1
|
Lo HR, Yan CH, Yan Y, Lo CC. Dimercaptosuccinic acid with membrane-targeting activity against Pseudomonas aeruginosa. Microb Pathog 2025; 204:107550. [PMID: 40194609 DOI: 10.1016/j.micpath.2025.107550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/07/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Multidrug resistant (MDR) gram-negative bacteria (GNB) are a serious health threat. GNB require divalent cations for the integrity of their outer membrane (OM), which can be inhibited by dimercaptosuccinic acid (DMSA), a sulfhydryl-containing metal chelator that has been used as an antidote to heavy metal toxicity. We aim to investigatethe effects and mechanisms of action of DMSA on Pseudomonas aeruginosa. MAIN METHODS The inhibition of P. aeruginosa strains by DMSA was determined using growth kinetics analysis. Biofilm formation was evaluated using crystal violet staining after incubation for 24 h. We determined the bacterial OM permeability and cell membrane potential using propidium iodide (PI) and bis-(1,3-dibutylbarbituric acid) trimethineoxonol (DiBAC4(3)) staining, respectively, following DMSA exposure. The bioenergetics-related activity of DMSA-treated bacteria was assessed by determining intracellular ATP levels, bacterial motility and N-phenyl-naphtylamide (NPN) efflux assay. RESULTS DMSA inhibited the growth of bacteria in a concentration-dependent manner and repressed biofilm formation by P. aeruginosa. DMSA-treated bacteria exhibited increased PI uptake and enhanced DiBAC4(3) fluorescence intensity compared with untreated cells. Treatment of P. aeruginosa with DMSA reduced the intracellular ATP levels, bacterial motility, and efflux activity in the tested cells. SIGNIFICANCE The antibacterial mechanisms of DMSA may be related to alterations in OM permeability, membrane depolarization, and impaired bioenergetics-related activity, which are essential for bacterial viability and infection.
Collapse
Affiliation(s)
- Horng-Ren Lo
- Department of Medical Laboratory Science and Biotechnology, Fooyin University, Kaohsiung, Taiwan.
| | - Cian-Hui Yan
- Department of Medical Laboratory Science and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| | - Ya Yan
- Department of Laboratory Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, Taiwan
| | - Chung-Cheng Lo
- Department of Internal Medicine, Pingtung Veterans General Hospital Longquan Branch, Pingtung, Taiwan
| |
Collapse
|
2
|
Li T, Wang Z, Zhang X, Hao Z, Guo Y, Shen J, Velkov T, Dai C. Natural Product Usnic Acid as an Antibacterial Therapeutic Agent: Current Achievements and Further Prospects. ACS Infect Dis 2025. [PMID: 40371696 DOI: 10.1021/acsinfecdis.5c00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Antimicrobial resistance (AMR) poses a significant global public health threat, endangering both human and animal health. In clinical environments, AMR often undermines the effectiveness of antibacterial treatments, underscoring the urgent need to discover and develop new antibacterial agents or alternatives to antibiotics. Usnic acid, a secondary metabolite derived from lichens, has emerged as a promising candidate owing to its diverse pharmacological properties, which include antibacterial, immune-regulating, antiaging, and anti-inflammatory activities. Extensive research has shown that usnic acid exhibits strong direct antibacterial effects against Gram-positive bacteria and acts as an antimicrobial adjuvant to enhance the therapeutic efficacy of antibiotic drugs against Gram-negative pathogens. Its mechanisms of action are multifaceted, encompassing the inhibition of RNA, DNA, and protein synthesis; suppression of bacterial efflux pump protein expression and membrane-localized drug-resistant enzyme activity; disruption of cell membrane integrity and metabolic homeostasis; and reduction of virulence factor production and biofilm formation. Despite its potential, the clinical application of usnic acid as an antibacterial agent faces significant challenges including poor aqueous solubility, low bioavailability, and dose-dependent toxicity. To overcome these limitations, nanodelivery systems such as liposomes and polymeric nanoparticles have been developed to enhance solubility, improve targeted delivery, and reduce toxicity, thereby expanding its therapeutic potential. Structural modification can also enhance the antibacterial activity and address solubility issues. This review systematically consolidates current knowledge on usnic acid's antibacterial properties, molecular mechanisms, and combinatorial therapies. It critically evaluates advancements in nanoformulation strategies, assesses safety and toxicity profiles, and identifies obstacles to its development as a clinically viable antibacterial agent. By addressing these aspects, this review aims to provide actionable insights, foster interdisciplinary dialogue, and catalyze further innovation in leveraging this natural product to combat AMR.
Collapse
Affiliation(s)
- Tian Li
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P.R. China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, P.R. China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, P. R. China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P.R. China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, P.R. China
| | - Xiuying Zhang
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, P. R. China
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Zhihui Hao
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P.R. China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, P.R. China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, P. R. China
| | - Yong Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan Province, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P.R. China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, P.R. China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, P. R. China
| | - Tony Velkov
- Department of Pharmacology, Biodiscovery Institute, Monash University, Melbourne, Victoria 3800, Australia
| | - Chongshan Dai
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P.R. China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, P.R. China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, P. R. China
| |
Collapse
|
3
|
Cernicchi G, Di Gregorio A, Felicetti T, Rampacci E, Casari G, Armeni T, Romaldi B, Zefaj E, Passamonti F, Massari S, Manfroni G, Barreca ML, Tabarrini O, Vignaroli C, Sabatini S. NorA Efflux Pump Inhibitors: Expanding SAR Knowledge of Pyrazolo[4,3-c][1,2]benzothiazine 5,5-Dioxide Derivatives. Arch Pharm (Weinheim) 2025; 358:e70000. [PMID: 40390184 DOI: 10.1002/ardp.70000] [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: 12/12/2024] [Revised: 03/19/2025] [Accepted: 04/07/2025] [Indexed: 05/21/2025]
Abstract
Antimicrobial resistance (AMR) represents a significant global concern, driven by the overuse of antibiotics. One of the principal mechanisms contributing to AMR is the activity of microbial efflux pumps (EPs), which expel diverse antibiotics out of bacterial cells, thereby rendering them ineffective. Our research aimed to expand the range of molecular classes that inhibit the Staphylococcus aureus EP NorA. In this study, starting from the hit compound pyrazolo[4,3-c][1,2]benzothiazine 5,5-dioxide 1, previously reported as a NorA efflux pump inhibitor (EPI), we undertook medicinal chemistry efforts, which involved the iterative combination of the design and synthesis of new analogues with data obtained through ethidium bromide efflux inhibition assays. Subsequent synergistic assays with ciprofloxacin (CPX) against the resistant strain SA-1199B led to the identification of three potent compounds (3, 10, and 19). The evaluation of these compounds in combination with CPX against NorA-overexpressing and NorA-knockout strains (SA-K2378 and SA-K1902, respectively) confirmed that the observed synergy with CPX is dependent on the presence of NorA. Additionally, the combination of NorA EPIs with CPX reduced biofilm production in NorA-overexpressing strains. These findings enhance our understanding of the structure-activity relationship of pyrazolobenzothiazine derivatives and support the use of EtBr efflux assays for rapid NorA inhibitors' identification.
Collapse
Affiliation(s)
- Giada Cernicchi
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Alessandra Di Gregorio
- Department of Life and Environmental Science, Università Politecnica delle Marche, Ancona, Italy
| | - Tommaso Felicetti
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Elisa Rampacci
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Giulia Casari
- Department of Specialized Clinical Sciences and Odontostomatology, Università Politecnica delle Marche, Torrette di Ancona, Italy
| | - Tatiana Armeni
- Department of Specialized Clinical Sciences and Odontostomatology, Università Politecnica delle Marche, Torrette di Ancona, Italy
| | - Brenda Romaldi
- Department of Specialized Clinical Sciences and Odontostomatology, Università Politecnica delle Marche, Torrette di Ancona, Italy
| | - Ermelinda Zefaj
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Fabrizio Passamonti
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Serena Massari
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Giuseppe Manfroni
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Maria Letizia Barreca
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Carla Vignaroli
- Department of Life and Environmental Science, Università Politecnica delle Marche, Ancona, Italy
| | - Stefano Sabatini
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
4
|
Rodriguez Velazquez D, Forte L, Varela Guerrero JA, Díaz Alvarado T, Elghandour MMMY, Maggiolino A, De Palo P, Salem AZM. Could Mesquite ( Prosopis juliflora) Help Control Gastrointestinal Parasites in Horses? Animals (Basel) 2025; 15:1245. [PMID: 40362060 PMCID: PMC12071051 DOI: 10.3390/ani15091245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Exploring new natural-origin antiparasitic alternatives is essential in addressing the resistance issues that have emerged due to the widespread use of chemical or synthetic antiparasitic compounds. This area remains relatively underexplored despite the increasing recognition of plants like Prosopis juliflora (mesquite) for their antiparasitic properties and nutritional value as a high-protein animal feed. This review aims to investigate the antiparasitic potential of P. juliflora against gastrointestinal parasites in horses. The review incorporates sources from 2005 to 2024. It was found that various phytochemical compounds from P. juliflora, particularly alkaloids, have demonstrated significant antiparasitic activity. Alkaloids are the most studied group due to their antibacterial, antiparasitic, and cytotoxic properties. According to the literature, the phytochemicals produced by P. juliflora hold promise as a potential solution for combating gastrointestinal parasites in horses. However, while preliminary findings suggest promising results, in vivo studies are necessary to evaluate the precise antiparasitic efficacy of these compounds and to determine the appropriate IC50 doses of P. juliflora alkaloids in horses, as well as to assess potential toxicity and side effects associated with their use.
Collapse
Affiliation(s)
- Desiderio Rodriguez Velazquez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca C.P. 50000, Estado de México, Mexico; (D.R.V.); (J.A.V.G.); (T.D.A.); (M.M.M.Y.E.)
| | - Lucrezia Forte
- Department of Veterinary Medicine, University of Bari, 70010 Valenzano, Italy; (A.M.); (P.D.P.)
| | - Jorge Antonio Varela Guerrero
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca C.P. 50000, Estado de México, Mexico; (D.R.V.); (J.A.V.G.); (T.D.A.); (M.M.M.Y.E.)
| | - Tonantzin Díaz Alvarado
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca C.P. 50000, Estado de México, Mexico; (D.R.V.); (J.A.V.G.); (T.D.A.); (M.M.M.Y.E.)
| | - Mona M. M. Y. Elghandour
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca C.P. 50000, Estado de México, Mexico; (D.R.V.); (J.A.V.G.); (T.D.A.); (M.M.M.Y.E.)
| | - Aristide Maggiolino
- Department of Veterinary Medicine, University of Bari, 70010 Valenzano, Italy; (A.M.); (P.D.P.)
| | - Pasquale De Palo
- Department of Veterinary Medicine, University of Bari, 70010 Valenzano, Italy; (A.M.); (P.D.P.)
| | - Abdelfattah Z. M. Salem
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca C.P. 50000, Estado de México, Mexico; (D.R.V.); (J.A.V.G.); (T.D.A.); (M.M.M.Y.E.)
- Dipartimento di Scienze del Suolo, della Pianta e degli Alimenti (Di.S.S.P.A.), Università degli Studi di Bari, Via Giovanni Amendola, 165/a, 70126 Bari, Italy
| |
Collapse
|
5
|
Yu XQ, Yang H, Feng HZ, Hou J, Tian JQ, Niu SM, You CG, Tao XY, Zhang SP, Wang ZP, He YX. Targeting efflux pumps prevents the multi-step evolution of high-level resistance to fluoroquinolone in Pseudomonas aeruginosa. Microbiol Spectr 2025; 13:e0298124. [PMID: 39982069 PMCID: PMC11960432 DOI: 10.1128/spectrum.02981-24] [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: 11/18/2024] [Accepted: 01/08/2025] [Indexed: 02/22/2025] Open
Abstract
Antibiotic resistance is emerging as a significant global health crisis, necessitating the urgent development of novel antibiotics or alternative therapies. Although it is recognized that bacteria require multiple mutations to develop resistance levels exceeding the mutant prevention concentration, the specific mutation combinations conferring high resistance have been largely undefined. Here, we investigated the multi-step evolution of fluoroquinolone resistance in Pseudomonas aeruginosa through experimental evolution and whole-genome sequencing coupled with proteomic approaches. We discovered that in low-dose and high-dose experimental evolution scenarios, combinations of mutations in the negative regulators of efflux pumps (nfxB/mexR) and DNA gyrases (gyrA/gyrB) contributed to the high-level resistance and some of these combinations were also prevalent in clinical isolates of P. aeruginosa. Notably, the selected nfxB mutation, which resulted in the overexpression of the MexCD-OprJ efflux pump, also exhibited collateral sensitivity to aminoglycosides and enhanced antibiotic tolerance. It was further revealed that the efflux pump inhibitor phenylalanine-arginine β-naphthylamide (PAβN) could effectively prevent evolution to high-level resistance for both laboratory and clinical P. aeruginosa strains. Our work highlights the critical role of efflux pump repressor-related mutations in the evolution of high-level antibiotic resistance and demonstrates the potential of targeting these mutations to impede the evolution toward high-level resistance.IMPORTANCEIn this study, we examined the stepwise evolution of fluoroquinolone resistance in Pseudomonas aeruginosa using experimental evolution, whole-genome sequencing, and proteomic analyses. Our findings revealed that under both low-dose and high-dose conditions, mutations in efflux pump regulators (nfxB/mexR) and DNA gyrase genes (gyrA/gyrB) synergistically contributed to high-level resistance. These mutation combinations were not only observed in experimental settings but also detected in clinical isolates of P. aeruginosa. This work underscores the pivotal role of efflux pump repressor-related mutations in the progression to high-level antibiotic resistance. It also highlights the promise of targeting efflux pumps as a strategy to prevent the multi-step evolution of resistance in P. aeruginosa.
Collapse
Affiliation(s)
- Xiao-Quan Yu
- Institute of Urology, Gansu Province Clinical Research Center for urinary system disease, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Hao Yang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Han-Zhong Feng
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jun Hou
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jun-Qiang Tian
- Institute of Urology, Gansu Province Clinical Research Center for urinary system disease, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Shao-Min Niu
- Institute of Urology, Gansu Province Clinical Research Center for urinary system disease, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Chong-Ge You
- Laboratory Medicine Center, The Second Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Xuan-Yu Tao
- Institute for Environmental Genomics, University of Oklahoma, Norman, Oklahoma, USA
| | - Si-Ping Zhang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Zhi-Ping Wang
- Institute of Urology, Gansu Province Clinical Research Center for urinary system disease, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yong-Xing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| |
Collapse
|
6
|
Zlotnikov ID, Belogurova NG, Kudryashova EV. Targeted Delivery Inside the Cells Directly Visualized with Förster Resonance Energy Transfer (FRET). Polymers (Basel) 2025; 17:790. [PMID: 40292615 PMCID: PMC11944702 DOI: 10.3390/polym17060790] [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: 02/08/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
We established a real-time Förster resonance energy transfer (FRET) based assay to evaluate targeted drug delivery using polymeric micelles. Red fluorescent protein (RFP)-expressing E. coli cells were used as a test system to monitor the delivery of drug-fluorophore such as curcumin and umbelliferones (MUmb and AMC) encapsulated in the polymeric micellar formulations. The efficiency of the drug delivery was quantified using the FRET efficiency, measured as the degree of energy transfer from the drug to the RFP. FRET efficiency directly provides the determination of the delivery efficacy, offering a versatile platform adaptable to various drugs and cell types. We used polymer micelles as a carrier for targeted delivery of fluorescent drugs to bacterial cells expressing RFP. The physicochemical characterization of the interaction between the drugs and the micelles including spectral properties, and the solubility and binding constants, were determined. We revealed a stronger affinity of MUmb for heparin-based micelles (Kd~10-5 M) compared to chitosan-based micelles (Kd~10-4 M), underscoring the influence of polymer composition on drug loading efficiency. For micelles containing MUmb, a FRET efficiency significantly exceeds (by three times) the efficiency for non-micellar MUmb, which have minimal penetration into bacterial cells. The most noticeable effect was observed with the use of the micellar curcumin providing pronounced activation of the RPF fluorescence signal, due to the interaction with curcumins (fluorophore-donor). Curcumin delivery using Chit5-OA micelle resulted in a 115% increase in RFP fluorescence intensity, and Hep-LA showed a significant seven-fold increase. These results highlight the significant effect of micellar composition on the effectiveness of drug delivery. In addition, we have developed a visual platform designed to evaluate the effectiveness of a pharmaceutical product through the visualization of the fluorescence of a bacterial culture on a Petri dish. This method allows us to quickly and accurately assess the penetration of a drug into bacteria, or those located inside other cells, such as macrophages, where the intercellular latent forms of the infection are located. Micellar formulations show enhanced antibacterial activity compared to free drugs, and formulations with Hep-OA micelles demonstrate the most significant reduction in E. coli viability. Synergistic effects were observed when combining curcumin and MUmb with moxifloxacin, resulting in a remarkable 40-50% increase in efficacy. The presented approach, based on the FRET test system with RFP expressed in the bacterial cells, establishes a powerful platform for development and optimizing targeted drug delivery systems.
Collapse
Affiliation(s)
| | | | - Elena V. Kudryashova
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia; (I.D.Z.)
| |
Collapse
|
7
|
de Sousa Silveira Z, Macêdo NS, Menezes Dantas DD, Rodrigues Dos Santos Barbosa C, Muniz DF, Morais Oliveira-Tintino CDD, Relison Tintino S, Alencar GG, Marinho ES, Rocha MND, Marinho MM, Santos HSD, Coutinho HDM, Cunha FABD, Silva MVD. Evaluation of the antibacterial and inhibitory activity of the NorA and TetK efflux pumps of Staphylococcus aureus by p-coumaric acid. Microb Pathog 2025; 200:107318. [PMID: 39848298 DOI: 10.1016/j.micpath.2025.107318] [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: 09/15/2024] [Revised: 12/25/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
The NorA and TetK efflux pumps mediate resistance to fluoroquinolone and tetracycline antibiotics by actively extruding these compounds and reducing their intracellular concentrations. Consequently, intense research has focused on inhibiting these efflux mechanisms using antimicrobial agents derived from natural or synthetic sources. This study used Fourier transform infrared spectroscopy (ATR-FTIR) to analyze the various functional groups present in p-coumaric acid. We also investigated the antibacterial activity of p-coumaric acid on strains of Staphylococcus aureus carrying the NorA and TetK efflux pumps, as well as the compound's ability to increase the fluorescence of ethidium bromide (EtBr) and Sytox Green. In addition, the interactions of this compound with NorA were analyzed using molecular docking, and its pharmacokinetic properties were evaluated using ADMET (Absorption, Distribution, Metabolism, Excretion and Toxicity) modeling. The results revealed that p-coumaric acid exhibited no direct antibacterial activity against the tested Staphylococcus aureus strains. However, significant reductions in the minimum inhibitory concentrations (MICs) of norfloxacin and EtBr, both used as NorA substrates, were observed when combined with p-coumaric acid. It was also observed that p-coumaric acid increased the fluorescence emission of EtBr and Sytox Green in strains 1199 and 1199B, suggesting the inhibition of the efflux mechanism and enhanced membrane permeabilization in S. aureus. The in silico analysis demonstrated that p-coumaric acid exhibits a favorable binding energy with NorA, comparable to that of chlorpromazine. These results position p-coumaric acid as a promising antibiotic adjuvant and efflux inhibitor in strains harboring NorA.
Collapse
Affiliation(s)
- Zildene de Sousa Silveira
- Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), 50670-901, Recife, PE, Brazil.
| | - Nair Silva Macêdo
- Postgraduate Program in Biological Chemistry (PPQB), Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil.
| | - Débora de Menezes Dantas
- Postgraduate Program in Biological Chemistry (PPQB), Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil.
| | | | - Débora Feitosa Muniz
- Postgraduate Program in Biological Chemistry (PPQB), Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil
| | | | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Cariri Regional University (URCA), Crato, CE, Brazil.
| | - Gabriel Gonçalves Alencar
- Laboratory of Microbiology and Molecular Biology (LMBM), Cariri Regional University (URCA), Crato, CE, Brazil.
| | - Emmanuel Silva Marinho
- State University of Ceará, Postgraduate Program in Natural Sciences, Natural Resources Bioprospecting and Monitoring Laboratory (LBMRN), Fortaleza, Ceará, Brazil.
| | - Matheus Nunes da Rocha
- State University of Ceará, Postgraduate Program in Natural Sciences, Natural Resources Bioprospecting and Monitoring Laboratory (LBMRN), Fortaleza, Ceará, Brazil.
| | - Marcia Machado Marinho
- State University of Ceará, Postgraduate Program in Natural Sciences, Natural Resources Bioprospecting and Monitoring Laboratory (LBMRN), Fortaleza, Ceará, Brazil.
| | - Hélcio Silva Dos Santos
- Northeast Biotechnology Network (RENORBIO-Nucleadora UECE), State University of Acaraú Valley (UVA), Sobral, CE, Brazil.
| | - Henrique Douglas Melo Coutinho
- Postgraduate Program in Biological Chemistry (PPQB), Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil.
| | | | - Márcia Vanusa da Silva
- Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), 50670-901, Recife, PE, Brazil.
| |
Collapse
|
8
|
Siritapetawee J, Hua Y, Talabnin C, Naewwan N, Charoenwattanasatien R, Phoovasawat C, Srichan S, Kantachot C. Potential for application of direct thrombin inhibitors isolated from Euphorbia resinifera O.Berg latex in fibrin clot formation. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1253:124480. [PMID: 39879729 DOI: 10.1016/j.jchromb.2025.124480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
Direct thrombin inhibitors (designated as EuRL-DTIs) were partially purified from ethanol extracts of Euphorbia resinifera O.Berg latex. The obtained EuRL-DTIs comprised four major compounds: two isomers of phenolic compounds (C19H26O12) and two amide compounds (tentatively identified as C24H44N4O4 and C36H66N6O6), as identified by liquid chromatography and electrospray ionisation quadrupole time-of-flight mass spectrometry (LC-ESI-QTOF-MS/MS), attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopy, and/or nuclear magnetic resonance (NMR) spectroscopy. The effects of EuRL-DTIs on human thrombin-induced fibrin clot production were analysed using thrombin time, sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE), synchrotron radiation X-ray tomographic microscopy (SRXTM), and scanning electron microscopy (SEM). Kinetic studies revealed that EuRL-DTIs inhibited human thrombin from cleaving the chromogenic substrate S2238, with a Ki of 3.7 μg/mL, in a non-competitive inhibition manner. All results supported the hypothesis that the EuRL-DTIs directly abolished thrombin activity in the production of fibrin clots without requiring a cofactor. The cytotoxicity test showed that EuRL-DTIs were nontoxic to normal human foetal lung fibroblasts (IMR-90). Thus, EuRL-DTIs have potential as antithrombotic agents for application as drugs for thrombosis treatments or in medical devices such as coating surgical sutures.
Collapse
Affiliation(s)
- Jaruwan Siritapetawee
- Biochemistry-Electrochemistry Research Unit, School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand.
| | - Yanling Hua
- The Center for Scientific and Technological Equipment, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Chutima Talabnin
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Nopporn Naewwan
- Translational Medicine Program, Institute of Medicine, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | | | | | - Supawan Srichan
- Synchrotron Light Research Institute (Public Organization), Nakhon Ratchasima, Thailand
| | - Chortip Kantachot
- Department of Biological Science, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, Thailand
| |
Collapse
|
9
|
Krzyżek P. Helicobacter pylori Efflux Pumps: A Double-Edged Sword in Antibiotic Resistance and Biofilm Formation. Int J Mol Sci 2024; 25:12222. [PMID: 39596287 PMCID: PMC11594842 DOI: 10.3390/ijms252212222] [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: 10/28/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Helicobacter pylori is a major pathogen associated with various gastric diseases. Despite decades of research, the treatment of H. pylori remains challenging. One of the primary mechanisms contributing to failures of therapies targeting this bacterium is genetic mutations in drug target sites, although the growing body of scientific data highlights that efflux pumps may also take part in this process. Efflux pumps are proteinaceous transporters actively expelling antimicrobial agents from the interior of the targeted cells and reducing the intracellular concentration of these compounds. Considering that efflux pumps contribute to both antimicrobial resistance and biofilm formation, an in-depth understanding of their properties may constitute a cornerstone in the development of novel therapeutics against H. pylori. In line with this, the aim of the current review is to describe the multitude of efflux pumps produced by H. pylori and present the data describing the involvement of these proteins in tolerance and/or resistance to various classes of antimicrobial substances.
Collapse
Affiliation(s)
- Paweł Krzyżek
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
10
|
Liu T, Xu H, Huang T, Liu G, Cao H, Lin Y, Li Y, Li Y, Yao X. Fuzheng Touxie Jiedu Huayu Decoction inhibits the MexAB-OprM efflux pump and quorum sensing-mediated biofilm formation in difficult-to-treat multidrug resistance Pseudomonas aeruginosa. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118365. [PMID: 38796070 DOI: 10.1016/j.jep.2024.118365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/06/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fuzheng Touxie Jiedu Huayu Decoction (FTJHD) is a commonly used clinical formula that has been found effective in resisting multidrug resistance-Pseudomonas aeruginosa in previous in vivo and in vitro studies. AIM OF THE STUDY To investigate the antimicrobial effects of FTJHD and its drug-containing serum alone or in combination with ceftazidime on difficult-to-treat multidrug resistance-P. aeruginosa (DTMDR-P. aeruginosa). MATERIALS AND METHODS The antibacterial effects of FTJHD and its drug-containing alone or in combination with ceftazidime against DTMDR-P. aeruginosa were examined by the tube dilution method and bacterial growth curves. The changes in the bacterial ultrastructure were examined by transmission electron microscopy. The biofilm formation ability of bacteria was examined by crystal violet staining and scanning electron microscopy. The expression of the MexAB-OprM efflux pump and quorum sensing system genes were validated through quantitative polymerase chain reaction. Molecular docking was used to evaluate the interaction between active components and the MexAB-OprM efflux pump. RESULTS FTJHD-containing serums at 1-, 2-, 4-, and 8-fold concentrations reduced the minimal inhibitory concentration (MIC) of ceftazidime against DTMDR-P. aeruginosa from 128 μg/mL to 64 μg/mL. Sub-inhibitory concentrations of ceftazidime in combination with FTJHD and FTJHD-containing serum prolonged the lag period of bacterial growth and reduced bacterial numbers. Additionally, 1/2 MIC of ceftazidime combined with FTJHD-containing serum significantly inhibited the activity of the MexAB-OprM efflux pump and quorum sensing system, thus reducing biofilm formation while causing more severe damage to the bacteria. Molecular docking revealed a strong affinity of quercetin, baicalein, luteolin, kaempferol, and β-sitosterol for the efflux pump regulatory proteins OprM and MexR. CONCLUSION FTJHD can exert synergistic anti-DTMDR-P. aeruginosa effects with ceftazidime by inhibiting biofilm formation mediated by the MexAB-OprM efflux pump and quorum sensing.
Collapse
Affiliation(s)
- Tong Liu
- Intensive Care Unit, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine (BUCM), Beijing, China
| | - Hongri Xu
- Emergency Department and Intensive Care Unit, BUCM Third Affiliated Hospital, Beijing, China.
| | - Tingxuan Huang
- Respiratory Department, BUCM Third Affiliated Hospital, Beijing, China
| | - Guoxing Liu
- Liu Zunji Chinese Medicine Clinic, Shannxi, China
| | - Hongyun Cao
- Emergency Department and Intensive Care Unit, BUCM Third Affiliated Hospital, Beijing, China
| | - Ying Lin
- Clinical Laboratory, Dongzhimen Hospital Affiliated to BUCM, Beijing, China
| | - Yali Li
- Emergency Department and Intensive Care Unit, BUCM Third Affiliated Hospital, Beijing, China
| | - Yan Li
- Intensive Care Unit, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine (BUCM), Beijing, China.
| | - Xingwei Yao
- Clinical Laboratory, Dongzhimen Hospital Affiliated to BUCM, Beijing, China.
| |
Collapse
|
11
|
Liu Y, Zhang J, Cheng D, Guo W, Liu X, Chen Z, Zhang Z, Ngo HH. Fate and mitigation of antibiotics and antibiotic resistance genes in microbial fuel cell and coupled systems. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 938:173530. [PMID: 38815818 DOI: 10.1016/j.scitotenv.2024.173530] [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: 04/09/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Microbial fuel cells (MFCs), known for their low energy consumption, high efficiency, and environmental friendliness, have been widely utilized for removing antibiotics from wastewater. Compared to conventional wastewater treatment methods, MFCs produce less sludge while exhibiting superior antibiotic removal capacity, effectively reducing the spread of antibiotic resistance genes (ARGs). This study investigates 1) the mechanisms of ARGs generation and proliferation in MFCs; 2) the influencing factors on the fate and removal of antibiotics and ARGs; and 3) the fate and mitigation of ARGs in MFC and MFC-coupled systems. It is indicated that high removal efficiency of antibiotics and minimal amount of sludge production contribute the mitigation of ARGs in MFCs. Influencing factors, such as cathode potential, electrode materials, salinity, initial antibiotic concentration, and additional additives, can lead to the selection of tolerant microbial communities, thereby affecting the abundance of ARGs carried by various microbial hosts. Integrating MFCs with other wastewater treatment systems can synergistically enhance their performance, thereby improving the overall removal efficiency of ARGs. Moreover, challenges and future directions for mitigating the spread of ARGs using MFCs are suggested.
Collapse
Affiliation(s)
- Yufei Liu
- College of Safety and Environmental Engineering, Shandong University of Science and Technology, Qingdao 266590, China; Institute of Yellow River Delta Earth Surface Processes and Ecological Integrity, Shandong University of Science and Technology, Qingdao 266590, China
| | - Jian Zhang
- College of Safety and Environmental Engineering, Shandong University of Science and Technology, Qingdao 266590, China; Institute of Yellow River Delta Earth Surface Processes and Ecological Integrity, Shandong University of Science and Technology, Qingdao 266590, China
| | - Dongle Cheng
- College of Safety and Environmental Engineering, Shandong University of Science and Technology, Qingdao 266590, China; Institute of Yellow River Delta Earth Surface Processes and Ecological Integrity, Shandong University of Science and Technology, Qingdao 266590, China.
| | - Wenshan Guo
- Centre for Technology in Water and Wastewater, School of Civil and Environmental Engineering, University of Technology Sydney, Sydney, NWS 2007, Australia
| | - Xiaoqing Liu
- Centre for Technology in Water and Wastewater, School of Civil and Environmental Engineering, University of Technology Sydney, Sydney, NWS 2007, Australia
| | - Zhijie Chen
- UNSW Water Research Centre, School of Civil and Environmental Engineering, The University New South Wales, Sydney, NSW 2052, Australia
| | - Zehao Zhang
- National Engineering Laboratory of Urban Sewage Advanced Treatment and Resource Utilization Technology, The College of Architecture and Civil Engineering, Beijing University of Technology, Beijing 100124, China
| | - Huu Hao Ngo
- College of Safety and Environmental Engineering, Shandong University of Science and Technology, Qingdao 266590, China; Institute of Yellow River Delta Earth Surface Processes and Ecological Integrity, Shandong University of Science and Technology, Qingdao 266590, China; Centre for Technology in Water and Wastewater, School of Civil and Environmental Engineering, University of Technology Sydney, Sydney, NWS 2007, Australia.
| |
Collapse
|
12
|
Fakhoury AA, Thompson TP, Rahman KM, Megaw J, McAteer MI, Skvortsov T, Kelly SA, Gilmore BF. Identification and characterisation of two functional antibiotic MATE efflux pumps in the archaeon Halorubrum amylolyticum. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:21. [PMID: 39843964 PMCID: PMC11721430 DOI: 10.1038/s44259-024-00036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 06/17/2024] [Indexed: 01/24/2025]
Abstract
Multidrug efflux pumps have been found to play a crucial role in drug resistance in bacteria and eukaryotes. In this study, we investigated the presence of functional multidrug and toxic compound extrusion (MATE) efflux pumps, inferred from whole genome sequencing, in the halophilic archaeon Halorubrum amylolyticum CSM52 using Hoechst 33342 dye accumulation and antimicrobial sensitivity tests in the presence and absence of efflux pump inhibitors (EPIs). The whole genome sequence of H. amylolyticum CSM52 contained two putative MATE-type efflux pump genes, which may contribute to the inherent resistance to conventional antimicrobial agents reported in archaea. Antimicrobial susceptibility of the wild-type H. amylolyticum CSM52 testing revealed a lack of sensitivity to a wide range of antimicrobials, including glycopeptides, aminoglycosides, macrolides, fluoroquinolones, tetracycline, and chloramphenicol. However, the presence of EPIs, such as thioridazine, fluoxetine, and chlorpromazine, significantly increased the susceptibility of H. amylolyticum CSM52 to a number of these antimicrobials, indicating the potential involvement of efflux pumps in the observed resistance. A molecular modelling study with EPIs and substrate antimicrobials provided important insights into the molecular interactions with the putative transporter. It suggests that the occupancy of the transporter channel by EPIs has the potential to impact the efflux of antimicrobials. Phylogenetic analysis of the amino acid sequences of both MATE pumps showed low similarity with bacterial representatives, suggesting the presence of novel and distinct MATE efflux pumps in archaea. Our findings provide the first experimental evidence of active antibiotic efflux mechanisms in archaea and their potential roles in antimicrobial resistance, broadening our understanding of mechanisms of archaeal antimicrobial resistance, an overlooked aspect of AMR research.
Collapse
Affiliation(s)
- Asma A Fakhoury
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | - Thomas P Thompson
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Khondaker Miraz Rahman
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - Julianne Megaw
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
| | - Matthew I McAteer
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Timofey Skvortsov
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Stephen A Kelly
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Brendan F Gilmore
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK.
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK.
| |
Collapse
|
13
|
Wei PW, Wang X, Wang C, Chen M, Liu MZ, Liu WX, He YL, Xu GB, Zheng XH, Zhang H, Liu HM, Wang B. Ginkgo biloba L. exocarp petroleum ether extract inhibits methicillin-resistant Staphylococcus aureus by modulating ion transport, virulence, and biofilm formation in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:117957. [PMID: 38493904 DOI: 10.1016/j.jep.2024.117957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 03/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As reported in the Ancient Chinese Medicinal Books, Ginkgo biloba L. fruit has been used as a traditional Chinese medicine for the treatment asthma and cough or as a disinfectant. Our previous study demonstrated that G. biloba exocarp extract (GBEE), an extract of a traditional Chinese herb, inhibits the formation of methicillin-resistant Staphylococcus aureus (MRSA) biofilms. However, GBEE is a crude extract that contains many components, and the underlying mechanisms of purified GBEE fractions extracted with solvents of different polarities are unknown. AIM OF THE STUDY This study aimed to investigate the different components in GBEE fractions extracted with solvents of different polarities and their antibacterial effects and mechanisms against MRSA and Staphylococcus haemolyticus biofilms both in vitro and in vivo. METHODS The components in different fractions were detected by high-performance liquid chromatography-high resolution mass spectrometry (HPLC-HRMS). Microbroth dilution assays and time growth curves were used to determine the antibacterial effects of the fractions on 15 clinical bacterial isolates. Crystal violet staining, scanning electron microscopy (SEM) and transmission electron microscopy (TEM) were utilized to identify the fractions that affected bacterial biofilm formation. The potential MRSA targets of the GBEE fraction obtained with petroleum ether (PE), denoted GBEE-PE, were screened by transcriptome sequencing, and the gene expression profile was verified by quantitative polymerase chain reaction (qPCR). RESULTS HPLC-HRMS analysis revealed that the four GBEE fractions (extracted with petroleum ether, ethyl acetate, n-butanol, and water) contained different ginkgo components, and the antibacterial effects decreased as the polarity of the extraction solvent increased. The antibacterial activity of GBEE-PE was greater than that of the GBEE fraction extracted with ethyl acetate (EA). GBEE-PE improved H. illucens survival and reduced MRSA colonization in model mouse organs. Crystal violet staining and SEM and TEM analyses revealed that GBEE-PE inhibited MRSA and S. haemolyticus biofilm formation. Transcriptional analysis revealed that GBEE-PE inhibits MRSA biofilms by altering ion transport, cell wall metabolism and virulence-related gene expression. In addition, the LO2 cell viability and H. illucens toxicity assay data showed that GBEE-PE at 20 mg/kg was nontoxic. CONCLUSION The GBEE fractions contained different components, and their antibacterial effects decreased with increases in the polarity of the extraction solvent. GBEE-PE limited MRSA growth and biofilm formation by affecting ion transport, cell wall synthesis, and virulence-related pathways. This research provides a more detailed overview of the mechanism by which GBEE-PE inhibits MRSA both in vitro and in vivo and suggests that GBEE-PE is a new prospective antimicrobial with the potential to be used in MRSA therapeutics in the future.
Collapse
Affiliation(s)
- Peng-Wei Wei
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Xu Wang
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Cong Wang
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Formulation (R&D) Department, Guiyang, 550001, China
| | - Ming Chen
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China
| | - Meng-Zhu Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China
| | - Wen-Xia Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, School of Basic Medical Science, Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Yan-Ling He
- Zhejiang Hisun Pharmaceutical Co., Ltd., Taizhou, 318000, Zhejiang, China
| | - Guo-Bo Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, Guizhou, China.
| | - Xiao-He Zheng
- Zhejiang Hisun Pharmaceutical Co., Ltd., Taizhou, 318000, Zhejiang, China
| | - Hua Zhang
- Department of Laboratory Medicine, Guizhou Provincial People's Hospital, Affiliated Hospital of Guizhou University, Guiyang, 550002, Guizhou, China.
| | - Hong-Mei Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China.
| | - Bing Wang
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China; Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, School of Basic Medical Science, Guizhou Medical University, Guiyang, 561113, Guizhou, China.
| |
Collapse
|
14
|
Luo L, Wang Z, Huang X, Gu JD, Yu C, Deng O. The fate of antibiotic resistance genes in wastewater containing microalgae treated by chlorination, ultra-violet, and Fenton reaction. WATER RESEARCH 2024; 254:121392. [PMID: 38430757 DOI: 10.1016/j.watres.2024.121392] [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: 10/23/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Antibiotic resistance genes (ARGs) and bacteria (ARBs) in the effluent of wastewater treatment plants (WWTPs) are of utmost importance for the dissemination of ARGs in natural aquatic environments. Therefore, there is an urgent need for effective technologies to eliminate WWTP ARGs/ARBs and mitigate the associated risks posed by the discharged ARG in aquatic environments. To test the effective technology for eliminating ARGs/ARBs, we compared the removal of ARGs and ARBs by three different tertiary treatments, namely ultra-violet (UV) disinfection, chlorination disinfection, and Fenton oxidation. Then, the treated wastewater was co-cultured with Chlorella vulgaris (representative of aquatic biota) to investigate the fate of discharged ARGs into the aquatic environment. The results demonstrated that chlorination (at a chlorine concentration of 15 mg/L) and Fenton (at pH 2.73, with 0.005 mol/L Fe2+ and 0.0025 mol/L H2O2) treatment showed higher efficacy in ARG removal (1.8 - 4.17 logs) than UV treatment (15 min) (1.29 - 3.87 logs). Moreover, chlorine at 15 mg/L and Fenton treatment effectively suppressed ARB regeneration while UV treatment for 15 min could not. Regardless of treatments tested in this study, the input of treated wastewater to the Chlorella system increased the number of ARGs and mobile genetic elements (MGEs), indicating the potential risk of ARG dissemination associated with WWTP discharge. Among the wastewater-Chlorella co-culture systems, chlorination resulted in less of an increase in the number of ARGs and MGEs compared to Fenton and UV treatment. When comparing the wastewater systems to the co-culture systems, it was observed that Chlorella vulgaris reduced the number of ARGs and MGEs in chlorination and UV-treated wastewater; however, Chlorella vulgaris promoted ARG survival in Fenton-treated water, suggesting that aquatic microalgae might act as a barrier to ARG dissemination. Overall, chlorination treatment not only effectively removes ARGs and inhibits ARB regeneration but also shows a lower risk of ARG dissemination. Therefore, chlorination is recommended for practical application in controlling the spread of discharged ARGs from WWTP effluent in natural aquatic environments.
Collapse
Affiliation(s)
- Ling Luo
- College of Environmental Sciences, Sichuan Agricultural University, Chengdu 611130, PR China.
| | - Zimu Wang
- College of Environmental Sciences, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Xin Huang
- College of Environmental Sciences, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Ji-Dong Gu
- Environmental Science and Engineering Research Group, Guangdong Technion - Israel Institute of Technology, Shantou 515063, PR China
| | - Chenxiao Yu
- College of Environmental Sciences, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Ouping Deng
- College of Environmental Sciences, Sichuan Agricultural University, Chengdu 611130, PR China; College of Resources, Sichuan Agricultural University, Chengdu 611130, PR China.
| |
Collapse
|
15
|
Jiang Q, Geng F, Shen J, Zhu P, Lu Z, Lu F, Zhou L. Blue light-mediated gene expression as a promising strategy to reduce antibiotic resistance in Escherichia coli. Biotechnol J 2024; 19:e2400023. [PMID: 38719589 DOI: 10.1002/biot.202400023] [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: 01/10/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 08/21/2024]
Abstract
The discovery of antibiotics has noticeably promoted the development of human civilization; however, antibiotic resistance in bacteria caused by abusing and overusing greatly challenges human health and food safety. Considering the worsening situation, it is an urgent demand to develop emerging nontraditional technologies or methods to address this issue. With the expanding of synthetic biology, optogenetics exhibits a tempting prospect for precisely regulating gene expression in many fields. Consequently, it is attractive to employ optogenetics to reduce the risk of antibiotic resistance. Here, a blue light-controllable gene expression system was established in Escherichia coli based on a photosensitive DNA-binding protein (EL222). Further, this strategy was successfully applied to repress the expression of β-lactamase gene (bla) using blue light illumination, resulting a dramatic reduction of ampicillin resistance in engineered E. coli. Moreover, blue light was utilized to induce the expression of the mechanosensitive channel of large conductance (MscL), triumphantly leading to the increase of streptomycin susceptibility in engineered E. coli. Finally, the increased susceptibility of ampicillin and streptomycin was simultaneously induced by blue light in the same E. coli cell, revealing the excellent potential of this strategy in controlling multidrug-resistant (MDR) bacteria. As a proof of concept, our work demonstrates that light can be used as an alternative tool to prolong the use period of common antibiotics without developing new antibiotics. And this novel strategy based on optogenetics shows a promising foreground to combat antibiotic resistance in the future.
Collapse
Affiliation(s)
- Qingwei Jiang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| | - Feng Geng
- College of Pharmacy, Binzhou Medical University, Yantai, PR China
| | - Juan Shen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| | - Ping Zhu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| | - Fengxia Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| | - Libang Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| |
Collapse
|
16
|
Jaiswal N, Kumar A. Modulators of Candida albicans Membrane Drug Transporters: A Lucrative Portfolio for the Development of Effective Antifungals. Mol Biotechnol 2024; 66:960-974. [PMID: 38206530 DOI: 10.1007/s12033-023-01017-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/01/2023] [Indexed: 01/12/2024]
Abstract
The escalating prevalence of membrane drug transporters and drug efflux pumps in pathogenic yeast like Candida albicans necessitates a comprehensive understanding of their roles in MDR. The overexpression of drug transporter families, ABC and MFS, implicated in MDR through drug efflux and poses a significant challenge in the diagnosis and treatment of fungal infection. Various mechanisms have been proposed for MDR; however, the upregulation of ABC and MFS superfamily transporters is most noticeable in MDR. The direct inhibition of these transporters seems an efficient strategy to overcome this problem. The goal of the article is to present an overview of the prospect of utilizing these modulators of C. albicans drug transports as effective antifungal molecules against MDR addressing a critical gap in the field. The review tries to address to prevent drug extrusion by modulating the expression of drug transporters of C. albicans. The review discussed the progress in identifying potent, selective, and non-toxic modulators of these transporters to develop some effective antifungals and overcome MDR. We reviewed major studies in this area and found that recent work has shifted toward the exploration of natural compounds as potential modulators to restore drug sensitivity in MDR fungal cells. The focus of this review is to survey and interpret current research information on modulators of C. albicans drug transporters from natural sources emphasizing those compounds that are potent antifungal agents.
Collapse
Affiliation(s)
- Neha Jaiswal
- Department of Biotechnology, National Institute of Technology, Raipur, CG, 492010, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, CG, 492010, India.
| |
Collapse
|
17
|
Ganjo AR, Balaky STJ, Mawlood AH, Smail SB, Shabila NP. Characterization of genes related to the efflux pump and porin in multidrug-resistant Escherichia coli strains isolated from patients with COVID-19 after secondary infection. BMC Microbiol 2024; 24:122. [PMID: 38600509 PMCID: PMC11005145 DOI: 10.1186/s12866-024-03283-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Escherichia coli (E. coli) is a multidrug resistant opportunistic pathogen that can cause secondary bacterial infections in patients with COVID-19. This study aimed to determine the antimicrobial resistance profile of E. coli as a secondary bacterial infection in patients with COVID-19 and to assess the prevalence and characterization of genes related to efflux pumps and porin. METHODS A total of 50 nonduplicate E. coli isolates were collected as secondary bacterial infections in COVID-19 patients. The isolates were cultured from sputum samples. Confirmation and antibiotic susceptibility testing were conducted by Vitek 2. PCR was used to assess the prevalence of the efflux pump and porin-related genes in the isolates. The phenotypic and genotypic evolution of antibiotic resistance genes related to the efflux pump was evaluated. RESULTS The E. coli isolates demonstrated high resistance to ampicillin (100%), cefixime (62%), cefepime (62%), amoxicillin-clavulanic acid (60%), cefuroxime (60%), and ceftriaxone (58%). The susceptibility of E. coli to ertapenem was greatest (92%), followed by imipenem (88%), meropenem (86%), tigecycline (80%), and levofloxacin (76%). Regarding efflux pump gene combinations, there was a significant association between the acrA gene and increased resistance to levofloxacin, between the acrB gene and decreased resistance to meropenem and increased resistance to levofloxacin, and between the ompF and ompC genes and increased resistance to gentamicin. CONCLUSIONS The antibiotics ertapenem, imipenem, meropenem, tigecycline, and levofloxacin were effective against E. coli in patients with COVID-19. Genes encoding efflux pumps and porins, such as acrA, acrB, and outer membrane porins, were highly distributed among all the isolates. Efflux pump inhibitors could be alternative antibiotics for restoring tetracycline activity in E. coli isolates.
Collapse
Affiliation(s)
- Aryan R Ganjo
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Salah Tofik Jalal Balaky
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq.
- Department of Medical Microbiology, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq.
| | - Ahang Hasan Mawlood
- Department of Medical Microbiology, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
- Department of Medical Laboratory Technique, College of Medical Technology, AL-Kitab University, Kirkuk, Iraq
| | | | - Nazar P Shabila
- College of Health Sciences, Catholic University in Erbil, Erbil, Kurdistan Region, Iraq
- Department of Community Medicine, College of Medicine, Hawler Medical University, Erbil, Iraq
| |
Collapse
|
18
|
Weigert Muñoz A, Zhao W, Sieber SA. Monitoring host-pathogen interactions using chemical proteomics. RSC Chem Biol 2024; 5:73-89. [PMID: 38333198 PMCID: PMC10849124 DOI: 10.1039/d3cb00135k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/09/2023] [Indexed: 02/10/2024] Open
Abstract
With the rapid emergence and the dissemination of microbial resistance to conventional chemotherapy, the shortage of novel antimicrobial drugs has raised a global health threat. As molecular interactions between microbial pathogens and their mammalian hosts are crucial to establish virulence, pathogenicity, and infectivity, a detailed understanding of these interactions has the potential to reveal novel therapeutic targets and treatment strategies. Bidirectional molecular communication between microbes and eukaryotes is essential for both pathogenic and commensal organisms to colonise their host. In particular, several devastating pathogens exploit host signalling to adjust the expression of energetically costly virulent behaviours. Chemical proteomics has emerged as a powerful tool to interrogate the protein interaction partners of small molecules and has been successfully applied to advance host-pathogen communication studies. Here, we present recent significant progress made by this approach and provide a perspective for future studies.
Collapse
Affiliation(s)
- Angela Weigert Muñoz
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 D-85748 Garching Germany
| | - Weining Zhao
- College of Pharmacy, Shenzhen Technology University Shenzhen 518118 China
| | - Stephan A Sieber
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 D-85748 Garching Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Germany
| |
Collapse
|
19
|
Chauhan SS, Gupta A, Srivastava A, Parthasarathi R. Discovering targeted inhibitors for Escherichia coli efflux pump fusion proteins using computational and structure-guided approaches. J Comput Chem 2024; 45:13-24. [PMID: 37656428 DOI: 10.1002/jcc.27215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/18/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Multidrug resistance pathogens causing infections and illness remain largely untreated clinically. Efflux pumps are one of the primary processes through which bacteria develop resistance by transferring antibiotics from the interior of their cells to the outside environment. Inhibiting these pumps by developing efficient derivatives appears to be a promising strategy for restoring antibiotic potency. This investigation explores literature-reported inhibitors of E. coli efflux pump fusion proteins AcrB-AcrA and identify potential chemical derivatives of these inhibitors to overcome the limitations. Using computational and structure-guided approaches, a study was conducted with the selected inhibitors (AcrA:25-AcrB:59) obtained by data mining and their derivatives (AcrA:857-AcrB:3891) to identify their inhibitory effect on efflux pump using virtual screening, molecular docking and density functional theory (DFT) calculations. The finding indicates that Compound 2 (ZINC000072136376) has shown better binding and a significant inhibitory effect on AcrA, while Compound 3 (ZINC000072266819) has shown stronger binding and substantial inhibition effect on both non-mutant and mutated AcrB subunits. The identified derivatives could exhibit a better inhibitor and provide a potential approach for restoring the actions of resistant antibiotics.
Collapse
Affiliation(s)
- Shweta Singh Chauhan
- Computational Toxicology Facility, Toxicoinformatics & Industrial Research, CSIR-Indian Institute of Toxicology Research, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anshika Gupta
- Computational Toxicology Facility, Toxicoinformatics & Industrial Research, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Aashna Srivastava
- Computational Toxicology Facility, Toxicoinformatics & Industrial Research, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Ramakrishnan Parthasarathi
- Computational Toxicology Facility, Toxicoinformatics & Industrial Research, CSIR-Indian Institute of Toxicology Research, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
20
|
Vieira Da Cruz A, Jiménez-Castellanos JC, Börnsen C, Van Maele L, Compagne N, Pradel E, Müller RT, Meurillon V, Soulard D, Piveteau C, Biela A, Dumont J, Leroux F, Deprez B, Willand N, Pos KM, Frangakis AS, Hartkoorn RC, Flipo M. Pyridylpiperazine efflux pump inhibitor boosts in vivo antibiotic efficacy against K. pneumoniae. EMBO Mol Med 2024; 16:93-111. [PMID: 38177534 PMCID: PMC10897476 DOI: 10.1038/s44321-023-00007-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 01/06/2024] Open
Abstract
Antimicrobial resistance is a global problem, rendering conventional treatments less effective and requiring innovative strategies to combat this growing threat. The tripartite AcrAB-TolC efflux pump is the dominant constitutive system by which Enterobacterales like Escherichia coli and Klebsiella pneumoniae extrude antibiotics. Here, we describe the medicinal chemistry development and drug-like properties of BDM91288, a pyridylpiperazine-based AcrB efflux pump inhibitor. In vitro evaluation of BDM91288 confirmed it to potentiate the activity of a panel of antibiotics against K. pneumoniae as well as revert clinically relevant antibiotic resistance mediated by acrAB-tolC overexpression. Using cryo-EM, BDM91288 binding to the transmembrane region of K. pneumoniae AcrB was confirmed, further validating the mechanism of action of this inhibitor. Finally, proof of concept studies demonstrated that oral administration of BDM91288 significantly potentiated the in vivo efficacy of levofloxacin treatment in a murine model of K. pneumoniae lung infection.
Collapse
Affiliation(s)
- Anais Vieira Da Cruz
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Juan-Carlos Jiménez-Castellanos
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Clara Börnsen
- Buchmann Institute for Molecular Life Sciences and Institute for Biophysics, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Laurye Van Maele
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Nina Compagne
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Elizabeth Pradel
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Reinke T Müller
- Institute of Biochemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
| | - Virginie Meurillon
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Daphnée Soulard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Catherine Piveteau
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Alexandre Biela
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Julie Dumont
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Florence Leroux
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, US 41-UAR 2014-PLBS, F-59000, Lille, France
| | - Benoit Deprez
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Nicolas Willand
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Klaas M Pos
- Institute of Biochemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany.
| | - Achilleas S Frangakis
- Buchmann Institute for Molecular Life Sciences and Institute for Biophysics, Goethe University Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany.
| | - Ruben C Hartkoorn
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France.
| | - Marion Flipo
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France.
| |
Collapse
|
21
|
Moreno-Chamba B, Salazar-Bermeo J, Navarro-Simarro P, Narváez-Asensio M, Martínez-Madrid MC, Saura D, Martí N, Valero M. Autoinducers modulation as a potential anti-virulence target of bacteria by phenolic compounds. Int J Antimicrob Agents 2023; 62:106937. [PMID: 37517626 DOI: 10.1016/j.ijantimicag.2023.106937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/05/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023]
Abstract
OBJECTIVE The goal of this study was to determine the effects of phenolic extracts from grape (GrPE), pomegranate (PoPE), and persimmon (PePE) by-products on bacterial virulence activities such as biofilms, motility, energy-dependent efflux pumps, and β-lactamase activity, which are modulated primarily by quorum sensing (QS), defining their potential applications. METHOD The microdilution method was used to determine the minimum inhibitory concentration (MIC) and sub-inhibitory concentrations (SICs) of the extracts against reference pathogenic bacteria. The antibacterial mode of action was determined by labelling bacterial cells in in vivo cell-tracking experiments. RESULTS Antibiograms showed that PoPE inhibited bacteria at lower concentrations, and PePE had a stronger effect against Klebsiella pneumoniae. Both extracts caused significant cell membrane damage (CMD), whereas GrPE did not. At SICs, all extracts showed anti-QS activity, especially PePE, which inhibited violacein and pyocyanin production at 1/128 × MIC. Additionally, QS autoinducers found in Chromobacterium violaceum and Pseudomonas aeruginosa were modulated by the extracts; PePE showed the highest modulation. Antibiofilm assays revealed that GrPE, at MIC and 2 × MIC, acted as a potent antibiofilm agent against biofilms of Pseudomonas putida, Bacillus cereus, and Staphylococcus aureus, which was related to disruption of swarming motility by GrPE. All extracts, especially PoPE, exerted a potent effect against the activation of efflux pumps of P. aeruginosa as well as β-lactamase activity in K. pneumoniae. CONCLUSION Results suggest that the anti-virulence potential of the extracts may be related to their effect as extracellular autoinducer modulators. This study allowed to define potential applications of these extracts.
Collapse
Affiliation(s)
- Bryan Moreno-Chamba
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche (UMH), Alicante, Spain; Instituto de Ingeniería de Alimentos para el Desarrollo, Universitat Politècnica de València, Valencia, Spain
| | - Julio Salazar-Bermeo
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche (UMH), Alicante, Spain; Instituto de Ingeniería de Alimentos para el Desarrollo, Universitat Politècnica de València, Valencia, Spain
| | - Pablo Navarro-Simarro
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche (UMH), Alicante, Spain
| | - Marta Narváez-Asensio
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche (UMH), Alicante, Spain
| | - María Concepción Martínez-Madrid
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche (UMH), Alicante, Spain
| | - Domingo Saura
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche (UMH), Alicante, Spain
| | - Nuria Martí
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche (UMH), Alicante, Spain
| | - Manuel Valero
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche (UMH), Alicante, Spain.
| |
Collapse
|
22
|
Peng B, Li H, Peng XX. Call for next-generation drugs that remove the uptake barrier to combat antibiotic resistance. Drug Discov Today 2023; 28:103753. [PMID: 37640151 DOI: 10.1016/j.drudis.2023.103753] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/15/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Existing antibacterial agents can be categorized into two generations, but bacterial insensitivity towards both of these generations poses a serious public health challenge worldwide. Thus, novel approaches and/or novel antibacterials are urgently needed to maintain a concentration of antibacterials that is lethal to bacteria that are resistant to existing antibiotic treatments. Metabolite(s)-based adjuvants that promote antibiotic uptake and enhance antibiotic efficacy are an effective strategy that is unlikely to develop resistance. Thus, we propose a metabolite(s)-based approach, in which metabolites and antibacterials are combined, as a promising strategy for the development of next-generation agents to combat a variety of antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Bo Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, People's Republic of China
| | - Hui Li
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, People's Republic of China
| | - Xuan-Xian Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, People's Republic of China.
| |
Collapse
|
23
|
Avakh A, Grant GD, Cheesman MJ, Kalkundri T, Hall S. The Art of War with Pseudomonas aeruginosa: Targeting Mex Efflux Pumps Directly to Strategically Enhance Antipseudomonal Drug Efficacy. Antibiotics (Basel) 2023; 12:1304. [PMID: 37627724 PMCID: PMC10451789 DOI: 10.3390/antibiotics12081304] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/26/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) poses a grave clinical challenge due to its multidrug resistance (MDR) phenotype, leading to severe and life-threatening infections. This bacterium exhibits both intrinsic resistance to various antipseudomonal agents and acquired resistance against nearly all available antibiotics, contributing to its MDR phenotype. Multiple mechanisms, including enzyme production, loss of outer membrane proteins, target mutations, and multidrug efflux systems, contribute to its antimicrobial resistance. The clinical importance of addressing MDR in P. aeruginosa is paramount, and one pivotal determinant is the resistance-nodulation-division (RND) family of drug/proton antiporters, notably the Mex efflux pumps. These pumps function as crucial defenders, reinforcing the emergence of extensively drug-resistant (XDR) and pandrug-resistant (PDR) strains, which underscores the urgency of the situation. Overcoming this challenge necessitates the exploration and development of potent efflux pump inhibitors (EPIs) to restore the efficacy of existing antipseudomonal drugs. By effectively countering or bypassing efflux activities, EPIs hold tremendous potential for restoring the antibacterial activity against P. aeruginosa and other Gram-negative pathogens. This review focuses on concurrent MDR, highlighting the clinical significance of efflux pumps, particularly the Mex efflux pumps, in driving MDR. It explores promising EPIs and delves into the structural characteristics of the MexB subunit and its substrate binding sites.
Collapse
Affiliation(s)
| | | | | | | | - Susan Hall
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4222, Australia; (A.A.); (G.D.G.); (M.J.C.); (T.K.)
| |
Collapse
|
24
|
Kadeřábková N, Mahmood AJS, Furniss RCD, Mavridou DAI. Making a chink in their armor: Current and next-generation antimicrobial strategies against the bacterial cell envelope. Adv Microb Physiol 2023; 83:221-307. [PMID: 37507160 PMCID: PMC10517717 DOI: 10.1016/bs.ampbs.2023.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Gram-negative bacteria are uniquely equipped to defeat antibiotics. Their outermost layer, the cell envelope, is a natural permeability barrier that contains an array of resistance proteins capable of neutralizing most existing antimicrobials. As a result, its presence creates a major obstacle for the treatment of resistant infections and for the development of new antibiotics. Despite this seemingly impenetrable armor, in-depth understanding of the cell envelope, including structural, functional and systems biology insights, has promoted efforts to target it that can ultimately lead to the generation of new antibacterial therapies. In this article, we broadly overview the biology of the cell envelope and highlight attempts and successes in generating inhibitors that impair its function or biogenesis. We argue that the very structure that has hampered antibiotic discovery for decades has untapped potential for the design of novel next-generation therapeutics against bacterial pathogens.
Collapse
Affiliation(s)
- Nikol Kadeřábková
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States
| | - Ayesha J S Mahmood
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States
| | - R Christopher D Furniss
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Despoina A I Mavridou
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States; John Ring LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
25
|
Szal T, Chauhan SS, Lewe P, Rachad FZ, Madre M, Paunina L, Witt S, Parthasarathi R, Windshügel B. Efflux Pump-Binding 4(3-Aminocyclobutyl)Pyrimidin-2-Amines Are Colloidal Aggregators. Biomolecules 2023; 13:1000. [PMID: 37371580 PMCID: PMC10296211 DOI: 10.3390/biom13061000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Efflux pumps are a relevant factor in antimicrobial resistance. In E. coli, the tripartite efflux pump AcrAB-TolC removes a chemically diverse set of antibiotics from the bacterium. Therefore, small molecules interfering with efflux pump function are considered adjuvants for improving antimicrobial therapies. Several compounds targeting the periplasmic adapter protein AcrA and the efflux pump AcrB have been identified to act synergistically with different antibiotics. Among those, several 4(3-aminocyclobutyl)pyrimidin-2-amines have been shown to bind to both proteins. In this study, we intended to identify analogs of these substances with improved binding affinity to AcrA using virtual screening followed by experimental validation. While we succeeded in identifying several compounds showing a synergistic effect with erythromycin on E. coli, biophysical studies suggested that 4(3-aminocyclobutyl)pyrimidin-2-amines form colloidal aggregates that do not bind specifically to AcrA. Therefore, these substances are not suited for further development. Our study emphasizes the importance of implementing additional control experiments to identify aggregators among bioactive compounds.
Collapse
Affiliation(s)
- Tania Szal
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525 Hamburg, Germany; (T.S.); (F.-Z.R.)
- School of Science, Constructor University, 28759 Bremen, Germany
| | - Shweta Singh Chauhan
- Computational Toxicology Facility, Toxicoinformatics & Industrial Research CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; (S.S.C.); (R.P.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Philipp Lewe
- Centre for Structural Systems Biology (CSSB), University Medical Center Hamburg-Eppendorf (UKE), 22607 Hamburg, Germany; (P.L.); (S.W.)
| | - Fatima-Zahra Rachad
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525 Hamburg, Germany; (T.S.); (F.-Z.R.)
| | - Marina Madre
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (M.M.); (L.P.)
| | - Laura Paunina
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia; (M.M.); (L.P.)
| | - Susanne Witt
- Centre for Structural Systems Biology (CSSB), University Medical Center Hamburg-Eppendorf (UKE), 22607 Hamburg, Germany; (P.L.); (S.W.)
| | - Ramakrishnan Parthasarathi
- Computational Toxicology Facility, Toxicoinformatics & Industrial Research CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; (S.S.C.); (R.P.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Björn Windshügel
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525 Hamburg, Germany; (T.S.); (F.-Z.R.)
- School of Science, Constructor University, 28759 Bremen, Germany
| |
Collapse
|
26
|
Zhang Q, Zhou L, Zhao Y, Gao S, Yang Y, Chen Q, Li W, Qi Q, Dong Q, Lei J, Guo X, Gao Q, Yang Y. Uncovering the virome and its interaction with antibiotic resistome during compost fertilization. JOURNAL OF HAZARDOUS MATERIALS 2023; 457:131763. [PMID: 37311294 DOI: 10.1016/j.jhazmat.2023.131763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/26/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023]
Abstract
Antibiotic resistance is a pressing global health issue, leading to increased illnesses and fatalities. The contribution of viruses to the acquisition, preservation, and dissemination of antibiotic resistance genes (ARGs) is not yet fully understood. By using a high-throughput functional gene-based microarray (GeoChip 5.0), this study examines the prevalence and relative abundance of bacteriophage and eukaryotic viral genes in swine manure, compost, compost-amended agricultural soil, and unamended soil from suburban regions of Beijing, China. Our findings reveal a significantly elevated presence of biomarker viral genes in compost-amended soils compared to unamended soils, suggesting potential health risks associated with compost fertilization. We also observed stronger ecological interactions between ARGs and viral genes in manure and compost than in soils. Network analysis identified arabinose efflux permeases and EmrB/QacA resistance genes, linked to CRISPR encoding sequences, as keystone nodes, indicating possible ARG acquisition via virus infections. Moreover, positive correlations were found between viral genes, antibiotic concentrations, and ARG diversity in manure, compost, and compost-amended soils, highlighting a likely pathway for virus-mediated ARG transfer. In summary, our results indicate that use of compost as a fertilizer in agricultural settings could facilitate the spread of ARGs through viral mechanisms, allowing for time-delayed genetic exchanges over broader temporal and spatial scales than ARGs within bacterial genomes.
Collapse
Affiliation(s)
- Qingxia Zhang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Lei Zhou
- Center for professional training and service, China Association for Science and Technology, China
| | - Yilong Zhao
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing 100084, China
| | - Shuhong Gao
- School of Civil and Environmental Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Yanjun Yang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Qingyun Chen
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wenhui Li
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Qi Qi
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qiang Dong
- Institute of Chemical Defense, Beijing 102205, China
| | - Jiesi Lei
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing 100084, China
| | - Xue Guo
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing 100084, China
| | - Qun Gao
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing 100084, China.
| | - Yunfeng Yang
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
27
|
Moore-Machacek A, Gloe A, O'Leary N, Reen FJ. Efflux, Signaling and Warfare in a Polymicrobial World. Antibiotics (Basel) 2023; 12:antibiotics12040731. [PMID: 37107093 PMCID: PMC10135244 DOI: 10.3390/antibiotics12040731] [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: 02/24/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
The discovery void of antimicrobial development has occurred at a time when the world has seen a rapid emergence and spread of antimicrobial resistance, the 'perfect storm' as it has often been described. While the discovery and development of new antibiotics has continued in the research sphere, the pipeline to clinic has largely been fed by derivatives of existing classes of antibiotics, each prone to pre-existing resistance mechanisms. A novel approach to infection management has come from the ecological perspective whereby microbial networks and evolved communities already possess small molecular capabilities for pathogen control. The spatiotemporal nature of microbial interactions is such that mutualism and parasitism are often two ends of the same stick. Small molecule efflux inhibitors can directly target antibiotic efflux, a primary resistance mechanism adopted by many species of bacteria and fungi. However, a much broader anti-infective capability resides within the action of these inhibitors, borne from the role of efflux in key physiological and virulence processes, including biofilm formation, toxin efflux, and stress management. Understanding how these behaviors manifest within complex polymicrobial communities is key to unlocking the full potential of the advanced repertoires of efflux inhibitors.
Collapse
Affiliation(s)
| | - Antje Gloe
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland
- Institute for Pharmaceutical Microbiology, University of Bonn, D-53113 Bonn, Germany
| | - Niall O'Leary
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland
| | - F Jerry Reen
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland
- Synthesis and Solid-State Pharmaceutical Centre, University College Cork, T12 K8AF Cork, Ireland
| |
Collapse
|
28
|
Guo T, Chen Y, Chen W, Semple SJ, Gu X, Polyak SW, Sun G, Venter H, Ma S. Design and synthesis of benzochromene derivatives as AcrB inhibitors for the reversal of bacterial multidrug resistance. Eur J Med Chem 2023; 249:115148. [PMID: 36709649 DOI: 10.1016/j.ejmech.2023.115148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/17/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
A series of novel benzo[h]chromene compounds were designed, synthesized and evaluated for their biological activity as AcrB inhibitors. The compounds were assessed for their ability to potentiate the effect of antibiotics. Compounds with antibiotic-potentiating effects were then evaluated for inhibition of Nile Red efflux, and for off-target effects including activity on the outer and inner bacterial membranes and toxicity. Six compounds were identified to reduce the MIC values of at least one of the tested antibiotics by at least 4-fold, and further reduced the MICs in the presence of a membrane permeabilizer. The identified compounds were also able to inhibit Nile Red efflux at concentrations between 50 μM and 200 μM. The compounds did not disrupt the bacterial outer membrane nor display toxicity in a nematode model (Caenorhabditis elegans). The 4-methoxyphenoxy)propoxy derivative compound G6 possessed the most potent antibacterial potentiation with erythromycin by 8-fold even without the presence of a membrane permeabilizer. Furthermore, H6, G6, G10 and G11 completely abolished the Nile Red efflux at a concentration of 50 μM. The 3,4-dihydro-2H-benzo[h]chromen-5-yl)(morpholino)methanone core appears to be a promising chemical skeleton to be further studied in the discovery of more putative AcrB inhibitors.
Collapse
Affiliation(s)
- Ting Guo
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Yang Chen
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Weijin Chen
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Susan J Semple
- Quality Use of Medicines and Pharmacy Research Centre, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Xiaotong Gu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Steven W Polyak
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Guanglin Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Henrietta Venter
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia.
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China.
| |
Collapse
|
29
|
Athar M, Gervasoni S, Catte A, Basciu A, Malloci G, Ruggerone P, Vargiu AV. Tripartite efflux pumps of the RND superfamily: what did we learn from computational studies? MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 36972322 DOI: 10.1099/mic.0.001307] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Bacterial resistance to antibiotics has been long recognized as a priority to address for human health. Among all micro-organisms, the so-called multi-drug resistant (MDR) bacteria, which are resistant to most, if not all drugs in our current arsenal, are particularly worrisome. The World Health Organization has prioritized the ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter species) pathogens, which include four Gram-negative bacterial species. In these bacteria, active extrusion of antimicrobial compounds out of the cell by means of 'molecular guns' known as efflux pumps is a main determinant of MDR phenotypes. The resistance-nodulation-cell division (RND) superfamily of efflux pumps connecting the inner and outer membrane in Gram-negative bacteria is crucial to the onset of MDR and virulence, as well as biofilm formation. Thus, understanding the molecular basis of the interaction of antibiotics and inhibitors with these pumps is key to the design of more effective therapeutics. With the aim to contribute to this challenge, and complement and inspire experimental research, in silico studies on RND efflux pumps have flourished in recent decades. Here, we review a selection of such investigations addressing the main determinants behind the polyspecificity of these pumps, the mechanisms of substrate recognition, transport and inhibition, as well as the relevance of their assembly for proper functioning, and the role of protein-lipid interactions. The journey will end with a perspective on the role of computer simulations in addressing the challenges posed by these beautifully complex machineries and in supporting the fight against the spread of MDR bacteria.
Collapse
Affiliation(s)
- Mohd Athar
- Physics Department, University of Cagliari, Cittadella Universitaria, SP 8 km 0.700, 09042, Monserrato (CA), Italy
| | - Silvia Gervasoni
- Physics Department, University of Cagliari, Cittadella Universitaria, SP 8 km 0.700, 09042, Monserrato (CA), Italy
| | - Andrea Catte
- Physics Department, University of Cagliari, Cittadella Universitaria, SP 8 km 0.700, 09042, Monserrato (CA), Italy
| | - Andrea Basciu
- Physics Department, University of Cagliari, Cittadella Universitaria, SP 8 km 0.700, 09042, Monserrato (CA), Italy
| | - Giuliano Malloci
- Physics Department, University of Cagliari, Cittadella Universitaria, SP 8 km 0.700, 09042, Monserrato (CA), Italy
| | - Paolo Ruggerone
- Physics Department, University of Cagliari, Cittadella Universitaria, SP 8 km 0.700, 09042, Monserrato (CA), Italy
| | - Attilio Vittorio Vargiu
- Physics Department, University of Cagliari, Cittadella Universitaria, SP 8 km 0.700, 09042, Monserrato (CA), Italy
| |
Collapse
|
30
|
Phan TV, Nguyen CHH, Nguyen VTV. 3D-Pharmacophore and Molecular Docking Studies for AcrAB-TolC Efflux Pump Potential Inhibitors from DrugBank and Traditional Chinese Medical Database. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.11081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Due to the widespread resistance to several antibiotics, the AcrAB-TolC tripartite efflux pump is the primary multi-drug efflux system of Escherichia coli. One of the most promising treatments since the discovery of efflux pump inhibitors is the combination of them with antibiotics.
AIM: Based on the efflux pump inhibitor database and the structure of AcrB, the research was created the virtual screening models with prediction capabilities for the efflux pump inhibitory effects of candidates from the DrugBank and Traditional Chinese Medical databank.
Methods: The pharmacophore models were developed by MOE 2015.10 software using a database of 119 efflux pump inhibitors discovered in 12 research publications and belonged to different structural classes. The binding site was found on the AcrB protein (PDB: 4DX7) by LeadIT 2.0.2 software that corresponds to the hydrophobic trap in the proximal pocket.
Results: The potential inhibitors which satisfied the pharmacophore model and had docking scores under -20 kJ.mol-1 have been established. In which, TCM_20290, DB00303, DB04642, DB08116, TCM_29530, and 2,5-dimethyl-3-O-D-glucopyranosyl-naphthol have the best docking scores of -32.76, -26.59, -26.14, -25.62, -24.88, and -22.82 kJ.mol-1, respectively.
Conclusions: After the screening, the result was obtained six compounds may be potential efflux pump inhibitors that can be used for additional studies. In the future, further in vitro and in vivo research should be required to confirm the effects of these compounds. The ongoing battle against antibiotic resistance shows promise with the finding on initiators that can obstruct AcrAB–TolC multidrug efflux pumps.
Keywords: AcrAB-TolC, inhibitors, Escherichia coli, pharmacophore, molecular docking.
Collapse
|
31
|
Influence of the Hydrophobicity of Pluronic Micelles Encapsulating Curcumin on the Membrane Permeability and Enhancement of Photoinduced Antibacterial Activity. Pharmaceutics 2022; 14:pharmaceutics14102137. [PMID: 36297572 PMCID: PMC9608470 DOI: 10.3390/pharmaceutics14102137] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Apart from its well-known activity as an antimicrobial agent, Curcumin (CURC) has recently started to arouse interest as a photosensitizer in the photodynamic therapy of bacterial infections. The aim of the present study was to evidence the influence of the encapsulation of Curcumin into polymeric micelles on the efficiency of photoinduced microbial inhibition. The influence of the hydrophobicity of the selected Pluronics (P84, P123, and F127) on the encapsulation, stability, and antimicrobial efficiency of CURC-loaded micelles was investigated. In addition, the size, morphology, and drug-loading capacity of the micellar drug delivery systems have been characterized. The influence of the presence of micellar aggregates and unassociated molecules of various Pluronics on the membrane permeability was investigated on both normal and resistant microbial strains of E. coli, S. aureus, and C. albicans. The antimicrobial efficiency on the common pathogens was assessed for CURC-loaded polymeric micelles in dark conditions and activated by blue laser light (470 nm). Significant results in the reduction of the microorganism’s growth were found in cultures of C. albicans, even at very low concentrations of surfactants and Curcumin. Unlike the membrane permeabilization effect of the monomeric solution of Pluronics, reported in the case of tumoral cells, a limited permeabilization effect was found on the studied microorganisms. Encapsulation of the Curcumin in Pluronic P84 and P123 at very low, nontoxic concentrations for photosensitizer and drug-carrier, produced CURC-loaded micelles that prove to be effective in the light-activated inhibition of resistant species of Gram-positive bacteria and fungi.
Collapse
|
32
|
do Socorro Costa M, da Silva ARP, Araújo NJS, Filho JMB, Tavares JF, de Freitas TS, Pereira Junior FN, de Sousa EO, Maia FPA, de Vasconcelos JEL, Pinheiro JCA, Coutinho HDM. Evaluation of the antibacterial and inhibitory activity of NorA and MepA efflux pumps from Staphylococcus aureus by diosgenin. Life Sci 2022; 308:120978. [PMID: 36122765 DOI: 10.1016/j.lfs.2022.120978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 10/31/2022]
Abstract
The increase in bacterial resistance to available antibiotics has driven several researchers to search for new agents with therapeutic properties. Diosgenin is a naturally occurring steroidal saponin that has demonstrated several pharmacological properties. In the present study, we report the antimicrobial activity of diosgenin against the standard and multidrug-resistant bacteria of Escherichia coli, Pseudomonas aeruginosa and Staphylococcus aureus, in addition to the efflux pump inhibitory activity against Staphylococcus aureus strains carrying NorA and MepA pumps. For this purpose, the broth microdilution method was used, from which the value of the Minimum Inhibitory Concentration (MIC) was obtained, and this was associated with subinhibitory concentration (MIC/8) with antibiotic of clinical use and ethidium bromide for strains carrier by efflux pump. Diosgenin showed antimicrobial activity for standard S. aureus bacteria and potentiating activity in association with gentamicin and ampicillin for P. aeruginosa multidrug-resistant bacteria, it also showed potentiation in association with norfloxacin against the E. coli strain and gentamicin against the S. aureus strain. Antimicrobial activity against efflux pump-bearing strains revealed that saponin did not interfere with the efflux pump mechanism or intervened antagonistically. Thus, saponin has shown to be very promising against bacterial resistance in association with aminoglycoside, fluoroquinolones and beta-lactam, however additional studies should be carried out to better elucidate the mechanism of action of diosgenin.
Collapse
Affiliation(s)
- Maria do Socorro Costa
- Graduate Program in Biotechnology, State University of Ceará, Fortaleza, Ceará, Brazil; Laboratory of Microbiology and Molecular Biology, Regional University of Cariri - LMBM, Crato, Ceará, Brazil
| | - Ana Raquel Pereira da Silva
- Graduate Program in Biotechnology, State University of Ceará, Fortaleza, Ceará, Brazil; Laboratory of Microbiology and Molecular Biology, Regional University of Cariri - LMBM, Crato, Ceará, Brazil
| | | | - José Maria Barbosa Filho
- Laboratory Technology Pharmaceutical, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Josean Fechine Tavares
- Laboratory Technology Pharmaceutical, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Thiago Sampaio de Freitas
- Laboratory of Microbiology and Molecular Biology, Regional University of Cariri - LMBM, Crato, Ceará, Brazil
| | | | | | | | | | - Jacqueline Cosmo Andrade Pinheiro
- Laboratory of Microbiology and Molecular Biology, Regional University of Cariri - LMBM, Crato, Ceará, Brazil; Graduate Program in Health Sciences, Federal University of Cariri, Barbalha, Ceará, Brazil
| | | |
Collapse
|
33
|
de Oliveira Silva JV, Meneguello JE, Formagio MD, de Freitas CF, Hioka N, Pilau EJ, Marchiosi R, Machinski Junior M, de Abreu Filho BA, Zanetti Campanerut-Sá PA, Graton Mikcha JM. Proteomic Investigation over the Antimicrobial Photodynamic Therapy Mediated by Rose Bengal Against Staphylococcus aureus. Photochem Photobiol 2022; 99:957-966. [PMID: 36054748 DOI: 10.1111/php.13707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/28/2022] [Indexed: 11/28/2022]
Abstract
In order, to understand the antimicrobial action of photodynamic therapy and how this technique can contribute to its application in the control of pathogens. The objective of the study was to employ a proteomic approach to investigate the protein profile of S. aureus after antimicrobial photodynamic therapy mediated by rose bengal (RB-aPDT). S. aureus was treated with RB (10 nmol/l) and illuminated with green LED (0.17 J/cm2 ) for cell viability evaluation. Afterward, proteomic analysis was employed for protein identification and bioinformatic tools to classify the differentially expressed proteins. The reduction of S. aureus after photoinactivation was ~2.5 log CFU/ml. A total of 12 proteins (four up-regulated and eight down-regulated), correspond exclusively to alteration by RB-aPDT. Functionally these proteins are distributed in protein binding, structural constituent of ribosome, proton transmembrane transporter activity, and ATPase activity. The effects of photodamage include alterations of levels of several proteins resulting in an activated stress response, altered membrane potential, and effects on energy metabolism. These 12 proteins required the presence of both light and RB suggesting a unique response to photodynamic effects. The information about this technique contributes valuable insights into bacterial mechanisms and the mode of action of photodynamic therapy.
Collapse
Affiliation(s)
| | - Jean Eduardo Meneguello
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Paraná, Brazil
| | - Maíra Dante Formagio
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Paraná, Brazil
| | - Camila Fabiano de Freitas
- Department of Chemistry, State University of Maringá, Paraná, Brazil.,Departament of Chemistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Noboru Hioka
- Department of Chemistry, State University of Maringá, Paraná, Brazil
| | | | - Rogério Marchiosi
- Department of Biochemistry, State University of Maringá, Paraná, Brazil
| | | | | | | | | |
Collapse
|
34
|
Yang JL, Sun H, Zhou X, Yang M, Zhan XY. Antimicrobial susceptibility profiles and tentative epidemiological cutoff values of Legionella pneumophila from environmental water and soil sources in China. Front Microbiol 2022; 13:924709. [PMID: 36312931 PMCID: PMC9597688 DOI: 10.3389/fmicb.2022.924709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/28/2022] [Indexed: 11/25/2022] Open
Abstract
Legionnaires’ disease (LD), caused by Legionella, including the most prevalent Legionella pneumophila, has been treated primarily with antibiotics. Environmental water and soil are the reservoirs for L. pneumophila. Studying antimicrobial susceptibility using a large number of isolates from various environmental sources and regions could provide an unbiased result. In the present study, antimicrobial susceptibility of 1464 environmental L. pneumophila isolates that were derived from various environmental water and soil sources of 12 cities in China to rifampin (RIF), erythromycin (ERY), clarithromycin (CLA), azithromycin (AZI), ciprofloxacin (CIP), moxifloxacin (MOX), levofloxacin (LEV), and doxycycline (DOX) was investigated, and minimum inhibitory concentration (MIC) data were obtained. We show that regarding macrolides, ERY was least active (MIC90 = 0.5 mg/L), while CLA was most active (MIC90 = 0.063 mg/L). A total of three fluoroquinolones have similar MICs on L. pneumophila. Among these antimicrobials, RIF was the most active agent, while DOX was the most inactive one. We observed different susceptibility profiles between serogroup 1 (sg1) and sg2-15 or between water and soil isolates from different regions. The ECOFFs were ERY and AZI (0.5 mg/L), RIF (0.002 mg/L), CIP, CLA and MOX (0.125 mg/L), LEV (0.063 mg/), and DOX (32 mg/L). Overall, two fluoroquinolone-resistant environmental isolates (0.14%) were first documented based on the wild-type MIC distribution. Not all azithromycin-resistant isolates (44/46, 95.65%) harbored the lpeAB efflux pump. The MICs of the ERY and CLA on the lpeAB + isolates were not elevated. These results suggested that the lpeAB efflux pump might be only responsible for AZI resistance, and undiscovered AZI-specific resistant mechanisms exist in L. pneumophila. Based on the big MIC data obtained in the present study, the same defense strategies, particularly against both CLA and RIF, may exist in L. pneumophila. The results determined in our study will guide further research on antimicrobial resistance mechanisms of L. pneumophila and could be used as a reference for setting clinical breakpoints and discovering antimicrobial-resistant isolates in the clinic, contributing to the antibiotic choice in the treatment of LD.
Collapse
|
35
|
New C-6 functionalized quinoline NorA inhibitors strongly synergize with ciprofloxacin against planktonic and biofilm growing resistant Staphylococcus aureus strains. Eur J Med Chem 2022; 241:114656. [DOI: 10.1016/j.ejmech.2022.114656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022]
|
36
|
Sionov RV, Steinberg D. Targeting the Holy Triangle of Quorum Sensing, Biofilm Formation, and Antibiotic Resistance in Pathogenic Bacteria. Microorganisms 2022; 10:1239. [PMID: 35744757 PMCID: PMC9228545 DOI: 10.3390/microorganisms10061239] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic and recurrent bacterial infections are frequently associated with the formation of biofilms on biotic or abiotic materials that are composed of mono- or multi-species cultures of bacteria/fungi embedded in an extracellular matrix produced by the microorganisms. Biofilm formation is, among others, regulated by quorum sensing (QS) which is an interbacterial communication system usually composed of two-component systems (TCSs) of secreted autoinducer compounds that activate signal transduction pathways through interaction with their respective receptors. Embedded in the biofilms, the bacteria are protected from environmental stress stimuli, and they often show reduced responses to antibiotics, making it difficult to eradicate the bacterial infection. Besides reduced penetration of antibiotics through the intricate structure of the biofilms, the sessile biofilm-embedded bacteria show reduced metabolic activity making them intrinsically less sensitive to antibiotics. Moreover, they frequently express elevated levels of efflux pumps that extrude antibiotics, thereby reducing their intracellular levels. Some efflux pumps are involved in the secretion of QS compounds and biofilm-related materials, besides being important for removing toxic substances from the bacteria. Some efflux pump inhibitors (EPIs) have been shown to both prevent biofilm formation and sensitize the bacteria to antibiotics, suggesting a relationship between these processes. Additionally, QS inhibitors or quenchers may affect antibiotic susceptibility. Thus, targeting elements that regulate QS and biofilm formation might be a promising approach to combat antibiotic-resistant biofilm-related bacterial infections.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Biofilm Research Laboratory, The Institute of Biomedical and Oral Research, The Faculty of Dental Medicine, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel;
| | | |
Collapse
|
37
|
Dashtbani-Roozbehani A, Brown MH. Efflux Pump Mediated Antimicrobial Resistance by Staphylococci in Health-Related Environments: Challenges and the Quest for Inhibition. Antibiotics (Basel) 2021; 10:antibiotics10121502. [PMID: 34943714 PMCID: PMC8698293 DOI: 10.3390/antibiotics10121502] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 01/04/2023] Open
Abstract
The increasing emergence of antimicrobial resistance in staphylococcal bacteria is a major health threat worldwide due to significant morbidity and mortality resulting from their associated hospital- or community-acquired infections. Dramatic decrease in the discovery of new antibiotics from the pharmaceutical industry coupled with increased use of sanitisers and disinfectants due to the ongoing COVID-19 pandemic can further aggravate the problem of antimicrobial resistance. Staphylococci utilise multiple mechanisms to circumvent the effects of antimicrobials. One of these resistance mechanisms is the export of antimicrobial agents through the activity of membrane-embedded multidrug efflux pump proteins. The use of efflux pump inhibitors in combination with currently approved antimicrobials is a promising strategy to potentiate their clinical efficacy against resistant strains of staphylococci, and simultaneously reduce the selection of resistant mutants. This review presents an overview of the current knowledge of staphylococcal efflux pumps, discusses their clinical impact, and summarises compounds found in the last decade from plant and synthetic origin that have the potential to be used as adjuvants to antibiotic therapy against multidrug resistant staphylococci. Critically, future high-resolution structures of staphylococcal efflux pumps could aid in design and development of safer, more target-specific and highly potent efflux pump inhibitors to progress into clinical use.
Collapse
|
38
|
Clinical Status of Efflux Resistance Mechanisms in Gram-Negative Bacteria. Antibiotics (Basel) 2021; 10:antibiotics10091117. [PMID: 34572699 PMCID: PMC8467137 DOI: 10.3390/antibiotics10091117] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/05/2021] [Accepted: 09/15/2021] [Indexed: 01/25/2023] Open
Abstract
Antibiotic efflux is a mechanism that is well-documented in the phenotype of multidrug resistance in bacteria. Efflux is considered as an early facilitating mechanism in the bacterial adaptation face to the concentration of antibiotics at the infectious site, which is involved in the acquirement of complementary efficient mechanisms, such as enzymatic resistance or target mutation. Various efflux pumps have been described in the Gram-negative bacteria most often encountered in infectious diseases and, in healthcare-associated infections. Some are more often involved than others and expel virtually all families of antibiotics and antibacterials. Numerous studies report the contribution of these pumps in resistant strains previously identified from their phenotypes. The authors characterize the pumps involved, the facilitating antibiotics and those mainly concerned by the efflux. However, today no study describes a process for the real-time quantification of efflux in resistant clinical strains. It is currently necessary to have at hospital level a reliable and easy method to quantify the efflux in routine and contribute to a rational choice of antibiotics. This review provides a recent overview of the prevalence of the main efflux pumps observed in clinical practice and provides an idea of the prevalence of this mechanism in the multidrug resistant Gram-negative bacteria. The development of a routine diagnostic tool is now an emergency need for the proper application of current recommendations regarding a rational use of antibiotics.
Collapse
|
39
|
Yi M, Wang H, Wang M, Cao J, Gao F, Ke X, Liu Z, Liu Y, Lu M. Efficient Inhibition of Streptococcus agalactiae by AIEgen-Based Fluorescent Nanomaterials. Front Chem 2021; 9:715565. [PMID: 34354981 PMCID: PMC8329347 DOI: 10.3389/fchem.2021.715565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/22/2021] [Indexed: 12/03/2022] Open
Abstract
Streptococcus agalactiae, referred to as group B streptococcus (GBS), is a prominent co-pathogenic bacterium causing the onset and death of human, animal, and aquatic products. Although antibiotics are efficient against GBS, antibiotic resistance through antibiotic overuse is an equally serious problem. Therefore, the treatment of GBS infection appears strongly dependent on nonantibiotic therapy, such as photodynamic therapy. Different from other photosensitizers (PSs), luminogens with aggregation-induced emission (AIEgen) can efficiently generate fluorescence and reactive oxygen species (ROS). Herein, TBP-1, an efficient AIE PSs, is chosen to resist GBS, and its antibacterial activity and the killing mechanism toward GBS are investigated. The ROS generation performance and the images of GBS treated with TBP-1 in the dark or under white light irradiation were investigated. TBP-1 with its high ROS generation ability can efficiently kill GBS and serve as a novel treatment strategy against GBS infection.
Collapse
Affiliation(s)
- Mengmeng Yi
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Aquatic Animal Immune Technology, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou, China
| | - He Wang
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Aquatic Animal Immune Technology, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou, China
| | - Miao Wang
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Aquatic Animal Immune Technology, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou, China
| | - Jianmeng Cao
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Aquatic Animal Immune Technology, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou, China
| | - Fengying Gao
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Aquatic Animal Immune Technology, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou, China
| | - Xiaoli Ke
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Aquatic Animal Immune Technology, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou, China
| | - Zhigang Liu
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Aquatic Animal Immune Technology, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou, China
| | - Ying Liu
- Key Laboratory of Environment Controlled Aquaculture, Ministry of Education, Dalian Ocean University, Dalian, China
| | - Maixin Lu
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Aquatic Animal Immune Technology, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou, China
| |
Collapse
|