diseases caused by these broad-host range species.
Collapse
Affiliation(s)
- Kiran Kumari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Yogender Aggarwal
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Rajnish Prakash Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India.
| |
Collapse
|
24
|
Jalal RS, Sonbol HS. Resistome Signature and Antibiotic Resistance Mechanisms in Rhizospheric Soil Bacteriomes of Mecca Region, Saudi Arabia: Insights into Impact on Human Health. Life (Basel) 2024; 14:928. [PMID: 39202671 PMCID: PMC11355665 DOI: 10.3390/life14080928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
The objective of this investigation is to ascertain the distinctive profile of the rhizospheric soil resistome within the Mecca region, while also evaluating the potential risks associated with the horizontal transfer of resistome determinants to the open environment and human clinical isolates. We have made metagenomic whole-genome shotgun sequencing for rhizospheric microbiomes of two endemic plants, namely Moringa oleifera and Abutilon fruticosum. The rhizospheric resistomes of the two plants and the abundance of antibiotic resistance genes (ARGs) were identified by cross-referencing encoded proteins with the comprehensive antibiotic resistance database (CARD). The identified ARGs were then analyzed for their antimicrobial resistance (AMR) mechanisms. Predominantly within this soil are the two bacterial species Pseudomonas aeruginosa and Mycobacterium tuberculosis. These opportunistic human pathogens are implicated in respiratory infections and are correlated with heightened mortality rates. The most prevalent array of ARGs existing in this soil comprises mexA, mexC, mexE, and cpxR, associated with mechanisms of antibiotic active efflux, along with ACC(2), ACC(3), AAC(6), and APH(6), in addition to arr1, arr3, arr4, iri, rphA, and rphB, implicated in antibiotic inactivation. Furthermore, vanS, vanR, and vanJ are identified for antibiotic target alteration, while rpoB2 and RbpA are noted for antibiotic target replacement and protection, respectively. These mechanisms confer resistance against a diverse spectrum of drug classes encompassing fluoroquinolones, aminoglycosides, glycopeptides, and rifampicins. This study underscores the potential hazards posed to human health by the presence of these pathogenic bacteria within the rhizospheric soil of the Mecca region, particularly in scenarios where novel ARGs prevalent in human populations are harbored and subsequently transmitted through the food chain to human clinical isolates. Consequently, stringent adherence to good agricultural and food transportation practices is imperative, particularly with regard to edible plant parts and those utilized in folkloric medicine.
Collapse
Affiliation(s)
- Rewaa S. Jalal
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah 21493, Saudi Arabia;
| | - Hana S. Sonbol
- Department of Biology, College of Sciences, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| |
Collapse
|
25
|
Li J, Qin Z, Zhang B, Wu X, Wu J, Peng L, Xiao Y. Development of transcriptional factor-based whole-cell biosensors to monitor and degrade antibiotics using mutant cells obtained via adaptive laboratory evolution. JOURNAL OF HAZARDOUS MATERIALS 2024; 473:134536. [PMID: 38759406 DOI: 10.1016/j.jhazmat.2024.134536] [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: 01/25/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/19/2024]
Abstract
With the widespread use of antibiotics and increasing environmental concerns regarding antibiotic abuse, the detection and degradation of antibiotic residues in various samples has become a pressing issue. Transcriptional factor (TF)-based whole-cell biosensors are low-cost, easy-to-use, and flexible tools for detecting chemicals and controlling bioprocesses. However, because of cytotoxicity caused by antibiotics, the application of such biosensors is limited in the presence of antibiotics. In this study, we used antibiotic-tolerant mutants obtained via adaptive laboratory evolution (ALE) to develop TF-based whole-cell biosensors for antibiotic monitoring and degradation. The biosensors had high performance and stability in detecting relatively high concentrations of tetracycline (Tc) and nisin. The ALE mutant-based Tc biosensor exhibited a 10-fold larger linear detection range than the wild-type strain-based biosensor. Then, the Tc biosensor was employed to detect residual amounts of Tc in mouse stool, serum, and urine samples and facilitate Tc biodegradation in mouse stool, demonstrating its high utility. Considering that ALE has been demonstrated to enhance cell tolerance to various toxic chemicals, our strategy might facilitate the development of whole-cell biosensors for most antibiotics and other toxic ligands.
Collapse
Affiliation(s)
- Jiawei Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Ziqing Qin
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Baohui Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Xiaofeng Wu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Jing Wu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Lifeng Peng
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Yi Xiao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China.
| |
Collapse
|
26
|
Nguyen A, Roy JJS, Kim JH, Yun KH, Lee W, Kim KK, Kim T, Chaurasia AK. Repeated Exposure of Vancomycin to Vancomycin-Susceptible Staphylococcus aureus (VSSA) Parent Emerged VISA and VRSA Strains with Enhanced Virulence Potentials. J Microbiol 2024; 62:535-553. [PMID: 38814539 DOI: 10.1007/s12275-024-00139-8] [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: 11/25/2023] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/31/2024]
Abstract
The emergence of resistance against the last-resort antibiotic vancomycin in staphylococcal infections is a serious concern for human health. Although various drug-resistant pathogens of diverse genetic backgrounds show higher virulence potential, the underlying mechanism behind this is not yet clear due to variability in their genetic dispositions. In this study, we investigated the correlation between resistance and virulence in adaptively evolved isogenic strains. The vancomycin-susceptible Staphylococcus aureus USA300 was exposed to various concentrations of vancomycin repeatedly as a mimic of the clinical regimen to obtain mutation(s)-accrued-clonally-selected (MACS) strains. The phenotypic analyses followed by expression of the representative genes responsible for virulence and resistance of MACS strains were investigated. MACS strains obtained under 2 and 8 µg/ml vancomycin, named Van2 and Van8, respectively; showed enhanced vancomycin minimal inhibitory concentrations (MIC) to 4 and 16 µg/ml, respectively. The cell adhesion and invasion of MACS strains increased in proportion to their MICs. The correlation between resistance and virulence potential was partially explained by the differential expression of genes known to be involved in both virulence and resistance in MACS strains compared to parent S. aureus USA300. Repeated treatment of vancomycin against vancomycin-susceptible S. aureus (VSSA) leads to the emergence of vancomycin-resistant strains with variable levels of enhanced virulence potentials.
Collapse
Affiliation(s)
- An Nguyen
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - J Jean Sophy Roy
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Ji-Hoon Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyung-Hee Yun
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Truc Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| | - Akhilesh Kumar Chaurasia
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| |
Collapse
|
27
|
Che J, Fang Q, Hu S, Liu B, Wang L, Fang X, Li L, Luo T, Bao B. The Impact of Vp-Porin, an Outer Membrane Protein, on the Biological Characteristics and Virulence of Vibrio Parahaemolyticus. BIOLOGY 2024; 13:485. [PMID: 39056680 PMCID: PMC11273978 DOI: 10.3390/biology13070485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024]
Abstract
Porins are crucial proteins located in the outer membrane that directly influence antimicrobial resistance mechanisms and virulence in bacteria. In this study, a porin gene (Vp-porin) was cloned in V. parahaemolyticus, and the function of Vp-Porin in biological characteristics and virulence was investigated. The results of sequence analysis showed that Vp-Porin is highly conserved in Vibrio spp., and the predicted 3D structure showed it could form a 20-strand transmembrane β-barrel domian. Membrane permeabilization provides evidence that the membrane integrity of ∆Vp-porin was damaged and the sensitivity to tetracycline, polymyxin B, rifampicin and cephalothin of ∆Vp-porin obviously increased. In addition, loss of Vp-porin damaged motility due to downregulated flagellar synthesis. In addition, ∆Vp-porin exhibited attenuated cytotoxicity to Tetrahymena. The relative survival rate of Tetrahymena infection with ∆Vp-porin was 86%, which is much higher than that with WT (49%). Taken together, the results of this study indicate that Vp-Porin in V. parahaemolyticus plays various roles in biological characteristics in membrane integrity, antimicrobial resistance and motility and contributes to virulence.
Collapse
Affiliation(s)
- Jinyuan Che
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China; (J.C.); (L.W.)
| | - Qitong Fang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; (Q.F.); (S.H.); (B.L.)
| | - Shaojie Hu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; (Q.F.); (S.H.); (B.L.)
| | - Binghong Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; (Q.F.); (S.H.); (B.L.)
| | - Lei Wang
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China; (J.C.); (L.W.)
| | - Xiu Fang
- Fujian Provincial Key Laboratory of Breeding Lateolabrax Japonicus, Fuding 355200, China;
| | - Lekang Li
- Jiujiang Academy of Fishery Sciences, Jiujiang 332000, China;
| | - Tuyan Luo
- Institute of Quality Standards and Testing Technology for Agro-Products, Fujian Academy of Agricultural Science, Fuzhou 350003, China
| | - Baolong Bao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; (Q.F.); (S.H.); (B.L.)
| |
Collapse
|
28
|
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
|
29
|
Patra S, Biswas P, Karmakar S, Biswas K. Repression of resistance mechanisms of Pseudomonas aeruginosa: implications of the combination of antibiotics and phytoconstituents. Arch Microbiol 2024; 206:294. [PMID: 38850339 DOI: 10.1007/s00203-024-04012-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/17/2024] [Accepted: 05/19/2024] [Indexed: 06/10/2024]
Abstract
Antimicrobial resistance is a prevalent problem witnessed globally and creating an alarming situation for the treatment of infections caused by resistant pathogens. Available armaments such as antibiotics often fail to exhibit the intended action against resistant pathogens, leading to failure in the treatments that are causing mortality. New antibiotics or a new treatment approach is necessary to combat this situation. P. aeruginosa is an opportunistic drug resistant pathogen and is the sixth most common cause of nosocomial infections. P. aeruginosa due to its genome organization and other factors are exhibiting resistance against drugs. Bacterial biofilm formation, low permeability of outer membrane, the production of the beta-lactamase, and the production of several efflux systems limits the antibacterial potential of several classes of antibiotics. Combination of phytoconstituents with antibiotics is a promising strategy to combat multidrug resistant P. aeruginosa. Phytoconstituents such as flavonoids, terpenoids, alkaloids, polypeptides, phenolics, and essential oils are well known antibacterial agents. In this review, the activity of combination of the phytoconstituents and antibiotics, and their corresponding mechanism of action was discussed elaborately. The combination of antibiotics and plant-derived compounds exhibited better efficacy compared to antibiotics alone against the antibiotic resistance P. aeruginosa infections.
Collapse
Affiliation(s)
- Susmita Patra
- Eminent College of Pharmaceutical Technology, Barbaria, Barasat, North 24 Parganas, Kolkata, West Bengal, 700126, India
| | - Poulomi Biswas
- Eminent College of Pharmaceutical Technology, Barbaria, Barasat, North 24 Parganas, Kolkata, West Bengal, 700126, India
| | - Sanmoy Karmakar
- Department of Pharmaceutical Technology, Jadavpur University, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Kaushik Biswas
- Eminent College of Pharmaceutical Technology, Barbaria, Barasat, North 24 Parganas, Kolkata, West Bengal, 700126, India.
| |
Collapse
|
30
|
Suarez SA, Martiny AC. Intraspecific variation in antibiotic resistance potential within E. coli. Microbiol Spectr 2024; 12:e0316223. [PMID: 38661581 PMCID: PMC11237723 DOI: 10.1128/spectrum.03162-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/15/2024] [Indexed: 04/26/2024] Open
Abstract
Intraspecific genomic diversity brings the potential for an unreported and diverse reservoir of cryptic antibiotic resistance genes in pathogens, as cryptic resistance can occur without major mutations and horizontal transmission. Here, we predicted the differences in the types of antibiotics and genes that induce cryptic and latent resistance between micro-diverse Escherichia coli strains. For example, we hypothesize that known resistance genes will be the culprit of latent resistance within clinical strains. We used a modified functional metagenomics method to induce expression in eight E. coli strains. We found a total of 66 individual genes conferring phenotypic resistance to 11 out of 16 antibiotics. A total of 14 known antibiotic resistance genes comprised 21% of total identified genes, whereas the majority (52 genes) were unclassified cryptic resistance genes. Between the eight strains, 1.2% of core orthologous genes were positive (conferred resistance in at least one strain). Sixty-four percent of positive orthologous genes conferred resistance to only one strain, demonstrating high intraspecific variability of latent resistance genes. Cryptic resistance genes comprised most resistance genes among laboratory and clinical strains as well as natural, semisynthetic, and synthetic antibiotics. Known antibiotic resistance genes primarily conferred resistance to multiple antibiotics from varying origins and within multiple strains. Hence, it is uncommon for E. coli to develop cross-cryptic resistance to antibiotics from multiple origins or within multiple strains. We have uncovered prospective and previously unknown resistance genes as well as antibiotics that have the potential to trigger latent antibiotic resistance in E. coli strains from varying origins.IMPORTANCEIntraspecific genomic diversity may be a driving force in the emergence of adaptive antibiotic resistance. Adaptive antibiotic resistance enables sensitive bacterial cells to acquire temporary antibiotic resistance, creating an optimal window for the development of permanent mutational resistance. In this study, we investigate cryptic resistance, an adaptive resistance mechanism, and unveil novel (cryptic) antibiotic resistance genes that confer resistance when amplified within eight E. coli strains derived from clinical and laboratory origins. We identify the potential of cryptic resistance genes to confer cross-resistance to antibiotics from varying origins and within multiple strains. We discern antibiotic characteristics that promote latent resistance in multiple strains, considering intraspecific diversity. This study may help detect novel resistance genes and functional genes that could become responsible for cryptic resistance among diverse strains and antibiotics, thus also identifying potential novel antibiotic targets and mechanisms.
Collapse
Affiliation(s)
- Stacy A. Suarez
- Department of Ecology and Evolutionary Biology, University of California, Irvine, California, USA
| | - Adam C. Martiny
- Department of Ecology and Evolutionary Biology, University of California, Irvine, California, USA
- Department of Earth System Science, University of California, Irvine, California, USA
| |
Collapse
|
31
|
Tandar ST, Aulin LBS, Leemkuil EMJ, Liakopoulos A, van Hasselt JGC. Semi-mechanistic modeling of resistance development to β-lactam and β-lactamase-inhibitor combinations. J Pharmacokinet Pharmacodyn 2024; 51:199-211. [PMID: 38008877 DOI: 10.1007/s10928-023-09895-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/27/2023] [Indexed: 11/28/2023]
Abstract
The use of β-lactam (BL) and β-lactamase inhibitor (BLI) combinations, such as piperacillin-tazobactam (PIP-TAZ) is an effective strategy to combat infections by extended-spectrum β-lactamase-producing bacteria. However, in Gram-negative bacteria, resistance (both mutational and adaptive) to BL-BLI combination can still develop through multiple mechanisms. These mechanisms may include increased β-lactamase activity, reduced drug influx, and increased drug efflux. Understanding the relative contribution of these mechanisms during resistance development helps identify the most impactful mechanism to target in designing a treatment to counter BL-BLI resistance. This study used semi-mechanistic mathematical modeling in combination with antibiotic sensitivity assays to assess the potential impact of different resistance mechanisms during the development of PIP-TAZ resistance in a Klebsiella pneumoniae isolate expressing CTX-M-15 and SHV-1 β-lactamases. The mathematical models were used to evaluate the potential impact of several cellular changes as a sole mediator of PIP-TAZ resistance. Our semi-mechanistic model identified 2 out of the 13 inspected mechanisms as key resistance mechanisms that may independently support the observed magnitude of PIP-TAZ resistance, namely porin loss and efflux pump up-regulation. Simulation using the resulting models also suggested the possible adjustment of PIP-TAZ dose outside its commonly used 8:1 dosing ratio. The current study demonstrated how theory-based mechanistic models informed by experimental data can be used to support hypothesis generation regarding potential resistance mechanisms, which may guide subsequent experimental studies.
Collapse
Affiliation(s)
- Sebastian T Tandar
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Linda B S Aulin
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department Clinical Pharmacy and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Eva M J Leemkuil
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Apostolos Liakopoulos
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Biology, Utrecht University, Utrecht, The Netherlands
| | - J G Coen van Hasselt
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
32
|
Laborda P, Molin S, Johansen HK, Martínez JL, Hernando-Amado S. Role of bacterial multidrug efflux pumps during infection. World J Microbiol Biotechnol 2024; 40:226. [PMID: 38822187 DOI: 10.1007/s11274-024-04042-7] [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: 04/03/2024] [Accepted: 05/29/2024] [Indexed: 06/02/2024]
Abstract
Multidrug efflux pumps are protein complexes located in the cell envelope that enable bacteria to expel, not only antibiotics, but also a wide array of molecules relevant for infection. Hence, they are important players in microbial pathogenesis. On the one hand, efflux pumps can extrude exogenous compounds, including host-produced antimicrobial molecules. Through this extrusion, pathogens can resist antimicrobial agents and evade host defenses. On the other hand, efflux pumps also have a role in the extrusion of endogenous compounds, such as bacterial intercommunication signaling molecules, virulence factors or metabolites. Therefore, efflux pumps are involved in the modulation of bacterial behavior and virulence, as well as in the maintenance of the bacterial homeostasis under different stresses found within the host. This review delves into the multifaceted roles that efflux pumps have, shedding light on their impact on bacterial virulence and their contribution to bacterial infection. These observations suggest that strategies targeting bacterial efflux pumps could both reinvigorate the efficacy of existing antibiotics and modulate the bacterial pathogenicity to the host. Thus, a comprehensive understanding of bacterial efflux pumps can be pivotal for the development of new effective strategies for the management of infectious diseases.
Collapse
Affiliation(s)
- Pablo Laborda
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, 9301, Denmark.
| | - Søren Molin
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Helle Krogh Johansen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, 9301, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
33
|
Ma K, Chinelo OR, Gu M, Kong F, Jiang Y, Wang H, Xue T. Role of ArcA in the regulation of antibiotic sensitivity in avian pathogenic Escherichia coli. Poult Sci 2024; 103:103686. [PMID: 38574461 PMCID: PMC11004985 DOI: 10.1016/j.psj.2024.103686] [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: 02/06/2024] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024] Open
Abstract
Avian pathogenic Escherichia coli (APEC) is one of the common extraintestinal infectious disease pathogens in chickens, geese, and other birds, inducing serious impediments to the development of the poultry industry. Hence, investigating how bacteria regulate themselves amidst different challenging conditions is immense essential in prevention and treatment for bacterial pathogen infections. The ArcA regulatory factor has been reported to regulate oxygen availability in strains, but its role in regulation of antibiotics resistance in APEC is unclear. This study delved into understanding how ArcA regulates antibiotic resistance in APEC. An E. coli APEC40 arcA knockout strain was constructed, and the regulatory mechanism of arcA on APEC antibiotic susceptibility was identified by drug sensitivity test, colony counting assay, real-time quantitative PCR, β-galactosidase assays and electrophoretic mobility shift assay (EMSA). The results showed that ArcA directly binds to the promoter region of the outer membrane protein OmpC/OmpW and regulates bacterial susceptibility to kanamycin and penicillin G. At the same time, the double knockout of ompW and ompW/arcA resulted in an increase in resistance to kanamycin compared to the deletion of the arcA gene. This outcome provided experimental proof suggesting that the outer membrane protein OmpW could serve as a crucial pathway for the ingress of kanamycin into cells. These results confirmed the important regulatory role of ArcA transcription factors under APEC antibiotic stress.
Collapse
Affiliation(s)
- Kai Ma
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Okoro Ruth Chinelo
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Mantian Gu
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Fanwenqing Kong
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ying Jiang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Hui Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Ting Xue
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
34
|
Villalba de la Peña M, Kronholm I. Antimicrobial resistance in the wild: Insights from epigenetics. Evol Appl 2024; 17:e13707. [PMID: 38817397 PMCID: PMC11134192 DOI: 10.1111/eva.13707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 03/04/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024] Open
Abstract
Spreading of bacterial and fungal strains that are resistant to antimicrobials poses a serious threat to the well-being of humans, animals, and plants. Antimicrobial resistance has been mainly investigated in clinical settings. However, throughout their evolutionary history microorganisms in the wild have encountered antimicrobial substances, forcing them to evolve strategies to combat antimicrobial action. It is well known that many of these strategies are based on genetic mechanisms, but these do not fully explain important aspects of the antimicrobial response such as the rapid development of resistance, reversible phenotypes, and hetero-resistance. Consequently, attention has turned toward epigenetic pathways that may offer additional insights into antimicrobial mechanisms. The aim of this review is to explore the epigenetic mechanisms that confer antimicrobial resistance, focusing on those that might be relevant for resistance in the wild. First, we examine the presence of antimicrobials in natural settings. Then we describe the documented epigenetic mechanisms in bacteria and fungi associated with antimicrobial resistance and discuss innovative epigenetic editing techniques to establish causality in this context. Finally, we discuss the relevance of these epigenetic mechanisms on the evolutionary dynamics of antimicrobial resistance in the wild, emphasizing the critical role of priming in the adaptation process. We underscore the necessity of incorporating non-genetic mechanisms into our understanding of antimicrobial resistance evolution. These mechanisms offer invaluable insights into the dynamics of antimicrobial adaptation within natural ecosystems.
Collapse
Affiliation(s)
| | - Ilkka Kronholm
- Department of Biological and Environmental ScienceUniversity of JyväskyläJyväskyläFinland
| |
Collapse
|
35
|
Branda F, Scarpa F. Implications of Artificial Intelligence in Addressing Antimicrobial Resistance: Innovations, Global Challenges, and Healthcare's Future. Antibiotics (Basel) 2024; 13:502. [PMID: 38927169 PMCID: PMC11200959 DOI: 10.3390/antibiotics13060502] [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: 04/30/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Antibiotic resistance poses a significant threat to global public health due to complex interactions between bacterial genetic factors and external influences such as antibiotic misuse. Artificial intelligence (AI) offers innovative strategies to address this crisis. For example, AI can analyze genomic data to detect resistance markers early on, enabling early interventions. In addition, AI-powered decision support systems can optimize antibiotic use by recommending the most effective treatments based on patient data and local resistance patterns. AI can accelerate drug discovery by predicting the efficacy of new compounds and identifying potential antibacterial agents. Although progress has been made, challenges persist, including data quality, model interpretability, and real-world implementation. A multidisciplinary approach that integrates AI with other emerging technologies, such as synthetic biology and nanomedicine, could pave the way for effective prevention and mitigation of antimicrobial resistance, preserving the efficacy of antibiotics for future generations.
Collapse
Affiliation(s)
- Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
36
|
Singh G, Rana A, Smriti. Decoding antimicrobial resistance: unraveling molecular mechanisms and targeted strategies. Arch Microbiol 2024; 206:280. [PMID: 38805035 DOI: 10.1007/s00203-024-03998-2] [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: 03/31/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
Antimicrobial resistance poses a significant global health threat, necessitating innovative approaches for combatting it. This review explores various mechanisms of antimicrobial resistance observed in various strains of bacteria. We examine various strategies, including antimicrobial peptides (AMPs), novel antimicrobial materials, drug delivery systems, vaccines, antibody therapies, and non-traditional antibiotic treatments. Through a comprehensive literature review, the efficacy and challenges of these strategies are evaluated. Findings reveal the potential of AMPs in combating resistance due to their unique mechanisms and lower propensity for resistance development. Additionally, novel drug delivery systems, such as nanoparticles, show promise in enhancing antibiotic efficacy and overcoming resistance mechanisms. Vaccines and antibody therapies offer preventive measures, although challenges exist in their development. Non-traditional antibiotic treatments, including CRISPR-Cas systems, present alternative approaches to combat resistance. Overall, this review underscores the importance of multifaceted strategies and coordinated global efforts to address antimicrobial resistance effectively.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Biosciences (UIBT), Chandigarh University, Punjab, 140413, India
| | - Anita Rana
- Department of Biosciences (UIBT), Chandigarh University, Punjab, 140413, India.
| | - Smriti
- Department of Biosciences (UIBT), Chandigarh University, Punjab, 140413, India
| |
Collapse
|
37
|
Wang C, Ma Q, Zhang J, Meng N, Xu D. Co-culture of benzalkonium chloride promotes the biofilm formation and decreases the antibiotic susceptibility of a Pseudomonas aeruginosa strain. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2024; 26:858-869. [PMID: 38687259 DOI: 10.1039/d4em00035h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Benzalkonium chloride (BAC) is a disinfectant with broad-spectrum antibacterial properties, yet despite its widespread use and detection in the environment, the effects of BAC exposure on microorganisms remain poorly documented. Herein, the impacts of BAC on a Pseudomonas aeruginosa strain Jade-X were systematically investigated. The results demonstrated that the minimum inhibitory concentration (MIC) of BAC against strain Jade-X was 64 mg L-1. Exposure to BAC concentrations of 8, 16, 32, and 64 mg L-1 significantly augmented biofilm formation by 2.03-, 2.43-, 2.96-, and 2.56-fold respectively. The swimming and twitching abilities, along with the virulence factor production, were inhibited. Consistently, quantitative reverse transcription PCR assays revealed significant downregulation of genes related to flagellate- and pili-mediated motilities (flgD, flgE, pilB, pilQ, and motB), as well as phzA and phzB genes involved in pyocyanin production. The results of disk diffusion and MIC assays demonstrated that BAC decreased the antibiotic susceptibility of ciprofloxacin, levofloxacin, norfloxacin, and tetracycline. Conversely, an opposite trend was observed for polymyxin B and ceftriaxone. Genomic analysis revealed that strain Jade-X harbored eleven resistance-nodulation-cell division efflux pumps, with mexCD-oprJ exhibiting significant upregulation while mexEF-oprN and mexGHI-opmD were downregulated. In addition, the quorum sensing-related regulators LasR and RhlR were also suppressed, implying that BAC might modulate the physiological and biochemical behaviors of strain Jade-X by attenuating the quorum sensing system. This study enhances our understanding of interactions between BAC and P. aeruginosa, providing valuable insights to guide the regulation and rational use of BAC.
Collapse
Affiliation(s)
- Caihong Wang
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| | - Qiao Ma
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| | - Jiaxin Zhang
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| | - Nan Meng
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| | - Dan Xu
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| |
Collapse
|
38
|
Wang C, Yang J, Xu Z, Lv L, Chen S, Hong M, Liu JH. Promoter regulatory mode evolution enhances the high multidrug resistance of tmexCD1-toprJ1. mBio 2024; 15:e0021824. [PMID: 38564664 PMCID: PMC11077950 DOI: 10.1128/mbio.00218-24] [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: 01/24/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
Antibiotic resistance could rapidly emerge from acquiring the mobile antibiotic resistance genes, which are commonly evolved from an intrinsic gene. The emergence of the plasmid-borne mobilized efflux pump gene cluster tmexCD1-toprJ1 renders the last-resort antibiotic tigecycline ineffective, although its evolutionary mechanism remains unclear. In this study, we investigate the regulatory mechanisms of the progenitor NfxB-MexCD-OprJ, a chromosomally encoded operon that does not mediate antibiotic resistance in the wild-type version, and its homologs, TNfxB1-TMexCD1-TOprJ1 mediating high-level tigecycline resistance, and TNfxB3-TMexCD3-TOprJ1. Mechanistic studies demonstrated that in nfxB-mexCD-oprJ, MexCD expression was under a weaker promoter, PmexC and inhibited by a strong repressor NfxB. For tmexCD1-toprJ1, TMexCD1 was highly expressed owing to the presence of a strong promoter, PtmexC1, and an inactive suppressor, TNfxB1, with a T39R mutation that rendered it unable to bind to promoter DNA. In tnfxB3-tmexCD3-toprJ1b, TMexCD3 expression was intermediate because of the local regulator TNfxB3, which binds to two inverted repeat sequences of PtmexC. Additionally, TNfxB3 exhibited lower protein expression and weaker DNA binding affinity than its ancestor NfxB, together with their promoter activities difference explaining the different expression levels of tmexCD-toprJ homologs. Distinct fitness burdens on these homologs-carrying bacteria were observed due to the corresponding expression level, which might be associated with their global prevalence. In summary, our data depict the mechanisms underlying the evolution and dissemination of an important mobile antibiotic resistance gene from an intrinsic chromosomal gene.IMPORTANCEAs antibiotic resistance seriously challenges global health, tigecycline is one of the few effective drugs in the pipeline against infections caused by multidrug-resistant pathogens. Our previous work identified a novel tigecycline resistance efflux pump gene cluster tmexCD1-toprJ1 in animals and humans, together with its various variants, a rising clinical concern. Herein, this study focused on how the local regulation modes of tmexCD1-toprJ1 evolved to a highly expressed efflux pump. Through comparative analysis between three tnfxB-tmexCD-toprJ homologs and their progenitor nfxB-mexCD-oprJ, modes, we demonstrated the evolutionary dynamics from a chromosomal silent gene to an active state. We found the de-repression of the local regulator and an increase of the promoter activity work together to promote a high production of drug efflux machines and enhance multidrug resistance. Our findings revealed that TMexCD1-TOprJ1 adopts a distinct evolutionary path to achieve higher multidrug resistance, urgently needing tight surveillance.
Collapse
Affiliation(s)
- Chengzhen Wang
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis of Ministry of Agricultural and Rural Affairs, National Risk Assessment Laboratory for Antimicrobial Resistant of Microorganisms in Animals, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, Guangdong, China
| | - Jun Yang
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis of Ministry of Agricultural and Rural Affairs, National Risk Assessment Laboratory for Antimicrobial Resistant of Microorganisms in Animals, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, Guangdong, China
| | - Zeling Xu
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, China
| | - Luchao Lv
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis of Ministry of Agricultural and Rural Affairs, National Risk Assessment Laboratory for Antimicrobial Resistant of Microorganisms in Animals, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, Guangdong, China
| | - Sheng Chen
- State Key Lab of Chemical Biology and Drug Discovery and the Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Mei Hong
- College of Life Sciences, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, South China Agricultural University, Guangzhou, China
| | - Jian-Hua Liu
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis of Ministry of Agricultural and Rural Affairs, National Risk Assessment Laboratory for Antimicrobial Resistant of Microorganisms in Animals, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
Omran BA, Tseng BS, Baek KH. Nanocomposites against Pseudomonas aeruginosa biofilms: Recent advances, challenges, and future prospects. Microbiol Res 2024; 282:127656. [PMID: 38432017 DOI: 10.1016/j.micres.2024.127656] [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: 10/26/2023] [Revised: 01/10/2024] [Accepted: 02/17/2024] [Indexed: 03/05/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic bacterial pathogen that causes life-threatening and persistent infections in immunocompromised patients. It is the culprit behind a variety of hospital-acquired infections owing to its multiple tolerance mechanisms against antibiotics and disinfectants. Biofilms are sessile microbial aggregates that are formed as a result of the cooperation and competition between microbial cells encased in a self-produced matrix comprised of extracellular polymeric constituents that trigger surface adhesion and microbial aggregation. Bacteria in biofilms exhibit unique features that are quite different from planktonic bacteria, such as high resistance to antibacterial agents and host immunity. Biofilms of P. aeruginosa are difficult to eradicate due to intrinsic, acquired, and adaptive resistance mechanisms. Consequently, innovative approaches to combat biofilms are the focus of the current research. Nanocomposites, composed of two or more different types of nanoparticles, have diverse therapeutic applications owing to their unique physicochemical properties. They are emerging multifunctional nanoformulations that combine the desired features of the different elements to obtain the highest functionality. This review assesses the recent advances of nanocomposites, including metal-, metal oxide-, polymer-, carbon-, hydrogel/cryogel-, and metal organic framework-based nanocomposites for the eradication of P. aeruginosa biofilms. The characteristics and virulence mechanisms of P. aeruginosa biofilms, as well as their devastating impact and economic burden are discussed. Future research addressing the potential use of nanocomposites as innovative anti-biofilm agents is emphasized. Utilization of nanocomposites safely and effectively should be further strengthened to confirm the safety aspects of their application.
Collapse
Affiliation(s)
- Basma A Omran
- Department of Biotechnology, Yeungnam University, Gyeongbuk, Gyeongsan 38541, Republic of Korea; Department of Processes Design & Development, Egyptian Petroleum Research Institute (EPRI), PO 11727, Nasr City, Cairo, Egypt
| | - Boo Shan Tseng
- School of Life Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA.
| | - Kwang-Hyun Baek
- Department of Biotechnology, Yeungnam University, Gyeongbuk, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
40
|
Li H, Xu H. Mechanisms of bacterial resistance to environmental silver and antimicrobial strategies for silver: A review. ENVIRONMENTAL RESEARCH 2024; 248:118313. [PMID: 38280527 DOI: 10.1016/j.envres.2024.118313] [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: 12/14/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
The good antimicrobial properties of silver make it widely used in food, medicine, and environmental applications. However, the release and accumulation of silver-based antimicrobial agents in the environment is increasing with the extensive use of silver-based antimicrobials, and the prevalence of silver-resistant bacteria is increasing. To prevent the emergence of superbugs, it is necessary to exercise rational and strict control over drug use. The mechanism of bacterial resistance to silver has not been fully elucidated, and this article provides a review of the progress of research on the mechanism of bacterial resistance to silver. The results indicate that bacterial resistance to silver can occur through inducing silver particles aggregation and Ag+ reduction, inhibiting silver contact with and entry into cells, efflux of silver particles and Ag+ in cells, and activation of damage repair mechanisms. We propose that the bacterial mechanism of silver resistance involves a combination of interrelated systems. Finally, we discuss how this information can be used to develop the next generation of silver-based antimicrobials and antimicrobial therapies. And some antimicrobial strategies are proposed such as the "Trojan Horse" - camouflage, using efflux pump inhibitors to reduce silver efflux, working with "minesweeper", immobilization of silver particles.
Collapse
Affiliation(s)
- Hui Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
41
|
Wu W, Huang J, Xu Z. Antibiotic influx and efflux in Pseudomonas aeruginosa: Regulation and therapeutic implications. Microb Biotechnol 2024; 17:e14487. [PMID: 38801351 PMCID: PMC11129675 DOI: 10.1111/1751-7915.14487] [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: 03/14/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
Pseudomonas aeruginosa is a notorious multidrug-resistant pathogen that poses a serious and growing threat to the worldwide public health. The expression of resistance determinants is exquisitely modulated by the abundant regulatory proteins and the intricate signal sensing and transduction systems in this pathogen. Downregulation of antibiotic influx porin proteins and upregulation of antibiotic efflux pump systems owing to mutational changes in their regulators or the presence of distinct inducing molecular signals represent two of the most efficient mechanisms that restrict intracellular antibiotic accumulation and enable P. aeruginosa to resist multiple antibiotics. Treatment of P. aeruginosa infections is extremely challenging due to the highly inducible mechanism of antibiotic resistance. This review comprehensively summarizes the regulatory networks of the major porin proteins (OprD and OprH) and efflux pumps (MexAB-OprM, MexCD-OprJ, MexEF-OprN, and MexXY) that play critical roles in antibiotic influx and efflux in P. aeruginosa. It also discusses promising therapeutic approaches using safe and efficient adjuvants to enhance the efficacy of conventional antibiotics to combat multidrug-resistant P. aeruginosa by controlling the expression levels of porins and efflux pumps. This review not only highlights the complexity of the regulatory network that induces antibiotic resistance in P. aeruginosa but also provides important therapeutic implications in targeting the inducible mechanism of resistance.
Collapse
Affiliation(s)
- Weiyan Wu
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research CentreSouth China Agricultural UniversityGuangzhouChina
| | - Jiahui Huang
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research CentreSouth China Agricultural UniversityGuangzhouChina
| | - Zeling Xu
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research CentreSouth China Agricultural UniversityGuangzhouChina
| |
Collapse
|
42
|
Manrique PD, Leus IV, López CA, Mehla J, Malloci G, Gervasoni S, Vargiu AV, Kinthada RK, Herndon L, Hengartner NW, Walker JK, Rybenkov VV, Ruggerone P, Zgurskaya HI, Gnanakaran S. Predicting permeation of compounds across the outer membrane of P. aeruginosa using molecular descriptors. Commun Chem 2024; 7:84. [PMID: 38609430 PMCID: PMC11015012 DOI: 10.1038/s42004-024-01161-y] [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: 10/04/2023] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
The ability Gram-negative pathogens have at adapting and protecting themselves against antibiotics has increasingly become a public health threat. Data-driven models identifying molecular properties that correlate with outer membrane (OM) permeation and growth inhibition while avoiding efflux could guide the discovery of novel classes of antibiotics. Here we evaluate 174 molecular descriptors in 1260 antimicrobial compounds and study their correlations with antibacterial activity in Gram-negative Pseudomonas aeruginosa. The descriptors are derived from traditional approaches quantifying the compounds' intrinsic physicochemical properties, together with, bacterium-specific from ensemble docking of compounds targeting specific MexB binding pockets, and all-atom molecular dynamics simulations in different subregions of the OM model. Using these descriptors and the measured inhibitory concentrations, we design a statistical protocol to identify predictors of OM permeation/inhibition. We find consistent rules across most of our data highlighting the role of the interaction between the compounds and the OM. An implementation of the rules uncovered in our study is shown, and it demonstrates the accuracy of our approach in a set of previously unseen compounds. Our analysis sheds new light on the key properties drug candidates need to effectively permeate/inhibit P. aeruginosa, and opens the gate to similar data-driven studies in other Gram-negative pathogens.
Collapse
Affiliation(s)
- Pedro D Manrique
- Physics Department, George Washington University, Washington, 20052, DC, USA.
| | - Inga V Leus
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, 73019, OK, USA
| | - César A López
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, 87545, NM, USA
| | - Jitender Mehla
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, 73019, OK, USA
| | - Giuliano Malloci
- Department of Physics, University of Cagliari, Monserrato, 20052, CA, Italy
| | - Silvia Gervasoni
- Department of Physics, University of Cagliari, Monserrato, 20052, CA, Italy
| | - Attilio V Vargiu
- Department of Physics, University of Cagliari, Monserrato, 20052, CA, Italy
| | - Rama K Kinthada
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, 63103, MO, USA
| | - Liam Herndon
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, 87545, NM, USA
| | - Nicolas W Hengartner
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, 87545, NM, USA
| | - John K Walker
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, 63103, MO, USA
| | - Valentin V Rybenkov
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, 73019, OK, USA
| | - Paolo Ruggerone
- Department of Physics, University of Cagliari, Monserrato, 20052, CA, Italy
| | - Helen I Zgurskaya
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, 73019, OK, USA
| | - S Gnanakaran
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, 87545, NM, USA.
| |
Collapse
|
43
|
Stevanovic M, Teuber Carvalho JP, Bittihn P, Schultz D. Dynamical model of antibiotic responses linking expression of resistance genes to metabolism explains emergence of heterogeneity during drug exposures. Phys Biol 2024; 21:036002. [PMID: 38412523 PMCID: PMC10988634 DOI: 10.1088/1478-3975/ad2d64] [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: 09/14/2023] [Revised: 01/25/2024] [Accepted: 02/27/2024] [Indexed: 02/29/2024]
Abstract
Antibiotic responses in bacteria are highly dynamic and heterogeneous, with sudden exposure of bacterial colonies to high drug doses resulting in the coexistence of recovered and arrested cells. The dynamics of the response is determined by regulatory circuits controlling the expression of resistance genes, which are in turn modulated by the drug's action on cell growth and metabolism. Despite advances in understanding gene regulation at the molecular level, we still lack a framework to describe how feedback mechanisms resulting from the interdependence between expression of resistance and cell metabolism can amplify naturally occurring noise and create heterogeneity at the population level. To understand how this interplay affects cell survival upon exposure, we constructed a mathematical model of the dynamics of antibiotic responses that links metabolism and regulation of gene expression, based on the tetracycline resistancetetoperon inE. coli. We use this model to interpret measurements of growth and expression of resistance in microfluidic experiments, both in single cells and in biofilms. We also implemented a stochastic model of the drug response, to show that exposure to high drug levels results in large variations of recovery times and heterogeneity at the population level. We show that stochasticity is important to determine how nutrient quality affects cell survival during exposure to high drug concentrations. A quantitative description of how microbes respond to antibiotics in dynamical environments is crucial to understand population-level behaviors such as biofilms and pathogenesis.
Collapse
Affiliation(s)
- Mirjana Stevanovic
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States of America
| | - João Pedro Teuber Carvalho
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States of America
| | - Philip Bittihn
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| | - Daniel Schultz
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States of America
| |
Collapse
|
44
|
Summer M, Ali S, Tahir HM, Abaidullah R, Fiaz U, Mumtaz S, Fiaz H, Hassan A, Mughal TA, Farooq MA. Mode of Action of Biogenic Silver, Zinc, Copper, Titanium and Cobalt Nanoparticles Against Antibiotics Resistant Pathogens. J Inorg Organomet Polym Mater 2024; 34:1417-1451. [DOI: 10.1007/s10904-023-02935-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/02/2023] [Indexed: 08/04/2024]
|
45
|
Buisson Y. [A multi-resistant bacterium before the era of antibiotics: the agent of melioidosis]. C R Biol 2024; 346:17-21. [PMID: 37655946 DOI: 10.5802/crbiol.109] [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: 01/11/2023] [Accepted: 01/25/2023] [Indexed: 09/02/2023]
Abstract
Melioidosis is an infectious, tropical and emerging disease, due to a bacterium of the hydrotelluric environment, Burkholderia pseudomallei, which is considered as a potential biological weapon because of its exceptional resistance and virulence capacities. Its worldwide spread, outside the original endemic foci of Southeast Asia and Australia, is favoured by global warming and the diabetes mellitus pandemic, which is the main predisposing factor.In humans, melioidosis is an opportunistic infection, following professional (rice farmers, soldiers) or accidental contamination, by inhalation or inoculation. B. pseudomallei is a facultative intracellular bacterium that can overcome host immune defences, induce acute, subacute, or chronic invasive infection, or remain latent for years. The acute infection is polymorphic, bacteraemic in more than 50% of cases, frequently complicated by shock, and revealed by visceral abscesses, most often pulmonary. It is fatal in 20 to 50% of cases, the prognosis depending on the delay before the establishment of effective first-line antibiotic therapy, using ceftazidime or carbapenems, and therefore on the speed of bacteriological diagnosis.B. pseudomallei is a saprophytic bacterium, resident of the rhizosphere where it has developed and accumulated capacities to overcome environmental stresses and competition with organisms living in such ecosystem. These adaptation mechanisms are also the virulence factors that make melioidosis serious, in particular the efflux pumps that are the main support for its multi-resistance to antibiotics.
Collapse
|
46
|
Fekri Kohan S, Nouhi Kararoudi A, Bazgosha M, Adelifar S, Hafezolghorani Esfahani A, Ghaderi Barmi F, Kouchakinejad R, Barzegari E, Shahriarinour M, Ranji N. Determining the potential targets of silybin by molecular docking and its antibacterial functions on efflux pumps and porins in uropathogenic E. coli. Int Microbiol 2024:10.1007/s10123-024-00488-9. [PMID: 38363383 DOI: 10.1007/s10123-024-00488-9] [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: 12/03/2023] [Revised: 01/11/2024] [Accepted: 01/29/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND One of the causes of antibiotic resistance is the reduced accumulation of antibiotics in bacterial cells through pumping out the drugs. Silybin, a key component of the Silybum marianum plant, exhibits various beneficial properties, including anti-bacterial, anti-inflammatory, antioxidant, and hepatoprotective effects. METHODS AND RESULTS Clinical isolates of E. coli were procured from 17 Shahrivar Children's Hospital in Rasht, Guilan, located in northern Iran. Their susceptibility to six antibiotics was assessed using disc diffusion and broth dilution (MIC) methods. The antibacterial effects of silybin-loaded polymersome nanoparticles (SPNs) were investigated with broth dilution (MIC) and biofilm assays. Molecular docking was utilized to evaluate silybin's (the antibacterial component) binding affinity to efflux pumps, porins, and their regulatory elements. Additionally, qRT-PCR analysis explored the expression patterns of acrA, acrB, tolC, ompC, and ompF genes in both SPNs (sub-MIC) and ciprofloxacin (sub-MIC)-treated and untreated E. coli isolates. The combined use of SPNs and ciprofloxacin exhibited a notable reduction in bacterial growth and biofilm formation, in ciprofloxacin-resistant isolates. The study identified eight overlapping binding sites of the AcrABZ-TolC efflux pump in association with silybin, demonstrating a binding affinity ranging from -7.688 to -10.33 Kcal/mol. Furthermore, the qRT-PCR analysis showed that silybin upregulated AcrAB-TolC efflux pump genes and downregulated ompC and ompF porin genes in combination with ciprofloxacin in transcriptional level in uropathogenic E. coli. CONCLUSIONS Silybin, a safe herbal compound, exhibits potential in inhibiting antibiotic resistance within bacterial isolates, potentially through the regulation of gene expression and plausible binding to target proteins.
Collapse
Affiliation(s)
- Shirin Fekri Kohan
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, P.O. Box: 41335-3516, Rasht, Iran
| | - Alireza Nouhi Kararoudi
- Department of Biology, Faculty of Sciences, Lahijan Branch, Islamic Azad University, Rasht, Iran
| | - Maryam Bazgosha
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, P.O. Box: 41335-3516, Rasht, Iran
| | - Somayeh Adelifar
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, P.O. Box: 41335-3516, Rasht, Iran
| | - Arman Hafezolghorani Esfahani
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, P.O. Box: 41335-3516, Rasht, Iran
| | - Fatemeh Ghaderi Barmi
- Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota, USA
| | - Reyhaneh Kouchakinejad
- Department of Chemistry, Faculty of Sciences, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Ebrahim Barzegari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahdi Shahriarinour
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, P.O. Box: 41335-3516, Rasht, Iran.
| | - Najmeh Ranji
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, P.O. Box: 41335-3516, Rasht, Iran.
| |
Collapse
|
47
|
Padhi AK, Maurya S. Uncovering the secrets of resistance: An introduction to computational methods in infectious disease research. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 139:173-220. [PMID: 38448135 DOI: 10.1016/bs.apcsb.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Antimicrobial resistance (AMR) is a growing global concern with significant implications for infectious disease control and therapeutics development. This chapter presents a comprehensive overview of computational methods in the study of AMR. We explore the prevalence and statistics of AMR, underscoring its alarming impact on public health. The role of AMR in infectious disease outbreaks and its impact on therapeutics development are discussed, emphasizing the need for novel strategies. Resistance mutations are pivotal in AMR, enabling pathogens to evade antimicrobial treatments. We delve into their importance and contribution to the spread of AMR. Experimental methods for quantitatively evaluating resistance mutations are described, along with their limitations. To address these challenges, computational methods provide promising solutions. We highlight the advantages of computational approaches, including rapid analysis of large datasets and prediction of resistance profiles. A comprehensive overview of computational methods for studying AMR is presented, encompassing genomics, proteomics, structural bioinformatics, network analysis, and machine learning algorithms. The strengths and limitations of each method are briefly outlined. Additionally, we introduce ResScan-design, our own computational method, which employs a protein (re)design protocol to identify potential resistance mutations and adaptation signatures in pathogens. Case studies are discussed to showcase the application of ResScan in elucidating hotspot residues, understanding underlying mechanisms, and guiding the design of effective therapies. In conclusion, we emphasize the value of computational methods in understanding and combating AMR. Integration of experimental and computational approaches can expedite the discovery of innovative antimicrobial treatments and mitigate the threat posed by AMR.
Collapse
Affiliation(s)
- Aditya K Padhi
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India.
| | - Shweata Maurya
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| |
Collapse
|
48
|
Bo L, Sun H, Li YD, Zhu J, Wurpel JND, Lin H, Chen ZS. Combating antimicrobial resistance: the silent war. Front Pharmacol 2024; 15:1347750. [PMID: 38420197 PMCID: PMC10899355 DOI: 10.3389/fphar.2024.1347750] [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: 12/01/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
Once hailed as miraculous solutions, antibiotics no longer hold that status. The excessive use of antibiotics across human healthcare, agriculture, and animal husbandry has given rise to a broad array of multidrug-resistant (MDR) pathogens, posing formidable treatment challenges. Antimicrobial resistance (AMR) has evolved into a pressing global health crisis, linked to elevated mortality rates in the modern medical era. Additionally, the absence of effective antibiotics introduces substantial risks to medical and surgical procedures. The dwindling interest of pharmaceutical industries in developing new antibiotics against MDR pathogens has aggravated the scarcity issue, resulting in an exceedingly limited pipeline of new antibiotics. Given these circumstances, the imperative to devise novel strategies to combat perilous MDR pathogens has become paramount. Contemporary research has unveiled several promising avenues for addressing this challenge. The article provides a comprehensive overview of these innovative therapeutic approaches, highlighting their mechanisms of action, benefits, and drawbacks.
Collapse
Affiliation(s)
- Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Haidong Sun
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yi-Dong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Jonathan Zhu
- Carle Place Middle and High School, Carle Place, NY, United States
| | - John N. D. Wurpel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Hanli Lin
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhe-Sheng Chen
- Institute for Biotechnology, St. John’s University, Queens, NY, United States
| |
Collapse
|
49
|
Li P, Zhang S, Wang J, Al-Shamiri MM, Luo K, Liu S, Mi P, Wu X, Liu H, Tian H, Han B, Lei J, Han S, Han L. The role of type VI secretion system genes in antibiotic resistance and virulence in Acinetobacter baumannii clinical isolates. Front Cell Infect Microbiol 2024; 14:1297818. [PMID: 38384301 PMCID: PMC10879597 DOI: 10.3389/fcimb.2024.1297818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024] Open
Abstract
Introduction The type VI secretion system (T6SS) is a crucial virulence factor in the nosocomial pathogen Acinetobacter baumannii. However, its association with drug resistance is less well known. Notably, the roles that different T6SS components play in the process of antimicrobial resistance, as well as in virulence, have not been systematically revealed. Methods The importance of three representative T6SS core genes involved in the drug resistance and virulence of A. baumannii, namely, tssB, tssD (hcp), and tssM was elucidated. Results A higher ratio of the three core genes was detected in drug-resistant strains than in susceptible strains among our 114 A. baumannii clinical isolates. Upon deletion of tssB in AB795639, increased antimicrobial resistance to cefuroxime and ceftriaxone was observed, alongside reduced resistance to gentamicin. The ΔtssD mutant showed decreased resistance to ciprofloxacin, norfloxacin, ofloxacin, tetracycline, and doxycycline, but increased resistance to tobramycin and streptomycin. The tssM-lacking mutant showed an increased sensitivity to ofloxacin, polymyxin B, and furazolidone. In addition, a significant reduction in biofilm formation was observed only with the ΔtssM mutant. Moreover, the ΔtssM strain, followed by the ΔtssD mutant, showed decreased survival in human serum, with attenuated competition with Escherichia coli and impaired lethality in Galleria mellonella. Discussion The above results suggest that T6SS plays an important role, participating in the antibiotic resistance of A. baumannii, especially in terms of intrinsic resistance. Meanwhile, tssM and tssD contribute to bacterial virulence to a greater degree, with tssM being associated with greater importance.
Collapse
Affiliation(s)
- Pu Li
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Sirui Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Jingdan Wang
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Mona Mohamed Al-Shamiri
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Kai Luo
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Shuyan Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Peng Mi
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Department of Laboratory Medicine, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Xiaokang Wu
- Department of Laboratory Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Haiping Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Department of Laboratory Medicine, Xi’an Daxing Hospital, Xi’an, China
| | - Huohuan Tian
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bei Han
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Jin’e Lei
- Department of Laboratory Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shaoshan Han
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lei Han
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| |
Collapse
|
50
|
Telhig S, Pham NP, Ben Said L, Rebuffat S, Ouellette M, Zirah S, Fliss I. Exploring the genetic basis of natural resistance to microcins. Microb Genom 2024; 10:001156. [PMID: 38407259 PMCID: PMC10926693 DOI: 10.1099/mgen.0.001156] [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: 03/29/2023] [Accepted: 11/28/2023] [Indexed: 02/27/2024] Open
Abstract
Enterobacteriaceae produce an arsenal of antimicrobial compounds including microcins, ribosomally produced antimicrobial peptides showing diverse structures and mechanisms of action. Microcins target close relatives of the producing strain to promote its survival. Their narrow spectrum of antibacterial activity makes them a promising alternative to conventional antibiotics, as it should decrease the probability of resistance dissemination and collateral damage to the host's microbiota. To assess the therapeutic potential of microcins, there is a need to understand the mechanisms of resistance to these molecules. In this study, we performed genomic analyses of the resistance to four microcins [microcin C, a nucleotide peptide; microcin J25, a lasso peptide; microcin B17, a linear azol(in)e-containing peptide; and microcin E492, a siderophore peptide] on a collection of 54 Enterobacteriaceae from three species: Escherichia coli, Salmonella enterica and Klebsiella pneumoniae. A gene-targeted analysis revealed that about half of the microcin-resistant strains presented mutations of genes involved in the microcin mechanism of action, especially those involved in their uptake (fhuA, fepA, cirA and ompF). A genome-wide association study did not reveal any significant correlations, yet relevant genetic elements were associated with microcin resistance. These were involved in stress responses, biofilm formation, transport systems and acquisition of immunity genes. Additionally, microcin-resistant strains exhibited several mutations within genes involved in specific metabolic pathways, especially for S. enterica and K. pneumoniae.
Collapse
Affiliation(s)
- Soufiane Telhig
- Food Science Department, Food and Agriculture Faculty, Laval University, Quebec, Canada
- Laboratoire Molécules de Communication et Adaptation des Microorganismes, Muséum national d’Histoire naturelle, Centre national de la Recherche scientifique, Paris, France
| | - Nguyen Phuong Pham
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Laila Ben Said
- Food Science Department, Food and Agriculture Faculty, Laval University, Quebec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec, Canada
| | - Sylvie Rebuffat
- Laboratoire Molécules de Communication et Adaptation des Microorganismes, Muséum national d’Histoire naturelle, Centre national de la Recherche scientifique, Paris, France
| | - Marc Ouellette
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Séverine Zirah
- Laboratoire Molécules de Communication et Adaptation des Microorganismes, Muséum national d’Histoire naturelle, Centre national de la Recherche scientifique, Paris, France
| | - Ismaïl Fliss
- Food Science Department, Food and Agriculture Faculty, Laval University, Quebec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec, Canada
| |
Collapse
|