1
|
Moulick S, Bera R, Roy DN. Bactericidal action of ginger (Zingiber officinale Roscoe) extract against Escherichia coli through synergistic modulation of the AcrAB-TolC efflux pump and inhibition of peptidoglycan synthesis: In vitro and in silico approaches. Microb Pathog 2025; 204:107624. [PMID: 40268150 DOI: 10.1016/j.micpath.2025.107624] [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: 11/13/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
The emergence of multidrug-resistant Escherichia coli is considered a severe threat to global health, largely attributed to the bacterium's ability to expel antibiotics via efflux pump systems. This study explores the antibacterial efficacy of a methanol extract derived from Zingiber officinale R. (ginger), a traditional medicinal spice, against an E. coli strain overexpressing the AcrAB-TolC efflux system. To evaluate the extract's efficacy, three E. coli strains were tested: AG100 (AcrAB-TolC+), AG100A (ΔAcrAB), and D22 (lpxC mutant). The ginger extract exhibited antibacterial activity against E. coli AG100A and D22, with minimal inhibitory concentrations (MIC) and minimal bactericidal concentrations (MBC) of 625 μg/mL. However, the extract showed no inhibitory effect against E. coli AG100, even at 10 mg/mL, suggesting the AcrAB-TolC system plays a key role in resistance. Notably, combining the extract with an efflux pump inhibitor (EPI) strongly enhanced its bactericidal effect, reducing the MBC for AG100 to 313 μg/mL. EtBr accumulation assays confirmed that the ginger extract, in combination with EPI, amplified intracellular drug retention, peaking fluorescence within 30 min and sustaining elevated levels over 60 min. Molecular docking further revealed that bioactive compounds such as 6-shogaol strongly bind within the binding domain of AcrB homotrimer, inhibiting pump function. Additionally, cell wall biosynthesis assays demonstrated 69-75 % inhibition when the ginger extract was used at 2-fold-4-fold its MIC in the presence of EPI, further intensifying bactericidal effects. These results underscore ginger's dual-action mechanisms, highlighting its potential as an effective natural antimicrobial agent against drug-resistant E. coli.
Collapse
Affiliation(s)
- Soumitra Moulick
- TCG Lifesciences Private Limited, Kolkata, West Bengal, India; Department of Biotechnology, National Institute of Technology - Raipur, Chhattisgarh, India
| | - Rammohan Bera
- TCG Lifesciences Private Limited, Kolkata, West Bengal, India
| | - Dijendra Nath Roy
- Department of Biotechnology, National Institute of Technology - Raipur, Chhattisgarh, India.
| |
Collapse
|
2
|
Masalane NS, Bester LA, Ismail A, Essack SY, Mbanga J. Drug resistant Klebsiella pneumoniae from patients and hospital effluent: a correlation? BMC Microbiol 2025; 25:284. [PMID: 40350461 PMCID: PMC12066053 DOI: 10.1186/s12866-025-03987-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND The application of wastewater-based epidemiology has gained traction as a cost effective tool in antimicrobial resistance (AMR) surveillance with studies showing a correlation between the presence of resistant bacteria from hospital sewage and patients. This study compared Klebsiella pneumoniae from patients and hospital effluent in terms of antibiotic resistance patterns, antibiotic resistance genes (ARGs), mobile genetic elements (MGEs) and phylogenomic relationships. RESULTS Pooled effluent samples were collected from the final effluent point of a regional hospital and K. pneumoniae isolates were identified on selective media. Clinical isolates were also collected from the same hospital. Antimicrobial susceptibility testing (AST) was performed using the VITEK® 2 system. DNA was extracted prior to whole genome sequencing (WGS). The resistome, mobilome, and phylogenetic lineages of sequenced isolates were assessed using bioinformatics analysis. A total of 10 randomly selected presumptive and 10 clinical K. pneumoniae constituted the sample and were subjected to AST. Total resistance was observed in the clinical samples to cefuroxime, cefotaxime, piperacillin/tazobactam, gentamicin, tobramycin and trimethoprim/sulfamethoxazole. The effluent isolates exhibited total susceptibility to most antibiotics but showed resistance to amoxicillin/clavulanic acid and piperacillin/tazobactam (100%), and tigecycline (10%). The effluent isolates did not exhibit a diverse resistome, while the clinical isolates harboured genes conferring resistance to aminoglycoside (aph(6)-Id, aph(3'')-Ib, aac(6')-Ib-cr, aadA16), ß-lactam (blaSVH group, blaOXA group, blaTEM group), and fluoroquinolone (oqxA, oqxB) antibiotics. Only class 1 integrons were identified. Phylogenetic analysis revealed that effluent isolates from this study were not closely related to the clinical isolates. CONCLUSION This study showed no correlation between the resistance profiles of the clinical and effluent isolates. The relationship between AMR in hospital effluent and clinical resistance may depend on the antimicrobial agents and bacterial species studied.
Collapse
Affiliation(s)
- Naledi S Masalane
- Antimicrobial Research Unit, College of Health Sciences, University of Kwazulu-Natal, Private Bag X54001, Durban, 4000, South Africa
| | - Linda A Bester
- Biomedical Resource Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Arshad Ismail
- Sequencing Core Facility, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, South Africa
| | - Sabiha Y Essack
- Antimicrobial Research Unit, College of Health Sciences, University of Kwazulu-Natal, Private Bag X54001, Durban, 4000, South Africa
- School of Pharmacy, University of Jordan, Amman, Jordan
| | - Joshua Mbanga
- Antimicrobial Research Unit, College of Health Sciences, University of Kwazulu-Natal, Private Bag X54001, Durban, 4000, South Africa.
- Department of Applied Biology and Biochemistry, National University of Science and Technology, AC939, Ascot, Bulawayo, Zimbabwe.
| |
Collapse
|
3
|
Abdelkhalig SM, Elmanakhly AR, Alblwi NAN, Alharbi NK, Alhomrani M, Alamri AS, Alshehri F, Mosbah RA, Safwat NA, AbdElrahman M, Bendary MM. Comparative analysis of diarrheagenic and uropathogenic Escherichia coli isolates: antimicrobial resistance, virulence, and genomic profiling. J Appl Microbiol 2025; 136:lxaf082. [PMID: 40216405 DOI: 10.1093/jambio/lxaf082] [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: 01/02/2025] [Revised: 02/16/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
AIMS Escherichia coli is a key pathogen causing gastrointestinal and urinary tract infections. Diarrheagenic E. coli (DEC) and uropathogenic E. coli (UPEC) are distinct major pathotypes linked to specific clinical outcomes. Therefore, this study aimed to compare DEC and UPEC isolates regarding distribution, antimicrobial resistance, serotypes, resistance, and virulence gene profiles. MATERIALS AND METHODS A total of 400 clinical samples (200 stools and 200 urine) were analyzed using phenotypic and genotypic methods. Antimicrobial resistance, serotyping, and detection of resistance and virulence genes were performed. Phylogenetic and correlation analyses were conducted to explore genetic relationships and interactions. RESULTS Of 97 E. coli isolates (24.25% prevalence), 56 DEC and 41 UPEC were detected. DEC isolates primarily included serotypes O26, O45, and O55, while UPEC predominantly featured O1 and O25. UPEC showed higher multidrug resistance, while DEC was more virulent. UPEC carried unique markers (ureC, papC), and DEC harbored stx and aggR genes associated with gastrointestinal infections. Phylogenetic analysis showed separate clustering for DEC and UPEC, with limited genetic overlap. Correlation analysis identified strong associations within resistance and virulence genes but a negative correlation between these traits. CONCLUSION This study compared the phenotypic and genetic features of DEC and UPEC, highlighting their distinct pathogenic traits. Limited genetic overlap suggests potential gene transfer, influencing adaptability, and evolution.
Collapse
Affiliation(s)
- Sozan M Abdelkhalig
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Airport Road, Al-Qirawan District, Riyadh 11597, Saudi Arabia
| | - Arwa R Elmanakhly
- Department of Microbiology and Immunology, Faculty of Pharmacy, Modern University of Technology and Information, South Teseen Street, Fifth Settlement, New Cairo, Cairo 11559, Egypt
| | - Noaf Abdullah N Alblwi
- Al Hadithah General Hospital, King Fahd Road, Al-Hadithah District, Al-Qurayyat 77431, Saudi Arabia
| | - Nada K Alharbi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Airport Road, Al-Narjis District, Riyadh 11671, Saudi Arabia
| | - Majid Alhomrani
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Science, Taif University, Al-Hawiyah District, Airport Road,Taif 26432, Saudi Arabia
- Research Center for health science, Deanship of Scientific Research, Taif University, Airport Road, Al-Hawiyah District, Taif 26432, Saudi Arabia
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Science, Taif University, Al-Hawiyah District, Airport Road,Taif 26432, Saudi Arabia
- Research Center for health science, Deanship of Scientific Research, Taif University, Airport Road, Al-Hawiyah District, Taif 26432, Saudi Arabia
| | - Fatma Alshehri
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Airport Road, Al-Narjis District, Riyadh 11671, Saudi Arabia
| | - Rasha A Mosbah
- Infection Control Unit, Zagazig University Hospital, University Street, Al Shark District, Zagazig 44511, Egypt
| | - Nesreen A Safwat
- Department of Microbiology and Immunology, Faculty of Pharmacy, Modern University of Technology and Information, South Teseen Street, Fifth Settlement, New Cairo, Cairo 11559, Egypt
| | - Mohamed AbdElrahman
- Clinical Pharmacy Department, College of Pharmacy, Al-Mustaqbal University, Hilla-Kifl Road, Babylon District, Babylon 51001, Iraq
- Clinical Pharmacy Department, Badr University Hospital, Faculty of Medicine, Helwan University, Badr City, Suez Road, Cairo, 11795, Egypt
| | - Mahmoud M Bendary
- Department of Microbiology and Immunology, Faculty of Pharmacy, Port Said University, Al-Dawahy District, 23 July Street, Port Said 42511, Egypt
| |
Collapse
|
4
|
Chauhan SS, Jamal T, Gupta A, Parthasarathi R. Co-development of efflux pump inhibitors with antibiotics on targeting structural and mutational aspects of AcrB subunit. Mol Divers 2025:10.1007/s11030-025-11204-8. [PMID: 40287901 DOI: 10.1007/s11030-025-11204-8] [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: 03/24/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
The rise of antimicrobial resistance (AMR) of the routinely used antibiotics is ineffective against drug-resistant pathogenic strains of Escherichia coli, set off with limited treatment choices, costs, and increasing mortality rates. Multidrug efflux pumps have been identified as crucial determinants of AMR, flushing numerous antibiotics from cells in a non-specific way, and have emerged as promising drug targets to overcome AMR. Herein, the work focuses on determining structural and mutational insights of tripartite efflux pump subunit AcrB by executing multiple sequence alignment (MSA); the residues 615 and 617 at the substrate-binding site were identified mutated from an aromatic amino acid, phenylalanine, to an aliphatic amino acid, alanine. The study proceeded with the co-development of AcrB antagonist's by applying pharmacokinetic parameters filters to 40,613 natural compounds and molecular docking of single compounds, multiple ligand simultaneous docking (MLSD), molecular dynamics (MD) simulations, principal component analysis (PCA), and free energy landscape (FEL) analysis by considering resistant antibiotics. The identified mutations in the AcrB subunit are responsible for upregulating the activity of the AcrAB-TolC efflux pump and leading to a reduced concentration of antibiotics in the bacterial cytoplasm, ultimately increasing antibiotic resistance. Furthermore, based upon compound screening against target AcrB, 3-Hydroxyfumiquinazoline A shows competitive interaction with the antibiotic Erythromycin. A similar interaction pattern was observed between Sungucine and Cheatoglobosin D with Novobiocin while Procheatoglobosin I and Chaetoglobosin Q with Fusidic acid. Our findings highlight a novel class of efflux pump inhibitors (EPIs) that effectively antagonize the AcrB subunit and could serve as novel adjuvant alternatives for reviving antibiotic activity in resistant bacteria.
Collapse
Affiliation(s)
- Shweta Singh Chauhan
- REACT-Computational Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Tanya Jamal
- REACT-Computational Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Anshika Gupta
- REACT-Computational Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Ramakrishnan Parthasarathi
- REACT-Computational Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| |
Collapse
|
5
|
Duda-Madej A, Viscardi S, Niezgódka P, Szewczyk W, Wińska K. The Impact of Plant-Derived Polyphenols on Combating Efflux-Mediated Antibiotic Resistance. Int J Mol Sci 2025; 26:4030. [PMID: 40362268 PMCID: PMC12071758 DOI: 10.3390/ijms26094030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The global healthcare system is increasingly challenged by the rising prevalence of multidrug-resistant bacteria and the limited therapeutic options for related infections. Efflux-mediated antibiotic resistance represents a significant obstacle, primarily due to the absence of drugs specifically designed to target bacterial efflux pumps. Recent research has identified polyphenols, a broad class of plant-derived organic compounds, as potential inhibitors of efflux pump activity. This review consolidates data on the inhibitory properties of eight widely distributed polyphenols: curcumin, quercetin, luteolin, tannic acid, naringenin, epigallocatechin-3-gallate, ellagic acid, and resveratrol. These compounds have demonstrated the capacity to inhibit efflux pumps, either through direct interference with bacterial protein function or by downregulating the expression of genes encoding pump subunits. Importantly, several polyphenols exhibit synergistic interactions with antibiotics, including colistin, ciprofloxacin, and tetracycline. For instance, quercetin has shown inhibitory potency comparable to that of established efflux pump inhibitors such as verapamil and reserpine. These findings suggest that polyphenols represent promising candidates for the development of novel efflux pump inhibitors. However, further research is required to validate their efficacy and safety and facilitate their translation into clinical applications for combating antibiotic resistance.
Collapse
Affiliation(s)
- Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 4, 50-368 Wrocław, Poland
| | - Szymon Viscardi
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.); (P.N.); (W.S.)
| | - Piotr Niezgódka
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.); (P.N.); (W.S.)
| | - Wiktoria Szewczyk
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.); (P.N.); (W.S.)
| | - Katarzyna Wińska
- Department of Food Chemistry and Biocatalysis, Faculty of Biotechnology and Food Science, Wrocław University of Environmental and Life Sciences, C. K. Norwida 25, 50-375 Wrocław, Poland;
| |
Collapse
|
6
|
Kerek Á, Román I, Szabó Á, Kovács D, Kardos G, Kovács L, Jerzsele Á. Antibiotic resistance genes in Escherichia coli - literature review. Crit Rev Microbiol 2025:1-35. [PMID: 40249005 DOI: 10.1080/1040841x.2025.2492156] [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: 11/04/2023] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
Antimicrobial resistance threatens humans and animals worldwide and is recognized as one of the leading global public health issues. Escherichia coli (E. coli) has an unquestionable role in carrying and transmitting antibiotic resistance genes (ARGs), which in many cases are encoded on plasmids or phage, thus creating the potential for horizontal gene transfer. In this literature review, the authors summarize the major antibiotic resistance genes occurring in E. coli bacteria, through the major antibiotic classes. The aim was not only listing the resistance genes against the clinically relevant antibiotics, used in the treatment of E. coli infections, but also to cover the entire resistance gene carriage in E. coli, providing a more complete picture. We started with the long-standing antibiotic groups (beta-lactams, aminoglycosides, tetracyclines, sulfonamides and diaminopyrimidines), then moved toward the newer groups (phenicols, peptides, fluoroquinolones, nitrofurans and nitroimidazoles), and in every group we summarized the resistance genes grouped by the mechanism of their action (enzymatic inactivation, antibiotic efflux, reduced permeability, etc.). We observed that the frequency of antibiotic resistance mechanisms changes in the different groups.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - István Román
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Ábel Szabó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Dóra Kovács
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Gábor Kardos
- One Health Institute, University of Debrecen, Debrecen, Hungary
- National Public Health Center, Budapest, Hungary
- Department of Gerontology, Faculty of Health Sciences, University of Debrecen, Nyíregyháza, Hungary
| | - László Kovács
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
- Department of Animal Hygiene, Herd Health and Mobile Clinic, University of Veterinary Medicine, Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
| |
Collapse
|
7
|
Chauhan R, Patel H, Bhardwaj B, Suryawanshi V, Rawat S. Copper induced augmentation of antibiotic resistance in Acinetobacter baumannii MCC 3114. Biometals 2025; 38:485-504. [PMID: 39708209 DOI: 10.1007/s10534-024-00657-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
Increasing antibiotic resistance among the common nosocomial pathogen i.e. Acinetobacter baumannii poses life threat to the health care workers as well as to the society. The dissemination of antibiotic resistance in this pathogen at an alarming rate could be not only due to the overuse of antibiotics but also due to the stress caused by exposure of bacterium to several environmental contaminants in their niches. In the present study, effect of copper stress on augmentation in the antibiotic resistance of A. baumannii MCC 3114 against three clinically used antibiotics was investigated along with the phenotypic and genotypic alterations in the cell. It induced 8, 44 and 22-fold increase in resistance against colistin, ciprofloxacin and levofloxacin, respectively. Moreover, the biofilm formation of adapted culture was significantly enhanced due to a dense EPS around the cell (as revealed by SEM images). The structural changes in EPS were demonstrated by FTIR spectroscopy. The adequate growth of adapted MCC 3114 despite increased level of ROS indicates its persistence in copper and ROS stress. The physiological alterations in cell viz., increased efflux pump activity and decreased membrane permeability was observed. Molecular analysis revealed increased expression of efflux pump related genes, oxidative stress genes, integron and antibiotic resistance genes. In sum, our study revealed that the exposure of the critical pathogen, A. baunmannii to copper in hospital settings and environmental reservoirs can impose adaptive pressure which may lead to genotypic as well phenotypic changes in cell resulting into the augmentation of antibiotic resistance.
Collapse
Affiliation(s)
- Ravi Chauhan
- Microbiology Lab, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, 382030, India
| | - Hardi Patel
- Microbiology Lab, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, 382030, India
| | - Bhavna Bhardwaj
- Microbiology Lab, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, 382030, India
| | - Vijay Suryawanshi
- Microbiology Lab, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, 382030, India
| | - Seema Rawat
- Microbiology Lab, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, 382030, India.
| |
Collapse
|
8
|
Zhang D, Kukkar D, Bhatt P, Kim KH, Kaur K, Wang J. Novel nanomaterials-based combating strategies against drug-resistant bacteria. Colloids Surf B Biointerfaces 2025; 248:114478. [PMID: 39778220 DOI: 10.1016/j.colsurfb.2024.114478] [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: 11/09/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Numerous types of contemporary antibiotic treatment regimens have become ineffective with the increasing incidence of drug tolerance. As a result, it is pertinent to seek novel and innovative solutions such as antibacterial nanomaterials (NMs) for the prohibition and treatment of hazardous microbial infections. Unlike traditional antibiotics (e.g., penicillin and tetracycline), the unique physicochemical characteristics (e.g., size dependency) of NMs endow them with bacteriostatic and bactericidal potential. However, it is yet difficult to mechanistically predict or decipher the networks of molecular interaction (e.g., between NMs and the biological systems) and the subsequent immune responses. In light of such research gap, this review outlines various mechanisms accountable for the inception of drug tolerance in bacteria. It also delineates the primary factors governing the NMs-induced molecular mechanisms against microbes, specifically drug-resistant bacteria along with the various NM-based mechanisms of antibacterial activity. The review also explores future directions and prospects for NMs in combating drug-resistant bacteria, while addressing challenges to their commercial viability within the healthcare industry.
Collapse
Affiliation(s)
- Daohong Zhang
- Yantai Key Laboratory of Nanoscience and Technology for Prepared Food, Yantai Engineering Research Center of Green Food Processing and Quality Control, College of Food Engineering, Ludong University, Yantai, Shandong 264025, China
| | - Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India.
| | - Poornima Bhatt
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, South Korea.
| | - Kamalpreet Kaur
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab 140406, India
| | - Jianlong Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
9
|
Guo Y, Yang B, Zhou X, Gong Z, Wang E, Pan Y, Zhao Y, Liu H. Proteomic Analysis Reveals the Phenotypic Heterogeneity and Tolerance Mechanisms of Halophilic Vibrio parahaemolyticus Under Dual Stress of Low Salinity and Bile Salts in the Human Intestine. Biomolecules 2025; 15:518. [PMID: 40305270 PMCID: PMC12024522 DOI: 10.3390/biom15040518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Vibrio parahaemolyticus, a halophilic Gram-negative bacterium commonly found in aquatic products, can colonize the human small intestine, causing gastroenteritis and potentially leukemia. As a major intestinal pathogen, it poses a significant threat to public health. This study aims to investigate the phenotypic heterogeneity of V. parahaemolyticus in the low-salinity and bile salt environments of the human intestinal tract and to elucidate its mechanisms of tolerance and pathogenicity using proteomics. The experimental results indicated that under the low salinity and bile salts conditions of the human intestinal environment, the growth, motility, and biofilm formation of the strains were significantly inhibited. Proteomics analysis revealed that, under these conditions, the energy metabolism, chemotaxis system, flagellar motor, and ribosome-related proteins of V. parahaemolyticus were significantly affected, thereby influencing its growth, motility, and biofilm formation. Furthermore, the activation of the secretion system, particularly the T2SS, enhanced the virulence of secreted factors on host cells. Additionally, the activation of the β-lactam resistance pathway increased resistance to the intestinal environment, thereby enhancing the pathogenicity of V. parahaemolyticus.
Collapse
Affiliation(s)
- Yingying Guo
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (Y.G.); (X.Z.); (Z.G.); (E.W.); (Y.P.)
| | - Bing Yang
- Shanghai Majorbio Bio-Pharm Technology Co., Ltd., Shanghai 201318, China;
| | - Xiaoyan Zhou
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (Y.G.); (X.Z.); (Z.G.); (E.W.); (Y.P.)
| | - Zhangxi Gong
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (Y.G.); (X.Z.); (Z.G.); (E.W.); (Y.P.)
| | - Enxiao Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (Y.G.); (X.Z.); (Z.G.); (E.W.); (Y.P.)
| | - Yingjie Pan
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (Y.G.); (X.Z.); (Z.G.); (E.W.); (Y.P.)
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai 201306, China
- Laboratory of Quality & Safety Risk Assessment for Aquatic Product on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai 201306, China
| | - Yong Zhao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (Y.G.); (X.Z.); (Z.G.); (E.W.); (Y.P.)
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai 201306, China
- Laboratory of Quality & Safety Risk Assessment for Aquatic Product on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai 201306, China
| | - Haiquan Liu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (Y.G.); (X.Z.); (Z.G.); (E.W.); (Y.P.)
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai 201306, China
- Laboratory of Quality & Safety Risk Assessment for Aquatic Product on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai 201306, China
- Engineering Research Center of Food Thermal-Processing Technology, Shanghai Ocean University, Shanghai 201306, China
- Food Industry Chain Ecological Recycling Research Institute, Food Science and Technology College, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
10
|
Sun T, Delaplace P, Li G, James A, Pan J, Zhang J. Novel insights into the effect of arbuscular mycorrhizal fungi inoculation in soils under long-term biosolids application: Emphasis on antibiotic and metal resistance genes, and mobile genetic elements. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 369:125846. [PMID: 39952592 DOI: 10.1016/j.envpol.2025.125846] [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: 11/25/2024] [Revised: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
The application of biosolids can improve soil fertility and crop productivity but also accompanies risks of heavy metals and antibiotics introduction. In the presence of heavy metals contamination, using arbuscular mycorrhizal fungi (AMF) is a promising strategy to enhance soil microbial community stability and plant tolerance resistance to heavy metals, and to reduce the spread of antibiotic resistance genes (ARGs). The present study investigated the impacts of AMF inoculation on soil and plant heavy metal contents, and soil microbial communities by pot experiments. The results showed that AMF inoculation significantly enhanced plant biomass, and reduced soil and plant heavy metals contents. While AMF inoculation did not alter bacterial and fungal community compositions, it increased bacterial diversity at higher biosolids concentrations. Notably, AMF inoculation enhanced microbial network complexity and increased keystone taxa abundance. Furthermore, several beneficial microorganisms with high resistance to heavy metals were enriched in AMF-inoculated soils. Metagenomic analysis revealed a reduction in the mobile genetic element (MGE) gene IS91 in AMF-inoculated soils and an increase in heavy metal resistance genes compared to soils without AMF. The possibility of reduction in MGE-mediated spread of ARGs is one of the key findings of this study. As a caution, this study also detected enrichment of few ARGs in high biosolids-amended soils with AMF inoculation. Overall, AMF inoculation could be a valuable strategy in agriculture for mitigating the environmental risks associated with biosolids, heavy metals and antibiotic resistance, thereby promoting sustainable soil management and health.
Collapse
Affiliation(s)
- Tao Sun
- State Key Laboratory of Efficient Utilization of Arable Land in China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing, 100081, China; Gembloux Agro Bio Tech, University of Liège, 5030, Belgium
| | | | - Guihua Li
- State Key Laboratory of Efficient Utilization of Arable Land in China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Anina James
- State Key Laboratory of Efficient Utilization of Arable Land in China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Junting Pan
- State Key Laboratory of Efficient Utilization of Arable Land in China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Jianfeng Zhang
- State Key Laboratory of Efficient Utilization of Arable Land in China, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences, Beijing, 100081, China; Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, 130112, China.
| |
Collapse
|
11
|
Baek S, Seo J, Yun T, Kim J, Shin Y, Choi J, Chang J, Kim I, Yang YH, Kim W, Lee W. Heavy metals promote the formation of multidrug-tolerant Staphylococcus aureus and Escherichia coli persisters. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 293:118014. [PMID: 40080939 DOI: 10.1016/j.ecoenv.2025.118014] [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: 11/21/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Bacterial persisters are dormant phenotypic variants that are tolerant to antibiotics, contributing to treatment failure and the emergence of antimicrobial resistance. Although the formation of persisters has been extensively studied in regards to bacterial infections and treatment, such as antibiotic exposure or intracellular survival within macrophages, the role of environmental stressors in persister formation remains largely unexplored. In this study, we investigate the role of environmental heavy metals, specifically arsenic (As), cadmium (Cd), and mercury (Hg), in promoting persister cell formation in Staphylococcus aureus and Escherichia coli. Log-phase cultures were exposed to heavy metals (5 mM As, 1.25 mM Cd, 4 µM Hg for S. aureus; 12.5 mM As, 2 mM Cd, and 15 µM Hg for E. coli) for 0.5 h to induce persister cells. We observed that exposure to these metals induced persister cell formation, confirmed by intracellular ATP levels through microscopy and luciferase assays, as well as by reactive oxygen species (ROS) levels using carboxy-H2DCFDA. Short-term heavy metal exposure strongly depleted intracellular ATP while generating ROS. Moreover, we observed enhanced expression of genes involved in the SOS response, including recA, umuC, dinB, rexA, rexB, sulA, rpoS, and soxR, as measured by qPCR. This response was likely induced by elevated ROS levels following heavy metal exposure. Furthermore, we demonstrate that heavy metal-induced bacterial persisters exhibited a substantially increased emergence of antibiotic resistance, as shown by ciprofloxacin resistance developing in the presence of heavy metals. Therefore, our results clearly demonstrate that heavy metals can induce persister cells by depleting cellular ATP and generating ROS, and these bacterial responses to heavy metals substantially contribute to antibiotic resistance. These findings highlight the intricate relationship between environmental heavy metals, bacterial persister formation, and antibiotic resistance, emphasizing the need for a "One Health" strategy to address the growing antibiotic resistance crisis.
Collapse
Affiliation(s)
- Seongeun Baek
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jinbeom Seo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Taegwan Yun
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jin Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - YuJin Shin
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jiwoo Choi
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - JuOae Chang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Inseo Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yung-Hun Yang
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
12
|
Zhang T, Yan L, Qi J, Su R, Li X, Sun S, Song Y, Wei M, Zhang D. Antibiotic resistance genes in the coastal atmosphere under varied weather conditions: Distribution, influencing factors, and transmission mechanisms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125695. [PMID: 39824332 DOI: 10.1016/j.envpol.2025.125695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Antibiotic resistance genes (ARGs) have escalated to levels of concern worldwide as emerging environmental pollutants. Increasing evidence suggests that non-antibiotic antimicrobial substances expedite the spread of ARGs. However, the drivers and mechanisms involved in the generation and spread of ARGs in the atmosphere remain inadequately elucidated. Co-occurrence networks, mantel test analysis, and partial least squares path modeling were used to analyze the symbiotic relationships of ARGs with meteorological conditions, atmospheric pollutants, water-soluble inorganic ions, bacteria, mobile genetic elements (MGEs), antibacterial biocide and metal resistance genes, and to identify the direct drivers of ARGs. The types and abundance of ARGs exhibited different seasonal distribution. Specifically, the types exhibited a strong alignment with the diversity of air masses terrestrial sources, while the abundance displayed a significant positive correlation with both biocide resistance genes (BRGs) and metal resistance genes (MRGs). The contribution of bacterial communities and MGEs to the generation and spread of ARGs was constrained by the low levels of antibiotics in the atmosphere and the existence of "viral intermediates". Conversely, antibacterial biocides and metals influenced mutation rates, cellular SOS responses, and oxidative stress of bacteria, consequently facilitating the generation and spread of ARGs. Moreover, the co-selection among their derivatives, resistance genes, ensured a stable presence of ARGs. The research highlighted the significant impact of residual antimicrobial substances on both the generation and spread of ARGs. Elucidating the sources of aerosols and the co-selection mechanism linking with ARGs, BRGs, and MRGs were crucial for preserving the stability of ARGs in the atmosphere.
Collapse
Affiliation(s)
- Ting Zhang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, PR China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, PR China
| | - Lingchong Yan
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, PR China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, PR China
| | - Jianhua Qi
- Key Laboratory of Marine Environment and Ecology, Ministry of Education, Ocean University of China, Qingdao, 266100, PR China
| | - Rongguo Su
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, PR China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, PR China
| | - Xianguo Li
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, PR China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, PR China
| | - Shaohua Sun
- Laoshan District Meteorological Bureau, Qingdao, 266107, PR China
| | - Yongzhong Song
- Jufeng Peak Tourist Management Service Center of Laoshan Scenic Spot, Qingdao, 266100, PR China
| | - Mingming Wei
- Laoshan District Meteorological Bureau, Qingdao, 266107, PR China
| | - Dahai Zhang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, PR China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, PR China.
| |
Collapse
|
13
|
Brunet M, Amin SA, Bodachivskyi I, Kuzhiumparambil U, Seymour JR, Raina JB. An atlas of metabolites driving chemotaxis in prokaryotes. Nat Commun 2025; 16:1242. [PMID: 39890791 PMCID: PMC11785958 DOI: 10.1038/s41467-025-56410-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: 03/18/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
Chemicals inducing chemotaxis have been characterised for over 60 years across hundreds of publications. Without any synthesis of these scattered results, our current understanding of the molecules affecting prokaryotic behaviours is fragmented. Here, we examined 341 publications to assemble a comprehensive database of prokaryotic chemoeffectors, compiling the effect (attractant, repellent or neutral) of 926 chemicals previously tested and the chemotactic behaviour of 394 strains. Our analysis reveals that (i) not all chemical classes trigger chemotaxis equally, in particular, amino acids and benzenoids are much stronger attractants than carbohydrates; (ii) over one-quarter of attractants tested are not used for growth but solely act as chemotactic signals; (iii) the prokaryote's origin matters, as terrestrial strains respond to 50% more chemicals than those originating from human or marine biomes; (iv) repellents affect cell behaviour at concentrations 10-fold higher than attractants; (v) the effect of large molecules and the behaviour of bacteria other than Proteobacteria have been largely overlooked. Taken together, our findings provide a unifying view of the chemical characteristics that affect prokaryotic behaviours globally.
Collapse
Affiliation(s)
- Maéva Brunet
- Climate Change Cluster, University of Technology Sydney, Ultimo, NSW, Australia
| | - Shady A Amin
- Marine Microbiomics Lab, Biology Program, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Iurii Bodachivskyi
- Department of Chemistry of Bioactive Nitrogen Containing Heterocyclic Bases, V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry of the NAS of Ukraine, Kyiv, Ukraine
| | | | - Justin R Seymour
- Climate Change Cluster, University of Technology Sydney, Ultimo, NSW, Australia
| | - Jean-Baptiste Raina
- Climate Change Cluster, University of Technology Sydney, Ultimo, NSW, Australia.
| |
Collapse
|
14
|
Wright M, Kaur M, Thompson LK, Cox G. A historical perspective on the multifunctional outer membrane channel protein TolC in Escherichia coli. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:6. [PMID: 39863731 PMCID: PMC11762307 DOI: 10.1038/s44259-025-00078-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Since its discovery nearly 60 years ago, TolC has been associated with various cellular functions in Escherichia coli, including the efflux of environmental stressors and virulence factors. It also acts as a receptor for specific bacteriophages and the colicin E1 toxin. This review highlights key discoveries over the past six decades and emphasizes the remaining gaps in understanding how TolC contributes to physiological functions in E. coli.
Collapse
Affiliation(s)
- Mallory Wright
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, ON, Canada
| | - Mandeep Kaur
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, ON, Canada
| | - Laura K Thompson
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, ON, Canada
| | - Georgina Cox
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, ON, Canada.
| |
Collapse
|
15
|
Hamami E, Huo W, Hernandez-Bird J, Castaneda A, Bai J, Syal S, Ortiz-Marquez JC, van Opijnen T, Geisinger E, Isberg RR. Identification of determinants that allow maintenance of high-level fluoroquinolone resistance in Acinetobacter baumannii. mBio 2025; 16:e0322124. [PMID: 39589129 PMCID: PMC11708032 DOI: 10.1128/mbio.03221-24] [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: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/27/2024] Open
Abstract
Acinetobacter baumannii is associated with multidrug-resistant infections in healthcare settings, with fluoroquinolones such as ciprofloxacin being currently ineffective. Clinical isolates largely harbor mutations in the GyrA and TopoIV fluoroquinolone targets, as well as mutations that increase expression of drug resistance-nodulation-division (RND) efflux pumps. Factors critical for maintaining fitness levels of pump overproducers are uncharacterized despite their prevalence in clinical isolates. We, here, identify proteins that contribute to the fitness of fluoroquinolone-resistant (FQR) strains overexpressing three known RND systems using high-density insertion mutagenesis. Overexpression of the AdeFGH efflux pump caused hypersensitization to defects in outer membrane homeostatic regulation, including lesions that reduced lipooligosaccharide (LOS) biosynthesis and blocked production of the major A. baumannii porin. In contrast, AdeAB pump hyperexpression, in the absence of elevated adeC expression (the outer membrane component of the pump), was relatively tolerant to loss of these functions, consistent with the outer membrane protein being the primary disruptive component. Surprisingly, overexpression of proton-transporting efflux pumps had little impact on cytosolic pH, consistent with a compensatory response to pump activity. The most striking transcriptional changes were associated with AdeFGH pump overexpression, including the activation of the phenylacetate (PAA) degradation regulon. Disruption of the PAA pathway resulted in cytosolic acidification and defective expression of genes involved in protection from oxidative stress. These results indicate that RND efflux pump overproduction is compensated by maintenance of outer membrane integrity in A. baumannii to facilitate fitness of FQR isolates.IMPORTANCEAcinetobacter baumannii is a pathogen that often causes multidrug-resistant infections in healthcare settings, presenting a threat to the efficacy of known therapeutic interventions. Fluoroquinolones such as ciprofloxacin are currently ineffective against a majority of clinical A. baumannii isolates, many of which express pumps that remove this antibiotic class from within the bacterium. Three of these pumps can be found in most clinical isolates, with one of the three often hyperproduced at all times. In this study, we identify proteins that are necessary for the fitness of pump hyperproducers. The identified proteins are necessary to stabilize the outer membrane and allow the cytoplasm to tolerate the accumulation of ions as a consequence of excess pump activity. These results point to strategies for developing therapies that combine known antibiotics with drugs that target proteins important for survival of strains hyper-expressing efflux pumps.
Collapse
Affiliation(s)
- Efrat Hamami
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Wenwen Huo
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Juan Hernandez-Bird
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Arnold Castaneda
- Department of Biology, Tufts University, Medford, Massachusetts, USA
| | - Jinna Bai
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Sapna Syal
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Juan C. Ortiz-Marquez
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
- Innovation Laboratory, Broad Institute, Cambridge, Massachusetts, USA
| | - Tim van Opijnen
- Innovation Laboratory, Broad Institute, Cambridge, Massachusetts, USA
| | - Edward Geisinger
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Ralph R. Isberg
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Azeem K, Fatima S, Ali A, Ubaid A, Husain FM, Abid M. Biochemistry of Bacterial Biofilm: Insights into Antibiotic Resistance Mechanisms and Therapeutic Intervention. Life (Basel) 2025; 15:49. [PMID: 39859989 PMCID: PMC11767195 DOI: 10.3390/life15010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Biofilms, composed of structured communities of bacteria embedded in a self-produced extracellular matrix, pose a significant challenge due to their heightened resistance to antibiotics and immune responses. This review highlights the mechanisms underpinning antibiotic resistance within bacterial biofilms, elucidating the adaptive strategies employed by microorganisms to withstand conventional antimicrobial agents. This encompasses the role of the extracellular matrix, altered gene expression, and the formation of persister cells, contributing to the recalcitrance of biofilms to eradication. A comprehensive understanding of these resistance mechanisms provides a for exploring innovative therapeutic interventions. This study explores promising avenues for future research, emphasizing the necessity of uncovering the specific genetic and phenotypic adaptations occurring within biofilms. The identification of vulnerabilities in biofilm architecture and the elucidation of key biofilm-specific targets emerge as crucial focal points for the development of targeted therapeutic strategies. In addressing the limitations of traditional antibiotics, this review discusses innovative therapeutic approaches. Nanomaterials with inherent antimicrobial properties, quorum-sensing inhibitors disrupting bacterial communication, and bacteriophages as biofilm-specific viral agents are highlighted as potential alternatives. The exploration of combination therapies, involving antimicrobial agents, biofilm-disrupting enzymes, and immunomodulators, is emphasized to enhance the efficacy of existing treatments and overcome biofilm resilience.
Collapse
Affiliation(s)
- Kashish Azeem
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Sadaf Fatima
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Asghar Ali
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
- Clinical Biochemistry Laboratory, Department of Biochemistry, School of Chemical and Life Science, Jamia Hamdard, New Delhi 110062, India
| | - Ayesha Ubaid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Abid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| |
Collapse
|
17
|
Rajput P, Nahar KS, Rahman KM. Evaluation of Antibiotic Resistance Mechanisms in Gram-Positive Bacteria. Antibiotics (Basel) 2024; 13:1197. [PMID: 39766587 PMCID: PMC11672434 DOI: 10.3390/antibiotics13121197] [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: 10/24/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The prevalence of resistance in Gram-positive bacterial infections is rapidly rising, presenting a pressing global challenge for both healthcare systems and economies. The WHO categorizes these bacteria into critical, high, and medium priority groups based on the urgency for developing new antibiotics. While the first priority pathogen list was issued in 2017, the 2024 list remains largely unchanged. Despite six years having passed, the progress that has been made in developing novel treatment approaches remains insufficient, allowing antimicrobial resistance to persist and worsen on a global scale. Various strategies have been implemented to address this growing threat by targeting specific resistance mechanisms. This review evaluates antimicrobial resistance (AMR) in Gram-positive bacteria, highlighting its critical impact on global health due to the rise of multidrug-resistant pathogens. It focuses on the unique cell wall structure of Gram-positive bacteria, which influences their identification and susceptibility to antibiotics. The review explores the mechanisms of AMR, including enzymatic inactivation, modification of drug targets, limiting drug uptake, and increased drug efflux. It also examines the resistance strategies employed by high-priority Gram-positive pathogens such as Staphylococcus aureus, Streptococcus pneumoniae, and Enterococcus faecium, as identified in the WHO's 2024 priority list.
Collapse
Affiliation(s)
- Pratiksing Rajput
- Institute of Pharmaceutical Science, King’s College London, 150 Stamford Street, London SE1 9NH, UK;
| | - Kazi S. Nahar
- Department of Natural Sciences, Faculty of Science & Technology, Middlesex University, The Burroughs, Hendon, London NW4 4BT, UK;
| | - Khondaker Miraz Rahman
- Institute of Pharmaceutical Science, King’s College London, 150 Stamford Street, London SE1 9NH, UK;
| |
Collapse
|
18
|
El-Demerdash AS, Kamel SA, Elariny EYT, Henidi H, Mahran Y, Alahdal H, Saleh AM, Ibrahim RA. Natural Inhibitors of Salmonella MDR Efflux Pumps AcrAB and AcrD: An Integrated In Silico, Molecular, and In Vitro Investigation. Int J Mol Sci 2024; 25:12949. [PMID: 39684663 DOI: 10.3390/ijms252312949] [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: 10/25/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Multidrug-resistant (MDR) Salmonella remains a significant global health threat. This study aimed to explore the potential of essential oil components as novel inhibitors of the Salmonella MDR efflux pumps AcrAB and AcrD. Salmonella isolates were characterized for serotype, antibiotic resistance, and efflux pump activity. Essential oil components were screened for inhibitory effects using phenotypic and genotypic methods. In silico docking and molecular dynamics simulations were conducted to investigate binding interactions and stability. Salmonella Typhimurium was the predominant serotype with high MDR rates. Efflux pump activity was prevalent. Cumin and cinnamon oils demonstrated promising inhibitory effects on these pumps. Molecular docking simulations revealed strong binding affinities of analyzed compounds to the AcrAB and AcrD binding pocket. The 2-methyl-1-(p-tolyl)propan-2-ol exhibited higher stability within the AcrAB binding pocket compared to (1S,3R,5R)-1-isopropyl-4-methylenebicyclo[3.1.0]hexan-3-ol within the AcrD binding pocket. Treatment with these oils significantly downregulated efflux pump genes (robA, acrB, mdtB, acrF, acrD, soxS, mdsB, marA). The novel approach of combining in silico and molecular dynamics simulations with precise gene expression analysis provides a valuable framework for future studies aimed at combating MDR Salmonella efflux pumps.
Collapse
Affiliation(s)
- Azza S El-Demerdash
- Laboratory of Biotechnology, Department of Microbiology, Agricultural Research Center (ARC), Animal Health Research Institute (AHRI), Zagazig 44516, Egypt
| | - Shimaa A Kamel
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Eman Y T Elariny
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Hanan Henidi
- Research Department, Natural and Health Sciences Research Center, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Yasmin Mahran
- Research Department, Natural and Health Sciences Research Center, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Hadil Alahdal
- Department of Biology, College of Sciences, Princess Nourah bint Abdulrahman University, Riyadh 13415, Saudi Arabia
| | - Abdulrahman M Saleh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, KasrEl-Aini Street, Cairo 11562, Egypt
- Infection Control and Epidemiology Surveillance Unit, Aweash El-Hagar Family Medicine Center, Ministry of Health and Population (MOHP), Mansoura 35711, Egypt
| | - Rehab A Ibrahim
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
19
|
Jin HW, Eom YB. Antibacterial and Anti-biofilm Effects of Thymoquinone Against Carbapenem-Resistant Uropathogenic Escherichia coli. Indian J Microbiol 2024; 64:1747-1756. [PMID: 39678958 PMCID: PMC11645355 DOI: 10.1007/s12088-024-01231-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/13/2024] [Indexed: 12/17/2024] Open
Abstract
Carbapenem antibiotics are widely used for their broad antibacterial effects, but the emergence of carbapenem-resistant Enterobacterales has recently become a global problem. To solve this problem, research is needed to find compounds that increase antibiotic activity. Therefore, this study aimed to validate the antibacterial and anti-biofilm effects, as well as the inhibition of gene expression of thymoquinone, an extract of Nigella sativa commonly used as a spice in many dishes. The minimum inhibitory concentration of carbapenem antibiotics and thymoquinone was determined. Phenotypic analysis was performed to confirm the effect of thymoquinone on motility, which is one of the virulence factors of carbapenem-resistant uropathogenic Escherichia coli (CR-UPEC). Furthermore, quantitative real-time polymerase chain reaction analysis was used to determine the expression levels of carbapenemase gene (bla KPC), efflux pump genes (acrA, acrB, acrD, tolC), as well as motility and adhesion genes (fliC, motA). In addition, biofilm inhibition and biofilm eradication assays were performed. All strains showed resistance to carbapenem antibiotics, while an antibacterial effect was confirmed at a concentration of 256 μg/mL of thymoquinone. Phenotypic analysis revealed a nearly 50% suppression in migration distance compared to the control group at 128 μg/mL of thymoquinone. Subsequent gene expression tests indicated the downregulation of carbapenemase-, efflux pump-, motility-, and adhesion genes by thymoquinone. Furthermore, our findings demonstrated that thymoquinone exhibits both biofilm formation inhibition and eradication effects. These findings suggest that thymoquinone may serve as a potential antibiotic adjuvant for treating CR-UPEC and could be a valuable resource in combating UTIs caused by multidrug-resistant bacteria. Graphical Abstract
Collapse
Affiliation(s)
- Hye-Won Jin
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, Chungnam 31538 Republic of Korea
| | - Yong-Bin Eom
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, Chungnam 31538 Republic of Korea
- Present Address: Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, 22 Soonchunhyang-ro, Sinchang-myeon, Asan-si, Chungcheongnam-do 31538 Republic of Korea
| |
Collapse
|
20
|
Yildirim M, Ozgeris B, Gormez A. The effect of novel β-lactam derivatives synthesized from substituted phenethylamines on resistance genes of MRSA isolates. J Antibiot (Tokyo) 2024; 77:802-811. [PMID: 39210001 DOI: 10.1038/s41429-024-00769-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
This study focuses on the activity of previously reported imine and β-lactam derivatives against methicillin-resistant Staphylococcus aureus (MRSA) isolates. The presence of mecA and blaZ genes in the isolates was determined, and the minimum inhibitory concentration (MIC) values were determined based on the antibacterial activity against these isolates. Active compounds were selected and their ability to act against resistant isolates in vitro was determined. Concurrently, biochemical (nitrocefin) and molecular (qRT-PCR) tests were used to investigate the ability of the compounds to induce resistance genes in MRSA isolates. The cytotoxicity of the compounds on human dermal fibroblasts (HDF) was investigated. The MIC values of compounds (10) and (12) against MSSA and MRSA isolates were 7.81 and 15.62 μg ml-1, respectively. The most active compounds were identified as (10) and (12), and it was observed that the isolates did not develop resistance to these compounds in vitro. These compounds were found to inhibit β-lactamase, reduce the expression of resistance genes, and exhibit reduced HDF cell toxicity in a dose-dependent manner. According to the findings of the study, it can be concluded that these compounds show promise as hits with an interesting mechanism of action for further chemical modifications to develop new MRSA inhibitors.
Collapse
Affiliation(s)
- Merve Yildirim
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, 25050, Turkey
| | - Bunyamin Ozgeris
- Department of Basic Sciences, Faculty of Science, Erzurum Technical University, Erzurum, 25050, Turkey
| | - Arzu Gormez
- Department of Biology, Faculty of Science, Dokuz Eylul University, Izmir, 35390, Turkey.
| |
Collapse
|
21
|
Li Y, Wilhelm MJ, Wu T, Hu XH, Ruiz ON, Dai HL. Quantifying bacterial efflux within subcellular domains of Pseudomonas aeruginosa. Appl Environ Microbiol 2024; 90:e0144724. [PMID: 39475289 DOI: 10.1128/aem.01447-24] [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: 07/19/2024] [Accepted: 09/27/2024] [Indexed: 11/21/2024] Open
Abstract
Molecular efflux is a mechanism through which bacteria actively expel undesirable substances. This is a crucial line of defense against toxic chemicals in harsh environments. Understanding how efflux works is critical for designing antimicrobial strategies. Though much is already known about efflux proteins, important details about the mechanisms of efflux (e.g., importance of specific subcellular domains and ejection rates) have yet to be experimentally quantified. Herein, we use the nonlinear optical technique, second harmonic light scattering, to simultaneously measure the efflux rates from the periplasm and cytosol of a Gram-negative bacterium. The influence of efflux on the uptake kinetics of a mild antimicrobial, malachite green (MG), by Pseudomonas aeruginosa was quantified. It is observed that efflux primarily occurs from the periplasm and is two orders of magnitude faster than from the cytosol. Efflux pumps activate to maintain MG concentrations in the periplasm below 1 µM, while efflux from the cytosol maintains MG concentration below 0.1 µM. Efflux pumps are shown to saturate when exogenous MG concentrations are greater than 25 µM, while the cytosol efflux function saturates at >15 µM. Finally, efflux pumps can simultaneously eject different compounds, as proven by experiments with both MG and hexane, a known effluxable compound.IMPORTANCEMolecular efflux pumps are a crucial defense mechanism that protects bacteria from an otherwise unchecked influx of toxic molecules present in the extracellular environment. The efflux functions constitute a significant hindrance to antimicrobial efficacy. While much is now known regarding the structure of these channels, knowledge of the influence of efflux in individual subcellular domains and the associated ejection rates is still lacking. Using the nonlinear optical technique, second-harmonic light scattering, we have measured the threshold concentrations for pump activation, saturation concentrations, and efflux rates from both the periplasm and cytosol in living Gram-negative bacteria. The quantified efflux data in the different subcellular compartments not only provide a clear mechanistic understanding but also are critical for developing antimicrobial strategies.
Collapse
Affiliation(s)
- Yujie Li
- Institute for Membranes and Interfaces, Temple University, Philadelphia, Pennsylvania, USA
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Michael J Wilhelm
- Institute for Membranes and Interfaces, Temple University, Philadelphia, Pennsylvania, USA
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Tong Wu
- Institute for Membranes and Interfaces, Temple University, Philadelphia, Pennsylvania, USA
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Xiao-Hua Hu
- Institute for Membranes and Interfaces, Temple University, Philadelphia, Pennsylvania, USA
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Oscar N Ruiz
- Biomaterials Branch, Materials & Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson AFB, Ohio, USA
| | - Hai-Lung Dai
- Institute for Membranes and Interfaces, Temple University, Philadelphia, Pennsylvania, USA
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Guedes RFDM, Guedes GMDM, Gomes FIF, Soares ACDCF, Pereira VC, Freitas AS, Amando BR, Sidrim JJC, Cordeiro RDA, Rocha MFG, Castelo-Branco DDSCM. Antimicrobial and antibiofilm effect of promethazine on bacterial isolates from canine otitis externa: an in vitro study. Microb Pathog 2024; 196:106993. [PMID: 39374884 DOI: 10.1016/j.micpath.2024.106993] [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: 06/12/2024] [Revised: 09/11/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Otitis externa is an inflammatory disease of the external ear canal of complex and multifactorial etiology associated with recurrent bacterial infection. This study aimed to assess the antimicrobial and antibiofilm activity of promethazine against bacterial isolates from dogs with otitis externa, as well as the effect of this compound on the dynamics of biofilm formation over 120 h. Planktonic bacterial susceptibility to promethazine was evaluated to determine the minimum inhibitory concentrations (MIC). The minimum biofilm eradication concentration (MBEC) was also determined by broth microdilution. To evaluate the effect on biofilm growth, promethazine was tested at three concentrations MIC, MIC/2 and MIC/8, with daily readings at 48, 72, 96 and 120 h. The MICs of promethazine ranged from 48.83 to 781.25 μg mL-1. Promethazine significantly (P < 0.05) reduced mature biofilm biomass, with MBECs ranging from 48.8 to 6250 μg mL-1 and reduced (P < 0.01) biofilm formation for up to the 120-h, at concentrations corresponding to the MIC obtained against each isolate. Promethazine was effective against microorganisms associated with canine otitis externa. The data suggest that promethazine presents antimicrobial and antibiofilm activity and is a potential alternative to treat and prevent recurrent bacterial otitis in dogs. These results emphasize the importance of drug repurposing in veterinary otology as an alternative to reduce antimicrobial resistance.
Collapse
Affiliation(s)
- Rodrigo Fonseca de Medeiros Guedes
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Gláucia Morgana de Melo Guedes
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Francisco Ivanilsom Firmiano Gomes
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Ana Carla de Castro Freitas Soares
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Vinicius Carvalho Pereira
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Alyne Soares Freitas
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Bruno Rocha Amando
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - José Júlio Costa Sidrim
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Rossana de Aguiar Cordeiro
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Marcos Fábio Gadelha Rocha
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil; Postgraduate Program in Veterinary Sciences, College of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
| | - Débora de Souza Collares Maia Castelo-Branco
- Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Group of Applied Medical Microbiology, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil; Department of Pathology and Legal Medicine, Postgraduate Program in Medical Microbiology, Specialized Medical Mycology Center, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315, Rodolfo Teófilo, CEP: 60430-275, Fortaleza, Ceará, Brazil.
| |
Collapse
|
23
|
Hamami E, Huo W, Hernandez-Bird J, Castaneda A, Bai J, Syal S, Ortiz-Marquez JC, van Opijnen T, Geisinger E, Isberg RR. Identification of Determinants that Allow Maintenance of High-Level Fluoroquinolone Resistance in Acinetobacter baumannii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.03.560562. [PMID: 38645180 PMCID: PMC11030222 DOI: 10.1101/2023.10.03.560562] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Acinetobacter baumannii is associated with multidrug resistant (MDR) infections in healthcare settings, with fluoroquinolones such as ciprofloxacin being currently ineffective. Clinical isolates largely harbor mutations in the GyrA and TopoIV fluoroquinolone targets, as well as mutations that increase expression of drug resistance-nodulation-division (RND) efflux pumps. Factors critical for maintaining fitness levels of pump overproducers are uncharacterized despite their prevalence in clinical isolates. We here identify proteins that contribute to the fitness of FQR strains overexpressing three known RND systems using high-density insertion mutagenesis. Overexpression of the AdeFGH efflux pump caused hypersensitization to defects in outer membrane homeostatic regulation, including lesions that reduced LOS biosynthesis and blocked production of the major A. baumannii porin. In contrast, AdeAB pump hyperexpression, in the absence of elevated adeC expression (the outer membrane component of the pump), was relatively tolerant to loss of these functions, consistent with the outer membrane protein being the primary disruptive component. Surprisingly, overexpression of proton-transporting efflux pumps had little impact on cytosolic pH, consistent with a compensatory response to pump activity. The most striking transcriptional changes were associated with AdeFGH pump overexpression, including the activation of the phenylacetate (PAA) degradation regulon. Disruption of the PAA pathway resulted in cytosolic acidification and defective expression of genes involved in protection from oxidative stress. These results indicate that RND efflux pump overproduction is compensated by maintenance of outer membrane integrity in A. baumannii to facilitate fitness of FQR isolates.
Collapse
Affiliation(s)
- Efrat Hamami
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 150 Harrison Ave., Boston, MA 02111, USA
| | - Wenwen Huo
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 150 Harrison Ave., Boston, MA 02111, USA
| | - Juan Hernandez-Bird
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 150 Harrison Ave., Boston, MA 02111, USA
| | | | - Jinna Bai
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Sapna Syal
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 150 Harrison Ave., Boston, MA 02111, USA
| | - Juan C Ortiz-Marquez
- Department of Biology, Boston College, Chestnut Hill, MA 02135, USA
- Innovation Laboratory, Broad Institute, Cambridge, MA 02412, USA
| | - Tim van Opijnen
- Innovation Laboratory, Broad Institute, Cambridge, MA 02412, USA
| | - Edward Geisinger
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 150 Harrison Ave., Boston, MA 02111, USA
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Ralph R Isberg
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 150 Harrison Ave., Boston, MA 02111, USA
| |
Collapse
|
24
|
Li W, Tao Z, Zhou M, Jiang H, Wang L, Ji B, Zhao Y. Antibiotic adjuvants against multidrug-resistant Gram-negative bacteria: important component of future antimicrobial therapy. Microbiol Res 2024; 287:127842. [PMID: 39032266 DOI: 10.1016/j.micres.2024.127842] [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: 04/24/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
The swift emergence and propagation of multidrug-resistant (MDR) bacterial pathogens constitute a tremendous global health crisis. Among these pathogens, the challenge of antibiotic resistance in Gram-negative bacteria is particularly pressing due to their distinctive structure, such as highly impermeable outer membrane, overexpressed efflux pumps, and mutations. Several strategies have been documented to combat MDR Gram-negative bacteria, including the structural modification of existing antibiotics, the development of antimicrobial adjuvants, and research on novel targets that MDR bacteria are sensitive to. Drugs functioning as adjuvants to mitigate resistance to existing antibiotics may play a pivotal role in future antibacterial therapy strategies. In this review, we provide a brief overview of potential antibacterial adjuvants against Gram-negative bacteria and their mechanisms of action, and discuss the application prospects and potential for bacterial resistance to these adjuvants, along with strategies to reduce this risk.
Collapse
Affiliation(s)
- Wenwen Li
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Zhen Tao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Motan Zhou
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Huilin Jiang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Liudi Wang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Bingjie Ji
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Yongshan Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
25
|
Oliveira M, Antunes W, Mota S, Madureira-Carvalho Á, Dinis-Oliveira RJ, Dias da Silva D. An Overview of the Recent Advances in Antimicrobial Resistance. Microorganisms 2024; 12:1920. [PMID: 39338594 PMCID: PMC11434382 DOI: 10.3390/microorganisms12091920] [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: 09/03/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Antimicrobial resistance (AMR), frequently considered a major global public health threat, requires a comprehensive understanding of its emergence, mechanisms, advances, and implications. AMR's epidemiological landscape is characterized by its widespread prevalence and constantly evolving patterns, with multidrug-resistant organisms (MDROs) creating new challenges every day. The most common mechanisms underlying AMR (i.e., genetic mutations, horizontal gene transfer, and selective pressure) contribute to the emergence and dissemination of new resistant strains. Therefore, mitigation strategies (e.g., antibiotic stewardship programs-ASPs-and infection prevention and control strategies-IPCs) emphasize the importance of responsible antimicrobial use and surveillance. A One Health approach (i.e., the interconnectedness of human, animal, and environmental health) highlights the necessity for interdisciplinary collaboration and holistic strategies in combating AMR. Advancements in novel therapeutics (e.g., alternative antimicrobial agents and vaccines) offer promising avenues in addressing AMR challenges. Policy interventions at the international and national levels also promote ASPs aiming to regulate antimicrobial use. Despite all of the observed progress, AMR remains a pressing concern, demanding sustained efforts to address emerging threats and promote antimicrobial sustainability. Future research must prioritize innovative approaches and address the complex socioecological dynamics underlying AMR. This manuscript is a comprehensive resource for researchers, policymakers, and healthcare professionals seeking to navigate the complex AMR landscape and develop effective strategies for its mitigation.
Collapse
Affiliation(s)
- Manuela Oliveira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal; (Á.M.-C.); (D.D.d.S.)
- UCIBIO—Research Unit on Applied Molecular Biosciences, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Wilson Antunes
- Instituto Universitário Militar, CINAMIL, Unidade Militar Laboratorial de Defesa Biológica e Química, Avenida Doutor Alfredo Bensaúde, 4 piso, do LNM, 1849-012 Lisbon, Portugal
| | - Salete Mota
- ULSEDV—Unidade Local De Saúde De Entre Douro Vouga, Unidade de Santa Maria da Feira e Hospital S. Sebastião, Rua Dr. Cândido Pinho, 4520-211 Santa Maria da Feira, Portugal
| | - Áurea Madureira-Carvalho
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal; (Á.M.-C.); (D.D.d.S.)
- UCIBIO—Applied Molecular Biosciences Unit, Forensics and Biomedical Sciences Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
- Department of Public Health and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal; (Á.M.-C.); (D.D.d.S.)
- UCIBIO—Research Unit on Applied Molecular Biosciences, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
- Department of Public Health and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- FOREN—Forensic Science Experts, Avenida Dr. Mário Moutinho 33-A, 1400-136 Lisbon, Portugal
| | - Diana Dias da Silva
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal; (Á.M.-C.); (D.D.d.S.)
- UCIBIO—Applied Molecular Biosciences Unit, Forensics and Biomedical Sciences Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
- REQUIMTE/LAQV, ESS, Polytechnic of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
26
|
Dulanto Chiang A, Dekker JP. Efflux pump-mediated resistance to new beta lactam antibiotics in multidrug-resistant gram-negative bacteria. COMMUNICATIONS MEDICINE 2024; 4:170. [PMID: 39210044 PMCID: PMC11362173 DOI: 10.1038/s43856-024-00591-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
The emergence and spread of bacteria resistant to commonly used antibiotics poses a critical threat to modern medical practice. Multiple classes of bacterial efflux pump systems play various roles in antibiotic resistance, and members of the resistance-nodulation-division (RND) transporter superfamily are among the most important determinants of efflux-mediated resistance in gram-negative bacteria. RND pumps demonstrate broad substrate specificities, facilitating extrusion of multiple chemical classes of antibiotics from the bacterial cell. Several newer beta-lactams and beta-lactam/beta-lactamase inhibitor combinations (BL/BLI) have been developed to treat infections caused by multidrug resistant bacteria. Here we review recent studies that suggest RND efflux pumps in clinically relevant gram-negative bacteria may play critical but underappreciated roles in the development of resistance to beta-lactams and novel BL/BLI combinations. Improved understanding of the genetic and structural basis of RND efflux pump-mediated resistance may identify new antibiotic targets as well as strategies to minimize the emergence of resistance.
Collapse
Affiliation(s)
- Augusto Dulanto Chiang
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
- Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN, 37232, USA
| | - John P Dekker
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA.
- National Institutes of Health Clinical Center, NIH, Bethesda, MD, USA.
| |
Collapse
|
27
|
Luo VC, Peczuh MW. Location, Location, Location: Establishing Design Principles for New Antibacterials from Ferric Siderophore Transport Systems. Molecules 2024; 29:3889. [PMID: 39202968 PMCID: PMC11357680 DOI: 10.3390/molecules29163889] [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: 07/18/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
This review strives to assemble a set of molecular design principles that enables the delivery of antibiotic warheads to Gram-negative bacterial targets (ESKAPE pathogens) using iron-chelating siderophores, known as the Trojan Horse strategy for antibiotic development. Principles are derived along two main lines. First, archetypical siderophores and their conjugates are used as case studies for native iron transport. They enable the consideration of the correspondence of iron transport and antibacterial target location. The second line of study charts the rationale behind the clinical antibiotic cefiderocol. It illustrates the potential versatility for the design of new Trojan Horse-based antibiotics. Themes such as matching the warhead to a location where the siderophore delivers its cargo (i.e., periplasm vs. cytoplasm), whether or not a cleavable linker is required, and the relevance of cheaters to the effectiveness and selectivity of new conjugates will be explored. The effort to articulate rules has identified gaps in the current understanding of iron transport pathways and suggests directions for new investigations.
Collapse
Affiliation(s)
| | - Mark W. Peczuh
- Department of Chemistry, University of Connecticut, 55 N. Eagleville Road, U3060, Storrs, CT 06269, USA;
| |
Collapse
|
28
|
Kumari K, Aggarwal Y, Singh RP. Molecular characterization and in-depth genomic analysis to unravel the pathogenic features of an environmental isolate Enterobacter sp. S-33. Int Microbiol 2024; 27:1095-1110. [PMID: 38044418 DOI: 10.1007/s10123-023-00461-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Enterobacter species represent widely distributed opportunistic pathogens, commonly associated with plants and humans. In the present study, we performed a detailed molecular characterization as well as genomic study of a type VI secretion system (T6SS) bacterium belonging to member of the family Enterobacteriaceae and named Enterobacter sp. S-33. The comparative sequence analysis of the 16S rRNA gene showed that the strain was closely related to other Enterobacter species. The complete genome of the strain with a genome size of 4.6 Mbp and GC-content of 55.63% was obtained through high-quality sequencing. The genomic analysis with online tools unravelled the various genes belonging to the bacterial secretion system, antibiotic resistance, virulence, efflux pumps, etc. The isolate showed the motility behavior that contributes to Enterobacter persistence in a stressed environment and further supports infections. PCR amplification and further sequencing confirmed the presence of drug-efflux genes acrA, acrB, and outer membrane genes, viz. OmpA, OmpC, and OmpF. The cell surface hydrophobicity and co-aggregation assay against different bacterial strains illustrated its putative pathogenic nature. Genome mining identified various biosynthetic gene clusters (BGCs) corresponding to non-ribosomal proteins (NRPS), siderophore, and arylpolyene production. Briefly, genome sequencing and detailed characterization of environmental Enterobacter isolate will assist in understanding the epidemiology of Enterobacter species, and the further prevention and treatment of infectious diseases caused by these broad-host range species.
Collapse
Affiliation(s)
- Kiran Kumari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Yogender Aggarwal
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Rajnish Prakash Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India.
| |
Collapse
|
29
|
Bhagwat AC, Saroj SD. Polyamine as a microenvironment factor in resistance to antibiotics. Crit Rev Microbiol 2024; 50:504-513. [PMID: 37339480 DOI: 10.1080/1040841x.2023.2223277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/30/2023] [Accepted: 06/05/2023] [Indexed: 06/22/2023]
Abstract
One of the main issues in modern medicine is the decrease in the efficacy of antibiotic therapy against resistant microorganisms. The advent of antimicrobial resistance has added significantly to the impact of infectious diseases, in number of infections, as well as added healthcare costs. The development of antibiotic tolerance and resistance is influenced by a variety of environmental variables, and it is important to identify these environmental factors as part of any strategy for combating antibiotic resistance. The review aims to emphasize that biogenic polyamines are one of such environmental cues that impacts the antibiotic resistance in bacteria. The biogenic polyamines can help bacteria acquire resistance to antibiotics either by regulating the level of number of porin channels in the outer membrane, by modifying the outer membrane liposaccharides or by protecting macromolecule from antibiotic stress. Thus, understanding the way polyamines function in bacteria can thus be beneficial while designing the drugs to combat diseases.
Collapse
Affiliation(s)
- Amrita C Bhagwat
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Sunil D Saroj
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
| |
Collapse
|
30
|
Ralte L, Sailo H, Kumar NS, Singh YT. Exploring the pharmacological potential of Lepionurus sylvestris blume: from folklore medicinal usage to modern drug development strategies using in vitro and in silico analyses. BMC Complement Med Ther 2024; 24:289. [PMID: 39080609 PMCID: PMC11289938 DOI: 10.1186/s12906-024-04567-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Lepionurus sylvestris Blume has a long history of folklore medicinal usage against various ailments. However, studies on these plants were neglected particularly their pharmacological potential. METHODS The crude extract was identified using LC-MS analysis. In vitro assays were carried out to determine the properties of antioxidant, anti-microbial, and anti-cancer. Further, network pharmacology was proposed to evaluate the potential targets of the compounds against breast cancer and type II diabetes. Molecular docking and molecular dynamic simulation were used to determine the potential compounds for the drug formulation of diabetes. RESULTS Various bioactive compounds were identfied using LC-MS and Galiposin, Fujikinetin, Boeravinone B, 4-Deoxybryaquinone, and Norbaeocystin were described for the first time from the plant. Determination of antioxidant potential showed that the IC50 value of ABTS, DPPH, and phosphomolybdate was 24.33 µg/ml, 37.81 µg/ml, 60.35 µg/ml, and reducing power assays 1.185. The antibacterial activity against Streptococcus pyogenes, Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli was determined, and the minimum inhibition concentration (MIC) was found to be 5.3 mg/ml, 3.47 mg/ml, 3.33 mg/ml, and 2.7 mg/ml respectively, revealing the extracts as effective antibacterial agents. The IC50 values for the plant extract were determined to be 26 µg/ml, 30.52 µg/ml, and 24.39 µg/ml for HeLa, MCF-7, and K-562 cells, respectively, and the increasing concentration of the plant extract increased LDH release. Furthermore, the in silico network pharmacology, molecular docking which had the highest docking score for GAPDH and HIF-1 target proteins of -9.3 kcal/mol, and - 11.3 kcal/mol binding affinities, and molecular dynamic simulation analysis revealed the bioactive compound Boeravinone B present in the plant was significant for the treatment of various ailments. CONCLUSION Based on our findings, plant extracts could be a promising option for developing new drug formulations.
Collapse
Affiliation(s)
- Laldinfeli Ralte
- Department of Botany, Mizoram University, Aizawl, Mizoram, 796004, India
| | - Hmingremhlua Sailo
- Department of Botany, Mizoram University, Aizawl, Mizoram, 796004, India
| | | | - Y Tunginba Singh
- Department of Botany, Mizoram University, Aizawl, Mizoram, 796004, India.
- Department of Life Sciences (Botany), Manipur University, Imphal, Manipur, 795003, India.
| |
Collapse
|
31
|
Kengne MF, Tsobeng OD, Dadjo BST, Kuete V, Mbaveng AT. Multidrug Resistant Enteric Bacteria from Cancer Patients Admitted in Douala Laquintinie Hospital, Littoral Region of Cameroon. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:2084884. [PMID: 39036470 PMCID: PMC11259499 DOI: 10.1155/2024/2084884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/16/2024] [Accepted: 06/22/2024] [Indexed: 07/23/2024]
Abstract
Patients with cancer have weakened immune systems, making them more vulnerable to infections. This study was carried out to determine the bacterial origins of enteric disorders in cancer patients and noncancer patients at the Oncology Department of Laquintinie Hospital in Douala. A cross-sectional study was conducted from October 2021 to March 2023. Stool samples from 307 cancer patients with enteric disorders and 200 noncancer patients with enteric disorders were examined to diagnose the presence of bacteria using various techniques. Among all participants in this study, 62.13% were female and 37.87% were male. The average age of the participants was 46.38 ± 15.81 years, with a minimum age of 10 years and a maximum age of 84 years. The average age of participants was significantly higher (p < 0.000) in cancer patients (49.54 ± 14.65 years) compared to noncancer patients (41.53 ± 16.33 years). Proteus mirabilis, Proteus vulgaris, Salmonella typhi, Enterobacter cloacae, Klebsiella pneumoniae, Yersinia intemedia, and Klebsiella oxytoca were more frequently isolated in cancer patients than in noncancer patients, with the respective percentages of 56.25% versus 43.75%, 50.00% versus 50.00%, 61.66% versus 38.34%, 66.66% versus 33.34%, 72.22% versus 27.78%, 80.00 versus 20.00%, and 100% versus 0.00%. Most isolates were sensitive to imipenem (IMP), gentamicin (GEN), and amikacin (AMK). Proteus vulgaris, the most prevalent isolate, showed significantly high resistance (with p < 0.05) in cancer patients compared to noncancer patients at amoxicillin/clavuranic acid (AMC) (89.13% versus 41.30%), ceftriaxone (CTR) (63.04% versus 39.13%), ciprofloxacin (CIP) (65.22% versus 34.18%), and tetracycline (TET) (93.48% versus 63.04%). Multidrug resistance was observed in cancer patients compared to noncancer patients for Klebsiella pneumoniae (85.00% versus 60.00%), Salmonella typhi (84.62% versus 60.00%), and Klebsiella oxytoca (86.49% versus 43.48%). The increase in the number of Gram-negative infections among cancer patients, as shown in the present study, highlights the need for broad-spectrum therapy and effective planning of control programs to reduce bacterial diseases among cancer patients.
Collapse
Affiliation(s)
- Michael F. Kengne
- Department of BiochemistryFaculty of ScienceUniversity of Dschang, Dschang, Cameroon
| | - Ornella D. Tsobeng
- Department of BiochemistryFaculty of ScienceUniversity of Dschang, Dschang, Cameroon
| | - Ballue S. T. Dadjo
- Department of BiochemistryFaculty of ScienceUniversity of Dschang, Dschang, Cameroon
| | - Victor Kuete
- Department of BiochemistryFaculty of ScienceUniversity of Dschang, Dschang, Cameroon
| | - Armelle T. Mbaveng
- Department of BiochemistryFaculty of ScienceUniversity of Dschang, Dschang, Cameroon
| |
Collapse
|
32
|
Zhai J, Wang Y, Tang B, Zheng S, He S, Zhao W, Lin J, Li F, Bao Y, Lancuo Z, Liu C, Wang W. A comparison of antibiotic resistance genes and mobile genetic elements in wild and captive Himalayan vultures. PeerJ 2024; 12:e17710. [PMID: 39006014 PMCID: PMC11243982 DOI: 10.7717/peerj.17710] [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: 10/31/2023] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
As the most widely distributed scavenger birds on the Qinghai-Tibetan Plateau, Himalayan vultures (Gyps himalayensis) feed on the carcasses of various wild and domestic animals, facing the dual selection pressure of pathogens and antibiotics and are suitable biological sentinel species for monitoring antibiotic resistance genes (ARGs). This study used metagenomic sequencing to comparatively investigate the ARGs and mobile genetic elements (MGEs) of wild and captive Himalayan vultures. Overall, the resistome of Himalayan vultures contained 414 ARG subtypes resistant to 20 ARG types, with abundances ranging from 0.01 to 1,493.60 ppm. The most abundant resistance type was beta-lactam (175 subtypes), followed by multidrug resistance genes with 68 subtypes. Decreases in the abundance of macrolide-lincosamide-streptogramin (MLS) resistance genes were observed in the wild group compared with the zoo group. A total of 75 genera (five phyla) of bacteria were predicted to be the hosts of ARGs in Himalayan vultures, and the clinical (102 ARGs) and high-risk ARGs (35 Rank I and 56 Rank II ARGs) were also analyzed. Among these ARGs, twenty-two clinical ARGs, nine Rank I ARG subtypes, sixteen Rank II ARG subtypes were found to differ significantly between the two groups. Five types of MGEs (128 subtypes) were found in Himalayan vultures. Plasmids (62 subtypes) and transposases (44 subtypes) were found to be the main MGE types. Efflux pump and antibiotic deactivation were the main resistance mechanisms of ARGs in Himalayan vultures. Decreases in the abundance of cellular protection were identified in wild Himalayan vultures compared with the captive Himalayan vultures. Procrustes analysis and the co-occurrence networks analysis revealed different patterns of correlations among gut microbes, ARGs, and MGEs in wild and captive Himalayan vultures. This study is the first step in describing the characterization of the ARGs in the gut of Himalayan vultures and highlights the need to pay more attention to scavenging birds.
Collapse
Affiliation(s)
- Jundie Zhai
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - You Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Boyu Tang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Sisi Zheng
- Animal Disease Prevention and Control Center of Qinghai Province, Xining, China
| | - Shunfu He
- Xining Wildlife Park of Qinghai Province, Xining, China
| | - Wenxin Zhao
- Xining Wildlife Park of Qinghai Province, Xining, China
| | - Jun Lin
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Feng Li
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Yuzi Bao
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Zhuoma Lancuo
- College of Finance and Economics, Qinghai University, Xining, China
| | - Chuanfa Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Wen Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| |
Collapse
|
33
|
Zuberi A, Ahmad N, Ahmad H, Saeed M, Ahmad I. Beyond antibiotics: CRISPR/Cas9 triumph over biofilm-associated antibiotic resistance infections. Front Cell Infect Microbiol 2024; 14:1408569. [PMID: 39035353 PMCID: PMC11257871 DOI: 10.3389/fcimb.2024.1408569] [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: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 07/23/2024] Open
Abstract
A complex structure known as a biofilm is formed when a variety of bacterial colonies or a single type of cell in a group sticks to a surface. The extracellular polymeric compounds that encase these cells, often consisting of proteins, eDNA, and polysaccharides, exhibit strong antibiotic resistance. Concerns about biofilm in the pharmaceutical industry, public health, and medical fields have sparked a lot of interest, as antibiotic resistance is a unique capacity exhibited by these biofilm-producing bacteria, which increases morbidity and death. Biofilm formation is a complicated process that is controlled by several variables. Insights into the processes to target for the therapy have been gained from multiple attempts to dissect the biofilm formation process. Targeting pathogens within a biofilm is profitable because the bacterial pathogens become considerably more resistant to drugs in the biofilm state. Although biofilm-mediated infections can be lessened using the currently available medications, there has been a lot of focus on the development of new approaches, such as bioinformatics tools, for both treating and preventing the production of biofilms. Technologies such as transcriptomics, metabolomics, nanotherapeutics and proteomics are also used to develop novel anti-biofilm agents. These techniques help to identify small compounds that can be used to inhibit important biofilm regulators. The field of appropriate control strategies to avoid biofilm formation is expanding quickly because of this spurred study. As a result, the current article addresses our current knowledge of how biofilms form, the mechanisms by which bacteria in biofilms resist antibiotics, and cutting-edge treatment approaches for infections caused by biofilms. Furthermore, we have showcased current ongoing research utilizing the CRISPR/Cas9 gene editing system to combat bacterial biofilm infections, particularly those brought on by lethal drug-resistant pathogens, concluded the article with a novel hypothesis and aspirations, and acknowledged certain limitations.
Collapse
Affiliation(s)
- Azna Zuberi
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
- Department of Obs & Gynae, Northwestern University, Chicago, IL, United States
| | - Nayeem Ahmad
- Department of Biophysics, All India Institute of Medical Science, New Delhi, India
- Department of Microbiology, Immunology, and Infectious Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Hafiz Ahmad
- Department of Medical Microbiology & Immunology, Ras Al Khaimah (RAK) College of Medical Sciences, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Mohd Saeed
- Department of Biology, College of Science University of Hail, Hail, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
34
|
Rathod S, Dey S, Pawar S, Dhavale R, Choudhari P, Rajakumara E, Mahuli D, Bhagwat D, Tamboli Y, Sankpal P, Mali S, More H. Identification of potential biogenic chalcones against antibiotic resistant efflux pump (AcrB) via computational study. J Biomol Struct Dyn 2024; 42:5178-5196. [PMID: 37340697 DOI: 10.1080/07391102.2023.2225099] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/09/2023] [Indexed: 06/22/2023]
Abstract
The cases of bacterial multidrug resistance are increasing every year and becoming a serious concern for human health. Multidrug efflux pumps are key players in the formation of antibiotic resistance, which transfer out a broad spectrum of drugs from the cell and convey resistance to the host. Efflux pumps have significantly reduced the efficacy of the previously available antibiotic armory, thereby increasing the frequency of therapeutic failures. In gram-negative bacteria, the AcrAB-TolC efflux pump is the principal transporter of the substrate and plays a major role in the formation of antibiotic resistance. In the current work, advanced computer-aided drug discovery approaches were utilized to find hit molecules from the library of biogenic chalcones against the bacterial AcrB efflux pump. The results of the performed computational studies via molecular docking, drug-likeness prediction, pharmacokinetic profiling, pharmacophore mapping, density functional theory, and molecular dynamics simulation study provided ZINC000004695648, ZINC000014762506, ZINC000014762510, ZINC000095099506, and ZINC000085510993 as stable hit molecules against the AcrB efflux pumps. Identified hits could successfully act against AcrB efflux pumps after optimization as lead molecules.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sanket Rathod
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Sreenath Dey
- Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, Sangareddy, Telangana, India
| | - Swaranjali Pawar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Rakesh Dhavale
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Prafulla Choudhari
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Eerappa Rajakumara
- Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, Sangareddy, Telangana, India
| | - Deepak Mahuli
- Department of Pharmacology, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Durgacharan Bhagwat
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| | - Yasinalli Tamboli
- King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Poournima Sankpal
- Department of Pharmaceutical Chemistry, Ashokrao Mane College of Pharmacy, Kolhapur, MS, India
| | - Sachin Mali
- Department of Pharmaceutics, Y. D. Mane College of Pharmacy, Kagal, MS, India Kolhapur
| | - Harinath More
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, MS, India
| |
Collapse
|
35
|
Mukherjee S, Shinde SV, Talukdar P, Haldar J. Unveiling the potent activity of a synthetic ion transporter against multidrug-resistant Gram-positive bacteria and biofilms. RSC Med Chem 2024; 15:2127-2137. [PMID: 38911153 PMCID: PMC11187549 DOI: 10.1039/d4md00002a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/25/2024] [Indexed: 06/25/2024] Open
Abstract
The increasing prevalence of drug-resistant infections caused by Gram-positive bacteria poses a significant threat to public healthcare. These pathogens exhibit not only smart resistance mechanisms but also form impenetrable biofilms on various surfaces, rendering them resilient to conventional therapies. In this study, we present the potent antibacterial activity of a synthetic ion transporter T against multi-drug resistant (MDR) Gram-positive pathogens, with minimum inhibitory concentration (MIC) values ranging from 0.5 to 2 μg mL-1. The compound demonstrates high selectivity with negligible toxicity towards mammalian cells (HC50 = 810 μg mL-1). It exhibits fast killing kinetics, completely eliminating >5 log bacterial cells within 12 h. Moreover, the compound displays efficacy against both planktonic bacteria and preformed biofilms of methicillin-resistant S. aureus (MRSA), reducing the bacterial burden within the biofilm by 2 log. Mechanistic investigations reveal that the ion transporter depolarizes the bacterial membrane potential and enhances membrane permeability. Additionally, it generates reactive oxygen species, contributing to its bactericidal activity. Notably, MRSA did not exhibit detectable resistance to the ion transporter even after serial passaging for 10 days. Collectively, this novel class of ion transporter holds promise as a therapeutic candidate for combating infections caused by multi-drug resistant Gram-positive bacteria.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, JNCASR Jakkur Bangalore-560064 India
| | - Sopan Valiba Shinde
- Department of Chemistry, Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, JNCASR Jakkur Bangalore-560064 India
- School of Advanced Materials, JNCASR Jakkur Bangalore-560064 India
| |
Collapse
|
36
|
Thakkar N, Gajera G, Mehta D, Kothari V. Silversol ® (a Colloidal Nanosilver Formulation) Inhibits Growth of Antibiotic-Resistant Staphylococcus aureus by Disrupting Its Physiology in Multiple Ways. Pharmaceutics 2024; 16:726. [PMID: 38931848 PMCID: PMC11206351 DOI: 10.3390/pharmaceutics16060726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Antibiotic-resistant strains of Staphylococcus aureus are being viewed as a serious threat by various public health agencies. Identifying novel targets in this important pathogen is crucial to the development of new effective antibacterial formulations. We investigated the antibacterial effect of a colloidal nanosilver formulation, Silversol®, against an antibiotic-resistant strain of S. aureus using appropriate in vitro assays. Moreover, we deciphered the molecular mechanisms underlying this formulation's anti-S. aureus activity using whole transcriptome analysis. Lower concentrations of the test formulation exerted a bacteriostatic effect against this pathogen, and higher concentrations exerted a bactericidal effect. Silversol® at sub-lethal concentration was found to disturb multiple physiological traits of S. aureus such as growth, antibiotic susceptibility, membrane permeability, efflux, protein synthesis and export, biofilm and exopolysaccharide production, etc. Transcriptome data revealed that the genes coding for transcriptional regulators, efflux machinery, transferases, β-lactam resistance, oxidoreductases, metal homeostasis, virulence factors, and arginine biosynthesis are expressed differently under the influence of the test formulation. Genes (argG and argH) involved in arginine biosynthesis emerged among the major targets of Silversol®'s antibacterial activity against S. aureus.
Collapse
Affiliation(s)
- Nidhi Thakkar
- Institute of Science, Nirma University, Ahmedabad 382481, India; (N.T.); (G.G.)
| | - Gemini Gajera
- Institute of Science, Nirma University, Ahmedabad 382481, India; (N.T.); (G.G.)
| | - Dilip Mehta
- Viridis BioPharma Pvt. Ltd., Mumbai 400043, India;
| | - Vijay Kothari
- Institute of Science, Nirma University, Ahmedabad 382481, India; (N.T.); (G.G.)
| |
Collapse
|
37
|
Wu W, Huang J, Xu Z. Antibiotic influx and efflux in Pseudomonas aeruginosa: Regulation and therapeutic implications. Microb Biotechnol 2024; 17:e14487. [PMID: 38801351 PMCID: PMC11129675 DOI: 10.1111/1751-7915.14487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
Pseudomonas aeruginosa is a notorious multidrug-resistant pathogen that poses a serious and growing threat to the worldwide public health. The expression of resistance determinants is exquisitely modulated by the abundant regulatory proteins and the intricate signal sensing and transduction systems in this pathogen. Downregulation of antibiotic influx porin proteins and upregulation of antibiotic efflux pump systems owing to mutational changes in their regulators or the presence of distinct inducing molecular signals represent two of the most efficient mechanisms that restrict intracellular antibiotic accumulation and enable P. aeruginosa to resist multiple antibiotics. Treatment of P. aeruginosa infections is extremely challenging due to the highly inducible mechanism of antibiotic resistance. This review comprehensively summarizes the regulatory networks of the major porin proteins (OprD and OprH) and efflux pumps (MexAB-OprM, MexCD-OprJ, MexEF-OprN, and MexXY) that play critical roles in antibiotic influx and efflux in P. aeruginosa. It also discusses promising therapeutic approaches using safe and efficient adjuvants to enhance the efficacy of conventional antibiotics to combat multidrug-resistant P. aeruginosa by controlling the expression levels of porins and efflux pumps. This review not only highlights the complexity of the regulatory network that induces antibiotic resistance in P. aeruginosa but also provides important therapeutic implications in targeting the inducible mechanism of resistance.
Collapse
Affiliation(s)
- Weiyan Wu
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research CentreSouth China Agricultural UniversityGuangzhouChina
| | - Jiahui Huang
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research CentreSouth China Agricultural UniversityGuangzhouChina
| | - Zeling Xu
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research CentreSouth China Agricultural UniversityGuangzhouChina
| |
Collapse
|
38
|
de Carvalho Matias EG, Bezerra KS, Costa AHL, Clemente Junior WS, Oliveira JIN, Ribeiro Junior LA, Galvão DS, Fulco UL. Quantum biochemical analysis of the TtgR regulator and effectors. Sci Rep 2024; 14:8519. [PMID: 38609407 PMCID: PMC11015042 DOI: 10.1038/s41598-024-58441-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
The recent expansion of multidrug-resistant (MDR) pathogens poses significant challenges in treating healthcare-associated infections. Although antibacterial resistance occurs by numerous mechanisms, active efflux of the drugs is a critical concern. A single species of efflux pump can produce a simultaneous resistance to several drugs. One of the best-studied efflux pumps is the TtgABC: a tripartite resistance-nodulation-division (RND) efflux pump implicated in the intrinsic antibiotic resistance in Pseudomonas putida DOT-T1E. The expression of the TtgABC gene is down-regulated by the HTH-type transcriptional repressor TtgR. In this context, by employing quantum chemistry methods based on the Density Functional Theory (DFT) within the Molecular Fragmentation with Conjugate Caps (MFCC) approach, we investigate the coupling profiles of the transcriptional regulator TtgR in complex with quercetin (QUE), a natural polyphenolic flavonoid, tetracycline (TAC), and chloramphenicol (CLM), two broad-spectrum antimicrobial agents. Our quantum biochemical computational results show the: [i] convergence radius, [ii] total binding energy, [iii] relevance (energetically) of the ligands regions, and [iv] most relevant amino acids residues of the TtgR-QUE/TAC/CLM complexes, pointing out distinctions and similarities among them. These findings improve the understanding of the binding mechanism of effectors and facilitate the development of new chemicals targeting TtgR, helping in the battle against the rise of resistance to antimicrobial drugs. These advances are crucial in the ongoing fight against rising antimicrobial drug resistance, providing hope for a future where healthcare-associated infections can be more beneficially treated.
Collapse
Affiliation(s)
- E G de Carvalho Matias
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal, RN, 59072-970, Brazil
| | - K S Bezerra
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal, RN, 59072-970, Brazil
| | - A H Lima Costa
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal, RN, 59072-970, Brazil
| | - W S Clemente Junior
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal, RN, 59072-970, Brazil
| | - J I N Oliveira
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal, RN, 59072-970, Brazil
| | - L A Ribeiro Junior
- Institute of Physics, University of Brasília, Brasília, 70919-970, Brazil.
| | - D S Galvão
- Applied Physics Department, University of Campinas, Campinas, São Paulo, Brazil
| | - U L Fulco
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal, RN, 59072-970, Brazil
| |
Collapse
|
39
|
Mahey N, Tambat R, Kalia R, Ingavale R, Kodesia A, Chandal N, Kapoor S, Verma DK, Thakur KG, Jachak S, Nandanwar H. Pyrrole-based inhibitors of RND-type efflux pumps reverse antibiotic resistance and display anti-virulence potential. PLoS Pathog 2024; 20:e1012121. [PMID: 38593161 PMCID: PMC11003683 DOI: 10.1371/journal.ppat.1012121] [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: 09/08/2023] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
Efflux pumps of the resistance-nodulation-cell division (RND) superfamily, particularly the AcrAB-TolC, and MexAB-OprM, besides mediating intrinsic and acquired resistance, also intervene in bacterial pathogenicity. Inhibitors of such pumps could restore the activities of antibiotics and curb bacterial virulence. Here, we identify pyrrole-based compounds that boost antibiotic activity in Escherichia coli and Pseudomonas aeruginosa by inhibiting their archetype RND transporters. Molecular docking and biophysical studies revealed that the EPIs bind to AcrB. The identified efflux pump inhibitors (EPIs) inhibit the efflux of fluorescent probes, attenuate persister formation, extend post-antibiotic effect, and diminish resistant mutant development. The bacterial membranes remained intact upon exposure to the EPIs. EPIs also possess an anti-pathogenic potential and attenuate P. aeruginosa virulence in vivo. The intracellular invasion of E. coli and P. aeruginosa inside the macrophages was hampered upon treatment with the lead EPI. The excellent efficacy of the EPI-antibiotic combination was evidenced in animal lung infection and sepsis protection models. These findings indicate that EPIs discovered herein with negligible toxicity are potential antibiotic adjuvants to address life-threatening Gram-negative bacterial infections.
Collapse
Affiliation(s)
- Nisha Mahey
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Rushikesh Tambat
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, India
| | - Ritu Kalia
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Mohali, India
| | - Rajnita Ingavale
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Mohali, India
| | - Akriti Kodesia
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
- Structural Biology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Nishtha Chandal
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Srajan Kapoor
- Structural Biology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Dipesh Kumar Verma
- Structural Biology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Krishan Gopal Thakur
- Structural Biology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sanjay Jachak
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Mohali, India
| | - Hemraj Nandanwar
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
40
|
Rosa DS, Oliveira SADS, Souza RDFS, de França CA, Pires IC, Tavares MRS, de Oliveira HP, da Silva Júnior FAG, Moreira MAS, de Barros M, de Menezes GB, Antunes MM, Azevedo VADC, Naue CR, da Costa MM. Antimicrobial and antibiofilm activity of highly soluble polypyrrole against methicillin-resistant Staphylococcus aureus. J Appl Microbiol 2024; 135:lxae072. [PMID: 38503568 DOI: 10.1093/jambio/lxae072] [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: 11/13/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 03/21/2024]
Abstract
AIMS The purpose was to evaluate the antimicrobial activity of highly soluble polypyrrole (Hs-PPy), alone or combined with oxacillin, as well as its antibiofilm potential against methicillin-resistant Staphylococcus aureus strains. Furthermore, the in silico inhibitory mechanism in efflux pumps was also investigated. METHODS AND RESULTS Ten clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA) and two reference strains were used. Antimicrobial activity was determined by broth microdilution, and the combination effect with oxacillin was evaluated by the checkerboard assay. The biofilm formation capacity of MRSA and the interference of Hs-PPy were evaluated. The inhibitory action of Hs-PPy on the efflux pump was evaluated in silico through molecular docking. Hs-PPy showed activity against the isolates, with inhibitory action between 62.5 and 125 µg ml-1 and bactericidal action at 62.5 µg ml-1, as well as synergism in association with oxacillin. The isolates ranged from moderate to strong biofilm producers, and Hs-PPy interfered with the formation of this structure, but not with mature biofilm. There was no in silico interaction with the efflux protein EmrD, the closest homolog to NorA. CONCLUSIONS Hs-PPy interferes with biofilm formation by MRSA, has synergistic potential, and is an efflux pump inhibitor.
Collapse
Affiliation(s)
- Danillo Sales Rosa
- Universidade Federal do Vale do São Francisco, Petrolina, Pernambuco 56300-000, Brazil
| | | | | | | | | | | | | | | | | | - Mariana de Barros
- Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil
| | | | - Maísa Mota Antunes
- Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | | | - Carine Rosa Naue
- Hospital Universitário da Universidade Federal do Vale do São Francisco, Petrolina, Pernambuco 56304-205, Brazil
| | | |
Collapse
|
41
|
Li J, Liao Q, Wang Y, Wang X, Liu J, Zha R, He JZ, Zhang M, Zhang W. Involvement of functional metabolism promotes the enrichment of antibiotic resistome in drinking water: Based on the PICRUSt2 functional prediction. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 356:120544. [PMID: 38471323 DOI: 10.1016/j.jenvman.2024.120544] [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: 12/26/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024]
Abstract
Biofilters are the important source and sink of antibiotic resistance genes (ARGs) and antibiotic resistance bacteria (ARB) in the drinking water. Current studies generally ascribed the prevalence of BAR in biofilter from the perspective of gene behavior, i.e. horizontal gene transfer (HGT), little attentions have been paid on the ARGs carrier- ARB. In this study, we proposed the hypothesis that ARB participating in pollutant metabolism processes and becoming dominant is an important way for the enrichment of ARGs. To verify this, the antibiotic resistome and bacterial functional metabolic pathways of a sand filter was profiled using heterotrophic bacterial plate counting method (HPC), high-throughput qPCR, Illumina Hiseq sequencing and PICRUSt2 functional prediction. The results illustrated a significant leakage of ARB in the effluent of the sand filter with an average absolute abundance of approximately 102-103 CFU/mL. Further contribution analysis revealed that the dominant genera, such as Acinetobacter spp., Aeromonas spp., Elizabethkingia spp., and Bacillus spp., were primary ARGs hosts, conferring resistance to multiple antibiotics including sulfamethoxazole, tetracycline and β-lactams. Notably, these ARGs hosts were involved in nitrogen metabolism, including extracellular nitrate/nitrite transport and nitrite reduction, which are crucial in nitrification and denitrification in biofilters. For example, Acinetobacter spp., the dominant bacteria in the filter (relative abundance 69.97 %), contributed the majority of ARGs and 53.79 % of nitrite reduction function. That is, ARB can predominate by participating in the nitrogen metabolism pathways, facilitating the enrichment of ARGs. These findings provide insights into the stable presence of ARGs in biofilters from a functional metabolism perspective, offering a significant supplementary to the mechanisms of the emergence, maintenance, and transmission of BARin drinking water.
Collapse
Affiliation(s)
- Jiabing Li
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Qiuyu Liao
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Yun Wang
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Xuansen Wang
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Jinchi Liu
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Ruibo Zha
- School of Cultural Tourism and Public Administration, Fujian Normal University, Fuzhou 350117, China
| | - Ji-Zheng He
- Key Laboratory for Humid Subtropical Eco-geographical Processes of the Ministry of Education, Fujian Normal University, Fuzhou 350007, China; Fujian Sanming Forest Ecosystem National Observation and Research Station, Fujian Normal University, Sanming 365002, China
| | - Menglu Zhang
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Key Laboratory for Humid Subtropical Eco-geographical Processes of the Ministry of Education, Fujian Normal University, Fuzhou 350007, China; Fujian Sanming Forest Ecosystem National Observation and Research Station, Fujian Normal University, Sanming 365002, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China.
| | - Weifang Zhang
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| |
Collapse
|
42
|
Ding X, Lan W, Li J, Deng M, Li Y, Katayama Y, Gu JD. Metagenomic insight into the pathogenic-related characteristics and resistome profiles within microbiome residing on the Angkor sandstone monuments in Cambodia. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 918:170402. [PMID: 38307295 DOI: 10.1016/j.scitotenv.2024.170402] [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: 12/23/2022] [Revised: 01/06/2024] [Accepted: 01/22/2024] [Indexed: 02/04/2024]
Abstract
To reveal the characteristics of indigenous microbiome including the pathogenic-related ones on Angkor monuments in Cambodia and the distribution pattern of resistome at different locations, several sites, namely Angkor Wat, Bayon of Angkor Thom, and Prasat Preah Vihear with different exposure levels to tourists were selected to conduct the metagenomic analysis in this study. The general characteristics of the microbiome on these monuments were revealed, and the association between the environmental geo-ecological feature and the indigenous microbiome was delineated. The most common microbial groups included 6 phyla, namely Acidobacteria, Actinobacteria, Gemmatimonadetes, Nitrospirae, Proteobacteria and Verrucomicrobia on the monuments, but Firmicutes and Chlamydiae were the most dominant phyla found in bats droppings. The taxonomic family of Chitinophagaceae could serve as a signature microbial group for Preah Vihear, the less visited site. More importantly, the pathogenic-related characteristics of the microbiome residing on Angkor monuments were uncovered. A set of specific antibiotic resistance genes (ARGs) with cross-niches dispersal capacity (between the environmental microbiome and the microbiome within warm blood fauna) was identified to be high by the source tracking analysis based on ARGs profile varies in this study. Among the 10 ARG-types detected in this study, 6 of them are confined to resistance mechanism of antibiotic efflux-pump. The findings of this study provide new a new direction on public health management and implication globally at archaeological sites for tourism.
Collapse
Affiliation(s)
- Xinghua Ding
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China; School of History and Culture, Hunan Normal University, 36 Lushan Road, Changsha 410000, Hunan, People's Republic of China
| | - Wensheng Lan
- Shenzhen R&D Key Laboratory of Alien Pest Detection Technology, The Shenzhen Academy of Inspection and Quarantine, Food Inspection and Quarantine Center of Shenzhen Custom, 1011 Fuqiang Road, Shenzhen 518045, People's Republic of China
| | - Jing Li
- School of Food and Biotechnology, Guangdong Industry Polytechnic, Guangzhou 510300, People's Republic of China
| | - Maocheng Deng
- School of Food and Biotechnology, Guangdong Industry Polytechnic, Guangzhou 510300, People's Republic of China
| | - Yiliang Li
- Department of Earth Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Yoko Katayama
- Tokyo National Research Institute for Cultural Properties, 13-43 Ueno Park, Taito-ku, Tokyo 110-8713, Japan
| | - Ji-Dong Gu
- Environmental Science and Engineering Research Group, Guangdong Technion - Israel Institute of Technology, 241 Daxue Road, Shantou, Guangdong 515063, People's Republic of China; Guangdong Provincial Key Laboratory of Materials and Technologies for Energy Conversion, Guangdong Technion - Israel Institute of Technology, 241 Daxue Road, Shantou, Guangdong 515063, People's Republic of China.
| |
Collapse
|
43
|
Verma T, Nandini SS, Singh V, Raghavan A, Annappa H, Bhaskarla C, Dubey AK, Nandi D. Divergent Roles of Escherichia Coli Encoded Lon Protease in Imparting Resistance to Uncouplers of Oxidative Phosphorylation: Roles of marA, rob, soxS and acrB. Curr Microbiol 2024; 81:98. [PMID: 38372817 DOI: 10.1007/s00284-024-03632-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
Uncouplers of oxidative phosphorylation dissipate the proton gradient, causing lower ATP production. Bacteria encounter several non-classical uncouplers in the environment, leading to stress-induced adaptations. Here, we addressed the molecular mechanisms responsible for the effects of uncouplers in Escherichia coli. The expression and functions of genes involved in phenotypic antibiotic resistance were studied using three compounds: two strong uncouplers, i.e., Carbonyl cyanide m-chlorophenyl hydrazone (CCCP) and 2,4-Dinitrophenol (DNP), and one moderate uncoupler, i.e., Sodium salicylate (NaSal). Quantitative expression studies demonstrated induction of transcripts encoding marA, soxS and acrB with NaSal and DNP, but not CCCP. Since MarA and SoxS are degraded by the Lon protease, we investigated the roles of Lon using a lon-deficient strain (Δlon). Compared to the wild-type strain, Δlon shows compromised growth upon exposure to NaSal or 2, 4-DNP. This sensitivity is dependent on marA but not rob and soxS. On the other hand, the Δlon strain shows enhanced growth in the presence of CCCP, which is dependent on acrB. Interestingly, NaSal and 2,4-DNP, but not CCCP, induce resistance to antibiotics, such as ciprofloxacin and tetracycline. This study addresses the effects of uncouplers and the roles of genes involved during bacterial growth and phenotypic antibiotic resistance. Strong uncouplers are often used to treat wastewater, and these results shed light on the possible mechanisms by which bacteria respond to uncouplers. Also, the rampant usage of some uncouplers to treat wastewater may lead to the development of antibiotic resistance.
Collapse
Affiliation(s)
- Taru Verma
- Department of Bioengineering, Indian Institute of Science, Bengaluru, 560012, India
| | - Santhi Sanil Nandini
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Varsha Singh
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Abinaya Raghavan
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Harshita Annappa
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Chetana Bhaskarla
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Ashim Kumar Dubey
- Undergraduate program, Indian Institute of Science, Bengaluru, 560012, India
| | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India.
| |
Collapse
|
44
|
Ronneau S, Michaux C, Giorgio RT, Helaine S. Intoxication of antibiotic persisters by host RNS inactivates their efflux machinery during infection. PLoS Pathog 2024; 20:e1012033. [PMID: 38421944 PMCID: PMC10903880 DOI: 10.1371/journal.ppat.1012033] [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: 10/15/2023] [Accepted: 02/07/2024] [Indexed: 03/02/2024] Open
Abstract
The host environment is of critical importance for antibiotic efficacy. By impacting bacterial machineries, stresses encountered by pathogens during infection promote the formation of phenotypic variants that are transiently insensitive to the action of antibiotics. It is assumed that these recalcitrant bacteria-termed persisters-contribute to antibiotic treatment failure and relapsing infections. Recently, we demonstrated that host reactive nitrogen species (RNS) transiently protect persisters against the action of β-lactam antibiotics by delaying their regrowth within host cells. Here, we discovered that RNS intoxication of persisters also collaterally sensitizing them to fluoroquinolones during infection, explaining the higher efficiency of fluoroquinolones against intramacrophage Salmonella. By reducing bacterial respiration and the proton-motive force, RNS inactivate the AcrAB efflux machinery of persisters, facilitating the accumulation of fluoroquinolones intracellularly. Our work shows that target inactivity is not the sole reason for Salmonella persisters to withstand antibiotics during infection, with active efflux being a major contributor to survival. Thus, understanding how the host environment impacts persister physiology is critical to optimize antibiotics efficacy during infection.
Collapse
Affiliation(s)
- Séverin Ronneau
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Charlotte Michaux
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rachel T. Giorgio
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sophie Helaine
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
45
|
Soni K, Kothamasi D, Chandra R. Municipal wastewater treatment plant showing a potential reservoir for clinically relevant MDR bacterial strains co-occurrence of ESBL genes and integron-integrase genes. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 351:119938. [PMID: 38171124 DOI: 10.1016/j.jenvman.2023.119938] [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: 08/18/2023] [Revised: 11/22/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024]
Abstract
Municipal wastewater treatment plants (MWWTPs) are a milieu for co-occurrence of multiple antibiotic resistance genes (ARGs). This facilitates mixing and genetic exchange; and promotes dissemination of multidrug resistance (MDR) to wastewater bacterial communities which is hazardous for the effluent receiving environment. This study investigated the co-occurrence of extended-spectrum beta-lactamase (ESBL) genes (blaTEM, blaCTX-M, blaSHV, blaOXA), and integron-integrase genes (intI1, intI2, intI3) in MDR bacteria isolated from the Bharwara MWWTP in Lucknow, India. Thirty-one MDR bacterial colonies resistant to three or more antibiotics were isolated from three treatment stages of this MWWTP. Six of these: Staphylococcus aureus, Serratia marcescens, Salmonella enterica, Shigella sonnei, Escherichia coli, and Bacillus sp. Had co-occurrence of ESBL and integron-integrase genes. These six isolates were examined for the occurrence of MDR efflux genes (qacA, acrB) and ARGs (aac(3)-1, qnrA1, tetA, vanA) and tested for resistance against 12 different antibiotics. The highest resistance was against penicillin-G (100%) and lowest for chloramphenicol (16.66%). Bacillus sp. Isolate BWKRC6 had the highest co-occurrence of antibiotic resistance-determining genes and was resistant to all the 12 antibiotics tested. The co-occurrence of ESBL, integron-integrase, antibiotic resistance-determining and MDR efflux genes in bacteria isolated from the Bharwara MWWTP indicates that the wastewaters of this treatment plant may have become a hotspot for MDR bacteria and may present human and environmental health hazards. Therefore, there is need for a rapid action to limit the spread of this threat. Public regulatory authorities must urgently implement measures to prevent MWWTPs becoming reservoirs for evolution of antibiotic resistance genes and development of antibiotic resistance.
Collapse
Affiliation(s)
- Kuldeep Soni
- Department of Environmental Microbiology, School for Environmental Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, 226025, Uttar Pradesh, India
| | - David Kothamasi
- Laboratory of Soil Biology and Microbial Ecology, Department of Environmental Studies, University of Delhi, Delhi, 110007, India; Strathclyde Centre for Environmental Law and Governance, University of Strathclyde, Glasgow, G4 0LT, United Kingdom
| | - Ram Chandra
- Department of Environmental Microbiology, School for Environmental Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, 226025, Uttar Pradesh, India.
| |
Collapse
|
46
|
Panchal J, Patel A, Patel S, Goswami D. Understanding mastitis: Microbiome, control strategies, and prevalence - A comprehensive review. Microb Pathog 2024; 187:106533. [PMID: 38171428 DOI: 10.1016/j.micpath.2023.106533] [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: 10/30/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Mastitis significantly affects the udder tissue in dairy cattle, leading to inflammation, discomfort, and a decline in both milk yield and quality. The condition can be attributed to an array of microbial agents that access the mammary gland through multiple pathways. The ramifications of this ailment are not merely confined to animal welfare but extend to the financial viability of the livestock industry. This review offers a historical lens on mastitis, tracing its documentation back to 1851, and examines its global distribution with a focus on regional differences in prevalence and antimicrobial resistance (AMR) patterns. Specific microbial genes and communities implicated in both mastitis and AMR are explored, including Staphylococcus aureus, Streptococcus agalactiae,Streptococcus dysagalactiae, Streptococcus uberis Escherichia coli, Klebsiella pneumoniae, Mycoplasma bovis, Corynebacterium bovis, among others. These microorganisms have evolved diverse strategies to elude host immune responses and neutralize commonly administered antibiotics, complicating management efforts. The review aims a comprehensive overview of the current knowledge and research gaps on mastitis and AMR, and to highlight the need for a One Health approach to address this global health issue. Such an approach entails multi-disciplinary cooperation to foster judicious antibiotic use, enhance preventive measures against mastitis, and bolster surveillance and monitoring of AMR in pathogens responsible for mastitis.
Collapse
Affiliation(s)
- Janki Panchal
- Department of Microbiology & Biotechnology, University School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Arun Patel
- Department of Veterinary Microbiology, College of Veterinary Sciences & Animal Husbandry, Sardarkrushinagar, 385505, Kamdhenu University, Gujarat, India
| | - Sandip Patel
- Department of Veterinary Microbiology, College of Veterinary Sciences & Animal Husbandry, Sardarkrushinagar, 385505, Kamdhenu University, Gujarat, India
| | - Dweipayan Goswami
- Department of Microbiology & Biotechnology, University School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India.
| |
Collapse
|
47
|
Hu EW, Lu HF, Lin YT, Yang TC, Li LH. Modulatory role of SmeQ in SmeYZ efflux pump-involved functions in Stenotrophomonas maltophilia. J Antimicrob Chemother 2024; 79:383-390. [PMID: 38134316 DOI: 10.1093/jac/dkad392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND SmeYZ is a constitutively expressed efflux pump in Stenotrophomonas maltophilia. Previous studies demonstrated that: (i) smeYZ inactivation causes compromised swimming, oxidative stress tolerance and aminoglycoside resistance; and (ii) the ΔsmeYZ-mediated pleiotropic defects, except aminoglycoside susceptibility, result from up-regulation of entSCEBB'FA and sbiAB operons, and decreased intracellular iron level. OBJECTIVES To elucidate the modulatory role of SmeQ, a novel cytoplasmic protein, in ΔsmeYZ-mediated pleiotropic defects. METHODS The presence of operons was verified using RT-PCR. The role of SmeQ in ΔsmeYZ-mediated pleiotropic defects was assessed using in-frame deletion mutants and functional assays. A bacterial adenylate cyclase two-hybrid assay was used to investigate the protein-protein interactions. Gene expression was quantified using quantitative RT-PCR (RT-qPCR). RESULTS SmeYZ and the downstream smeQ formed an operon. SmeQ inactivation in the WT KJ decreased aminoglycoside resistance but did not affect swimming and tolerance to oxidative stress or iron depletion. However, smeQ inactivation in the smeYZ mutant rescued the ΔsmeYZ-mediated pleiotropic defects, except for aminoglycoside susceptibility. In the WT KJ, SmeQ positively modulated SmeYZ pump function by transcriptionally up-regulating the smeYZQ operon. Nevertheless, in the smeYZ mutant, SmeQ exerted its modulatory role by up-regulating entSCEBB'FA and sbiAB operons, decreasing intracellular iron levels, and causing ΔsmeYZ-mediated pleiotropic defects, except for aminoglycoside susceptibility. CONCLUSIONS SmeQ is the first small protein identified to be involved in efflux pump function in S. maltophilia. It exerts modulatory effect by transcriptionally altering the expression of target genes, which are the smeYZQ operon in the WT KJ, and smeYZQ, entSCEBB'FA and sbiAB operons in smeYZ mutants.
Collapse
Affiliation(s)
- En-Wei Hu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| | - Hsu-Feng Lu
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan, Republic of China
| | - Yi-Tsung Lin
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| | - Tsuey-Ching Yang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| | - Li-Hua Li
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, Republic of China
| |
Collapse
|
48
|
Ricaurte D, Huang Y, Sheth RU, Gelsinger DR, Kaufman A, Wang HH. High-throughput transcriptomics of 409 bacteria-drug pairs reveals drivers of gut microbiota perturbation. Nat Microbiol 2024; 9:561-575. [PMID: 38233648 PMCID: PMC11287798 DOI: 10.1038/s41564-023-01581-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 12/08/2023] [Indexed: 01/19/2024]
Abstract
Many drugs can perturb the gut microbiome, potentially leading to negative health consequences. However, mechanisms of most microorganism-drug responses have not been elucidated at the genetic level. Using high-throughput bacterial transcriptomics, we systematically characterized the gene expression profiles of prevalent human gut bacteria exposed to the most frequently prescribed orally administered pharmaceuticals. Across >400 drug-microorganism pairs, significant and reproducible transcriptional responses were observed, including pathways involved in multidrug resistance, metabolite transport, tartrate metabolism and riboflavin biosynthesis. Importantly, we discovered that statin-mediated upregulation of the AcrAB-TolC efflux pump in Bacteroidales species enhances microbial sensitivity to vitamin A and secondary bile acids. Moreover, gut bacteria carrying acrAB-tolC genes are depleted in patients taking simvastatin, suggesting that drug-efflux interactions generate collateral toxicity that depletes pump-containing microorganisms from patient microbiomes. This study provides a resource to further understand the drivers of drug-mediated microbiota shifts for better informed clinical interventions.
Collapse
Affiliation(s)
- Deirdre Ricaurte
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, NY, USA
| | - Yiming Huang
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, NY, USA
| | - Ravi U Sheth
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, NY, USA
| | | | - Andrew Kaufman
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Harris H Wang
- Department of Systems Biology, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
49
|
Behera DU, Gaur M, Sahoo M, Subudhi E, Subudhi BB. Development of pharmacophore models for AcrB protein and the identification of potential adjuvant candidates for overcoming efflux-mediated colistin resistance. RSC Med Chem 2024; 15:127-138. [PMID: 38283226 PMCID: PMC10809322 DOI: 10.1039/d3md00483j] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/26/2023] [Indexed: 01/30/2024] Open
Abstract
Growing multi-drug resistance (MDR) among ESKAPE pathogens is a huge challenge. Increased resistance to last-resort antibiotics, like colistin, has further aggravated this. Efflux is identified as a major route of colistin resistance. So, finding an FDA-approved efflux inhibitor for potential application as an adjuvant to colistin was the primary objective of this study. E. coli-AcrB pump inhibitors and substrates were used to develop and validate the pharmacophoric model. Drugs confirming this pharmacophore were subjected to molecular docking to identify hits for the AcrB binding pocket. The efflux inhibition potential of the top hit was validated through the in vitro evaluation of the minimum inhibitory concentration (MIC) in combination with colistin. The checkerboard assay was done to demonstrate synergism, which was further corroborated by the Time-kill assay. Ten common pharmacophore hypotheses were successfully generated using substrate/inhibitors. Following enrichment analysis, AHHNR.100 was identified as the top-ranked hypothesis, and 207 unique compounds were found to conform to this hypothesis. The multi-step docking of these compounds against the AcrB protein revealed argatroban as the top non-antibiotic hit. This significantly inhibited the efflux activity of colistin-resistant clinical isolates K. pneumoniae (n = 1) and M. morganii (n = 2). Further, their combination with colistin enhanced the susceptibility of these isolates, and the effect was found to be synergistic. Accordingly, the time-kill assay of this combination showed 8-log and 2-log reductions against K. pneumoniae and M. morganii, respectively. In conclusion, this study found argatroban as a bacterial efflux inhibitor that can be potentially used to overcome efflux-mediated resistance.
Collapse
Affiliation(s)
- Dibyajyoti Uttameswar Behera
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar-751003 Odisha India +91 9861075829
| | - Mahendra Gaur
- Drug Development and Analysis Laboratory, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar-751003 Odisha India +91 7978085389
- Department of Biotechnology & Food Technology, Punjabi University Patiala 147002 India
| | - Maheswata Sahoo
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar-751003 Odisha India +91 9861075829
| | - Enketeswara Subudhi
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar-751003 Odisha India +91 9861075829
| | - Bharat Bhusan Subudhi
- Drug Development and Analysis Laboratory, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar-751003 Odisha India +91 7978085389
| |
Collapse
|
50
|
He W, Jiang M, Li Y, Ge X. Identification of the Major Facilitator Superfamily Efflux Pump KpsrMFS in Klebsiella pneumoniae That Is Down-Regulated in the Presence of Multi-Stress Factors. Int J Mol Sci 2024; 25:1466. [PMID: 38338743 PMCID: PMC10855805 DOI: 10.3390/ijms25031466] [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: 12/15/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Efflux pumps play important roles in bacterial detoxification and some of them are stress-response elements that are up-regulated when the host is treated with antibiotics. However, efflux pumps that are down-regulated by stimulations are rarely discovered. Herein, we analyzed multiple transcriptome data and discovered a special (Major Facilitator Superfamily) MFS efflux pump, KpsrMFS, from Klebsiella pneumoniae, which was down-regulated when treated with antibiotics or extra carbon sources. Interestingly, overexpression of kpsrmfs resulted in halted cell growth in normal conditions, while the viable cells were rarely affected. The function of KpsrMFS was further analyzed and this efflux pump was determined to be a proton-driven transporter that can reduce the intracellular tetracycline concentration. In normal conditions, the expression of kpsrmfs was at a low level, while artificial overexpression of it led to increased endogenous reactive oxygen species (ROS) production. Moreover, by comparing the functions of adjacent genes of kpsrmfs, we further discovered another four genes that can confer similar phenotypes, indicating a special regulon that regulates cell growth. Our work provides new insights into the roles of efflux pumps and suggests a possible regulon that may regulate cell growth and endogenous ROS levels.
Collapse
Affiliation(s)
| | | | - Ying Li
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | | |
Collapse
|