1
|
Vivekanandan K, Kumar PV, Jaysree R, Rajeshwari T. Exploring molecular mechanisms of drug resistance in bacteria and progressions in CRISPR/Cas9-based genome expurgation solutions. Glob Med Genet 2025; 12:100042. [PMID: 40051841 PMCID: PMC11883354 DOI: 10.1016/j.gmg.2025.100042] [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: 01/15/2025] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Antibiotic resistance in bacteria is a critical global health challenge, driven by molecular mechanisms such as genetic mutations, efflux pumps, enzymatic degradation of antibiotics, target site modifications, and biofilm formation. Horizontal gene transfer (HGT) further accelerates the spread of resistance genes across bacterial populations. These mechanisms contribute to the emergence of multidrug-resistant (MDR) strains, rendering conventional antibiotics ineffective. Recent advancements in CRISPR/Cas9-based genome editing offer innovative solutions to combat drug resistance. CRISPR/Cas9 enables precise targeting of resistance genes, facilitating their deletion or inactivation, and provides a potential method to eliminate resistance-carrying plasmids. Furthermore, phage-delivered CRISPR systems show promise in selectively killing resistant bacteria while leaving susceptible strains unaffected. Despite challenges such as efficient delivery, off-target effects, and potential bacterial resistance to CRISPR itself, ongoing research and technological innovations hold promise for using CRISPR-based antimicrobials to reverse bacterial drug resistance and develop more effective therapies. These abstract highlights the molecular mechanisms underlying bacterial drug resistance and explores how CRISPR/Cas9 technology could revolutionize treatment strategies against resistant pathogens.
Collapse
Affiliation(s)
- K.E. Vivekanandan
- Department of Microbiology, PSG College of Arts and Science, Civil Aerodrome Post, Avinashi Road, Coimbatore, Tamil Nadu 641014, India
| | - P. Vinoth Kumar
- Department of Microbiology, Shri Nehru Maha Vidyalaya, Shri Gambhirmal Bafna Nagar, Malumachampatti, Coimbatore 641050, India
| | - R.C. Jaysree
- Department of Biotechnology, Nehru Arts and Science College, Thirumalayampalayam, Coimbatore 641105, India
| | - T. Rajeshwari
- Department of Biotechnology, Dhanalakshmi Srinivasan College of Arts and Science for Women, Thuraiyur, Perambalur, Tamilnadu 621212, India
| |
Collapse
|
2
|
Chen J, Kuang R, Liu H, Yang L, Li J, Li J, Xia J, Huang Y, Cui M, Zou L, Han X. Emergence of multidrug resistant Escherichia coli coharboring fosA3 and ESBL genes from retail ducks along slaughter line. Int J Food Microbiol 2025; 435:111185. [PMID: 40179607 DOI: 10.1016/j.ijfoodmicro.2025.111185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
The excessive and indiscriminate use of antimicrobial agents in poultry production has increased antimicrobial resistance in E. coli, posing a significant threat to public health. This study investigated the prevalence of multidrug-resistant (MDR) E. coli co-harboring fosA3 and extended-spectrum β-lactamases (ESBLs) genes in large-scale retail duck slaughterhouses in Sichuan Province, China. The antimicrobial susceptibility to 9 categories and 16 types of antimicrobial agents was assessed by using the broth microdilution method. Phylogenetic grouping and pulsed-field gel electrophoresis (PFGE) were used to investigate the phylogenetic groups and genetic characteristics of the isolates. Moreover, whole-genome sequencing (WGS) was performed on 35 representative E. coli isolates. Among 1, 059 samples collected, 895 E. coli were isolated by MacConkey (MAC) agar and eosin methylene blue (EMB) agar and PCR identification, and 150 strains of MDR E. coli co-carrying fosA3 and ESBL genes were finally screened for further analysis. The results indicated that 141 isolates were resistant to fosfomycin and exhibited high resistance to β-lactam antibiotics. All isolates demonstrated MDR patterns, with 148 isolates resistant to six or more classes of antimicrobial agents. The majority (70/150, 46.67 %) belonged to phylogenetic group B1. Based on an 85 % similarity threshold, all isolates were categorized into 40 distinct PFGE types. Genomic analysis revealed 27 different serotypes and 19 sequence types (STs), with O8 (14.29 %, 5/35) and ST155 (22.86 %, 8/35) being the most common. The IncFIB (AP001918) was the most prevalent plasmid replicon type, identified in 74.29 % (26/35) of the isolates. In addition, 49 acquired antimicrobial resistance genes (ARGs) associated with resistance to 11 types of antimicrobial agents were identified and chromosomal mutation of p.S83L in gyrA gene (88.57 %, 31/35) was the most common. Furthermore, 110 virulence factors (VFs) were identified, with those related to iron uptake and storage, adhesion, secretion and endotoxin production being the most prevalent. This study underscored the importance of rational use of antimicrobial agents in poultry farming and provided critical insights into the distribution of ARGs and VFs among retail duck-derived MDR E. coli.
Collapse
Affiliation(s)
- Jialan Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ruyi Kuang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Haotian Liu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lei Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jiarui Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junlin Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jing Xia
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of China, Chengdu, China; Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Yong Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of China, Chengdu, China; Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Min Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of China, Chengdu, China; Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Likou Zou
- College of Resources, Sichuan Agricultural University, Chengdu, China
| | - Xinfeng Han
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of China, Chengdu, China; Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China.
| |
Collapse
|
3
|
Li J, Chang J, Ma J, Zhou W, Yang Y, Wu J, Guan C, Yuan X, Xu L, Yu B, Su F, Ye S, Chen Y, Zhao G, Tang B. Genome-based assessment of antimicrobial resistance of Escherichia coli recovered from diseased swine in eastern China for a 12-year period. mBio 2025:e0065125. [PMID: 40243369 DOI: 10.1128/mbio.00651-25] [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: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
The global rise of antimicrobial resistance (AMR), driven by antibiotic use in healthcare and agriculture, poses a major public health threat. While AMR in clinical settings is well studied, there is a gap in understanding the resistance profiles of Escherichia coli from diseased livestock, particularly regarding zoonotic transmission. This study analyzes 114 E. coli isolates from diseased swine over 12 years, revealing that 99.12% were multidrug-resistant. Resistance was highest for ampicillin and amoxicillin/clavulanic acid (100%), followed by ciprofloxacin (96.49%) and tetracycline (94.74%). Furthermore, 21.05% of isolates were resistant to colistin, and 1.75% to tigecycline. A total of 76 antimicrobial resistance genes (ARGs) were identified, with mcr-1 found in 18.42%, mcr-3 in 4.39%, and tet(X4) in 1.75%. Significant co-occurrence of ARGs and plasmids suggests potential for co-selective dissemination. This study is the first to report enterotoxigenic E. coli (ETEC) strains carrying both mcr-1 and mcr-3 genes. After the 2017 colistin ban in China, mcr-1 detection rates significantly decreased, while florfenicol resistance rates increased in 2018-2021 (94.29%) compared to 2010-2017 (79.55%). This work provides valuable insights into the AMR profiles of E. coli from diseased swine and highlights trends that can inform strategies for monitoring and controlling public health risks associated with zoonotic E. coli transmission.IMPORTANCEThis study highlights the critical role of diseased and deceased swine in the spread of antimicrobial resistance (AMR), providing new insights into the transmission of resistance genes in zoonotic contexts. By analyzing E. coli from diseased swine, we identify key resistance genes such as mcr-1, mcr-3, and tet(X4), which pose significant public health risks, especially regarding last-resort antibiotics like colistin. Moreover, the study identifies novel transmission patterns of mcr genes, including ETEC strains carrying the mcr-3 gene and strains harboring both mcr-1 and mcr-3 genes. The role of plasmids in horizontal gene transfer is also revealed, facilitating rapid AMR spread across species. The long-term persistence of resistant strains highlights the challenges in controlling AMR in livestock. These findings underscore the need for enhanced surveillance and a One Health approach to mitigate AMR risks across animal, human, and environmental health.
Collapse
Affiliation(s)
- Junxing Li
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Jiang Chang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Jiangang Ma
- Xianghu Laboratory, Hangzhou, Zhejiang, China
| | - Wei Zhou
- Zhejiang Provincial Center for Animal Disease Prevention and Control, Hangzhou, China
| | - Yue Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Jing Wu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Chunjiu Guan
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiufang Yuan
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Lihua Xu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Bin Yu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Fei Su
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Shiyi Ye
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Yijie Chen
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Guoping Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
- National Genomics Data Center & Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Biao Tang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Sherry NL, Lee JYH, Giulieri SG, Connor CH, Horan K, Lacey JA, Lane CR, Carter GP, Seemann T, Egli A, Stinear TP, Howden BP. Genomics for antimicrobial resistance-progress and future directions. Antimicrob Agents Chemother 2025:e0108224. [PMID: 40227048 DOI: 10.1128/aac.01082-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025] Open
Abstract
Antimicrobial resistance (AMR) is a critical global public health threat, with bacterial pathogens of primary concern. Pathogen genomics has revolutionized the study of bacterial pathogens and provided deep insights into the mechanisms and dissemination of AMR, with the precision of whole-genome sequencing informing better control strategies. However, generating actionable data from genomic surveillance and diagnostic efforts requires integration at the public health and clinical interface that goes beyond academic efforts to identify resistance mechanisms, undertake post hoc analyses of outbreaks, and share data after research publications. In addition to timely genomics data, consideration also needs to be given to epidemiological sampling frames, analysis, and reporting mechanisms that meet International Organization for Standardization (ISO) standards and generation of reports that are interpretable and actionable for public health and clinical "end-users." Importantly, ensuring all countries have equitable access to data and technology is critical, through timely data sharing following the FAIR principles (findable, accessible, interoperable, and re-usable). In this review, we describe (i) advances in genomic approaches for AMR research and surveillance to understand emergence, evolution, and transmission of AMR and the key requirements to enable this work and (ii) discuss emerging and future applications of genomics at the clinical and public health interface, including barriers to implementation. Harnessing advances in genomics-enhanced AMR research and embedding robust and reproducible workflows within clinical and public health practice promises to maximize the impact of pathogen genomics for AMR globally in the coming decade.
Collapse
Affiliation(s)
- Norelle L Sherry
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Antimicrobial Resistance, Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases and Immunology, Austin Health, Heidelberg, Victoria, Australia
| | - Jean Y H Lee
- Centre for Pathogen Genomics, University of Melbourne, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Monash Health, Clayton, Victoria, Australia
| | - Stefano G Giulieri
- Centre for Pathogen Genomics, University of Melbourne, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service, Doherty Institute for Infection and Immunity, The Royal Melbourne Hospital, , Melbourne, Victoria, Australia
| | - Christopher H Connor
- Centre for Pathogen Genomics, University of Melbourne, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Kristy Horan
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jake A Lacey
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Courtney R Lane
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Antimicrobial Resistance, Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Centre for Pathogen Genomics, University of Melbourne, Melbourne, Victoria, Australia
| | - Glen P Carter
- Centre for Pathogen Genomics, University of Melbourne, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Torsten Seemann
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Centre for Pathogen Genomics, University of Melbourne, Melbourne, Victoria, Australia
| | - Adrian Egli
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Timothy P Stinear
- Centre for Pathogen Genomics, University of Melbourne, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Benjamin P Howden
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Antimicrobial Resistance, Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases and Immunology, Austin Health, Heidelberg, Victoria, Australia
- Centre for Pathogen Genomics, University of Melbourne, Melbourne, Victoria, Australia
- Microbiology Department, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
5
|
Tasnim Y, Rahman MK, Abdul-Hamid C, Awosile B. Beta-lactamase-producing Escherichia coli in migratory geese at West Texas recreational parks. Comp Immunol Microbiol Infect Dis 2025; 118:102320. [PMID: 39951929 DOI: 10.1016/j.cimid.2025.102320] [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: 10/16/2024] [Revised: 01/22/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
This study aimed to determine the prevalence, and the genomic characteristics of beta-lactamase-Resistant Escherichia coli isolated from the feces of migratory geese at one health interface in West Texas. A descriptive study was conducted. We collected geese feces (n = 165), water (n = 118), and soil (n = 74) from 22 recreational parks in West Texas. We used Chromogenic agar to isolate extended-spectrum beta-lactamase (ESBL)-Resistant-E. coli. We used the whole genome sequencing (WGS) method to determine the genomic characteristics of selected E. coli isolates. Among 357 samples, 12.61 % (95 %CI: 9.34-16.50) were positive for ESBL- Resistant-E. coli. From WGS of 20 E. coli isolates, 19 isolates harbored at least 1 beta-lactamase gene including blaCTX-M-1, blaCTX-M-65, blaCTX-M-14, blaCTX-M-15, blaCTX-M-27, blaCTX-M-55, blaCTX-M-32, blaTEM-1A, blaTEM-1B. Most of the isolates carried genes conferring resistance to tetracyclines-(tet(A), tet(B)), aminoglycosides-(aac(3)-IIa, aph(6)-Id, aph(3')-Ia, aadA1), sulfonamides-(sul1,sul2), amphenicol-(floR), trimethoprim-(dfrA1, dfrA14, dfrA17) and streptogramin-B(MLSB) agent-(mph(A)). 13 isolates showed chromosomal mutations in the promoter region G of the ampC beta-lactamase gene. We detected sixteen incompatibility plasmid groups and 60 virulence genes, which are related to adherence, exotoxin, invasion, and nutrition/metabolic factors. Genome analysis showed that all isolates were genetically similar to human E. coli isolates. The study showed that migratory geese at recreational parks can be reservoirs of resistant bacteria with diverse serotypes and sequence types of E. coli isolates. Based on the findings, the detection of a multidrug-resistant E. coli strain reinforces the importance of adequate hygiene practices for humans and pet animals after visiting recreational parks.
Collapse
Affiliation(s)
- Yamima Tasnim
- Texas Tech University School of Veterinary Medicine, Amarillo, TX 79106, United States.
| | - Md Kaisar Rahman
- Texas Tech University School of Veterinary Medicine, Amarillo, TX 79106, United States.
| | - Cherissa Abdul-Hamid
- Zoonosis Control Program, Texas Department of State Health Services, Lubbock, TX, USA.
| | - Babafela Awosile
- Texas Tech University School of Veterinary Medicine, Amarillo, TX 79106, United States.
| |
Collapse
|
6
|
Jousserand N, Auvray F, Chagneau C, Cavalié L, Maurey C, Drut A, Lavoué R, Oswald E. Zoonotic potential of uropathogenic Escherichia coli lineages from companion animals. Vet Res 2025; 56:69. [PMID: 40140916 PMCID: PMC11948896 DOI: 10.1186/s13567-025-01493-0] [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: 11/26/2024] [Accepted: 02/13/2025] [Indexed: 03/28/2025] Open
Abstract
Escherichia coli is responsible for urinary tract infections (UTI) in humans and pets. This study aims to provide data on the virulome and resistome of E. coli strains isolated during bacteriuria in companion animals and to assess their zoonotic potential. 135 E. coli strains prospectively collected from urine samples of 44 cats and 91 dogs in three French veterinary teaching hospitals were analyzed via antibiotic susceptibility tests and whole genome sequencing. Phylogroup B2 was overrepresented and several sequence types (STs) associated with human extra-intestinal pathogenic E. coli (ExPEC) were found. These included ST12, ST127 and ST141 (8 strains each), which were characterized by genetic homogeneity, and ST73 (23 strains) which contained several serotype-delineated sublineages with distinct distributions in pets and humans. Single nucleotide polymorphism (SNP) analysis further revealed the existence of highly related human and companion animal clones among these STs, indicative of a zoonotic potential. By contrast, other major human ExPEC STs (e.g. ST131, ST10, ST69, ST95 and ST1193) were rarely found (2 strains each), suggesting they might be less adapted to cats and dogs. Of note, ST372 (21 strains) was predominant and exclusively found in dogs. Pet E. coli UTI strains carried virulence genes commonly found in human E. coli UTI isolates. 15.6% of strains were predicted as multi-drug resistant. The major canine and feline ExPEC lineages were not associated with extended spectrum beta lactamase and AmpC production. Only one strain (from ST131) carried the blaCTX-M-15 gene. Persistent clones of E. coli isolated from five cats and nine dogs with recurrent infection had genetic traits similar to strains from other animals. Approximately one-third of the E. coli UTI strains from pets exhibited genetic similarities to those responsible for UTI in humans, suggesting a potential for zoonotic transmission. This study underscores the continued need to monitor and control antimicrobial resistance in companion animals.
Collapse
Affiliation(s)
- Nicolas Jousserand
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Frédéric Auvray
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Camille Chagneau
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- Service de Bactériologie-Hygiène, CHU Toulouse, Hôpital Purpan, Toulouse, France
| | - Laurent Cavalié
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- Service de Bactériologie-Hygiène, CHU Toulouse, Hôpital Purpan, Toulouse, France
| | - Christelle Maurey
- UMR Virologie, INRAE, ENVA, ANSES, Université Paris-Est, Maisons-Alfort, France
- Department of Internal Medicine, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Amandine Drut
- Oniris VetAgroBio Nantes, Université de Nantes, Nantes, France
- MIHA Team, INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Rachel Lavoué
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- InTheRes, Université de Toulouse, INRAE, ENVT, UPS, Toulouse, France
| | - Eric Oswald
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France.
- Service de Bactériologie-Hygiène, CHU Toulouse, Hôpital Purpan, Toulouse, France.
| |
Collapse
|
7
|
Chang TH, Pourtois JD, Haddock NL, Furukawa D, Kelly KE, Amanatullah DF, Burgener E, Milla C, Banaei N, Bollyky PL. Prophages are infrequently associated with antibiotic resistance in Pseudomonas aeruginosa clinical isolates. mSphere 2025; 10:e0090424. [PMID: 39945525 PMCID: PMC11934324 DOI: 10.1128/msphere.00904-24] [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: 10/23/2024] [Accepted: 01/16/2025] [Indexed: 02/19/2025] Open
Abstract
Lysogenic bacteriophages can integrate their genome into the bacterial chromosome in the form of a prophage and can promote genetic transfer between bacterial strains in vitro. However, the contribution of lysogenic bacteriophages to the incidence of antimicrobial resistance (AMR) in clinical settings is poorly understood. Here, in a set of 186 clinical isolates of Pseudomonas aeruginosa collected from respiratory cultures from 82 patients with cystic fibrosis, we evaluate the links between prophage counts and both genomic and phenotypic resistance to six anti-pseudomonal antibiotics: tobramycin, colistin, ciprofloxacin, meropenem, aztreonam, and piperacillin-tazobactam. We identified 239 different prophages in total. We find that P. aeruginosa isolates contain on average 3.06 ± 1.84 (SD) predicted prophages. We find no significant association between the number of prophages per isolate and the minimum inhibitory concentration for any of these antibiotics. We then investigate the relationship between particular prophages and AMR. We identify a single lysogenic phage associated with phenotypic resistance to the antibiotic tobramycin and, consistent with this association, we observe that AMR genes associated with resistance to tobramycin are more likely to be found when this prophage is present. However, we find that they are not encoded directly on prophage sequences. Additionally, we identify a single prophage statistically associated with ciprofloxacin resistance but do not identify any genes associated with ciprofloxacin phenotypic resistance. These findings suggest that prophages are only infrequently associated with the AMR genes in clinical isolates of P. aeruginosa.IMPORTANCEAntibiotic-resistant infections of Pseudomonas aeruginosa (Pa), a leading pathogen in patients with cystic fibrosis (CF), are a global health threat. While lysogenic bacteriophages are known to facilitate horizontal gene transfer, their role in promoting antibiotic resistance in clinical settings remains poorly understood. In our analysis of 186 clinical isolates of P. aeruginosa from CF patients, we find that prophage abundance does not predict phenotypic resistance to key antibiotics but that specific prophages are infrequently associated with tobramycin resistance genes. In addition, we do not find antimicrobial resistance (AMR) genes encoded directly on prophages. These results highlight that while phages can be associated with AMR, phage-mediated AMR transfer may be rare in clinical isolates and difficult to identify. This work is important for future efforts on mitigating AMR in CFCF and other vulnerable populations affected by Pa infections and advances our understanding of bacterial-phage dynamics in clinical infections.
Collapse
Affiliation(s)
- Tony H. Chang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Julie D. Pourtois
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Naomi L. Haddock
- University of California San Francisco Medical School, San Francisco, California, USA
| | - Daisuke Furukawa
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Kate E. Kelly
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Derek F. Amanatullah
- Department of Orthopaedic Surgery, Stanford Hospital and Clinics, Redwood City, California, USA
| | - Elizabeth Burgener
- Division of Pediatric Pulmonology and Sleep Medicine, Children’s Hospital Los Angeles, Keck School of Medicine at USC, Los Angeles, California, USA
| | - Carlos Milla
- Cystic Fibrosis Clinic, Department of Medicine, Lucille Packard Children’s Hospital, Stanford University School of Medicine, Stanford, California, USA
| | - Niaz Banaei
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
8
|
Tamai S, Okuno M, Ogura Y, Suzuki Y. Genetic diversity of dissolved free extracellular DNA compared to intracellular DNA in wastewater treatment plants. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 970:178989. [PMID: 40048953 DOI: 10.1016/j.scitotenv.2025.178989] [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: 12/20/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
Dissolved free extracellular DNA (free-exDNA) coexists with intracellular DNA (inDNA) in aquatic environments. Free-exDNA can be taken up by bacteria through transformation, and wastewater treatment plants (WWTPs) are positioned as potential hot spots for genetic contamination. However, studies comparing the composition of free-exDNA and inDNA is limited. This study employed colloidal adsorption and foam concentration method to recover free-exDNA from different WWTP stages and compared its diversity with inDNA via metagenomic analysis. Free-exDNA concentrations were observed to increase after chlorination. Genetic analysis revealed a higher abundance of specific genes following chlorination, suggesting that free-exDNA in effluent originated from bacterial death in secondary treated water. This result indicates that free-exDNA, which increases due to chlorination, is subsequently released into the catchment. Additionally, several high-risk antibiotic-resistance genes (ARGs) were detected that colocalized with mobile genetic elements. These ARGs were expected to have a high potential for gene transfer via transformation, and the risk was highlighted. Overall, these findings deepen our understanding of horizontal gene transfer risks in WWTPs.
Collapse
Affiliation(s)
- Soichiro Tamai
- Department of Environment and Resource Sciences, Interdisciplinary Graduate School of Agriculture and Engineering, University of Miyazaki, Gakuen Kibanadai-Nishi 1-1, Miyazaki 889-2192, Japan.
| | - Miki Okuno
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan.
| | - Yoshitoshi Ogura
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan.
| | - Yoshihiro Suzuki
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Miyazaki, Gakuen Kibanadai-Nishi 1-1, Miyazaki 889-2192, Japan.
| |
Collapse
|
9
|
Haq SU, Ling W, Aqib AI, Danmei H, Aleem MT, Fatima M, Ahmad S, Gao F. Exploring the intricacies of antimicrobial resistance: Understanding mechanisms, overcoming challenges, and pioneering innovative solutions. Eur J Pharmacol 2025; 998:177511. [PMID: 40090539 DOI: 10.1016/j.ejphar.2025.177511] [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: 11/15/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/18/2025]
Abstract
Antimicrobial resistance (AMR) poses a growing global threat. This review examines AMR from diverse angles, tracing the story of antibiotic resistance from its origins to today's crisis. It explores the rise of AMR, from its historical roots to the urgent need to counter this escalating menace. The review explores antibiotic classes, mechanisms, resistance profiles, and genetics. It details bacterial resistance mechanisms with illustrative examples. Multidrug-resistant bacteria spotlight AMR's resilience. Modern AMR control offers hope through precision medicine, stewardship, combination therapy, surveillance, and international cooperation. Converging traditional and innovative treatments presents an exciting frontier as novel compounds seek to enhance antibiotic efficacy. This review calls for global unity and proactive engagement to address AMR collectively, emphasizing the quest for innovative solutions and responsible antibiotic use. It underscores the interconnectedness of science, responsibility, and action in combatting AMR. Humanity faces a choice between antibiotic efficacy and obsolescence. The call is clear: unite, innovate, and prevail against AMR.
Collapse
Affiliation(s)
- Shahbaz Ul Haq
- Department of Pharmacology, Shantou University Medical College, Shantou, 515041, China.
| | - Wang Ling
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou, 730050, China
| | - Amjad Islam Aqib
- Department of Medicine, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63100, Pakistan
| | - Huang Danmei
- Department of Pharmacology, Shantou University Medical College, Shantou, 515041, China
| | - Muhammad Tahir Aleem
- Department of Pharmacology, Shantou University Medical College, Shantou, 515041, China
| | - Mahreen Fatima
- Faculty of Biosciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63100, Pakistan
| | - Saad Ahmad
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
10
|
Raju NP, Ansari A, Patil G, Sheeraz MS, Kukade S, Kumar S, Kapley A, Qureshi A. Antibiotic Resistance Dissemination and Mapping in the Environment Through Surveillance of Wastewater. J Basic Microbiol 2025; 65:e2400330. [PMID: 39676299 DOI: 10.1002/jobm.202400330] [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: 06/04/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 12/17/2024]
Abstract
Antibiotic resistance is one of the major health threat for humans, animals, and the environment, according to the World Health Organization (WHO) and the Global Antibiotic-Resistance Surveillance System (GLASS). In the last several years, wastewater/sewage has been identified as potential hotspots for the dissemination of antibiotic resistance and transfer of resistance genes. However, systematic approaches for mapping the antibiotic resistance situation in sewage are limited and underdeveloped. The present review has highlighted all possible perspectives by which the dynamics of ARBs/ARGs in the environment may be tracked, quantified and assessed spatio-temporally through surveillance of wastewater. Moreover, application of advanced methods like wastewater metagenomics for determining the community distribution of resistance at large has appeared to be promising. In addition, monitoring wastewater for antibiotic pollution at various levels, may serve as an early warning system and enable policymakers to take timely measures and build infrastructure to mitigate health crises. Thus, by understanding the alarming presence of antibiotic resistance in wastewater, effective action plans may be developed to address this global health challenge and its associated environmental risks.
Collapse
Affiliation(s)
- Neenu P Raju
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Aamir Ansari
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Gandhali Patil
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Mohammed Shahique Sheeraz
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Sushrut Kukade
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Shailendra Kumar
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Atya Kapley
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Asifa Qureshi
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| |
Collapse
|
11
|
Li Z, Li Z, Peng Y, Zhang M, Wen Y, Lu X, Kan B. Genomic diversity of mcr-carrying plasmids and the role of type IV secretion systems in IncI2 plasmids conjugation. Commun Biol 2025; 8:342. [PMID: 40025288 PMCID: PMC11873049 DOI: 10.1038/s42003-025-07748-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025] Open
Abstract
The rapid dissemination of colistin resistance via mcr-carrying plasmids (pMCRs) poses a significant public health challenge. This study examined the genomic diversity and conjugation mechanisms of pMCRs, with a particular focus on the role of type IV secretion systems (T4SS) in IncI2 plasmids. The 868 complete plasmid sequences revealed various replicon types of pMCRs, with IncI2 as the primary epidemic type, and the co-transfer risk of multidrug resistance genes associated with IncHI2. T4SS was identified in 89.9% of pMCRs, with the T4SS sequence exclusively carried by IncI2 being conserved and typical of the VirB/D4 type, consisting of 12 subunits. Conjugation assays confirmed the essential role of the pilus subunit VirB2 and the significant impact of VirB5P3 on conjugation. This was further validated in the in vivo intra-species competitive conjugation of Escherichia coli. Structural predictions show that a hypervariable region at the C-terminus of the pentameric VirB5 co-evolves in sequence with VirB6, and the conserved N-terminal may act as a potential drug target to inhibit the plasmid transfer channel. This study will deepen the understanding of the pMCR epidemic patterns and provide additional insights for controlling the spread of resistant plasmids.
Collapse
Affiliation(s)
- Zhe Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhenpeng Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yao Peng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Mengke Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, China
| | - Yuanxi Wen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xin Lu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| | - Biao Kan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| |
Collapse
|
12
|
Louca S. National antibiotic consumption is strongly related to the prevalence of antibiotic resistance across bacterial clades. iScience 2025; 28:111712. [PMID: 39898032 PMCID: PMC11787492 DOI: 10.1016/j.isci.2024.111712] [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: 05/22/2024] [Revised: 10/26/2024] [Accepted: 12/27/2024] [Indexed: 02/04/2025] Open
Abstract
The impact of societal antibiotic consumption on the prevalence of antibiotic resistance across microbial taxa in natural environments has not yet been assessed at global scales. Here, I examine the prevalence of 155 antibiotic resistance genes (ARGs) in 300,209 bacterial genomes, from non-clinical non-human-associated terrestrial environments at over 9,600 locations in 44 countries. I then compare ARG prevalences to nationwide antibiotic consumption rates, distinguishing between different ARG types. I find that depending on country and ARG type, ARG prevalences can be extremely high; for example, the probability that a given quinolone resistance gene is present in a given strain in Thailand was estimated at 42%. Further, I find strong positive correlations between nationwide antibiotic consumption rates and mean ARG prevalences for nearly all ARG types. Thus, national antibiotic consumption leaves a signal on the prevalence of ARGs across the bacterial tree, even in non-clinical environments.
Collapse
Affiliation(s)
- Stilianos Louca
- Department of Biology, University of Oregon, Eugene, OR 97403, USA
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
13
|
Khodaparast L, Khodaparast L, Duran-Romaña R, Wu G, Houben B, Duverger W, De Vleeschouwer M, Konstantoulea K, Nysen F, Schalck T, Curwen DJ, Martin LL, Carpentier S, Scorneaux B, Michiels J, Schymkowitz J, Rousseau F. Co-translational protein aggregation and ribosome stalling as a broad-spectrum antibacterial mechanism. Nat Commun 2025; 16:1561. [PMID: 39939597 PMCID: PMC11821998 DOI: 10.1038/s41467-025-56873-z] [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: 03/11/2024] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
Drug-resistant bacteria pose an urgent global health threat, necessitating the development of antibacterial compounds with novel modes of action. Protein biosynthesis accounts for up to half of the energy expenditure of bacterial cells, and consequently inhibiting the efficiency or fidelity of the bacterial ribosome is a major target of existing antibiotics. Here, we describe an alternative mode of action that affects the same process: allowing translation to proceed but causing co-translational aggregation of the nascent peptidic chain. We show that treatment with an aggregation-prone peptide induces formation of polar inclusion bodies and activates the SsrA ribosome rescue pathway in bacteria. The inclusion bodies contain ribosomal proteins and ribosome hibernation factors, as well as mRNAs and cognate nascent chains of many proteins in amyloid-like structures, with a bias for membrane proteins with a fold rich in long-range beta-sheet interactions. The peptide is bactericidal against a wide range of pathogenic bacteria in planktonic growth and in biofilms, and reduces bacterial loads in mouse models of Escherichia coli and Acinetobacter baumannii infections. Our results indicate that disrupting protein homeostasis via co-translational aggregation constitutes a promising strategy for development of broad-spectrum antibacterials.
Collapse
Affiliation(s)
- Laleh Khodaparast
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ladan Khodaparast
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ramon Duran-Romaña
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Guiqin Wu
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Bert Houben
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Wouter Duverger
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Matthias De Vleeschouwer
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Katerina Konstantoulea
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Fleur Nysen
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Thomas Schalck
- Centre of Microbial and Plant Genetics;KU Leuven, Leuven, Belgium
- Center for Microbiology;VIB-KU Leuven, Leuven, Belgium
| | - Daniel J Curwen
- School of Chemistry, Monash University, Clayton, Vic, Australia
| | | | - Sebastien Carpentier
- Systems Biology based Mass Spectrometry Laboratory (SyBioMa), KULeuven, Leuven, Belgium
| | | | - Jan Michiels
- Centre of Microbial and Plant Genetics;KU Leuven, Leuven, Belgium
- Center for Microbiology;VIB-KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium.
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| | - Frederic Rousseau
- Switch Laboratory, VIB Center for Brain and Disease Research, Leuven, Belgium.
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
14
|
Dewan I, Uecker H. Evolutionary rescue of bacterial populations by heterozygosity on multicopy plasmids. J Math Biol 2025; 90:26. [PMID: 39909926 PMCID: PMC11799102 DOI: 10.1007/s00285-025-02182-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/07/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025]
Abstract
Bacterial plasmids and other extrachromosomal DNA elements frequently carry genes with important fitness effects for their hosts. Multicopy plasmids can additionally carry distinct alleles of host-fitness-relevant genes on different plasmid copies, allowing for heterozygosity not possible for loci on haploid chromosomes. Plasmid-mediated heterozygosity may increase the fitness of bacterial cells in circumstances where there is an advantage to having multiple distinct alleles (heterozyogote advantage); however, plasmid-mediated heterozygosity is also subject to constant loss due to random segregation of plasmid copies on cell division. We analyze a multitype branching process model to study the evolution and maintenance of plasmid-mediated heterozygosity under a heterozygote advantage. We focus on an evolutionary rescue scenario in which a novel mutant allele on a plasmid must be maintained together with the wild-type allele to allow population persistance (although our results apply more generally to the maintenance of heterozygosity due to heterozygote advantage). We determine the probability of rescue and derive an analytical expression for the threshold on the fitness of heterozygotes required to overcome segregation and make rescue possible; this threshold decreases with increasing plasmids copy number. We further show that the formation of cointegrates from the fusion of plasmid copies increases the probability of rescue. Overall, our results provide a rigorous quantitative assessment of the conditions under which bacterial populations can adapt to multiple stressors through plasmid-mediated heterozygosity. Many of the results are furthermore applicable to the related problem of the maintenance of incompatible plasmids in the same cell under selection for both.
Collapse
Affiliation(s)
- Ian Dewan
- Research Group Stochastic Evolutionary Dynamics, Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, 24306, Plön, Germany.
| | - Hildegard Uecker
- Research Group Stochastic Evolutionary Dynamics, Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, 24306, Plön, Germany
| |
Collapse
|
15
|
Hempstead SC, Gensler CA, Haley CA, Wiedenheft AM, Robertson JB, Fedorka-Cray PJ, Jacob ME. Prevalence and Characterization of Salmonella Species on U.S. Swine Sites as Part of the NAHMS 2021 Swine Enteric Study. J Food Prot 2025; 88:100435. [PMID: 39701448 DOI: 10.1016/j.jfp.2024.100435] [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: 07/29/2024] [Revised: 12/02/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Salmonella species are an important cause of systemic and gastrointestinal disease in animals and humans worldwide; they are also increasingly resistant to multiple classes of antimicrobials which may aid in their treatment and control. Salmonella can also be shed asymptomatically. The aim of this study was to survey the U.S. swine industry to determine and update estimates of Salmonella prevalence, antimicrobial resistance (AMR), replicon distribution, and biofilm potential as part of the United States Department of Agriculture (USDA) National Animal Health Monitoring System (NAHMS) 2021 Swine Study. A total of 1,043 fecal samples were collected from finishing swine 20 weeks and older on 39 sites, from November 2021 through April 2022. Salmonella detection was culture-based and antimicrobial susceptibility was determined by broth microdilution; replicon typing was completed by PCR. Biofilm potential was assessed using a crystal violet assay and normalized to a positive control strain. Overall, the prevalence of Salmonella in swine feces was low (3.9%; 41 of 1,043) and comprised of a broad range of serotypes including S. Derby (34.1%) and S. I 4,[5],12:i:- (22.0%), also found previously. Resistance to at least one antimicrobial agent was detected in 61.0% of isolates with the most prevalent resistance to tetracycline (53.7%). Multidrug resistance (≥3 antimicrobial classes) was found in 26.8% of isolates. The most prevalent replicon types detected were IncHI2 (24.4%) and IncA/C (24.4%), both of which have previously been associated with Salmonella isolated from swine. The majority (85.4%) of isolates formed some biofilm in vitro, characterized as weak (36.6%), moderate (39.0%), or strong (9.8%), with no major associations among common serotype or AMR pattern. Surveillance programs updating the prevalence and characteristics of Salmonella in the U.S. swine population will inform ongoing farm-level and public health efforts to mitigate animal and human health risks.
Collapse
Affiliation(s)
- Stephanie C Hempstead
- Department of Population Health & Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Catherine A Gensler
- Department of Population Health & Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA; Department of Agricultural and Human Sciences, College of Agricultural and Life Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Charles A Haley
- National Animal Health Monitoring System, Veterinary Services, Animal and Plant Health Inspection Service, United States Department of Agriculture, Fort Collins, Colorado, USA
| | - Alyson M Wiedenheft
- National Animal Health Monitoring System, Veterinary Services, Animal and Plant Health Inspection Service, United States Department of Agriculture, Fort Collins, Colorado, USA; Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - James B Robertson
- Department of Population Health & Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Paula J Fedorka-Cray
- Department of Population Health & Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Megan E Jacob
- Department of Population Health & Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
16
|
Bustamante M, Mei S, Daras IM, van Doorn G, Falcao Salles J, de Vos MG. An eco-evolutionary perspective on antimicrobial resistance in the context of One Health. iScience 2025; 28:111534. [PMID: 39801834 PMCID: PMC11719859 DOI: 10.1016/j.isci.2024.111534] [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: 01/16/2025] Open
Abstract
The One Health approach musters growing concerns about antimicrobial resistance due to the increased use of antibiotics in healthcare and agriculture, with all of its consequences for human, livestock, and environmental health. In this perspective, we explore the current knowledge on how interactions at different levels of biological organization, from genetic to ecological interactions, affect the evolution of antimicrobial resistance. We discuss their role in different contexts, from natural systems with weak selection, to human-influenced environments that impose a strong pressure toward antimicrobial resistance evolution. We emphasize the need for an eco-evolutionary approach within the One Health framework and highlight the importance of horizontal gene transfer and microbiome interactions for increased understanding of the emergence and spread of antimicrobial resistance.
Collapse
Affiliation(s)
| | - Siyu Mei
- University of Groningen – GELIFES, Groningen, the Netherlands
| | - Ines M. Daras
- University of Groningen – GELIFES, Groningen, the Netherlands
| | - G.S. van Doorn
- University of Groningen – GELIFES, Groningen, the Netherlands
| | | | | |
Collapse
|
17
|
Mohany M, Aslam J, Ali MA, Khattak B, Fozia F, Ahmad I, Khan MD, Al-Rejaie SS, Ziaullah Z, Milošević M. Wastewater microbiology: occurrence and prevalence of antibiotic-resistant extended-spectrum β-lactamase-producing Enterobacteriaceae in the district wastewater system. JOURNAL OF WATER AND HEALTH 2025; 23:26-42. [PMID: 39882852 DOI: 10.2166/wh.2024.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 12/08/2024] [Indexed: 01/31/2025]
Abstract
Extended spectrum β-lactamase (ESBL)-producing Enterobacteriaceae, including Klebsiella pneumoniae and Escherichia coli, pose a serious risk to human health because of antibiotic resistance. Wastewater serves as a reservoir for these bacteria, contributing to the evolution and transmission of antibiotic-resistant strains. The research aims to identify ESBL bacterium in wastewater samples from District Kohat. K. pneumoniae and E. coli were confirmed as ESBL-producing bacteria through a comprehensive array of diagnostic procedures, including Gram staining, biochemical analyses, and antibiotic susceptibility testing. Fecal coliform count (FCC) analyses revealed varying microorganism levels. Both E. coli and K. pneumoniae isolates showed ESBL enzyme expression, indicating antibiotic resistance. Resistance patterns included ciprofloxacin, ampicillin, cefotaxime, cefoxitin, and amoxicillin-clavulanic acid for both species. E. coli displayed higher sensitivity for chloramphenicol, trimethoprim- sulfamethoxazole, and gentamicin. Ceftazidime minimum inhibitory concentration results showed E. coli's higher resistance. The study accentuates the presence of antibiotic-resistant strains, emphasizing the value of effective wastewater treatment. The study provides crucial insights into microbial characteristics, fecal contamination, ESBL production, and antibiotic resistance in E. coli and K. pneumoniae isolates, advocating for monitoring and mitigation strategies.
Collapse
Affiliation(s)
- Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh 11451, Saudi Arabia
| | - Jawad Aslam
- Department of Microbiology, Kohat University of Science and Technology, Kohat, KP 26000, Pakistan
| | - Muhammad Adnan Ali
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore 19200, Pakistan
| | - Baharullah Khattak
- Department of Microbiology, Kohat University of Science and Technology, Kohat, KP 26000, Pakistan
| | - Fozia Fozia
- Department of Biochemistry, KMU Institute of Dental Sciences, Kohat, KP 26000, Pakistan E-mail:
| | - Ijaz Ahmad
- Department of Chemistry, Kohat University of Science and Technology, Kohat 26000, Pakistan
| | - Muhammad Daud Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat 26000, Pakistan
| | - Salim S Al-Rejaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh 11451, Saudi Arabia
| | - Ziaullah Ziaullah
- College of Professional Studies, Northeastern University, Boston, MA, USA
| | - Marija Milošević
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Kragujevac 34000, Serbia
| |
Collapse
|
18
|
Harris SL, Dutta S, Liu N, Wollenberg T, Wang X. Extended structure-activity relationship studies of the [1,2,5]oxadiazolo[3,4-b]pyrazine-containing colistin adjuvants. Bioorg Med Chem Lett 2025; 115:130008. [PMID: 39481690 DOI: 10.1016/j.bmcl.2024.130008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Antimicrobial resistance (AMR) is a formidable global health challenge. Multidrug-resistant (MDR) Gram-negative bacterial infections are of primary concern due to diminishing treatment options and high morbidity and mortality. Colistin, a polymyxin family antibiotic, is a last-resort treatment for MDR Gram-negative infections, but its wider use has resulted in escalating resistance. In 2022, using a screening approach, we discovered that a [1,2,5]oxadiazolo[3,4-b]pyrazine (ODP)-containing compound selectively re-sensitized various MDR Gram-negative bacteria to colistin. Initial structure-activity relationship (SAR) studies confirmed that bisanilino ODP compounds are colistin adjuvants with low mammalian toxicity. Herein, we report our extended SAR studies on a wide range of ODP analogs bearing alkyl- or arylalkylamines. Specifically, we discovered two new compounds, 5q and 8g, with potent colistin-potentiating activity and low mammalian toxicity in a wide range of clinically relevant pathogens.
Collapse
Affiliation(s)
| | | | - Nianzi Liu
- Department of Chemistry, Boulder, CO 80309, USA
| | | | - Xiang Wang
- Department of Chemistry, Boulder, CO 80309, USA.
| |
Collapse
|
19
|
Notake S, Terada N, Tayama K, Sugie A, Abe M, Ikeda S, Iinuma H, Toeda Y, Ueda A, Nakamura K, Kurihara Y, Hitomi S. Extended-spectrum β-lactamase-producing Plesiomonas shigelloides isolated from the stool of a Japanese traveler returning from Rwanda: A case report. J Infect Chemother 2025; 31:102449. [PMID: 38906374 DOI: 10.1016/j.jiac.2024.06.012] [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: 04/17/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
A 21-year-old previously healthy Japanese woman visited an outpatient clinic because of abdominal pain, watery diarrhea, vomiting, and mild fever that had started on the previous day. She traveled to rural and urban areas of Rwanda and returned to Japan 3 days before. Stool culture yielded the Plesiomonas shigelloides strain TMCH301018, against which minimum inhibitory concentrations of cefotaxime and cefotaxime-clavulanate were 128 and ≤0.12/4 μg/mL, respectively. The strain had the blaCTX-M-27 gene and an IncA/C replicon-type plasmid. Moreover, a transformant produced by introduction of an IncA/C plasmid extracted from TMCH301018 into Escherichia coli DH5α was positive for the blaCTX-M-27 gene and fulfilled the criteria of extended-spectrum β-lactamase (ESBL) production described by the Clinical and Laboratory Standards Institute, indicating that TMCH301018 produced ESBL of CTX-M-27 and the ESBL-encoding gene was located on an IncA/C plasmid. Pathogenicity of TMCH301018 for the patient's complaints was uncertain because a molecular assay detected other enteropathogens in the stool specimen and the symptoms improved within 2 days with administration of oral ciprofloxacin, to which TMCH301018 was not susceptible. To our knowledge, this is the first report describing the isolation of ESBL-producing P. shigelloides.
Collapse
Affiliation(s)
- Shigeyuki Notake
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan.
| | - Norihiko Terada
- Division of Infectious Diseases, Department of Medicine, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan; Department of Infectious Diseases, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 3058576, Japan
| | - Kodai Tayama
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan
| | - Asami Sugie
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan
| | - Mariko Abe
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan
| | - Shiori Ikeda
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan
| | - Hiroatsu Iinuma
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan
| | - Yoshihiro Toeda
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan
| | - Atsuo Ueda
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan
| | - Koji Nakamura
- Department of Clinical Laboratory, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan
| | - Yoko Kurihara
- Division of Infectious Diseases, Department of Medicine, Tsukuba Medical Center Hospital, 1-3-1 Amakubo, Tsukuba, Ibaraki, 3058558, Japan; Department of Infectious Diseases, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 3058576, Japan
| | - Shigemi Hitomi
- Department of Infectious Diseases, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Department of Infectious Diseases, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki, 3058576, Japan
| |
Collapse
|
20
|
Zhang S, Shu Y, Yang Z, Zhong Z, Wang M, Jia R, Chen S, Liu M, Zhu D, Zhao X, Wu Y, Yang Q, Huang J, Ou X, Sun D, Tian B, Wu Z, He Y, Cheng A. Decoding the enigma: unveiling the transmission characteristics of waterfowl-associated bla NDM-5-positive Escherichia coli in select regions of China. Front Microbiol 2024; 15:1501594. [PMID: 39717269 PMCID: PMC11663885 DOI: 10.3389/fmicb.2024.1501594] [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: 09/25/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Escherichia coli (E. coli) serves as a critical indicator microorganism for assessing the prevalence and dissemination of antibiotic resistance, notably harboring various antibiotic-resistant genes (ARGs). Among these, the emergence of the bla NDM gene represents a significant threat to public health, especially since carbapenem antibiotics are vital for treating severe infections caused by Gram-negative bacteria. This study aimed to characterize the antibiotic resistance features of bla NDM-5-positive E. coli strains isolated from waterfowl in several regions of China and elucidate the dissemination patterns of the bla NDM-5 gene. We successfully isolated 103 bla NDM-5-positive E. coli strains from 431 intestinal fecal samples obtained from waterfowl across five provincial-level units in China, with all strains exhibiting multidrug resistance (MDR). Notably, the bla NDM-5 gene was identified on plasmids, which facilitate efficient and stable horizontal gene transfer (HGT). Our adaptability assays indicated that while the bla NDM-5-positive plasmid imposed a fitness cost on the host bacteria, the NDM-5 protein was successfully induced and purified, exhibiting significant enzymatic activity. One strain, designated DY51, exhibited a minimum inhibitory concentration (MIC) for imipenem of 4 mg/L, which escalated to 512 mg/L following exposure to increasing imipenem doses. This altered strain demonstrated stable resistance to imipenem alongside improved adaptability, correlating with elevated relative expression levels of the bla NDM-5 and overexpression of efflux pumps. Collectively, this study highlights the horizontal dissemination of the bla NDM-5 plasmid among E. coli strains, confirms the associated fitness costs, and provides insights into the mechanisms underlying the stable increase in antibiotic resistance to imipenem. These findings offer a theoretical framework for understanding the dissemination dynamics of bla NDM-5 in E. coli, which is essential for developing effective strategies to combat carbapenem antibiotic resistance.
Collapse
Affiliation(s)
- Shaqiu Zhang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Yanxi Shu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhechen Yang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Renyong Jia
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Shun Chen
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Mafeng Liu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Dekang Zhu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Xinxin Zhao
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Ying Wu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Qiao Yang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Juan Huang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Xumin Ou
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Di Sun
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Bin Tian
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Zhen Wu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Yu He
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Anchun Cheng
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| |
Collapse
|
21
|
Kidenya BR, Mboowa G, Sserwadda I, Kanyerezi S, Nakafu E, Akaro IL, Mkinze B, Joloba ML, Seni J. Whole genome-based characterization of extended-spectrum β-lactamase-producing Enterobacter cloacae from orthopedic patients and environment of a tertiary referral hospital in Tanzania. New Microbes New Infect 2024; 62:101486. [PMID: 39386352 PMCID: PMC11462357 DOI: 10.1016/j.nmni.2024.101486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
Objectives We investigated the genomic epidemiology of extended-spectrum β-lactamase-producing Enterobacter cloacae (ESBL-Ec) isolates from patients and hospital environment to better understand their distribution to help devising effective strategies for infection prevention and control. Methods We screened ESBL-Ec at Bugando Medical Center (BMC) in Mwanza, Tanzania. Rectal swabs from orthopedic patients on admission and swabs from the neighboring inanimate environment were collected. Following microbial culture, DNA was extracted from pure ESBL-Ec, and whole-genome sequencing was done. Sequence typing (ST), plasmid replicons, drug resistance, and virulence genes were deciphered using the Rapid Microbial Analysis Pipeline (rMAP). Results We obtained 209 ESBL isolates, of which 15 (7.2 %) were ESBL-Ec [8 (53.3 %) from patients and 7 (46.7 %) from the environment]. Seven isolates were novel and eight were diverse, each with a unique ST. All isolates harbored two to five β-lactamase genes, with the predominance of bla CTX-M-15 (15/15), bla OXA-1 (14/15), bla TEM (14/15) and bla ACT (12/15). The most common non β-lactam drug resistance genes were aac(3)-IIa (14/15), aac(6')-Ib-cr (14/15), fosA (14/15), and qnrB1 (12/15), aph(3″)-Ib (10/15) and aph(6)-Id (10/15). Eleven different types of plasmid replicons were identified in 14/15 of the isolates, harboring one to five plasmids, with the most common plasmids being IncFII (11/15) and IncFIB (10/15). All isolates harbored the outer membrane protein (ompA), and curli protein (csg) was in 14/15 isolates. Conclusion Admitted orthopedic patients and the hospital environment act as a reservoir of ESBL-Ec with diverse STs and endowed with drug resistance and arsenals of virulence genes, calling for their routine screening on admission for mitigation of potential subsequent infections.
Collapse
Affiliation(s)
- Benson R. Kidenya
- Department of Biochemistry and Molecular Biology, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, P.O. Box 1464, Mwanza, Tanzania
| | - Gerald Mboowa
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P. O Box 7072, Kampala, Uganda
- African Center of Excellence in Bioinformatics and Data-Intensive Sciences, The Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O Box 22418, Kampala, Uganda
- Africa Centres for Disease Control and Prevention, African Union Commission, Roosevelt Street, P.O. Box 3243, W21 K19, Addis Ababa, Ethiopia
| | - Ivan Sserwadda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P. O Box 7072, Kampala, Uganda
- African Center of Excellence in Bioinformatics and Data-Intensive Sciences, The Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O Box 22418, Kampala, Uganda
- Department of Biochemistry and Bioinformatics, School of Pure and Applied Sciences, Pwani University, P.O Box 195-80108, Kilifi, Kenya
| | - Stephen Kanyerezi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P. O Box 7072, Kampala, Uganda
- Africa Centres for Disease Control and Prevention, African Union Commission, Roosevelt Street, P.O. Box 3243, W21 K19, Addis Ababa, Ethiopia
| | - Esther Nakafu
- College of Veterinary Medicine, Animal Resources and Bio-security (COVAB), Makerere University, P. O Box 7062, Kampala, Uganda
| | - Inyasi Lawrence Akaro
- Department of Orthopedic Surgery, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, P.O. Box 1464, Mwanza, Tanzania
| | - Baraka Mkinze
- Department of Orthopedic Surgery, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, P.O. Box 1464, Mwanza, Tanzania
| | - Moses L. Joloba
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P. O Box 7072, Kampala, Uganda
| | - Jeremiah Seni
- Department of Microbiology and Immunology, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, P.O. Box 1464, Mwanza, Tanzania
| |
Collapse
|
22
|
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
|
23
|
Yusuf F, Ahmed SM, Dy D, Baney K, Waseem H, Gilbride KA. Occurrence and characterization of plasmid-encoded qnr genes in quinolone-resistant bacteria across diverse aquatic environments in southern Ontario. Can J Microbiol 2024; 70:492-506. [PMID: 39083844 DOI: 10.1139/cjm-2024-0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Antimicrobial resistance is an ever-increasing threat. The widespread usage of ciprofloxacin has led to the manifestation of resistance due to chromosomal mutations or the acquisition of plasmid-mediated quinolone resistance (PMQR) traits. Some particular PMQR traits, qnr genes, have been identified globally in clinical and environmental isolates. This study aimed to determine the prevalence of ciprofloxacin-resistant bacteria in aquatic environments in southern Ontario and investigate the extent of dissemination of ciprofloxacin resistance traits among the bacterial communities. We surveyed the prevalence of plasmid encoding qnr genes using a multiplex PCR assay of associated PMQR genes, qnrA, qnrB, and qnrS, on 202 isolates. Despite the absence of significant impacts on minimum inhibitory concentration levels, the presence of qnr genes correlates with heightened resistance to quinolones and nalidixic acid in some isolates. Taxonomic analysis highlights distinct differences in the composition and diversity of ciprofloxacin-sensitive (CipS) and ciprofloxacin-resistant (CipR) populations, with Proteobacteria dominating both groups. Importantly, CipR populations exhibit lower genetic diversity but higher prevalence of multiple antibiotic resistances, suggesting co-selection mechanisms. Co-occurrence analysis highlights significant associations between ciprofloxacin resistance and other antibiotic resistances, implicating complex genetic linkages. The results of our study signified the critical role of environmental monitoring in public health.
Collapse
Affiliation(s)
- Farhan Yusuf
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, Canada
| | - Saher M Ahmed
- Urban Water, Toronto Metropolitan University, Toronto, ON, Canada
| | - Danica Dy
- Department of Molecular and Cell Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katherine Baney
- Department of Cell Biology and Physiology, University of California, Berkeley, CA, USA
| | - Hassan Waseem
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, Canada
| | - Kimberley A Gilbride
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, Canada
- Urban Water, Toronto Metropolitan University, Toronto, ON, Canada
| |
Collapse
|
24
|
Samuel B, Mittelman K, Croitoru SY, Ben Haim M, Burstein D. Diverse anti-defence systems are encoded in the leading region of plasmids. Nature 2024; 635:186-192. [PMID: 39385022 PMCID: PMC11541004 DOI: 10.1038/s41586-024-07994-w] [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: 02/20/2023] [Accepted: 08/27/2024] [Indexed: 10/11/2024]
Abstract
Plasmids are major drivers of gene mobilization by means of horizontal gene transfer and play a key role in spreading antimicrobial resistance among pathogens1,2. Despite various bacterial defence mechanisms such as CRISPR-Cas, restriction-modification systems and SOS-response genes that prevent the invasion of mobile genetic elements3, plasmids robustly transfer within bacterial populations through conjugation4,5. Here we show that the leading region of plasmids, the first to enter recipient cells, is a hotspot for an extensive repertoire of anti-defence systems, encoding anti-CRISPR, anti-restriction, anti-SOS and other counter-defence proteins. We further identified in the leading region a prevalence of promoters known to allow expression from single-stranded DNA6, potentially facilitating rapid protection against bacterial immunity during the early stages of plasmid establishment. We demonstrated experimentally the importance of anti-defence gene localization in the leading region for efficient conjugation. These results indicate that focusing on the leading region of plasmids could lead to the discovery of diverse anti-defence genes. Combined, our findings show a new facet of plasmid dissemination and provide theoretical foundations for developing efficient conjugative delivery systems for natural microbial communities.
Collapse
Affiliation(s)
- Bruria Samuel
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Karin Mittelman
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Shirly Ynbal Croitoru
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Maya Ben Haim
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - David Burstein
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
25
|
Allain M, Morel-Journel T, Condamine B, Gibeaux B, Gachet B, Gschwind R, Denamur E, Landraud L. IncC plasmid genome rearrangements influence the vertical and horizontal transmission tradeoff in Escherichia coli. Antimicrob Agents Chemother 2024; 68:e0055424. [PMID: 39194203 PMCID: PMC11459957 DOI: 10.1128/aac.00554-24] [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: 04/17/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
It has been shown that an evolutionary tradeoff between vertical (host growth rate) and horizontal (plasmid conjugation) transmissions contributes to global plasmid fitness. As conjugative IncC plasmids are important for the spread of multidrug resistance (MDR), in a broad range of bacterial hosts, we investigated vertical and horizontal transmissions of two multidrug-resistant IncC plasmids according to their backbones and MDR-region rearrangements, upon plasmid entry into a new host. We observed plasmid genome deletions after conjugation in three diverse natural Escherichia coli clinical strains, varying from null to high number depending on the plasmid, all occurring in the MDR region. The plasmid burden on bacterial fitness depended more on the strain background than on the structure of the MDR region, with deletions appearing to have no impact. Besides, we observed an increase in plasmid transfer rate, from ancestral host to new clinical recipient strains, when the IncC plasmid was rearranged. Finally, using a second set of conjugation experiments, we investigated the evolutionary tradeoff of the IncC plasmid during the critical period of plasmid establishment in E. coli K-12, by correlating the transfer rates of deleted or non-deleted IncC plasmids and their costs on the recipient strain. Plasmid deletions strongly improved conjugation efficiency with no negative growth effect. Our findings indicate that the flexibility of the MDR-region of the IncC plasmids can promote their dissemination, and provide diverse opportunities to capture new resistance genes. In a broader view, they suggest that the vertical-horizontal transmission tradeoff can be manipulated by the plasmid to improve its fitness.
Collapse
Affiliation(s)
- Margaux Allain
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, IAME, Paris, France
- AP-HP, Laboratoire de Microbiologie Hygiène, Hôpital Louis Mourier, Colombes, France
| | - Thibaut Morel-Journel
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, IAME, Paris, France
| | - Bénédicte Condamine
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, IAME, Paris, France
| | - Benoist Gibeaux
- AP-HP, Laboratoire de Microbiologie Hygiène, Hôpital Louis Mourier, Colombes, France
| | - Benoit Gachet
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, IAME, Paris, France
| | - Rémi Gschwind
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, IAME, Paris, France
| | - Erick Denamur
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, IAME, Paris, France
- AP-HP, Laboratoire de Génétique Moléculaire, Hôpital Bichat, Paris, France
| | - Luce Landraud
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, IAME, Paris, France
- AP-HP, Laboratoire de Microbiologie Hygiène, Hôpital Louis Mourier, Colombes, France
| |
Collapse
|
26
|
Siame A, Yamba K, Samutela M, Mukubesa A, Mulundu G. Carriage and antimicrobial susceptibility patterns of rectal ESBL E. coli in surgical patients at the University Teaching Hospitals in Lusaka, Zambia. JAC Antimicrob Resist 2024; 6:dlae159. [PMID: 39386376 PMCID: PMC11462439 DOI: 10.1093/jacamr/dlae159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Background Surgical site infections (SSIs) are on the rise and are a global concern as they complicate the recovery of patients postoperatively. Bacterial colonization of the patient's skin and alimentary tract are known to be major contributing sources to SSIs. However, Zambia lacks data relating to carriage rates of antibiotic-resistant rectal Escherichia coli among surgical patients. Methods This was a cross-sectional study aimed at determining the preoperative (preop) and postoperative (postop) carriage and antimicrobial susceptibility patterns of rectal ESBL-producing E. coli (ESBL-Ec) in elective surgery patients at the highest tertiary hospital in Lusaka, Zambia. Phenotypic methods were used in the identification of E. coli. Antibiotic susceptibility patterns and identification of ESBL-Ec was determined by Kirby-Bauer disc diffusion. Results A total of 120 study participants were recruited, of which 75 were followed up at least 72 h after surgery. From 195 rectal swabs cultured, 177 (90.8%) were positive for E. coli, of which 53 (29.9%) were ESBL-Ec, with a significantly (P < 0.0001) higher proportion in postop (47.9%) than preop (17.3%) participants. Overall, ESBL-Ec isolates showed higher resistance in postop than preop to cefotaxime (100% versus 88.9%, respectively), ampicillin (100% versus 94.4%), ciprofloxacin (88.3% versus 83.3%), amoxicillin/clavulanic acid (80% versus 66.7%) and cefepime (80% versus 77.8%). MDR ESBL-Ec strains were more frequent in postop than in preop participants (91.4% versus 88.9%). Conclusions The study showed a significantly higher rate of antimicrobial-resistant rectal E. coli in postop than preop participants. There is a need to ascertain the source of the resistance and to institute robust infection control measures, preop screening of surgical patients for ESBL-Ec, and to raise awareness on prudent use of antibiotics.
Collapse
Affiliation(s)
- Amon Siame
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Kaunda Yamba
- Department of Pathology and Microbiology, University Teaching Hospitals, Lusaka, Zambia
| | - Mulemba Samutela
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | - Andrew Mukubesa
- Department of Disease Control, School of Veterinary Medicine, Lusaka, Zambia
| | - Gina Mulundu
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| |
Collapse
|
27
|
Machulin AV, Abramov VM, Kosarev IV, Deryusheva EI, Priputnevich TV, Panin AN, Manoyan AM, Chikileva IO, Abashina TN, Blumenkrants DA, Ivanova OE, Papazyan TT, Nikonov IN, Suzina NE, Melnikov VG, Khlebnikov VS, Sakulin VK, Samoilenko VA, Gordeev AB, Sukhikh GT, Uversky VN, Karlyshev AV. A Novel Bifidobacterium longum Subsp. longum T1 Strain from Cow's Milk: Homeostatic and Antibacterial Activity against ESBL-Producing Escherichia coli. Antibiotics (Basel) 2024; 13:924. [PMID: 39452191 PMCID: PMC11505560 DOI: 10.3390/antibiotics13100924] [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: 08/28/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: The global emergence of antibiotic-resistant zooanthroponotic Escherichia coli strains, producing extended-spectrum beta-lactamases (ESBL-E) and persisting in the intestines of farm animals, has now led to the development of a pandemic of extra-intestinal infectious diseases in humans. The search for innovative probiotic microorganisms that eliminate ESBL-E from the intestines of humans and animals is relevant. Previously, we received three isolates of bifidobacteria: from milk of a calved cow (BLLT1), feces of a newborn calf (BLLT2) and feces of a three-year-old child who received fresh milk from this calved cow (BLLT3). Our goal was to evaluate the genetic identity of BLLT1, BLLT2, BLLT3 isolates using genomic DNA fingerprinting (GDF), to study the tolerance, adhesion, homeostatic and antibacterial activity of BLLT1 against ESBL-E. Methods: We used a complex of microbiological, molecular biological, and immunological methods, including next generation sequencing (NGS). Results: GDF showed that DNA fragments of BLLT2 and BLLT3 isolates were identical in number and size to DNA fragments of BLLT1. These data show for the first time the possibility of natural horizontal transmission of BLLT1 through with the milk of a calved cow into the intestines of a calf and the intestines of a child. BLLT1 was resistant to gastric and intestinal stresses and exhibited high adhesive activity to calf, pig, chicken, and human enterocytes. This indicates the unique ability of BLLT1 to inhabit the intestines of animals and humans. We are the first to show that BLLT1 has antibacterial activity against ESBL-E strains that persist in humans and animals. BLLT1 produced 145 ± 8 mM of acetic acid, which reduced the pH of the nutrient medium from 6.8 to 5.2. This had an antibacterial effect on ESBL-E. The genome of BLLT1 contains ABC-type carbohydrate transporter gene clusters responsible for the synthesis of acetic acid with its antibacterial activity against ESBL-E. BLLT1 inhibited TLR4 mRNA expression induced by ESBL-E in HT-29 enterocytes, and protected the enterocyte monolayers used in this study as a bio-model of the intestinal barrier. BLLT1 increased intestinal alkaline phosphatase (IAP) as one of the main molecular factors providing intestinal homeostasis. Conclusions: BLLT1 shows promise for the creation of innovative functional nutritional products for humans and feed additives for farm animals that will reduce the spread of ESBL-E strains in the food chain.
Collapse
Affiliation(s)
- Andrey V. Machulin
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia (N.E.S.)
| | - Vyacheslav M. Abramov
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia; (T.V.P.); (A.B.G.)
| | - Igor V. Kosarev
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia; (T.V.P.); (A.B.G.)
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Tatiana V. Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia; (T.V.P.); (A.B.G.)
| | - Alexander N. Panin
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Ashot M. Manoyan
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Irina O. Chikileva
- Blokhin National Research Center of Oncology, Ministry of Health, 115478 Moscow, Russia
| | - Tatiana N. Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia (N.E.S.)
| | - Dmitriy A. Blumenkrants
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Olga E. Ivanova
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | | | - Ilia N. Nikonov
- Federal State Budgetary Educational Institution of Higher Education, St. Petersburg State University of Veterinary Medicine, 196084 Saint Petersburg, Russia
| | - Nataliya E. Suzina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia (N.E.S.)
| | - Vyacheslav G. Melnikov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia
| | | | - Vadim K. Sakulin
- Institute of Immunological Engineering, 142380 Lyubuchany, Russia
| | - Vladimir A. Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia (N.E.S.)
| | - Alexey B. Gordeev
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia; (T.V.P.); (A.B.G.)
| | - Gennady T. Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia; (T.V.P.); (A.B.G.)
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Andrey V. Karlyshev
- Department of Biomolecular Sciences, School of Life Sciences, Chemistry and Pharmacy, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston upon Thames KT1 2EE, UK;
| |
Collapse
|
28
|
Gelalcha BD, Mohamed RI, Gelgie AE, Kerro Dego O. Molecular epidemiology of extended-spectrum beta-lactamase-producing- Klebsiella species in East Tennessee dairy cattle farms. Front Microbiol 2024; 15:1439363. [PMID: 39380685 PMCID: PMC11458399 DOI: 10.3389/fmicb.2024.1439363] [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: 05/27/2024] [Accepted: 07/30/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction The rising prevalence of Extended-Spectrum Beta-Lactamase (ESBL)-producing Klebsiella species (spp.) poses a significant threat to human and animal health and environmental safety. To address this pressing issue, a comprehensive study was undertaken to elucidate the burden and dissemination mechanisms of ESBL-Klebsiella spp. in dairy cattle farms. Methods Fifty-seven Klebsiella species were isolated on CHROMagar™ ESBL plates and confirmed with MADLI-TOF MS and whole genome sequenced from 14 dairy farms. Results and discussion Six families of beta-lactamase (bla) (bla CTX-M, bla SHV, bla TEM, bla OXY, bla OXA, and bla SED) were detected in ESBL-Klebsiella spp. genomes. Most (73%) of isolates had the first three types of beta-lactamase genes, with bla SHV being the most frequent, followed by bla CTX-M. Most (93%) isolates harbored two or more bla genes. The isolates were genotypically MDR, with 26 distinct types of antibiotic resistance genes (ARGs) and point mutations in gyrA, gyrB, and parC genes. The genomes also harbored 22 different plasmid replicon types, including three novel IncFII. The IncFII and Col440I plasmids were the most frequent and were associated with bla CTXM-27 and qnrB19 genes, respectively. Eighteen distinct sequence types (STs), including eight isolates with novel STs of K. pneumoniae, were detected. The most frequently occurring STs were ST353 (n = 8), ST469 (n = 6), and the novel ST7501 (n = 6). Clusters of ESBL-Klebsiella strains with identical STs, plasmids, and ARGs were detected in multiple farms, suggesting possible clonal expansion. The same ESBL variant was linked to identical plasmids in different Klebsiella STs in some farms, suggesting horizontal spread of the resistance gene. The high burden and dual spread mechanism of ESBL genes in Klebsiella species, combined with the emergence of novel sequence types, could swiftly increase the prevalence of ESBL-Klebsiella spp., posing significant risks to human, animal, and environmental health. Immediate action is needed to implement rigorous surveillance and control measures to mitigate this risk.
Collapse
Affiliation(s)
- Benti D. Gelalcha
- Department of Animal Science, The University of Tennessee, Knoxville, TN, United States
| | - Ruwaa I. Mohamed
- Department of Genome Science and Technology, The University of Tennessee, Knoxville, TN, United States
| | - Aga Edema Gelgie
- Department of Animal Science, The University of Tennessee, Knoxville, TN, United States
| | - Oudessa Kerro Dego
- Department of Animal Science, The University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
29
|
Yang L, Wu X, Wu G, Wu Y, Li H, Shao B. Association analysis of antibiotic and disinfectant resistome in human and foodborne E. coli in Beijing, China. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 944:173888. [PMID: 38866143 DOI: 10.1016/j.scitotenv.2024.173888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/21/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
The widespread use of chemical disinfectants and antibiotics poses a major threat to food safety and human health. However, the mechanisms of co-transmission of antimicrobial resistance genes (ARGs) and biocides and metal resistance genes (BMRGs) of foodborne pathogens in the food chain is still unclear. This study isolated 343 E. coli strains from animal-derived foods in Beijing and incorporated online data of human-derived E. coli strains from NCBI. Our results demonstrated a relatively uniform distribution of strains from various regions in Beijing, indicating a lack of region-specific clustering. Additionally, predominant sequence types varied between food- and human-derived strains, suggesting a preference for different hosts and environments. Phenotypic association analysis showed that the chlorine disinfectants peroxides had a significant positive correlation with tetracyclines. Many more ARGs and BMRGs were enriched in human-associated E. coli compared with those in chicken- and pork-origin. The quaternary ammonium compounds (QACs) resistance gene qacEΔ1 had a strong correlation with aminoglycoside resistance gene aadA5, folate pathway antagonist resistance gene dfrA17, sul1 and macrolide resistance gene mph(A). The correlation results indicated a significant association between the copper resistance gene cluster pco and the silver resistance gene cluster sil. Coexistence of many resistance genes was observed within the qacEΔ1 gene structure, with qacEΔ1-sul1 being the most common combination. Our findings demonstrated that the epidemiological spread of resistance is affected by a combination of heavy metals, disinfectants and antibiotic use, suggesting that the prevention and control strategies of antimicrobial resistance need to be multifaceted and comprehensive.
Collapse
Affiliation(s)
- Lu Yang
- Shanghai Anti-doping Laboratory, Shanghai University of Sport, Shanghai 200438, China; Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China
| | - Xuan Wu
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; School of Public Health, Capital Medical University, Beijing 100069, China
| | - Guoquan Wu
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yige Wu
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Hui Li
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Bing Shao
- Shanghai Anti-doping Laboratory, Shanghai University of Sport, Shanghai 200438, China; Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
30
|
Sung K, Nawaz M, Park M, Chon J, Khan SA, Alotaibi K, Khan AA. Comprehensive Genomic Analysis of Uropathogenic E. coli: Virulence Factors, Antimicrobial Resistance, and Mobile Genetic Elements. Pathogens 2024; 13:794. [PMID: 39338985 PMCID: PMC11434687 DOI: 10.3390/pathogens13090794] [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: 08/09/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Our whole-genome sequencing analysis of sixteen uropathogenic E. coli isolates revealed a concerning picture of multidrug resistance and potentially virulent bacteria. All isolates belonged to four distinct clonal groups, with the highly prevalent ST131 lineage being associated with extensive antibiotic resistance and virulence factors. Notably, all isolates exhibited multidrug resistance, with some resistant to as many as 12 antibiotics. Fluoroquinolone resistance stemmed primarily from efflux pumps and mutations in gyrase and topoisomerase genes. Additionally, we identified genes encoding resistance to extended-spectrum cephalosporins, trimethoprim/sulfamethoxazole, and various heavy metals. The presence of diverse plasmids and phages suggests the potential for horizontal gene transfer and the dissemination of virulence factors. All isolates harbored genomic islands containing virulence factors associated with adhesion, biofilm formation, and invasion. Genes essential for iron acquisition, flagella biosynthesis, secretion systems, and toxin production were also prevalent. Adding further complexity to understanding the isolates' genetic makeup, we identified CRISPR-Cas systems. This study underscores the need for continued genomic surveillance in understanding the pathogenic mechanisms and resistance profiles of uropathogenic E. coli to aid in developing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Kidon Sung
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.N.); (M.P.); (S.A.K.); (K.A.); (A.A.K.)
| | - Mohamed Nawaz
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.N.); (M.P.); (S.A.K.); (K.A.); (A.A.K.)
| | - Miseon Park
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.N.); (M.P.); (S.A.K.); (K.A.); (A.A.K.)
| | - Jungwhan Chon
- Department of Companion Animal Health, Inje University, Gimhae 50834, Republic of Korea;
| | - Saeed A. Khan
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.N.); (M.P.); (S.A.K.); (K.A.); (A.A.K.)
| | - Khulud Alotaibi
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.N.); (M.P.); (S.A.K.); (K.A.); (A.A.K.)
| | - Ashraf A. Khan
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (M.N.); (M.P.); (S.A.K.); (K.A.); (A.A.K.)
| |
Collapse
|
31
|
Gonçalves Pereira J, Fernandes J, Mendes T, Gonzalez FA, Fernandes SM. Artificial Intelligence to Close the Gap between Pharmacokinetic/Pharmacodynamic Targets and Clinical Outcomes in Critically Ill Patients: A Narrative Review on Beta Lactams. Antibiotics (Basel) 2024; 13:853. [PMID: 39335027 PMCID: PMC11428226 DOI: 10.3390/antibiotics13090853] [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: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Antimicrobial dosing can be a complex challenge. Although a solid rationale exists for a link between antibiotic exposure and outcome, conflicting data suggest a poor correlation between pharmacokinetic/pharmacodynamic targets and infection control. Different reasons may lead to this discrepancy: poor tissue penetration by β-lactams due to inflammation and inadequate tissue perfusion; different bacterial response to antibiotics and biofilms; heterogeneity of the host's immune response and drug metabolism; bacterial tolerance and acquisition of resistance during therapy. Consequently, either a fixed dose of antibiotics or a fixed target concentration may be doomed to fail. The role of biomarkers in understanding and monitoring host response to infection is also incompletely defined. Nowadays, with the ever-growing stream of data collected in hospitals, utilizing the most efficient analytical tools may lead to better personalization of therapy. The rise of artificial intelligence and machine learning has allowed large amounts of data to be rapidly accessed and analyzed. These unsupervised learning models can apprehend the data structure and identify homogeneous subgroups, facilitating the individualization of medical interventions. This review aims to discuss the challenges of β-lactam dosing, focusing on its pharmacodynamics and the new challenges and opportunities arising from integrating machine learning algorithms to personalize patient treatment.
Collapse
Affiliation(s)
- João Gonçalves Pereira
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
- Serviço de Medicina Intensiva, Hospital Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
| | - Joana Fernandes
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Serviço de Medicina Intensiva, Centro Hospitalar de Trás-os-Montes e Alto Douro, 5000-508 Vila Real, Portugal
| | - Tânia Mendes
- Serviço de Medicina Interna, Hospital Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
| | - Filipe André Gonzalez
- Serviço de Medicina Intensiva, Hospital Garcia De Orta, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Susana M Fernandes
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Serviço de Medicina Intensiva, Hospital Santa Maria, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
32
|
Mahmud Z, Manik MRK, Rahman A, Karim MM, Islam LN. Impact of untreated tannery wastewater in the evolution of multidrug-resistant bacteria in Bangladesh. Sci Rep 2024; 14:20379. [PMID: 39223208 PMCID: PMC11369239 DOI: 10.1038/s41598-024-71472-6] [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: 03/06/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
The tannery industry produces one of the worst contaminants, and unsafe disposal in nearby waterbodies and landfills has become an imminent threat to public health, especially when the resulting multidrug-resistant bacteria and heavy metals enter community settings and animal food chains. In this study, we have collected 10 tannery wastewater (TWW) samples and 10 additional non-tannery wastewater (NTW) samples to compare the chemical oxygen demand (COD), pH, biological oxygen demand (BOD), dissolved oxygen (DO), total dissolved solids (TDS), chromium concentration, bacterial load, and antibiotic resistance profiles. While COD, pH, and chromium concentration data were previously published from our lab, this part of the study uncovers that TWW samples had a significantly higher bacterial load, compared to the non-tannery wastewater samples (5.89 × 104 and 9.38 × 103 cfu/mL, respectively), higher BOD and TDS values, and significantly lower DO values. The results showed that 53.4, 46.7, 40.0, and 40.0% of the TWW isolates were resistant to ceftriaxone, erythromycin, nalidixic acid, and azithromycin, respectively. On the other hand, 20.0, 30.0, 50.0, and 40.0% of the NTW isolates were resistant to the same antibiotics, respectively. These findings suggest that the TWW isolates were more resistant to antibiotics than the NTW isolates. Moreover, the TWW isolates exhibited higher multidrug resistance than the NTW isolates, 33.33, and 20.00%, respectively. Furthermore, spearman correlation analysis depicts that there is a negative correlation between BOD and bacterial load up to a certain level (r = - 0.7749, p = 0.0085). In addition, there is also a consistent negative correlation between COD and bacterial load (r = - 0.7112, p = 0.0252) and TDS and bacterial load (r = - 0.7621, p = 0.0104). These findings suggest that TWW could pose a significant risk to public health and the environment and highlight the importance of proper wastewater treatment in tannery industries.
Collapse
Affiliation(s)
- Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Md Rasel Khan Manik
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Adua Rahman
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | | | - Laila N Islam
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| |
Collapse
|
33
|
Baqar Z, Sinwat N, Prathan R, Chuanchuen R. Meat ducks as carriers of antimicrobial-resistant Escherichia coli harboring transferable R plasmids. J Vet Sci 2024; 25:e62. [PMID: 39231787 PMCID: PMC11450392 DOI: 10.4142/jvs.24074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/28/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024] Open
Abstract
IMPORTANCE Antimicrobial resistance (AMR) is a serious public health threat. AMR bacteria and their resistance determinants in food can be transmitted to humans through the food chain and by direct contact and disseminate directly to the environment. OBJECTIVE This study examined the AMR characteristics and transferable R plasmids in Escherichia coli isolated from meat ducks raised in an open-house system. METHODS One hundred seventy-seven (n = 177) commensal E. coli were examined for their antimicrobial susceptibilities and horizontal resistance transfer. The plasmids were examined by PCR-based plasmid replicon typing (PBRT) and plasmid multi-locus sequence typing (pMLST). RESULTS The highest resistance rate was found against ampicillin (AMP, 83.0%) and tetracycline (TET, 81.9%), and most isolates exhibited multidrug resistance (MDR) (86.4%). The R plasmids were conjugally transferred when TET (n = 4), AMP (n = 3), and chloramphenicol (n = 3) were used as a selective pressure. The three isolates transferred resistance genes either in AMP or TET. The blaCTX-M1 gene resided on conjugative plasmids. Five replicon types were identified, of which Inc FrepB was most common in the donors (n = 13, 38.4%) and transconjugants (n = 16, 31.2%). Subtyping F plasmids revealed five distinct replicons combinations, including F47:A-:B- (n = 2), F29:A-:B23 (n = 1), F29:A-:B- (n = 1), F18:A-B:- (n = 1), and F4:A-:B- (n = 1). The chloramphenicol resistance was significantly correlated with the other AMR phenotypes (p < 0.05). CONCLUSIONS AND RELEVANCE The meat ducks harbored MDR E. coli and played an important role in the environmental dissemination of AMR bacteria and its determinants. This confirms AMR as a health issue, highlighting the need for routine AMR monitoring and surveillance of meat ducks.
Collapse
Affiliation(s)
- Zulqarnain Baqar
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
- Research Unit for Microbial Food Safety and Antimicrobial Resistance, Department of Veterinary Public Health, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Center for Antimicrobial Resistance Monitoring in Food-borne Pathogens, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nuananong Sinwat
- Departments of Farm Resources and Production Medicine, Faculty of Veterinary Medicine, Kasetsart University, Nakhon Pathom 73140, Thailand
| | - Rangsiya Prathan
- Research Unit for Microbial Food Safety and Antimicrobial Resistance, Department of Veterinary Public Health, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Center for Antimicrobial Resistance Monitoring in Food-borne Pathogens, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Rungtip Chuanchuen
- Research Unit for Microbial Food Safety and Antimicrobial Resistance, Department of Veterinary Public Health, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Center for Antimicrobial Resistance Monitoring in Food-borne Pathogens, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
34
|
Iwan E, Zając M, Bomba A, Olejnik M, Skarżyńska M, Wasiński B, Wieczorek K, Tłuścik K, Wasyl D. Phylogenetics and Mobilization of Genomic Traits of Cephalosporin-Resistant Escherichia coli Originated from Retail Meat. Pathogens 2024; 13:700. [PMID: 39204300 PMCID: PMC11357031 DOI: 10.3390/pathogens13080700] [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: 07/08/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Contaminations with cephalosporin-resistant Escherichia coli across the food chain may pose a significant threat to public health because those antimicrobials are critically important in human medicine. The impact of the presented data is especially significant concerning Poland's role as one of the leading food producers in the EU. This work aimed to characterize the genomic contents of cephalosporin-resistant Escherichia coli (n = 36) isolated from retail meat to expand the official AMR monitoring reported by EFSA. The ESBL mechanism was predominant (via blaCTX-M-1 and blaSHV-12), with the AmpC-type represented by the blaCMY-2 variant. The strains harbored multiple resistance genes, mainly conferring resistance to aminoglycosides, sulfonamides, trimethoprim, tetracyclines. In some isolates, virulence factors-including intimin (eae) and its receptor (tir) were detected, indicating significant pathogenic potential. Resistance genes showed a link with IncI1 and IncB/O/K/Z plasmids. Cephalosporinases were particularly linked to ISEc9/ISEc1 (blaCTX-M-1 and blaCMY-2). The association of virulence with mobile elements was less common-mostly with IncF plasmids. The analysis of E. coli isolated from retail meat indicates accumulation of ARGs and their association with various mobile genetic elements, thus increasing the potential for the transmission of resistance across the food chain.
Collapse
Affiliation(s)
- Ewelina Iwan
- Department of Omics Analyses, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland (M.O.); (D.W.)
| | - Magdalena Zając
- Department of Microbiology, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland (B.W.)
| | - Arkadiusz Bomba
- Department of Omics Analyses, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland (M.O.); (D.W.)
| | - Małgorzata Olejnik
- Department of Omics Analyses, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland (M.O.); (D.W.)
- Faculty of Biological and Veterinary Sciences, Department of Basic and Preclinical Sciences, Nicolaus Copernicus University in Torun, 11 Gagarina St., 87-100 Torun, Poland
| | - Magdalena Skarżyńska
- Department of Microbiology, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland (B.W.)
| | - Bernard Wasiński
- Department of Microbiology, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland (B.W.)
| | - Kinga Wieczorek
- Department of Food of Safety, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland;
| | - Katarzyna Tłuścik
- Department of Omics Analyses, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland (M.O.); (D.W.)
| | - Dariusz Wasyl
- Department of Omics Analyses, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland (M.O.); (D.W.)
- Department of Microbiology, National Veterinary Research Institute, 57 Partyzantow, 24-100 Pulawy, Poland (B.W.)
| |
Collapse
|
35
|
Nyhoegen C, Bonhoeffer S, Uecker H. The many dimensions of combination therapy: How to combine antibiotics to limit resistance evolution. Evol Appl 2024; 17:e13764. [PMID: 39100751 PMCID: PMC11297101 DOI: 10.1111/eva.13764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/30/2024] [Accepted: 07/14/2024] [Indexed: 08/06/2024] Open
Abstract
In combination therapy, bacteria are challenged with two or more antibiotics simultaneously. Ideally, separate mutations are required to adapt to each of them, which is a priori expected to hinder the evolution of full resistance. Yet, the success of this strategy ultimately depends on how well the combination controls the growth of bacteria with and without resistance mutations. To design a combination treatment, we need to choose drugs and their doses and decide how many drugs get mixed. Which combinations are good? To answer this question, we set up a stochastic pharmacodynamic model and determine the probability to successfully eradicate a bacterial population. We consider bacteriostatic and two types of bactericidal drugs-those that kill independent of replication and those that kill during replication. To establish results for a null model, we consider non-interacting drugs and implement the two most common models for drug independence-Loewe additivity and Bliss independence. Our results show that combination therapy is almost always better in limiting the evolution of resistance than administering just one drug, even though we keep the total drug dose constant for a 'fair' comparison. Yet, exceptions exist for drugs with steep dose-response curves. Combining a bacteriostatic and a bactericidal drug which can kill non-replicating cells is particularly beneficial. Our results suggest that a 50:50 drug ratio-even if not always optimal-is usually a good and safe choice. Applying three or four drugs is beneficial for treatment of strains with large mutation rates but adding more drugs otherwise only provides a marginal benefit or even a disadvantage. By systematically addressing key elements of treatment design, our study provides a basis for future models which take further factors into account. It also highlights conceptual challenges with translating the traditional concepts of drug independence to the single-cell level.
Collapse
Affiliation(s)
- Christin Nyhoegen
- Research Group Stochastic Evolutionary Dynamics, Department of Theoretical BiologyMax Planck Institute for Evolutionary BiologyPlonGermany
| | - Sebastian Bonhoeffer
- Department of Environmental Systems Science, Institute of Integrative BiologyETH ZurichZurichSwitzerland
| | - Hildegard Uecker
- Research Group Stochastic Evolutionary Dynamics, Department of Theoretical BiologyMax Planck Institute for Evolutionary BiologyPlonGermany
| |
Collapse
|
36
|
Al-Rabia MW, Asfour HZ, Alhakamy NA, Bazuhair MA, Ibrahim TS, Abbas HA, Mansour B, Hegazy WAH, Seleem NM. Cilostazol is a promising anti-pseudomonal virulence drug by disruption of quorum sensing. AMB Express 2024; 14:87. [PMID: 39090255 PMCID: PMC11294311 DOI: 10.1186/s13568-024-01740-1] [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: 04/01/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
Resistance to antibiotics is a critical growing public health problem that desires urgent action to combat. To avoid the stress on bacterial growth that evokes the resistance development, anti-virulence agents can be an attractive strategy as they do not target bacterial growth. Quorum sensing (QS) systems play main roles in controlling the production of diverse virulence factors and biofilm formation in bacteria. Thus, interfering with QS systems could result in mitigation of the bacterial virulence. Cilostazol is an antiplatelet and a vasodilator FDA approved drug. This study aimed to evaluate the anti-virulence activities of cilostazol in the light of its possible interference with QS systems in Pseudomonas aeruginosa. Additionally, the study examines cilostazol's impact on the bacterium's ability to induce infection in vivo, using sub-inhibitory concentrations to minimize the risk of resistance development. In this context, the biofilm formation, the production of virulence factors and influence on the in vivo ability to induce infection were assessed in the presence of cilostazol at sub-inhibitory concentration. Furthermore, the outcome of combination with antibiotics was evaluated. Cilostazol interfered with biofilm formation in P. aeruginosa. Moreover, swarming motility, biofilm formation and production of virulence factors were significantly diminished. Histopathological investigation revealed that liver, spleen and kidney tissues damage was abolished in mice injected with cilostazol-treated bacteria. Cilostazol exhibited a synergistic outcome when used in combination with antibiotics. At the molecular level, cilostazol downregulated the QS genes and showed considerable affinity to QS receptors. In conclusion, Cilostazol could be used as adjunct therapy with antibiotics for treating Pseudomonal infections. This research highlights cilostazol's potential to combat bacterial infections by targeting virulence mechanisms, reducing the risk of antibiotic resistance, and enhancing treatment efficacy against P. aeruginosa. These findings open avenues for repurposing existing drugs, offering new, safer, and more effective infection control strategies.
Collapse
Affiliation(s)
- Mohammed W Al-Rabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Hani Z Asfour
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Mohammed A Bazuhair
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Tarek S Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Hisham A Abbas
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Basem Mansour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
- Department of Pharmaceutical Chemistry, Kut University College, Al Kut, Wasit, 52001, Iraq
| | - Wael A H Hegazy
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
- Department of Pharmaceutical Sciences, Pharmacy Program, College of Health Sciences, 113, Muscat, Oman.
| | - Noura M Seleem
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
37
|
Ammar AM, Abd El-Aziz NK, Aggour MG, Ahmad AAM, Abdelkhalek A, Muselin F, Smuleac L, Pascalau R, Attia FA. A Newly Incompatibility F Replicon Allele (FIB81) in Extensively Drug-Resistant Escherichia coli Isolated from Diseased Broilers. Int J Mol Sci 2024; 25:8347. [PMID: 39125914 PMCID: PMC11312129 DOI: 10.3390/ijms25158347] [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: 06/21/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Multiple drug resistance (MDR) has gained pronounced attention among Enterobacterales. The transfer of multiple antimicrobial resistance genes, frequently carried on conjugative incompatibility F (IncF) plasmids and facilitating interspecies resistance transmission, has been linked to Salmonella spp. and E. coli in broilers. In Egypt, the growing resistance is exacerbated by the limited clinical efficacy of many antimicrobials. In this study, IncF groups were screened and characterized in drug-resistant Salmonella spp. and E. coli isolated from broilers. The antimicrobial resistance profile, PCR-based replicon typing of bacterial isolates pre- and post-plasmid curing, and IncF replicon allele sequence typing were investigated. Five isolates of E. coli (5/31; 16.13%) and Salmonella spp. (5/36; 13.89%) were pan-susceptible to the examined antimicrobial agents, and 85.07% of tested isolates were MDR and extensively drug-resistant (XDR). Twelve MDR and XDR E. coli and Salmonella spp. isolates were examined for the existence of IncF replicons (FII, FIA, and FIB). They shared resistance to ampicillin, ampicillin/sulbactam, amoxicillin/clavulanate, doxycycline, cefotaxime, and colistin. All isolates carried from one to two IncF replicons. The FII-FIA-FIB+ and FII-FIA+FIB- were the predominant replicon patterns. FIB was the most frequently detected replicon after plasmid curing. Three XDR E. coli isolates that were resistant to 12-14 antimicrobials carried a newly FIB replicon allele with four nucleotide substitutions: C99→A, G112→T, C113→T, and G114→A. These findings suggest that broilers are a significant reservoir of IncF replicons with highly divergent IncF-FIB plasmid incompatibility groups circulating among XDR Enterobacterales. Supporting these data with additional comprehensive epidemiological studies involving replicons other than the IncF can provide insights for implementing efficient policies to prevent the spreading of new replicons to humans.
Collapse
Affiliation(s)
- Ahmed M. Ammar
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt; (A.M.A.); (A.A.M.A.)
| | - Norhan K. Abd El-Aziz
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt; (A.M.A.); (A.A.M.A.)
| | | | - Adel A. M. Ahmad
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt; (A.M.A.); (A.A.M.A.)
| | - Adel Abdelkhalek
- Food Safety, Hygiene and Technology Department, Faculty of Veterinary Medicine, Badr University in Cairo (BUC), Badr 11829, Egypt;
| | - Florin Muselin
- Department of Toxicology, Faculty of Veterinary Medicine, University of Life Sciences “King Michael I” from Timisoara, 300645 Timisoara, Romania;
| | - Laura Smuleac
- Department of Sustainable Development and Environmental Engineering, Faculty of Agriculture, University of Life Sciences “King Mihai I” from Timisoara, 300645 Timisoara, Romania;
| | - Raul Pascalau
- Department of Agricultural Technologies, Faculty of Agriculture, University of Life Sciences “ King Mihai I” from Timisoara, 300645 Timisoara, Romania
| | | |
Collapse
|
38
|
Sarkar S, Okafor CC. Impact of Veterinary Feed Directive Rules Changes on the Prevalence of Antibiotic Resistance Bacteria Isolated from Cecal Samples of Food-Producing Animals at US Slaughterhouses. Pathogens 2024; 13:631. [PMID: 39204232 PMCID: PMC11357339 DOI: 10.3390/pathogens13080631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
This study examined the impact of the 2017 Veterinary Feed Directive (VFD) rule changes on the prevalence of tetracycline-resistant and erythromycin-resistant bacteria (Salmonella spp., Campylobacter spp., and Escherichia coli) in cecal samples of food animals (cattle, swine, chicken, and turkey) at US slaughterhouses. Multivariable logistic regression was used to analyze 2013-2019 cecal samples of food-producing animals surveillance data from the National Antimicrobial Resistance Monitoring System (NARMS) in the U.S. The variables included year (used to evaluate VFD rule changes), host, and quarter of year. The analysis of surveillance data showed that the VFD rule changes have varying effects on tetracycline-resistant and erythromycin-resistant bacteria in food animals. For example, the odds of detecting tetracycline-resistant Salmonella spp. decreased in cattle but increased in chickens following the implementation of the VFD rule changes. Similarly, the odds of detecting tetracycline-resistant Escherichia coli decreased in chickens but increased in swine after the VFD rule changes. The odds of detecting erythromycin-resistant Campylobacter spp. increased in cattle but decreased in chickens after the VFD rule changes. In conclusion, the implementation of VFD rule changes has been beneficial in reducing the odds of detecting tetracycline-resistant Escherichia coli and erythromycin-resistant Campylobacter spp. in chickens, as well as tetracycline-resistant Salmonella spp. in cattle at US slaughterhouses.
Collapse
Affiliation(s)
| | - Chika C. Okafor
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA;
| |
Collapse
|
39
|
Aborode AT, Kumar N, Olowosoke CB, Ibisanmi TA, Ayoade I, Umar HI, Jamiu AT, Bolarinwa B, Olapade Z, Idowu AR, Adelakun IO, Onifade IA, Akangbe B, Abacheng M, Ikhimiukor OO, Awaji AA, Adesola RO. Predictive identification and design of potent inhibitors targeting resistance-inducing candidate genes from E. coli whole-genome sequences. FRONTIERS IN BIOINFORMATICS 2024; 4:1411935. [PMID: 39132675 PMCID: PMC11310021 DOI: 10.3389/fbinf.2024.1411935] [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: 04/03/2024] [Accepted: 06/12/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction: This work utilizes predictive modeling in drug discovery to unravel potential candidate genes from Escherichia coli that are implicated in antimicrobial resistance; we subsequently target the gidB, MacB, and KatG genes with some compounds from plants with reported antibacterial potentials. Method: The resistance genes and plasmids were identified from 10 whole-genome sequence datasets of E. coli; forty two plant compounds were selected, and their 3D structures were retrieved and optimized for docking. The 3D crystal structures of KatG, MacB, and gidB were retrieved and prepared for molecular docking, molecular dynamics simulations, and ADMET profiling. Result: Hesperidin showed the least binding energy (kcal/mol) against KatG (-9.3), MacB (-10.7), and gidB (-6.7); additionally, good pharmacokinetic profiles and structure-dynamics integrity with their respective protein complexes were observed. Conclusion: Although these findings suggest hesperidin as a potential inhibitor against MacB, gidB, and KatG in E. coli, further validations through in vitro and in vivo experiments are needed. This research is expected to provide an alternative avenue for addressing existing antimicrobial resistances associated with E. coli's MacB, gidB, and KatG.
Collapse
Affiliation(s)
| | - Neeraj Kumar
- Department of Pharmaceutical Chemistry, Bhupal Nobles’ College of Pharmacy, Udaipur, Rajasthan, India
| | - Christopher Busayo Olowosoke
- Department of Biotechnology, Federal University of Technology, Akure, Nigeria
- Institute of Bioinformatics and Molecular Therapeutics, Osogbo, Nigeria
| | | | - Islamiyyah Ayoade
- Computer-Aided Therapeutic Discovery and Design Platform, Federal University of Technology, Akure, Nigeria
- Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Haruna Isiyaku Umar
- Computer-Aided Therapeutic Discovery and Design Platform, Federal University of Technology, Akure, Nigeria
- Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Abdullahi Temitope Jamiu
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Basit Bolarinwa
- College of Medicine, Richmond Gabriel University, Richmond, Saint Vincent and the Grenadines
| | - Zainab Olapade
- Department of Biology, Lamar University, Lamar, TX, United States
| | - Abidemi Ruth Idowu
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| | - Ibrahim O. Adelakun
- Department of Chemistry, State University of New York Albany, Albany, NY, United States
| | | | - Benjamin Akangbe
- Department of Epidemiology, School of Public Health, Georgia State University, Atlanta, GA, United States
| | - Modesta Abacheng
- School of Public Health, Georgia State University, Atlanta, GA, United States
| | - Odion O. Ikhimiukor
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, United States
| | - Aeshah A. Awaji
- Department of Biology, Faculty of Science, University College of Taymaa, University of Tabuk, Tabuk, Saudi Arabia
| | - Ridwan Olamilekan Adesola
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
40
|
Prieto A, Miró L, Margolles Y, Bernabeu M, Salguero D, Merino S, Tomas J, Corbera JA, Perez-Bosque A, Huttener M, Fernández LÁ, Juarez A. Targeting plasmid-encoded proteins that contain immunoglobulin-like domains to combat antimicrobial resistance. eLife 2024; 13:RP95328. [PMID: 39046772 PMCID: PMC11268884 DOI: 10.7554/elife.95328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024] Open
Abstract
Antimicrobial resistance (AMR) poses a significant threat to human health. Although vaccines have been developed to combat AMR, it has proven challenging to associate specific vaccine antigens with AMR. Bacterial plasmids play a crucial role in the transmission of AMR. Our recent research has identified a group of bacterial plasmids (specifically, IncHI plasmids) that encode large molecular mass proteins containing bacterial immunoglobulin-like domains. These proteins are found on the external surface of the bacterial cells, such as in the flagella or conjugative pili. In this study, we show that these proteins are antigenic and can protect mice from infection caused by an AMR Salmonella strain harboring one of these plasmids. Furthermore, we successfully generated nanobodies targeting these proteins, that were shown to interfere with the conjugative transfer of IncHI plasmids. Considering that these proteins are also encoded in other groups of plasmids, such as IncA/C and IncP2, targeting them could be a valuable strategy in combating AMR infections caused by bacteria harboring different groups of AMR plasmids. Since the selected antigens are directly linked to AMR itself, the protective effect extends beyond specific microorganisms to include all those carrying the corresponding resistance plasmids.
Collapse
Affiliation(s)
- Alejandro Prieto
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Luïsa Miró
- Department of Biochemistry and Physiology, Universitat de BarcelonaBarcelonaSpain
- Institut de Nutrició i Seguretat Alimentària, Universitat de BarcelonaBarcelonaSpain
| | - Yago Margolles
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC)MadridSpain
| | - Manuel Bernabeu
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - David Salguero
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Susana Merino
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Joan Tomas
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Juan Alberto Corbera
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Facultad de Veterinaria, Universidad de Las Palmas de Gran Canaria (ULPGC), Campus Universitario de ArucasLas PalmasSpain
| | - Anna Perez-Bosque
- Department of Biochemistry and Physiology, Universitat de BarcelonaBarcelonaSpain
- Institut de Nutrició i Seguretat Alimentària, Universitat de BarcelonaBarcelonaSpain
| | - Mario Huttener
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC)MadridSpain
| | - Antonio Juarez
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
- Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and TechnologyBarcelonaSpain
| |
Collapse
|
41
|
Fahy S, O’Connor JA, Sleator RD, Lucey B. From Species to Genes: A New Diagnostic Paradigm. Antibiotics (Basel) 2024; 13:661. [PMID: 39061343 PMCID: PMC11274079 DOI: 10.3390/antibiotics13070661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/05/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Molecular diagnostics has the potential to revolutionise the field of clinical microbiology. Microbial identification and nomenclature have, for too long, been restricted to phenotypic characterisation. However, this species-level view fails to wholly account for genetic heterogeneity, a result of lateral gene transfer, mediated primarily by mobile genetic elements. This genetic promiscuity has helped to drive virulence development, stress adaptation, and antimicrobial resistance in several important bacterial pathogens, complicating their detection and frustrating our ability to control them. We argue that, as clinical microbiologists at the front line, we must embrace the molecular technologies that allow us to focus specifically on the genetic elements that cause disease rather than the bacterial species that express them. This review focuses on the evolution of microbial taxonomy since the introduction of molecular sequencing, the role of mobile genetic elements in antimicrobial resistance, the current and emerging assays in clinical laboratories, and the comparison of phenotypic versus genotypic analyses. In essence, it is time now to refocus from species to genes as part of a new diagnostic paradigm.
Collapse
Affiliation(s)
- Sinead Fahy
- Department of Microbiology, Mercy University Hospital, T12 WE28 Cork, Ireland;
- Department of Biological Sciences, Munster Technological University, T12 P928 Cork, Ireland; (J.A.O.); (B.L.)
| | - James A. O’Connor
- Department of Biological Sciences, Munster Technological University, T12 P928 Cork, Ireland; (J.A.O.); (B.L.)
| | - Roy D. Sleator
- Department of Biological Sciences, Munster Technological University, T12 P928 Cork, Ireland; (J.A.O.); (B.L.)
| | - Brigid Lucey
- Department of Biological Sciences, Munster Technological University, T12 P928 Cork, Ireland; (J.A.O.); (B.L.)
| |
Collapse
|
42
|
Leclercq SO, Bochereau P, Foubert I, Baumard Y, Travel A, Doublet B, Baucheron S. Persistence of commensal multidrug-resistant Escherichia coli in the broiler production pyramid is best explained by strain recirculation from the rearing environment. Front Microbiol 2024; 15:1406854. [PMID: 39035436 PMCID: PMC11259971 DOI: 10.3389/fmicb.2024.1406854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/07/2024] [Indexed: 07/23/2024] Open
Abstract
Despite the success of mitigation policies in several countries to reduce the use of antibiotics in veterinary medicine, pathogenic and commensal bacteria resistant to antibiotics are still circulating in livestock animals. However, factors contributing the most to antimicrobial resistance (AMR) persistence in these settings are yet not clearly identified. The broiler production, with its highly segmented, pyramidal structure offers an ideal context to understand and control the spread of resistant bacteria. By taking advantage of an experimental facility reproducing the whole broiler production pyramid, we demonstrate that resistant E. coli persist in our system primarily though recirculation of a few commensal clones surviving in the rearing environment. No vertical transmission from hens to offspring nor strain acquisition at the hatchery were detected, while import of new strains from outside the facility seems limited. Moreover, each clone carries its own resistance-conferring plasmid(s), and a single putative plasmid horizontal transfer could have been inferred. These results, observed for now in a small experimental facility with high level of biosecurity, must be confirmed in a commercial farm context but still provide invaluable information for future mitigation policies.
Collapse
|
43
|
Shu G, Gan T, Lin Z, Liu Y, Chen J, Wang C, Deng L, Li C, Chang LJ, Zhang W, Li H, Xu F, Fu H, Lin J. The resistance patterns and molecular characteristics of ESBL/AmpC-producing Escherichia coli from captive panda ecosystem in China. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116395. [PMID: 38728939 DOI: 10.1016/j.ecoenv.2024.116395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024]
Abstract
Escherichia coli (E. coli) plays an important ecological role, and is a useful bioindicator to recognize the evolution of resistance in human, animal and environment. Recently, extended-spectrum β-lactamases (ESBL) producing E.coli has posed a threat to public health. Generally, captive healthy giant pandas are not exposed to antibiotics; however, they still acquire antimicrobial resistant bacteria. In order to understand whether there is an exchange of resistance genes within the ecosystems of captive giant pandas, this study explored resistance characteristics of 330 commensal E. coli isolates from feces of giant pandas, the surroundings, and breeders. Isolates from different sources showed similar resistance phenotype, and ESBL/AmpC-producing isolates showed more profound resistance to antibiotics than non-ESBL/AmpC-producing isolates (P<0.05). Furthermore, the occurrence of broad-spectrum β-lactamase related resistance genes and colistin resistance genes was detected, and isolates phylogenetic typing and multilocus sequence typing (MLST) were applied in this study. Seven different β-lactamase resistance genes (blaCTX-M-55, blaCTX-M-15, blaCTX-M-27, blaCTX-M-65, blaTEM-1, blaOXA-1 and blaCMY) and mcr-1 were found in 68 ESBL/AmpC-producing isolates. blaCTX-M-55 (48.53 %) was found the most predominant resistance genes, followed by blaTEM-1 (19.12 %) and blaCTX-M-27 (16.18 %). Nonetheless, blaCTX-M-55 was commonly detected in the isolates from giant pandas (63.16 %), the surroundings (43.48 %), and breeders (33.33 %). However, there were no carbapenemase genes detected in this study. mcr-1 was harbored in only one isolate from giant panda. Forty-five tansconjugants were successfully obtained in the conjugation experiments. The presence of antimicrobial resistance and related resistance genes tested were observed in the transconjugants. The results indicated that 52.63 % of the isolates from giant panda 73.91 % of the isolates from surroundings, and 100 % of the isolates from breeders were phylogroup A. Total of 27 sequence types (ST) were recognized from the isolate by MLST and found that ST48 (19/68; 27.94 %) was the predominant ST type, especially in the isolates from giant pandas and the surroundings. In conclusion, commensal ESBL/AmpC-producing E. coli becomes a reservoir of ESBL resistance genes, which is a potential threaten to health of giant pandas. The interaction between giant pandas, surroundings and breeders contribute to development of resistant phenotypes and genotypes which might transfer across species or the surroundings easily; hence, strict monitoring based on a "One Health" approach is recommended.
Collapse
Affiliation(s)
- Gang Shu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Ting Gan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Ziqin Lin
- The Chinese university of Hongkong (Shenzhen), Faculty of Medicine, Shenzhen 518172, PR China
| | - Ying Liu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Jingyi Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Chengdong Wang
- China Conservation and Research Centre for the Giant Panda, Key Laboratory of SFGA on The Giant Panda, Chengdu, Sichuan 610081, PR China
| | - Linhua Deng
- China Conservation and Research Centre for the Giant Panda, Key Laboratory of SFGA on The Giant Panda, Chengdu, Sichuan 610081, PR China
| | - Caiwu Li
- China Conservation and Research Centre for the Giant Panda, Key Laboratory of SFGA on The Giant Panda, Chengdu, Sichuan 610081, PR China
| | - Li-Jen Chang
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, WA 20541, USA
| | - Wei Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Haohuan Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Funeng Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Hualing Fu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Juchun Lin
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.
| |
Collapse
|
44
|
Bhushan G, Castano V, Wong Fok Lung T, Chandler C, McConville TH, Ernst RK, Prince AS, Ahn D. Lipid A modification of colistin-resistant Klebsiella pneumoniae does not alter innate immune response in a mouse model of pneumonia. Infect Immun 2024; 92:e0001624. [PMID: 38771050 PMCID: PMC11237409 DOI: 10.1128/iai.00016-24] [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: 01/26/2024] [Accepted: 04/26/2024] [Indexed: 05/22/2024] Open
Abstract
Polymyxin resistance in carbapenem-resistant Klebsiella pneumoniae bacteria is associated with high morbidity and mortality in vulnerable populations throughout the world. Ineffective antimicrobial activity by these last resort therapeutics can occur by transfer of mcr-1, a plasmid-mediated resistance gene, causing modification of the lipid A portion of lipopolysaccharide (LPS) and disruption of the interactions between polymyxins and lipid A. Whether this modification alters the innate host immune response or carries a high fitness cost in the bacteria is not well established. To investigate this, we studied infection with K. pneumoniae (KP) ATCC 13883 harboring either the mcr-1 plasmid (pmcr-1) or the vector control (pBCSK) ATCC 13883. Bacterial fitness characteristics of mcr-1 acquisition were evaluated. Differentiated human monocytes (THP-1s) were stimulated with KP bacterial strains or purified LPS from both parent isolates and isolates harboring mcr-1. Cell culture supernatants were analyzed for cytokine production. A bacterial pneumonia model in WT C57/BL6J mice was used to monitor immune cell recruitment, cytokine induction, and bacterial clearance in the bronchoalveolar lavage fluid (BALF). Isolates harboring mcr-1 had increased colistin MIC compared to the parent isolates but did not alter bacterial fitness. Few differences in cytokines were observed with purified LPS from mcr-1 expressing bacteria in vitro. However, in a mouse pneumonia model, no bacterial clearance defect was observed between pmcr-1-harboring KP and parent isolates. Consistently, no differences in cytokine production or immune cell recruitment in the BALF were observed, suggesting that other mechanisms outweigh the effect of these lipid A mutations in LPS.
Collapse
Affiliation(s)
- Gitanjali Bhushan
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Victor Castano
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Tania Wong Fok Lung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Courtney Chandler
- Department of Microbial Pathogenesis, University of Maryland, School of Dentistry, Baltimore, Maryland, USA
| | - Thomas H. McConville
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland, School of Dentistry, Baltimore, Maryland, USA
| | - Alice S. Prince
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Danielle Ahn
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
45
|
Bendary MM, Ali MAM, Abdel Halim AS, Boufahja F, Chaudhary AA, Elkelish A, Soliman RHM, Hegazy WAH. Investigating Sulforaphane's anti-virulence and anti-quorum sensing properties against Pseudomonas aeruginosa. Front Pharmacol 2024; 15:1406653. [PMID: 38835668 PMCID: PMC11148281 DOI: 10.3389/fphar.2024.1406653] [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: 03/25/2024] [Accepted: 04/29/2024] [Indexed: 06/06/2024] Open
Abstract
Background P. aeruginosa, a significant bacterium, can cause severe illness and resistance to antibiotics. Quorum sensing (QS) systems regulate virulence factors production. Targeting QS could reduce bacteria pathogenicity and prevent antibiotic resistance. Cruciferous vegetables contain sulforaphane, known for its anti-inflammatory, antioxidant, anticancer, and antimicrobial properties. Aim We aimed to examine the inhibitory influences of sulforaphane, at a sub-inhibitory concentration (¼ minimum inhibitory concentration, MIC), on virulence and QS in P. aeruginosa. Materials and methods The sulforaphane's anti-virulence actions at sub-inhibitory concentrations were explored in vitro and in vivo. A sub-MIC concentration of sulforaphane was combined with anti-pseudomonal drugs, and the results of this combination were assessed. The virtual affinity of sulforaphane for the receptors of QS was studied, and its effect on the expression of QS genes was quantified. Results Sulforaphane significantly decreased the biofilm formation, motility, ability to withstand oxidative stress, and the synthesis of virulence extracellular enzymes such as proteases, hemolysins, and elastase, as well as other virulence factors like pyocyanin. In addition, sulforaphane lessened the severity of P. aeruginosa infection in mice. Sulforaphane reduced the antipseudomonal antibiotics' MICs when used together, resulting in synergistic effects. The observed anti-virulence impacts were attributed to the ability of sulforaphane to inhibit QS via suppressing the QS genes' expression. Conclusion Sulforaphane shows promise as a potent anti-virulence and anti-QS agent that can be used alongside conventional antimicrobials to manage severe infections effectively. Furthermore, this study paves the way for further investigation of sulforaphane and similar structures as pharmacophores for anti-QS candidates.
Collapse
Affiliation(s)
- Mahmoud M Bendary
- Department of Microbiology and Immunology, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Mohamed A M Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Alyaa S Abdel Halim
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Fehmi Boufahja
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Amr Elkelish
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
- Department of Botany and Microbiology, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Rania H M Soliman
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Wael A H Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
- Pharmacy Program, Department of Pharmaceutical Sciences, Oman College of Health Sciences, Muscat, Oman
| |
Collapse
|
46
|
Latif H, Pazra DF, Basri C, Wibawan IWT, Rahayu P. Whole genome sequencing analysis on antibiotic-resistant Escherichia coli isolated from pig farms in Banten Province, Indonesia. J Vet Sci 2024; 25:e44. [PMID: 38834513 PMCID: PMC11156600 DOI: 10.4142/jvs.24031] [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] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 06/06/2024] Open
Abstract
IMPORTANCE The emergence and rapid increase in the incidence of multidrug-resistant (MDR) bacteria in pig farms has become a serious concern and reduced the choice of effective antibiotics. OBJECTIVE This study analyzed the phylogenetics and diversity of antibiotic resistance genes (ARGs) and molecularly identified the source of ARGs in antibiotic-resistant Escherichia coli isolated from pig farms in Banten Province, Indonesia. METHODS Forty-four antibiotic-resistant E. coli isolates from fecal samples from 44 pig farms in Banten Province, Indonesia, were used as samples. The samples were categorized into 14 clusters. Sequencing was performed using the Oxford Nanopore Technologies MinION platform, with barcoding before sequencing with Nanopore Rapid sequencing gDNA-barcoding (SQK-RBK110.96) according to manufacturing procedures. ARG detection was conducted using ResFinder, and the plasmid replicon was determined using PlasmidFinder. RESULTS Three phylogenetic leaves of E. coli were identified in the pig farming cluster in Banten Province. The E. coli isolates exhibited potential resistance to nine classes of antibiotics. Fifty-one ARGs were identified across all isolates, with each cluster carrying a minimum of 10 ARGs. The ant(3'')-Ia and qnrS1 genes were present in all isolates. ARGs in the E. coli pig farming cluster originated mainly from plasmids, accounting for an average of 89.4%. CONCLUSIONS AND RELEVANCE The elevated potential for MDR events, coupled with the dominance of ARGs originating from plasmids, increases the risk of ARG spread among bacterial populations in animals, humans, and the environment.
Collapse
Affiliation(s)
- Hadri Latif
- Veterinery Public Health and Epidemology Division, School of Veterinary Medicine and Biomedical Sciences (SVMBS), IPB University, Bogor 16680, Indonesia
| | - Debby Fadhilah Pazra
- Animal Health Division, Bogor Agricultural Development Polytechnic, Bogor 16730, Indonesia.
| | - Chaerul Basri
- Veterinery Public Health and Epidemology Division, School of Veterinary Medicine and Biomedical Sciences (SVMBS), IPB University, Bogor 16680, Indonesia
| | - I Wayan Teguh Wibawan
- Medical Microbiology Division, School of Veterinary Medicine and Biomedical Sciences (SVMBS), IPB University, Bogor 16680, Indonesia
| | - Puji Rahayu
- Quality Control Laboratory and Certification of Animal Products, Bogor 16161, Indonesia
| |
Collapse
|
47
|
Elfaky MA, Okairy HM, Abdallah HM, Koshak AE, Mohamed GA, Ibrahim SR, Alzain AA, Hegazy WA, Khafagy ES, Seleem NM. Assessing the antibacterial potential of 6-gingerol: Combined experimental and computational approaches. Saudi Pharm J 2024; 32:102041. [PMID: 38558886 PMCID: PMC10981156 DOI: 10.1016/j.jsps.2024.102041] [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: 04/04/2024] Open
Abstract
The rise of antibiotic resistance in bacteria is becoming a global concern, particularly due to the dwindling supply of new antibiotics. This situation mandates the discovery of new antimicrobial candidates. Plant-derived natural compounds have historically played a crucial role in the development of antibiotics, serving as a rich source of substances possessing antimicrobial properties. Numerous studies have supported the reputation of 6-gingerol, a prominent compound found in the ginger family, for its antibacterial properties. In this study, the antibacterial activities of 6-gingerol were evaluated against Gram-negative bacteria, Acinetobacter baumannii and Klebsiella pneumoniae, with a particular focus on the clinically significant Gram-negative Pseudomonas aeruginosa and Gram-positive bacteria Staphylococcus aureus. Furthermore, the anti-virulence activities were assessed in vitro, in vivo, and in silico. The current findings showed that 6-gingerol's antibacterial activity is due to its significant effect on the disruption of the bacterial cell membrane and efflux pumps, as it significantly decreased the efflux and disrupted the cell membrane of S. aureus and P. aeruginosa. Furthermore, 6-gingerol significantly decreased the biofilm formation and production of virulence factors in S. aureus and P. aeruginosa in concentrations below MICs. The anti-virulence properties of 6-gingerol could be attributed to its capacity to disrupt bacterial virulence-regulating systems; quorum sensing (QS). 6-Gingerol was found to interact with QS receptors and downregulate the genes responsible for QS. In addition, molecular docking, and molecular dynamics (MD) simulation results indicated that 6-gingerol showed a comparable binding affinity to the co-crystalized ligands of different P. aeruginosa QS targets as well as stable interactions during 100 ns MD simulations. These findings suggest that 6-gingerol holds promise as an anti-virulence agent that can be combined with antibiotics for the treatment of severe infections.
Collapse
Affiliation(s)
- Mahmoud A. Elfaky
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hassan M. Okairy
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hossam M. Abdallah
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulrahman E. Koshak
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Gamal A. Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sabrin R.M. Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah 21442, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Abdulrahim A. Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Wael A.H. Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
- Pharmacy Program, Department of Pharmaceutical Sciences, Oman College of Health Sciences, Muscat 113, Oman
| | - El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Noura M. Seleem
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
48
|
Tetteh FKM, Ablordey A, Obeng-Nkrumah N, Opintan JA. Extended-spectrum beta-lactamases in clinical isolates of Escherichia coli and Klebsiella pneumoniae recovered from patients at the Tamale Teaching Hospital, Ghana. PLoS One 2024; 19:e0300596. [PMID: 38578750 PMCID: PMC10997077 DOI: 10.1371/journal.pone.0300596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 02/27/2024] [Indexed: 04/07/2024] Open
Abstract
INTRODUCTION Extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli and Klebsiella pneumoniae are pathogens of significant public health interest for which new antibiotics are urgently needed. AIM To determine the prevalence of ESBLs in E. coli and K. pneumoniae isolates from patients attending the Tamale Teaching Hospital (TTH) in Ghana. METHODOLOGY The study was a cross-sectional study involving convenience sampling of E. coli and K. pneumoniae isolates from consenting patients' clinical specimens, between April and June 2015. Antimicrobial susceptibility test was performed, and ESBL-producer phenotypes were further screened for BlaTEM, BlaSHV, and BlaCTX-M genes. Patients' clinical data were additionally collected using a structured questionnaire. RESULTS Of the 150 non-duplicate E. coli and K. pneumoniae isolates identified, 140 were confirmed as E. coli (84%, n = 117) and K. pneumoniae (16%, n = 23). Of these, sixty-two (44%) [E. coli (84%; n = 52); K. pneumoniae (16%; n = 10)] phenotypically expressed ESBLs. The proportion of ESBL-producing isolates was higher in adults (15-65 years) than in neonates (< 28 days) (p = 0.14). Most of the isolates showed a high percentage resistance to ampicillin (96%) and tetracycline (89%), but a relatively lower resistance to amikacin (36%). No isolate was resistant to meropenem. More ESBL producers were multidrug resistant compared to non-ESBL-producers [23% (14/62) versus 18% (14/78); p = 0.573]. Overall, 74% (n = 46) of the ESBL genotypes expressed BlaCTX-M-1 genes, followed by 63% (n = 39) BlaTEM, and 16% (n = 10) BlaSHV. The study showed a high prevalence of ESBL-positive E. coli and K. pneumoniae, mostly CTX-M-1 producers at TTH. CONCLUSION Routine laboratory ESBL screening is warranted to inform patient management.
Collapse
Affiliation(s)
- Francis Kwame Morgan Tetteh
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
- Pathology Division, 37 Military Hospital, Accra, Ghana
| | - Anthony Ablordey
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Noah Obeng-Nkrumah
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | | |
Collapse
|
49
|
Shang Y, Zhang Y, Wang R, Peng Y, Ding B, Liu Y, Li C, Feng L, Liu H, Yang C, Tang Y. Deciphering the molecular and functional basis of TMexCD1: the plasmid-encoded efflux pump of resistance-nodulation-division superfamily. Antimicrob Agents Chemother 2024; 68:e0167823. [PMID: 38477539 PMCID: PMC10989000 DOI: 10.1128/aac.01678-23] [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: 12/20/2023] [Accepted: 02/15/2024] [Indexed: 03/14/2024] Open
Abstract
Horizontal gene transfer has been demonstrated to be an important driver for the emergency of multidrug-resistant pathogens. Recently, a transferable gene cluster tmexCD1-toprJ1 of the resistance-nodulation-division (RND) superfamily was identified in the plasmids of animal-derived Klebsiella pneumoniae strains, with a higher efflux capacity for various drugs than the Escherichia coli AcrAB-TolC homolog system. In this study, we focused on the differences in the inner membrane pump of these two systems and identified some key residues that contribute to the robust efflux activity of the TMexCD1 system. With the aid of homologous modeling and molecular docking, eight residues from the proximal binding pocket (PBP) and nine from the distal binding pocket (DBP) were selected and subjected to site-directed mutagenesis. Several of them, such as S134, I139, D181, and A290, were shown to be important for substrate binding in the DBP region, and all residues in PBP and DBP showed certain substrate preferences. Apart from the conservative switch loop (L613-623TMexD1) previously identified in the E. coli AcrB (EcAcrB), a relatively unconservative loop (L665-675TMexD1) at the bottom of PBP was proposed as a critical element for the robust activity of TMexD1, due to variations at sites E669, G670, N673, and S674 compared to EcAcrAB, and the significantly altered efflux activity due to their mutations. The conservation and flexibility of these key factors can contribute to the evolution of the RND efflux pumps and thus serve as potential targets for developing inhibitors to block the widespread of the TMexCD1 system.
Collapse
Affiliation(s)
- Yan Shang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
- Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Ye Zhang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
| | - Ruimin Wang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
| | - Yishu Peng
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
| | - Bo Ding
- Shandong Institute for Food and Drug Control, Jinan, China
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yuanxiang Liu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
| | - Chongzhou Li
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
| | - Luhua Feng
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
| | - Honglei Liu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
| | - Chunyu Yang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
| | - Yajie Tang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
50
|
Ulrich JU, Epping L, Pilz T, Walther B, Stingl K, Semmler T, Renard BY. Nanopore adaptive sampling effectively enriches bacterial plasmids. mSystems 2024; 9:e0094523. [PMID: 38376263 PMCID: PMC10949517 DOI: 10.1128/msystems.00945-23] [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: 09/04/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Bacterial plasmids play a major role in the spread of antibiotic resistance genes. However, their characterization via DNA sequencing suffers from the low abundance of plasmid DNA in those samples. Although sample preparation methods can enrich the proportion of plasmid DNA before sequencing, these methods are expensive and laborious, and they might introduce a bias by enriching only for specific plasmid DNA sequences. Nanopore adaptive sampling could overcome these issues by rejecting uninteresting DNA molecules during the sequencing process. In this study, we assess the application of adaptive sampling for the enrichment of low-abundant plasmids in known bacterial isolates using two different adaptive sampling tools. We show that a significant enrichment can be achieved even on expired flow cells. By applying adaptive sampling, we also improve the quality of de novo plasmid assemblies and reduce the sequencing time. However, our experiments also highlight issues with adaptive sampling if target and non-target sequences span similar regions. IMPORTANCE Antimicrobial resistance causes millions of deaths every year. Mobile genetic elements like bacterial plasmids are key drivers for the dissemination of antimicrobial resistance genes. This makes the characterization of plasmids via DNA sequencing an important tool for clinical microbiologists. Since plasmids are often underrepresented in bacterial samples, plasmid sequencing can be challenging and laborious. To accelerate the sequencing process, we evaluate nanopore adaptive sampling as an in silico method for the enrichment of low-abundant plasmids. Our results show the potential of this cost-efficient method for future plasmid research but also indicate issues that arise from using reference sequences.
Collapse
Affiliation(s)
- Jens-Uwe Ulrich
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany
- Department of Mathematics and Computer Science, Free University of Berlin, Berlin, Germany
- Phylogenomics Unit, Center for Artificial Intelligence in Public Health Research, Robert Koch Institute, Wildau, Germany
| | - Lennard Epping
- Genome Sequencing and Genomic Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Tanja Pilz
- Genome Sequencing and Genomic Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Birgit Walther
- Advanced Light and Electron Microscopy, Robert Koch Institute, Berlin, Germany
| | - Kerstin Stingl
- National Reference Laboratory for Campylobacter, Department of Biological Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Torsten Semmler
- Genome Sequencing and Genomic Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Bernhard Y. Renard
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany
| |
Collapse
|