1
|
Rowaiye A, Ibeanu GC, Bur D, Nnadi S, Morikwe U, Ogugua AJ, Chukwudi CU. Phyto-molecules show potentials to combat drug-resistance in bacterial cell membranes. Microb Pathog 2025:107723. [PMID: 40398642 DOI: 10.1016/j.micpath.2025.107723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/17/2024] [Accepted: 05/19/2025] [Indexed: 05/23/2025]
Abstract
The global rise in antibiotic resistance and the emergence of infectious diseases have intensified the need for novel antimicrobial therapies. As a result, there is a growing demand to validate the ethnomedicinal claims that plant extracts possess antibacterial properties. This validation requires the characterization of specific phytoconstituents, including anti-infective compounds and antimicrobial peptides. This study explores the progress made in identifying and producing anti-infectives derived from plants, with a focus on their mechanisms of action, current applications, and future potentials. One key area of investigation is the therapeutic potential of phyto-molecules, that target bacterial cell membranes. These molecules which include phenols, alkaloids, terpenoids, saponins, and peptides, have shown significant ability to disrupt bacterial cell membranes through various molecular mechanisms. By impairing membrane integrity, inhibiting efflux pumps, and altering membrane permeability, phyto-molecules offer a novel strategy for combating drug-resistant bacterial strains. This disruption not only enhances the efficacy of conventional antibiotics but also provides standalone antimicrobial activity. In conclusion, phyto-molecules represent a promising solution to overcoming antibiotic resistance, with their ability to target structural and functional components of bacterial membranes offering new pathways for therapeutic development. However, further research is needed to assess the comparative effectiveness and safety of these plant-based molecules in relation to traditional membrane-disrupting antibiotics.
Collapse
Affiliation(s)
- Adekunle Rowaiye
- Department of Agricultural Biotechnology, National Biotechnology Development Agency, Abuja, Nigeria; Department of Pharmaceutical Science, North Carolina Central University, Durham, North Carolina, USA
| | - Gordon C Ibeanu
- Department of Pharmaceutical Science, North Carolina Central University, Durham, North Carolina, USA
| | - Doofan Bur
- Department of Medical Biotechnology, National Biotechnology Development Agency, Abuja, Nigeria
| | - Sandra Nnadi
- Plant Biology Department, University of Vermont, Burlington, Vermont, USA
| | - Ugonna Morikwe
- Department of Biology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; Department of Pharmaceutical Microbiology Biotechnology, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Akwoba Joseph Ogugua
- Department of Veterinary Public Health and Preventive Medicine, University of Nigeria, Nsukka, Enugu State, Nigeria; Vaccine Research Centre, University of Nigeria, 80 Coal City Garden Estate, Enugu, 400102, Nigeria.
| | - Chinwe Uzoma Chukwudi
- Vaccine Research Centre, University of Nigeria, 80 Coal City Garden Estate, Enugu, 400102, Nigeria; Department of Veterinary Pathology, University of Nigeria, Nsukka, Enugu State, Nigeria
| |
Collapse
|
2
|
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
|
3
|
da Cunha IV, da Silva Oliveira DD, Calefi GG, Silva NBS, Martins CHG, Rezende Júnior CDO, Tsubone TM. Photosensitizer associated with efflux pump inhibitors as a strategy for photodynamic therapy against bacterial resistance. Eur J Med Chem 2025; 284:117197. [PMID: 39731789 DOI: 10.1016/j.ejmech.2024.117197] [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: 10/04/2024] [Revised: 12/01/2024] [Accepted: 12/08/2024] [Indexed: 12/30/2024]
Abstract
Antimicrobial resistance is currently one of the biggest challenges in controlling infectious diseases and was listed among the top 10 threats to global health by the World Health Organization (WHO) in 2023. The antibiotics misuse has led to the widespread emergence of antimicrobial resistance, marking the beginning of the alarming increase in antibiotic resistance. In this context, Antimicrobial Photodynamic Therapy (aPDT) has garnered significant attention from the scientific community due to its potential to effectively eliminate multidrug-resistant pathogenic bacteria and its low propensity to induce drug resistance, which bacteria can quickly develop against traditional antibiotic treatments. However, some efflux pumps can expel diverse substrates from inside the cell, including photosensitizers used in aPDT, contributing to multidrug-resistance mechanisms. Efflux Pump Inhibitors are potential solutions to combat resistance mediated by these pumps and can play a crucial role in enhancing aPDT's effectiveness against multidrug-resistant bacteria. Therefore, combining efflux pumps inhibitors with photosensitizers can possible to eliminate the pathogen more efficiently. This review summarizes the mechanisms in which bacteria resist conventional antibiotic treatment, with a particular emphasis on efflux pump-mediated resistance, and present aPDT as a promising strategy to combat antibiotic resistance. Additionally, we highlighted several molecules of photosensitizer associated with efflux pump inhibitors as potential strategies to optimize aPDT, aiming to offer a perspective on future research directions on aPDT for overcoming the limitations of antibiotic resistance.
Collapse
Affiliation(s)
- Ieda Vieira da Cunha
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | - Gabriel Guimarães Calefi
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | | | | | - Tayana Mazin Tsubone
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
4
|
Zorman M, Kokot M, Zdovc I, Senerovic L, Mandic M, Zidar N, Cotman AE, Durcik M, Peterlin Mašič L, Minovski N, Anderluh M, Hrast Rambaher M. Enhancing Antibacterial Efficacy: Combining Novel Bacterial Topoisomerase Inhibitors with Efflux Pump Inhibitors and Other Agents Against Gram-Negative Bacteria. Antibiotics (Basel) 2024; 13:1081. [PMID: 39596774 PMCID: PMC11591509 DOI: 10.3390/antibiotics13111081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The novel bacterial topoisomerase inhibitors (NBTIs) developed in our laboratory show potent on-target enzyme inhibition but suffer from low activity against Gram-negative bacteria. METHODS With the aim of improving the antibacterial activity of our compounds against Gram-negative bacteria, we tested them in combination with different efflux pump inhibitors (EPIs), a strategy that showed promise in several other classes of antimicrobials. We also investigated the combined effect of NBTIs with ATP-competitive inhibitors of bacterial type II topoisomerases (ACIs), as well as the antibiofilm properties of our compounds and the combination with EPIs against early and mature Acietobacter baumannii biofilm. RESULTS Our results demonstrate that combinations of NBTIs with EPI Phenylalanine-arginyl-β-naphthylamide significantly reduce the corresponding NBTIs' minimal inhibitory concentration values and show potentiation of A. baumannii biofilm inhibition as compared to NBTIs alone. Although combinations of NBITs and ACIs did not show synergistic effects, the FIC index value calculations revealed additive effects for all the combinations of a selected NBTI in combination with three ACIs in all the assayed Gram-negative bacteria from the ESKAPE group. CONCLUSIONS These results show for the first time that combinations of NBTIs with either EPIs or a different class of the topoisomerase inhibitors may be a beneficial strategy to combat difficult-to-treat bacterial infections.
Collapse
Affiliation(s)
- Maša Zorman
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia; (M.Z.); (M.K.); (N.M.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia; (N.Z.); (A.E.C.); (M.D.); (L.P.M.); (M.A.)
| | - Maja Kokot
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia; (M.Z.); (M.K.); (N.M.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia; (N.Z.); (A.E.C.); (M.D.); (L.P.M.); (M.A.)
| | - Irena Zdovc
- Institute of Microbiology and Parasitology, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia;
| | - Lidija Senerovic
- Laboratory for Microbial Molecular Genetics and Ecology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (L.S.); (M.M.)
| | - Mina Mandic
- Laboratory for Microbial Molecular Genetics and Ecology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (L.S.); (M.M.)
| | - Nace Zidar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia; (N.Z.); (A.E.C.); (M.D.); (L.P.M.); (M.A.)
| | - Andrej Emanuel Cotman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia; (N.Z.); (A.E.C.); (M.D.); (L.P.M.); (M.A.)
| | - Martina Durcik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia; (N.Z.); (A.E.C.); (M.D.); (L.P.M.); (M.A.)
| | - Lucija Peterlin Mašič
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia; (N.Z.); (A.E.C.); (M.D.); (L.P.M.); (M.A.)
| | - Nikola Minovski
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia; (M.Z.); (M.K.); (N.M.)
| | - Marko Anderluh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia; (N.Z.); (A.E.C.); (M.D.); (L.P.M.); (M.A.)
| | - Martina Hrast Rambaher
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia; (N.Z.); (A.E.C.); (M.D.); (L.P.M.); (M.A.)
| |
Collapse
|
5
|
Belay WY, Getachew M, Tegegne BA, Teffera ZH, Dagne A, Zeleke TK, Abebe RB, Gedif AA, Fenta A, Yirdaw G, Tilahun A, Aschale Y. Mechanism of antibacterial resistance, strategies and next-generation antimicrobials to contain antimicrobial resistance: a review. Front Pharmacol 2024; 15:1444781. [PMID: 39221153 PMCID: PMC11362070 DOI: 10.3389/fphar.2024.1444781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Antibacterial drug resistance poses a significant challenge to modern healthcare systems, threatening our ability to effectively treat bacterial infections. This review aims to provide a comprehensive overview of the types and mechanisms of antibacterial drug resistance. To achieve this aim, a thorough literature search was conducted to identify key studies and reviews on antibacterial resistance mechanisms, strategies and next-generation antimicrobials to contain antimicrobial resistance. In this review, types of resistance and major mechanisms of antibacterial resistance with examples including target site modifications, decreased influx, increased efflux pumps, and enzymatic inactivation of antibacterials has been discussed. Moreover, biofilm formation, and horizontal gene transfer methods has also been included. Furthermore, measures (interventions) taken to control antimicrobial resistance and next-generation antimicrobials have been discussed in detail. Overall, this review provides valuable insights into the diverse mechanisms employed by bacteria to resist the effects of antibacterial drugs, with the aim of informing future research and guiding antimicrobial stewardship efforts.
Collapse
Affiliation(s)
- Wubetu Yihunie Belay
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Melese Getachew
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu Teffera
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Dagne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Tirsit Ketsela Zeleke
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Rahel Belete Abebe
- Department of clinical pharmacy, College of medicine and health sciences, University of Gondar, Gondar, Ethiopia
| | - Abebaw Abie Gedif
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Fenta
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getasew Yirdaw
- Department of environmental health science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Tilahun
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
6
|
Cui XD, Liu XK, Ma XY, Li SH, Zhang JK, Han RJ, Yi KF, Liu JH, Pan YS, He DD, Hu GZ, Zhai YJ. Restoring colistin sensitivity in colistin-resistant Salmonella and Escherichia coli: combinatorial use of berberine and EDTA with colistin. mSphere 2024; 9:e0018224. [PMID: 38738873 PMCID: PMC11332338 DOI: 10.1128/msphere.00182-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/07/2024] [Indexed: 05/14/2024] Open
Abstract
The appearance and prevalence of multidrug-resistance (MDR) Gram-negative bacteria (GNB) have limited our antibiotic capacity to control bacterial infections. The clinical efficacy of colistin (COL), considered as the "last resort" for treating GNB infections, has been severely hindered by its increased use as well as the emergence and prevalence of mobile colistin resistance (MCR)-mediated acquired drug resistance. Identifying promising compounds to restore antibiotic activity is becoming an effective strategy to alleviate the crisis of increasing MDR. We first demonstrated that the combination of berberine (BBR) and EDTA substantially restored COL sensitivity against COL-resistant Salmonella and Escherichia coli. Molecular docking indicated that BBR can interact with MCR-1 and the efflux pump system AcrAB-TolC, and BBR combined with EDTA downregulated the expression level of mcr-1 and tolC. Mechanically, BBR combined with EDTA could increase bacterial membrane damage, inhibit the function of multidrug efflux pump, and promote oxidative damage, thereby boosting the action of COL. In addition, transcriptome analysis found that the combination of BBR and EDTA can accelerate the tricarboxylic acid cycle, inhibit cationic antimicrobial peptide (CAMP) resistance, and attenuate Salmonella virulence. Notably, the combination of BBR and EDTA with COL significantly reduced the bacterial load in the liver and spleen of a mice model infected with Salmonella. Our findings revealed that BBR and EDTA can be used as adjuvants collectively with COL to synergistically reverse the COL resistance of bacteria. IMPORTANCE Colistin is last-resort antibiotic used to treat serious clinical infections caused by MDR bacterial pathogens. The recent emergence of transferable plasmid-mediated COL resistance gene mcr-1 has raised the specter of a rapid worldwide spread of COL resistance. Coupled with the fact of barren antibiotic development pipeline nowadays, a critical approach is to revitalize existing antibiotics using antibiotic adjuvants. Our research showed that berberine combined with EDTA effectively reversed COL resistance both in vivo and in vitro through multiple modes of action. The discovery of berberine in combination with EDTA as a new and safe COL adjuvant provides a therapeutic regimen for combating Gram-negative bacteria infections. Our findings provide a potential therapeutic option using existing antibiotics in combination with antibiotic adjuvants and address the prevalent infections caused by MDR Gram-negative pathogens worldwide.
Collapse
Affiliation(s)
- Xiao-die Cui
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiao-kang Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiao-yuan Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Shuai-hua Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Jun-kai Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Rong-jia Han
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Kai-fang Yi
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Jian-hua Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yu-shan Pan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Dan-dan He
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Gong-zheng Hu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ya-jun Zhai
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
7
|
Lopes AA, Vendrell-Fernández S, Deschamps J, Georgeault S, Cokelaer T, Briandet R, Ghigo JM. Bile-induced biofilm formation in Bacteroides thetaiotaomicron requires magnesium efflux by an RND pump. mBio 2024; 15:e0348823. [PMID: 38534200 PMCID: PMC11078008 DOI: 10.1128/mbio.03488-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Bacteroides thetaiotaomicron is a prominent member of the human gut microbiota contributing to nutrient exchange, gut function, and maturation of the host's immune system. This obligate anaerobe symbiont can adopt a biofilm lifestyle, and it was recently shown that B. thetaiotaomicron biofilm formation is promoted by the presence of bile. This process also requires a B. thetaiotaomicron extracellular DNase, which is not, however, regulated by bile. Here, we showed that bile induces the expression of several Resistance-Nodulation-Division (RND) efflux pumps and that inhibiting their activity with a global competitive efflux inhibitor impaired bile-dependent biofilm formation. We then showed that, among the bile-induced RND-efflux pumps, only the tripartite BT3337-BT3338-BT3339 pump, re-named BipABC [for Bile Induced Pump A (BT3337), B (BT3338), and C (BT3339)], is required for biofilm formation. We demonstrated that BipABC is involved in the efflux of magnesium to the biofilm extracellular matrix, which leads to a decrease of extracellular DNA concentration. The release of magnesium in the biofilm matrix also impacts biofilm structure, potentially by modifying the electrostatic repulsion forces within the matrix, reducing interbacterial distance and allowing bacteria to interact more closely and form denser biofilms. Our study therefore, identified a new molecular determinant of B. thetaiotaomicron biofilm formation in response to bile salts and provides a better understanding on how an intestinal chemical cue regulates biofilm formation in a major gut symbiont.IMPORTANCEBacteroides thetaiotaomicron is a prominent member of the human gut microbiota able to degrade dietary and host polysaccharides, altogether contributing to nutrient exchange, gut function, and maturation of the host's immune system. This obligate anaerobe symbiont can adopt a biofilm community lifestyle, providing protection against environmental factors that might, in turn, protect the host from dysbiosis and dysbiosis-related diseases. It was recently shown that B. thetaiotaomicron exposure to intestinal bile promotes biofilm formation. Here, we reveal that a specific B. thetaiotaomicron membrane efflux pump is induced in response to bile, leading to the release of magnesium ions, potentially reducing electrostatic repulsion forces between components of the biofilm matrix. This leads to a reduction of interbacterial distance and strengthens the biofilm structure. Our study, therefore, provides a better understanding of how bile promotes biofilm formation in a major gut symbiont, potentially promoting microbiota resilience to stress and dysbiosis events.
Collapse
Affiliation(s)
- Anne-Aurélie Lopes
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Department of Microbiology, Paris, France
- Pediatric Emergency, AP-HP, Necker-Enfants-Malades University Hospital, Paris, France
| | - Sol Vendrell-Fernández
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Department of Microbiology, Paris, France
| | - Julien Deschamps
- INRAE, AgroParisTech, Université Paris-Saclay Institut Micalis, Paris, France
| | - Sonia Georgeault
- Plateforme IBiSA des Microscopies, Université et CHRU de Tours, Tours, France
| | - Thomas Cokelaer
- Institut Pasteur, Université Paris Cité, Plate-forme Technologique Biomics, Center for Technological Resources and Research, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Center for Technological Resources and Research, Paris, France
| | - Romain Briandet
- INRAE, AgroParisTech, Université Paris-Saclay Institut Micalis, Paris, France
| | - Jean-Marc Ghigo
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Department of Microbiology, Paris, France
| |
Collapse
|
8
|
A Malik A, Dangroo NA, Kaur P, Attery S, A Rather M, Khan A, Ara T, Nandanwar H. Discovery of novel dihydronaphthalene-imidazole ligands as potential inhibitors of Staphylococcus aureus multidrug resistant NorA efflux pump: A combination of experimental and in silico molecular docking studies. Microb Pathog 2024; 190:106627. [PMID: 38521473 DOI: 10.1016/j.micpath.2024.106627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/22/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024]
Abstract
Overexpression of the efflux pump is a predominant mechanism by which bacteria show antimicrobial resistance (AMR) and leads to the global emergence of multidrug resistance (MDR). In this work, the inhibitory potential of library of dihydronapthyl scaffold-based imidazole derivatives having structural resemblances with some known efflux pump inhibitors (EPI) were designed, synthesized and evaluated against efflux pump inhibitor against overexpressing bacterial strains to study the synergistic effect of compounds and antibiotics. Out of 15 compounds, four compounds (Dz-1, Dz-3, Dz-7, and Dz-8) were found to be highly active. DZ-3 modulated the MIC of ciprofloxacin, erythromycin, and tetracycline by 128-fold each against 1199B, XU212 and RN4220 strains of S. aureus respectively. DZ-3 also potentiated tetracycline by 64-fold in E. coli AG100 strain. DZ-7 modulated the MIC of both tetracycline and erythromycin 128-fold each in S. aureus XU212 and S. aureus RN4220 strains. DZ-1 and DZ-8 showed the moderate reduction in MIC of tetracycline in E. coli AG100 only by 16-fold and 8-fold, respectively. DZ-3 was found to be the potential inhibitor of NorA as determined by ethidium bromide efflux inhibition and accumulation studies employing NorA overexpressing strain SA-1199B. DZ-3 displayed EPI activity at non-cytotoxic concentration to human cells and did not possess any antibacterial activity. Furthermore, molecular docking studies of DZ-3 was carried out in order to understand the possible binding sites of DZ-3 with the active site of the protein. These studies indicate that dihydronaphthalene scaffolds could serve as valuable cores for the development of promising EPIs.
Collapse
Affiliation(s)
- Asif A Malik
- Department of Chemistry, National Institute of Technology, Srinagar, J&K, 190006, India
| | - Nisar A Dangroo
- Department of Chemistry, Islamic University of Science and Technology, Awantipora, J &K, 192122, India.
| | - Parminder Kaur
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Shobit Attery
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Manzoor A Rather
- Department of Chemistry, Islamic University of Science and Technology, Awantipora, J &K, 192122, India.
| | - Abrar Khan
- Department of Chemistry, National Institute of Technology, Srinagar, J&K, 190006, India
| | - Tabassum Ara
- Department of Chemistry, National Institute of Technology, Srinagar, J&K, 190006, India.
| | - Hemraj Nandanwar
- Clinical Microbiology & Antimicrobial Research Laboratory, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India.
| |
Collapse
|
9
|
Korczak L, Majewski P, Iwaniuk D, Sacha P, Matulewicz M, Wieczorek P, Majewska P, Wieczorek A, Radziwon P, Tryniszewska E. Molecular mechanisms of tigecycline-resistance among Enterobacterales. Front Cell Infect Microbiol 2024; 14:1289396. [PMID: 38655285 PMCID: PMC11035753 DOI: 10.3389/fcimb.2024.1289396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/27/2024] [Indexed: 04/26/2024] Open
Abstract
The global emergence of antimicrobial resistance to multiple antibiotics has recently become a significant concern. Gram-negative bacteria, known for their ability to acquire mobile genetic elements such as plasmids, represent one of the most hazardous microorganisms. This phenomenon poses a serious threat to public health. Notably, the significance of tigecycline, a member of the antibiotic group glycylcyclines and derivative of tetracyclines has increased. Tigecycline is one of the last-resort antimicrobial drugs used to treat complicated infections caused by multidrug-resistant (MDR) bacteria, extensively drug-resistant (XDR) bacteria or even pan-drug-resistant (PDR) bacteria. The primary mechanisms of tigecycline resistance include efflux pumps' overexpression, tet genes and outer membrane porins. Efflux pumps are crucial in conferring multi-drug resistance by expelling antibiotics (such as tigecycline by direct expelling) and decreasing their concentration to sub-toxic levels. This review discusses the problem of tigecycline resistance, and provides important information for understanding the existing molecular mechanisms of tigecycline resistance in Enterobacterales. The emergence and spread of pathogens resistant to last-resort therapeutic options stands as a major global healthcare concern, especially when microorganisms are already resistant to carbapenems and/or colistin.
Collapse
Affiliation(s)
- Lukasz Korczak
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Majewski
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Dominika Iwaniuk
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Pawel Sacha
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | | | - Piotr Wieczorek
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | | | - Anna Wieczorek
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Radziwon
- Regional Centre for Transfusion Medicine, Bialystok, Poland
| | - Elzbieta Tryniszewska
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
10
|
Dwivedi GR, Pathak N, Tiwari N, Negi AS, Kumar A, Pal A, Sharma A, Darokar MP. Synergistic Antibacterial Activity of Gallic Acid Based Chalcone Indl 2 by Inhibiting Efflux Pump Transporters. Chem Biodivers 2024; 21:e202301820. [PMID: 38372508 DOI: 10.1002/cbdv.202301820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 02/20/2024]
Abstract
As a part of novel discovery of drugs from natural resources, present study was undertaken to explore the antibacterial potential of chalcone Indl-2 in combination with different group of antibiotics. MIC of antibiotics was reduced up to eight folds against the different cultures of E. coli by both chalcones. Among the two compounds, the i. e. 1-(3', 4,'5'-trimethoxyphenyl)-3-(3-Indyl)-prop-2-enone (6, Indl-2), a chalcone derivative of gallic acid (Indl-2) was better along with tetracycline (TET) worked synergistically and was found to inhibit efflux transporters as obvious by ethidium bromide efflux confirmed by ATPase assays and docking studies. In combination, Indl-2 kills the MDREC-KG4 cells, post-antibiotic effect (PAE) of TET was prolonged and mutant prevention concentration (MPC) of TET was also decreased. In-vivo studies revealed that Indl-2 reduces the concentration of TNF-α. In acute oral toxicity study, Indl-2 was non-toxic and well tolerated up-to dose of 2000 mg/kg. Perhaps, the study is going to report gallic acid derived chalcone as synergistic agent acting via inhibiting the primary efflux pumps.
Collapse
Affiliation(s)
- Gaurav Raj Dwivedi
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow, 226015, India
- Microbiology Department, ICMR-Regional Medical Research Centre, Gorakhpur, 273013, U.P., India
| | - Nandini Pathak
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow, 226015, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. - 201002, India
| | - Nimisha Tiwari
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow, 226015, India
| | - Arvind Singh Negi
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow, 226015, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. - 201002, India
| | - Akhil Kumar
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow, 226015, India
| | - Anirban Pal
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow, 226015, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. - 201002, India
| | - Ashok Sharma
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow, 226015, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. - 201002, India
| | - Mahendra P Darokar
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow, 226015, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. - 201002, India
| |
Collapse
|
11
|
Zhang L, Tian X, Sun L, Mi K, Wang R, Gong F, Huang L. Bacterial Efflux Pump Inhibitors Reduce Antibiotic Resistance. Pharmaceutics 2024; 16:170. [PMID: 38399231 PMCID: PMC10892612 DOI: 10.3390/pharmaceutics16020170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Bacterial resistance is a growing problem worldwide, and the number of deaths due to drug resistance is increasing every year. We must pay great attention to bacterial resistance. Otherwise, we may go back to the pre-antibiotic era and have no drugs on which to rely. Bacterial resistance is the result of several causes, with efflux mechanisms widely recognised as a significant factor in the development of resistance to a variety of chemotherapeutic and antimicrobial medications. Efflux pump inhibitors, small molecules capable of restoring the effectiveness of existing antibiotics, are considered potential solutions to antibiotic resistance and have been an active area of research in recent years. This article provides a review of the efflux mechanisms of common clinical pathogenic bacteria and their efflux pump inhibitors and describes the effects of efflux pump inhibitors on biofilm formation, bacterial virulence, the formation of bacterial persister cells, the transfer of drug resistance among bacteria, and mismatch repair. Numerous efforts have been made in the past 20 years to find novel efflux pump inhibitors which are known to increase the effectiveness of medicines against multidrug-resistant strains. Therefore, the application of efflux pump inhibitors has excellent potential to address and reduce bacterial resistance.
Collapse
Affiliation(s)
- Lan Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Z.); (X.T.); (L.S.); (K.M.); (R.W.); (F.G.)
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaoyuan Tian
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Z.); (X.T.); (L.S.); (K.M.); (R.W.); (F.G.)
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Lei Sun
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Z.); (X.T.); (L.S.); (K.M.); (R.W.); (F.G.)
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Kun Mi
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Z.); (X.T.); (L.S.); (K.M.); (R.W.); (F.G.)
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Ru Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Z.); (X.T.); (L.S.); (K.M.); (R.W.); (F.G.)
- MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Fengying Gong
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Z.); (X.T.); (L.S.); (K.M.); (R.W.); (F.G.)
- MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Lingli Huang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Z.); (X.T.); (L.S.); (K.M.); (R.W.); (F.G.)
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
- MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
12
|
Farnia P, Besharati S, Farina P, Ayoubi S, Marjani M, Ghanavi J, Tabarsi P, Velayati AA. The Role of Efflux Pumps transporter in Multi-drug Resistant Tuberculosis: Mycobacterial memberane protein(MmpL5). Int J Mycobacteriol 2024; 13:7-14. [PMID: 38771273 DOI: 10.4103/ijmy.ijmy_37_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/04/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND The overexpression of efflux pumps (Eps) was reported to contribute to multidrug resistant tuberculosis (MDR-TB). Increases in Eps that expel structurally unrelated drugs contribute to reduced susceptibility by decreasing the intracellular concentration of antibiotics. In the present study, an association of mycobacterial membrane protein (MmpS5-MmpL5) Ep and its gene regulator (Rv0678) was investigated in MDR-tuberculosis isolates. METHODS MTB strains were isolated from patients at two different intervals, i.e., once when they had persistent symptoms despite 3-15 ≥ months of treatment and once when they had started new combination therapy ≥2-3 months. Sputum specimens were subjected to Xpert MTB/rifampicin test and then further susceptibility testing using proportional method and multiplex polymerase chain reaction (PCR) were performed on them. The isolates were characterized using both 16S-23S RNA and hsp65 genes spacer (PCR-restriction fragment length polymorphism). Whole-genome sequencing (WGS) was investigated on two isolates from culture-positive specimen per patient. The protein structure was simulated using the SWISS-MODEL. The input format used for this web server was FASTA (amino acid sequence). Protein structure was also analysis using Ramachandran plot. RESULTS WGS documented deletion, insertion, and substitution in transmembrane transport protein MmpL5 (Rv0676) of Eps. Majority of the studied isolates (n = 12; 92.3%) showed a unique deletion mutation at three positions: (a) from amino acid number 771 (isoleucine) to 776 (valine), (b) from amino acid number 785 (valine) to 793 (histidine), and (c) from amino acid number 798 (leucine) to 806 (glycine)." One isolate (7.6%) had no deletion mutation. In all isolates (n = 13; 100%), a large insertion mutation consisting of 94 amino acid was observed "from amino acid number 846 (isoleucine) to amino acid number 939 (leucine)". Thirty-eight substitutions in Rv0676 were detected, of which 92.3% were identical in the studied isolates. WGS of mycobacterial membrane proteins (MmpS5; Rv0677) and its gene regulator (Rv0678) documented no deletion, insertion, and substitution. No differences were observed between MmpS5-MmpL5 and its gene regulator in isolates that were collected at different intervals. CONCLUSIONS Significant genetic mutation like insertion, deletion, and substitution within transmembrane transport protein MmpL5 (Rv0676) can change the functional balance of Eps and cause a reduction in drug susceptibility. This is the first report documenting a unique amino acid mutation (insertion and deletion ≥4-94) in Rv0676 among drug-resistant MTB. We suggest the changes in Mmpl5 (Rv0676) might occurred due to in-vivo sub-therapeutic drug stress within the host cell. Changes in MmpL5 are stable and detected through subsequent culture-positive specimens.
Collapse
Affiliation(s)
- Parissa Farnia
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Besharati
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Poopak Farina
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saman Ayoubi
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Marjani
- Clinical Tuberculosis and Epidemiology Research Centre, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalaledin Ghanavi
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Payam Tabarsi
- Clinical Tuberculosis and Epidemiology Research Centre, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Velayati
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Nanjan P, Bose V. Efflux-mediated Multidrug Resistance in Critical Gram-negative Bacteria and Natural Efflux Pump Inhibitors. Curr Drug Res Rev 2024; 16:349-368. [PMID: 38288795 DOI: 10.2174/0125899775271214240112071830] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/01/2023] [Accepted: 12/18/2023] [Indexed: 09/04/2024]
Abstract
Multidrug Resistance mechanisms in microorganisms confer the slackness of the existing drugs, leading to added difficulty in treating infections. As a consequence, efficient novel drugs and innovative therapies to treat MDR infections are necessarily required. One of the primary contributors to the emergence of multidrug resistance in gram-negative bacteria has been identified as the efflux pumps. These transporter efflux pumps reduce the intracellular concentration of antibiotics and aid bacterial survival in suboptimal low antibiotic concentration environments that may cause treatment failure. The reversal of this resistance via inhibition of the efflux mechanism is a promising method for increasing the effectiveness of antibiotics against multidrug-resistant pathogens. Such EPI, in combination with antibiotics, can make it easier to reintroduce traditional antibiotics into clinical practice. This review mostly examines efflux-mediated multidrug resistance in critical gram-negative bacterial pathogens and EPI of plant origin that have been reported over previous decades.
Collapse
Affiliation(s)
- Praveena Nanjan
- Department of Biochemistry, School of Life Science, Jss Academy of Higher Education and Research, Longwood Campus, Mysuru Road, Ooty, India
| | - Vanitha Bose
- Department of Biochemistry, School of Life Science, Jss Academy of Higher Education and Research, Longwood Campus, Mysuru Road, Ooty, India
| |
Collapse
|
14
|
Unnikrishnan VK, Sundaramoorthy NS, Nair VG, Ramaiah KB, Roy JS, Rajendran M, Srinath S, Kumar S, S PS, S SM, Nagarajan S. Genome analysis of triple phages that curtails MDR E. coli with ML based host receptor prediction and its evaluation. Sci Rep 2023; 13:23040. [PMID: 38155176 PMCID: PMC10754912 DOI: 10.1038/s41598-023-49880-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 12/13/2023] [Indexed: 12/30/2023] Open
Abstract
Infections by multidrug resistant bacteria (MDR) are becoming increasingly difficult to treat and alternative approaches like phage therapy, which is unhindered by drug resistance, are urgently needed to tackle MDR bacterial infections. During phage therapy phage cocktails targeting different receptors are likely to be more effective than monophages. In the present study, phages targeting carbapenem resistant clinical isolate of E. coli U1007 was isolated from Ganges River (U1G), Cooum River (CR) and Hospital waste water (M). Capsid architecture discerned using TEM identified the phage families as Podoviridae for U1G, Myoviridae for CR and Siphoviridae for M phage. Genome sequencing showed the phage genomes varied in size U1G (73,275 bp) CR (45,236 bp) and M (45,294 bp). All three genomes lacked genes encoding tRNA sequence, antibiotic resistant or virulent genes. A machine learning (ML) based multi-class classification model using Random Forest, Logistic Regression, and Decision Tree were employed to predict the host receptor targeted by receptor binding protein of all 3 phages and the best performing algorithm Random Forest predicted LPS O antigen, LamB or OmpC for U1G; FhuA, OmpC for CR phage; and FhuA, LamB, TonB or OmpF for the M phage. OmpC was validated as receptor for U1G by physiological experiments. In vivo intramuscular infection study in zebrafish showed that cocktail of dual phages (U1G + M) along with colsitin resulted in a significant 3.5 log decline in cell counts. Our study highlights the potential of ML tool to predict host receptor and proves the utility of phage cocktail to restrict E. coli U1007 in vivo.
Collapse
Affiliation(s)
- Vineetha K Unnikrishnan
- Center for Research On Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
- Antimicrobial Resistance Lab, ASK-I-312, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Niranjana Sri Sundaramoorthy
- Center for Research On Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
- Translational Health Sciences Technology Institute, Faridabad, India
| | - Veena G Nair
- Center for Research On Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
- Antimicrobial Resistance Lab, ASK-I-312, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Kavi Bharathi Ramaiah
- Center for Research On Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
- Antimicrobial Resistance Lab, ASK-I-312, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Jean Sophy Roy
- Center for Research On Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
| | - Malarvizhi Rajendran
- Center for Research On Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
| | - Sneha Srinath
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
| | - Santhosh Kumar
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
| | - Prakash Sankaran S
- Center for Research On Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
| | - Suma Mohan S
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India.
| | - Saisubramanian Nagarajan
- Center for Research On Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India.
- Antimicrobial Resistance Lab, ASK-I-312, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India.
| |
Collapse
|
15
|
Zlotnikov ID, Krylov SS, Semenova MN, Semenov VV, Kudryashova EV. Triphenylphosphine Derivatives of Allylbenzenes Express Antitumor and Adjuvant Activity When Solubilized with Cyclodextrin-Based Formulations. Pharmaceuticals (Basel) 2023; 16:1651. [PMID: 38139778 PMCID: PMC10747112 DOI: 10.3390/ph16121651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Allylbenzenes (apiol, dillapiol, myristicin and allyltetramethoxybenzene) are individual components of plant essential oils that demonstrate antitumor activity and can enhance the antitumor activity of cytotoxic drugs, such as paclitaxel, doxorubicin, cisplatin, etc. Triphenylphosphine (PPh3) derivatives of allylbenzenes are two to three orders of magnitude more potent than original allylbenzenes in terms of IC50. The inhibition of efflux pumps has been reported for allylbenzenes, and the PPh3 moiety is deemed to be responsible for preferential mitochondrial accumulation and the depolarization of mitochondrial membranes. However, due to poor solubility, the practical use of these substances has never been an option. Here, we show that this problem can be solved by using a complex formation with cyclodextrin (CD-based molecular containers) and polyanionic heparin, stabilizing the positive charge of the PPh3 cation. Such containers can solubilize both allylbenzenes and their PPh3 derivatives up to 0.4 mM concentration. Furthermore, we have observed that solubilized PPh3 derivatives indeed work as adjuvants, increasing the antitumor activity of paclitaxel against adenocarcinomic human alveolar basal epithelial cells (A549) by an order of magnitude (in terms of IC50) in addition to being quite powerful cytostatics themselves (IC50 in the range 1-10 µM). Even more importantly, CD-solubilized PPh3 derivatives show pronounced selectivity, being highly toxic for the A549 tumor cell line and minimally toxic for HEK293T non-tumor cells, red blood cells and sea urchin embryos. Indeed, in many cancers, the mitochondrial membrane is more prone to depolarization compared to normal cells, which probably explains the observed selectivity of our compounds, since PPh3 derivatives are known to act as mitochondria-targeting agents. According to the MTT test, 100 µM solution of PPh3 derivatives of allylbenzenes causes the death of up to 85% of A549 cancer cells, while for HEK293T non-cancer cells, only 15-20% of the cells died. The hemolytic index of the studied substances did not exceed 1%, and the thrombogenicity index was < 1.5%. Thus, this study outlines the experimental foundation for developing combined cytostatic medications, where effectiveness and selectivity are achieved through decreased concentration of the primary ingredient and the inclusion of adjuvants, which are safe or practically harmless substances.
Collapse
Affiliation(s)
- Igor D. Zlotnikov
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia;
| | - Sergey S. Krylov
- N. D. Zelinsky Institute of Organic Chemistry RAS, 47 Leninsky Prospect, 119991 Moscow, Russia
| | - Marina N. Semenova
- N. K. Koltzov Institute of Developmental Biology RAS, 26 Vavilov Street, 119334 Moscow, Russia
| | - Victor V. Semenov
- N. D. Zelinsky Institute of Organic Chemistry RAS, 47 Leninsky Prospect, 119991 Moscow, Russia
| | - Elena V. Kudryashova
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia;
| |
Collapse
|
16
|
Martin ALAR, Pereira RLS, Rocha JE, Farias PAM, Freitas TS, de Lemos Caldas FR, Figueredo FG, Sampaio NFL, Ribeiro-Filho J, Menezes IRDA, Brancaglion GA, de Paulo DC, Carvalho DT, Lima MA, Coutinho HDM, Fonteles MMF. In vitro and in silico evidences about the inhibition of MepA efflux pump by coumarin derivatives. Microb Pathog 2023; 182:106246. [PMID: 37454945 DOI: 10.1016/j.micpath.2023.106246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
The discovery of antibiotics has significantly transformed the outcomes of bacterial infections in the last decades. However, the development of antibiotic resistance mechanisms has allowed an increasing number of bacterial strains to overcome the action of antibiotics, decreasing their effectiveness against infections they were developed to treat. This study aimed to evaluate the antibacterial activity of synthetic coumarins Staphylococcus aureus in vitro and analyze their interaction with the MepA efflux pump in silico. The Minimum Inhibitory Concentration (MIC) determination showed that none of the test compounds have antibacterial activity. However, all coumarin derivatives decreased the MIC of the standard efflux inhibitor ethidium bromide, indicating antibacterial synergism. On the other hand, the C14 derivative potentiated the antibacterial activity of ciprofloxacin against the resistant strain. In silico analysis showed that C9, C11, and C13 coumarins showed the most favorable interaction with the MepA efflux pump. Nevertheless, due to the present in silico and in vitro investigation limitations, further experimental research is required to confirm the therapeutic potential of these compounds in vivo.
Collapse
Affiliation(s)
- Ana Luíza A R Martin
- Federal University of Ceará - UFC, Brazil; Faculty of Medicine Estácio Juazeiro do Norte - Estácio Juazeiro do Norte, Brazil
| | | | | | - Pablo A M Farias
- Faculty of Medicine Estácio Juazeiro do Norte - Estácio Juazeiro do Norte, Brazil
| | | | | | - Fernando G Figueredo
- Regional University of Cariri - URCA, Brazil; Faculty of Medicine Estácio Juazeiro do Norte - Estácio Juazeiro do Norte, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Nagy EZ, Kovács ÁB, Wehmann E, Bekő K, Földi D, Bányai K, Kreizinger Z, Gyuranecz M. Phenotypic and genetic insights into efflux pump mechanism in Mycoplasma anserisalpingitidis. Front Microbiol 2023; 14:1216893. [PMID: 37502405 PMCID: PMC10371760 DOI: 10.3389/fmicb.2023.1216893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
Introduction Mycoplasma anserisalpingitidis is one of the most important waterfowl-pathogenic mycoplasmas. Due to inadequate antibiotic treatment, many strains with high minimal inhibitory concentration (MIC) values for multiple drugs have been isolated lately. Decreased antibiotic susceptibility in several Mycoplasma species are known to be associated with mutations in topoisomerase and ribosomal genes, but other strategies such as active efflux pump mechanisms were also described. The scope of this study was the phenotypic and genetic characterization of the active efflux mechanism in M. anserisalpingitidis. Methods We measured the MIC values in the presence and absence of different efflux pump inhibitors (EPIs), such as carbonyl cyanide m-chlorophenylhydrazine (CCCP), orthovanadate (OV), and reserpine (RSP). Moreover, bioinformatic tools were utilized to detect putative regulatory sequences of membrane transport proteins coding genes, while comparative genome analysis was performed to reveal potential markers of antibiotic resistance. Results Out of the three examined EPIs, CCCP decreased the MICs at least two-fold below the original MICs (in 23 cases out of 36 strains). In the presence of OV or RSP, MIC value differences could be seen only if modified dilution series (10% decrease steps were used instead of two-fold dilutions) were applied (in 24/36 cases with OV and 9/36 with RSP). During comparative genome analysis, non-synonymous single nucleotide polymorphisms (nsSNPs) were identified in genes encoding ABC membrane transport proteins, which were displayed in higher percentages in M. anserisalpingitidis strains with increased MICs. In terms of other genes, a nsSNP was identified in DNA gyrase subunit A (gyrA) gene which can be related to decreased susceptibility to enrofloxacin. The present study is the first to highlight the importance of efflux pump mechanisms in M. anserisalpingitidis. Discussion Considering the observed effects of the EPI CCCP against this bacterium, it can be assumed, that the use of EPIs would increase the efficiency of targeted antibiotic therapy in the future control of this pathogen. However, further research is required to obtain a more comprehensive understanding of efflux pump mechanism in this bacterium.
Collapse
Affiliation(s)
- Eszter Zsófia Nagy
- Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
| | - Áron Botond Kovács
- Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
| | - Enikő Wehmann
- Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
| | - Katinka Bekő
- Veterinary Medical Research Institute, Budapest, Hungary
| | - Dorottya Földi
- Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
| | - Krisztián Bányai
- Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
| | - Zsuzsa Kreizinger
- Veterinary Medical Research Institute, Budapest, Hungary
- MolliScience Kft., Biatorbágy, Hungary
| | - Miklós Gyuranecz
- Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
- MolliScience Kft., Biatorbágy, Hungary
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, Budapest, Hungary
| |
Collapse
|
18
|
Hajiagha MN, Kafil HS. Efflux pumps and microbial biofilm formation. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023:105459. [PMID: 37271271 DOI: 10.1016/j.meegid.2023.105459] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/25/2023] [Accepted: 05/27/2023] [Indexed: 06/06/2023]
Abstract
Biofilm-related infections are resistant forms of pathogens that are regarded as a medical problem, particularly due to the spread of multiple drug resistance. One of the factors associated with biofilm drug resistance is the presence of various types of efflux pumps in bacteria. Efflux pumps also play a role in biofilm formation by influencing Physical-chemical interactions, mobility, gene regulation, quorum sensing (QS), extracellular polymeric substances (EPS), and toxic compound extrusion. According to the findings of studies based on efflux pump expression analysis, their role in the anatomical position within the biofilm will differ depending on the biofilm formation stage, encoding gene expression level, the type and concentration of substrate. In some cases, the function of the efflux pumps can overlap with each other, so it seems necessary to accurate identify the efflux pumps of biofilm-forming bacteria along with their function in this process. Such studies will help to choose treatment strategy, at least in combination with antibiotics. Furthermore, if the goal of treatment is an efflux pump manipulation, we should not limit it to inhibition.
Collapse
Affiliation(s)
- Mahdyeh Neghabi Hajiagha
- Department of Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
Gonçalves ASC, Leitão MM, Simões M, Borges A. The action of phytochemicals in biofilm control. Nat Prod Rep 2023; 40:595-627. [PMID: 36537821 DOI: 10.1039/d2np00053a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Covering: 2009 to 2021Antimicrobial resistance is now rising to dangerously high levels in all parts of the world, threatening the treatment of an ever-increasing range of infectious diseases. This has becoming a serious public health problem, especially due to the emergence of multidrug-resistance among clinically important bacterial species and their ability to form biofilms. In addition, current anti-infective therapies have low efficacy in the treatment of biofilm-related infections, leading to recurrence, chronicity, and increased morbidity and mortality. Therefore, it is necessary to search for innovative strategies/antibacterial agents capable of overcoming the limitations of conventional antibiotics. Natural compounds, in particular those obtained from plants, have been exhibiting promising properties in this field. Plant secondary metabolites (phytochemicals) can act as antibiofilm agents through different mechanisms of action from the available antibiotics (inhibition of quorum-sensing, motility, adhesion, and reactive oxygen species production, among others). The combination of different phytochemicals and antibiotics have revealed synergistic or additive effects in biofilm control. This review aims to bring together the most relevant reports on the antibiofilm properties of phytochemicals, as well as insights into their structure and mechanistic action against bacterial pathogens, spanning December 2008 to December 2021.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Miguel M Leitão
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
20
|
Rayane Correia de Oliveira M, Gabriely de Lima Silva M, Datiane de Morais Oliveira-Tintino C, Relison Tintino S, Esmeraldo Rocha J, Ernani Alves Magalhães F, Henrique Sousa da Costa R, Torres Pessoa R, Sousa Alcântara I, Oliveira Brito Pereira Bezerra Martins A, Douglas Melo Coutinho H, Raposo A, Carrascosa C, Raduan Jaber J, Aquino Saraiva R, Rose Alencar de Menezes I. Antibacterial effect, efflux pump inhibitory (NorA, TetK and MepA) of Staphylococcus aureus and in silico prediction of α, β and δ-damascone compounds. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2022.104482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
21
|
Tavares-Carreon F, De Anda-Mora K, Rojas-Barrera IC, Andrade A. Serratia marcescens antibiotic resistance mechanisms of an opportunistic pathogen: a literature review. PeerJ 2023; 11:e14399. [PMID: 36627920 PMCID: PMC9826615 DOI: 10.7717/peerj.14399] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/25/2022] [Indexed: 01/07/2023] Open
Abstract
Serratia marcescens is a ubiquitous bacterium from order Enterobacterales displaying a high genetic plasticity that allows it to adapt and persist in multiple niches including soil, water, plants, and nosocomial environments. Recently, S. marcescens has gained attention as an emerging pathogen worldwide, provoking infections and outbreaks in debilitated individuals, particularly newborns and patients in intensive care units. S. marcescens isolates recovered from clinical settings are frequently described as multidrug resistant. High levels of antibiotic resistance across Serratia species are a consequence of the combined activity of intrinsic, acquired, and adaptive resistance elements. In this review, we will discuss recent advances in the understanding of mechanisms guiding resistance in this opportunistic pathogen.
Collapse
Affiliation(s)
- Faviola Tavares-Carreon
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Karla De Anda-Mora
- Departamento de Microbiología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Idalia C. Rojas-Barrera
- Environmental Genomics Group, Max Planck Institute for Evolutionary Biology, Plön, Germany,Christian-Albrechts-University Kiel, Kiel, Germany
| | - Angel Andrade
- Departamento de Microbiología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| |
Collapse
|
22
|
Seukep AJ, Mbuntcha HG, Kuete V, Chu Y, Fan E, Guo MQ. What Approaches to Thwart Bacterial Efflux Pumps-Mediated Resistance? Antibiotics (Basel) 2022; 11:antibiotics11101287. [PMID: 36289945 PMCID: PMC9598416 DOI: 10.3390/antibiotics11101287] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 12/03/2022] Open
Abstract
An effective response that combines prevention and treatment is still the most anticipated solution to the increasing incidence of antimicrobial resistance (AMR). As the phenomenon continues to evolve, AMR is driving an escalation of hard-to-treat infections and mortality rates. Over the years, bacteria have devised a variety of survival tactics to outwit the antibiotic’s effects, yet given their great adaptability, unexpected mechanisms are still to be discovered. Over-expression of efflux pumps (EPs) constitutes the leading strategy of bacterial resistance, and it is also a primary driver in the establishment of multidrug resistance (MDR). Extensive efforts are being made to develop antibiotic resistance breakers (ARBs) with the ultimate goal of re-sensitizing bacteria to medications to which they have become unresponsive. EP inhibitors (EPIs) appear to be the principal group of ARBs used to impair the efflux system machinery. Due to the high toxicity of synthetic EPIs, there is a growing interest in natural, safe, and innocuous ones, whereby plant extracts emerge to be excellent candidates. Besides EPIs, further alternatives are being explored including the development of nanoparticle carriers, biologics, and phage therapy, among others. What roles do EPs play in the occurrence of MDR? What weapons do we have to thwart EP-mediated resistance? What are the obstacles to their development? These are some of the core questions addressed in the present review.
Collapse
Affiliation(s)
- Armel Jackson Seukep
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 437004, China
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Buea, Buea P.O. Box 63, Cameroon
- Sino-Africa Joint Research Center, Chinese Academy of Sciences, Wuhan 437004, China
- Innovation Academy for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
| | - Helene Gueaba Mbuntcha
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang P.O. Box 67, Cameroon
| | - Victor Kuete
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang P.O. Box 67, Cameroon
| | - Yindi Chu
- State Key Laboratory of Medical Molecular Biology, Department of Microbiology and Parasitology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences/School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Enguo Fan
- State Key Laboratory of Medical Molecular Biology, Department of Microbiology and Parasitology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences/School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
- College of Life Sciences, Linyi University, Linyi 276005, China
- Correspondence: (E.F.); (M.-Q.G.)
| | - Ming-Quan Guo
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 437004, China
- Sino-Africa Joint Research Center, Chinese Academy of Sciences, Wuhan 437004, China
- Innovation Academy for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- Correspondence: (E.F.); (M.-Q.G.)
| |
Collapse
|
23
|
Genome-Wide Association Study of Nucleotide Variants Associated with Resistance to Nine Antimicrobials in Mycoplasma bovis. Microorganisms 2022; 10:microorganisms10071366. [PMID: 35889084 PMCID: PMC9320666 DOI: 10.3390/microorganisms10071366] [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: 06/17/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 12/03/2022] Open
Abstract
Antimicrobial resistance (AMR) studies of Mycoplasma bovis have generally focused on specific loci versus using a genome-wide association study (GWAS) approach. A GWAS approach, using two different models, was applied to 194 Mycoplasma bovis genomes. Both a fixed effects linear model (FEM) and a linear mixed model (LMM) identified associations between nucleotide variants (NVs) and antimicrobial susceptibility testing (AST) phenotypes. The AMR phenotypes represented fluoroquinolones, tetracyclines, phenicols, and macrolides. Both models identified known and novel NVs associated (Bonferroni adjusted p < 0.05) with AMR. Fluoroquinolone resistance was associated with multiple NVs, including previously identified mutations in gyrA and parC. NVs in the 30S ribosomal protein 16S were associated with tetracycline resistance, whereas NVs in 5S rRNA, 23S rRNA, and 50S ribosomal proteins were associated with phenicol and macrolide resistance. For all antimicrobial classes, resistance was associated with NVs in genes coding for ABC transporters and other membrane proteins, tRNA-ligases, peptidases, and transposases, suggesting a NV-based multifactorial model of AMR in M. bovis. This study was the largest collection of North American M. bovis isolates used with a GWAS for the sole purpose of identifying novel and non-antimicrobial-target NVs associated with AMR.
Collapse
|
24
|
Hou H, Li Y, Jin Y, Chen S, Long J, Duan G, Yang H. The crafty opponent: the defense systems of Staphylococcus aureus and response measures. Folia Microbiol (Praha) 2022; 67:233-243. [PMID: 35149955 DOI: 10.1007/s12223-022-00954-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/29/2022] [Indexed: 11/29/2022]
Abstract
Staphylococcus aureus is a serious threat to public health. S. aureus infection can cause acute or long-term persistent infections that are often resistant to antibiotics and are associated with high morbidity and death. Understanding the defensive systems of S. aureus can help clinicians make the best use of antimicrobial drugs and can also help with antimicrobial stewardship. The mechanisms and clinical implications of S. aureus defense systems, as well as potential response systems, were discussed in this study. Because resistance to all currently available antibiotics is unavoidable, new medicines are always being developed. Alternative techniques, such as anti-virulence and bacteriophage therapies, are being researched and may become major tools in the fight against staphylococcal infections in the future, in addition to the development of new small compounds that affect cell viability.
Collapse
Affiliation(s)
- Hongjie Hou
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Yang Li
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Yuefei Jin
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Shuaiyin Chen
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Jinzhao Long
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Guangcai Duan
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, No. 100 of Science Avenue, Zhengzhou, 450001, China.
| |
Collapse
|
25
|
Pilot Study on Poultry Meat from Antibiotic Free and Conventional Farms: Can Metagenomics Detect Any Difference? Foods 2022; 11:foods11030249. [PMID: 35159402 PMCID: PMC8834493 DOI: 10.3390/foods11030249] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
Antibiotic free farms are increasing in the poultry sector in order to address new EU regulations and consumer concerns. In this pilot study, we investigated whether the efforts of raising chickens without the use antibiotics make any difference in the microbiome of poultry meat eaten by consumers. To this aim we compared the microbiomes characterizing caeca and the corresponding carcasses of two groups of chickens reared, one reared on a conventional farm and one on an antibiotic-free intensive farm. The results showed a clear separation between the taxonomic, functional and antibiotic resistant genes in the caeca of the birds reared on the conventional and antibiotic free farm. However, that separation was completely lost on carcasses belonging to the two groups. The antibiotic-free production resulted in statistically significant lower antimicrobial resistance load in the caeca in comparison to the conventional production. Moreover, the antimicrobial resistance load on carcasses was much higher than in the caeca, without any significant difference between carcasses coming from the two types of farms. All in all, the results of this research highlighted the need to reduce sources of microbial contamination and antimicrobial resistance not only at the farm level but also at the post-harvest one.
Collapse
|
26
|
Genomic and Metabolic Characteristics of the Pathogenicity in Pseudomonas aeruginosa. Int J Mol Sci 2021; 22:ijms222312892. [PMID: 34884697 PMCID: PMC8657582 DOI: 10.3390/ijms222312892] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 01/22/2023] Open
Abstract
In recent years, the effectiveness of antimicrobials in the treatment of Pseudomonas aeruginosa infections has gradually decreased. This pathogen can be observed in several clinical cases, such as pneumonia, urinary tract infections, sepsis, in immunocompromised hosts, such as neutropenic cancer, burns, and AIDS patients. Furthermore, Pseudomonas aeruginosa causes diseases in both livestock and pets. The highly flexible and versatile genome of P. aeruginosa allows it to have a high rate of pathogenicity. The numerous secreted virulence factors, resulting from its numerous secretion systems, the multi-resistance to different classes of antibiotics, and the ability to produce biofilms are pathogenicity factors that cause numerous problems in the fight against P. aeruginosa infections and that must be better understood for an effective treatment. Infections by P. aeruginosa represent, therefore, a major health problem and, as resistance genes can be disseminated between the microbiotas associated with humans, animals, and the environment, this issue needs be addressed on the basis of an One Health approach. This review intends to bring together and describe in detail the molecular and metabolic pathways in P. aeruginosa's pathogenesis, to contribute for the development of a more targeted therapy against this pathogen.
Collapse
|
27
|
Rodrigues Dos Santos Barbosa C, Feitosa Muniz D, Silvino Pereira P, Maria de Arruda Lima S, Datiane de Morais Oliveira Tintino C, Cintia Alexandrino de Souza V, Mariana Assis da Silva J, Henrique Sousa da Costa R, Cosmo Andrade Pinheiro J, Maria Lobo Soares de Matos Y, Rose Alencar Menezes I, Gonçalves da Silva T, Manoella de Souza Lima G, Cristina Leal Balbino T, Pinto Siqueira-Júnior J, Assis Bezerra da Cunha F, Douglas Melo Coutinho H, Relison Tintino S. Evaluation of Elaiophylin extracted from Streptomyces hygroscopicus as a potential inhibitor of the NorA efflux protein in Staphylococcus aureus: An in vitro and in silico approach. Bioorg Med Chem Lett 2021; 50:128334. [PMID: 34425202 DOI: 10.1016/j.bmcl.2021.128334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
Compounds capable of inhibiting the efflux pump mechanism are a promising alternative against bacterial resistance because, when combined with antibiotics, they can increase the effectiveness of these drugs by inhibiting active efflux. Elaiophylin, derived from Streptomyces hygroscopicus, is a natural antibiotic that exhibits a variety of biological activities, including antibacterial activity. However, its potential as an inhibitor of the bacterial efflux mechanism has not been investigated. This study evaluated the ability of Elaiophylin to inhibit the NorA efflux pump in Staphylococcus aureus strains. Therefore, tests were performed to obtain the Minimum Inhibitory Concentration (MIC) and to verify the ability of Elaiophylin to potentiate the MIC of the antibiotic Norfloxacin and Ethidium Bromide (EtBr), known substrates of NorA efflux. Real-time PCR and molecular docking assays were also performed to assess the potential of Elaiophylin against NorA. The strains SA-1199 (wild type) and SA-1199B (NorA over-expressed) of S. aureus were used for this study. The results showed that Elaiophylin significantly decreased the MIC of Norfloxacin and EtBr, increasing the activity of these substrates against S. aureus, which carries the efflux protein NorA. However, Elaiophylin provided a non-significant reduction in norA gene expression, however, molecular docking demonstrated a high binding affinity between Elaiophylin and NorA efflux protein, indicating that Elaiophylin can act as a potential NorA in S. aureus.
Collapse
Affiliation(s)
| | - Débora Feitosa Muniz
- Laboratory of Microbiology and Molecular Biology (LMBM), Department of Biological Chemistry/CCBS/URCA, Brazil
| | | | | | | | | | | | | | | | | | - Irwin Rose Alencar Menezes
- Laboratory of Microbiology and Molecular Biology (LMBM), Department of Biological Chemistry/CCBS/URCA, Brazil
| | | | | | | | | | | | | | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Department of Biological Chemistry/CCBS/URCA, Brazil
| |
Collapse
|
28
|
Pinel-Cabello M, Chapon V, Ruiz-Fresneda MA, Alpha-Bazin B, Berthomieu C, Armengaud J, Merroun ML. Delineation of cellular stages and identification of key proteins for reduction and biotransformation of Se(IV) by Stenotrophomonas bentonitica BII-R7. JOURNAL OF HAZARDOUS MATERIALS 2021; 418:126150. [PMID: 34111750 DOI: 10.1016/j.jhazmat.2021.126150] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/30/2021] [Accepted: 05/14/2021] [Indexed: 06/12/2023]
Abstract
The widespread use of selenium (Se) in technological applications (e.g., solar cells and electronic devices) has led to an accumulation of this metalloid in the environment to toxic levels. The newly described bacterial strain Stenotrophomonas bentonitica BII-R7 has been demonstrated to reduce mobile Se(IV) to Se(0)-nanoparticles (Se(0)NPs) and volatile species. Amorphous Se-nanospheres are reported to aggregate to form crystalline nanostructures and trigonal selenium. We investigated the molecular mechanisms underlying the biotransformation of Se(IV) to less toxic forms using differential shotgun proteomics analysis of S. bentonitica BII-R7 grown with or without sodium selenite for three different time-points. Results showed an increase in the abundance of several proteins involved in Se(IV) reduction and stabilization of Se(0)NPs, such as glutathione reductase, in bacteria grown with Se(IV), in addition to many proteins with transport functions, including RND (resistance-nodulation-division) systems, possibly facilitating Se uptake. Notably proteins involved in oxidative stress defense (e.g., catalase/peroxidase HPI) were also induced by Se exposure. Electron microscopy analyses confirmed the biotransformation of amorphous nanospheres to trigonal Se. Overall, our results highlight the potential of S. bentonitica in reducing the bioavailability of Se, which provides a basis both for the development of bioremediation strategies and the eco-friendly synthesis of biotechnological nanomaterials.
Collapse
Affiliation(s)
- M Pinel-Cabello
- Department of Microbiology, University of Granada, Campus Fuentenueva s/n, 18071 Granada, Spain.
| | - V Chapon
- CEA, CNRS, Aix-Marseille Université, BIAM, IPM, 13108 Saint-Paul-lez-Durance, France
| | - M A Ruiz-Fresneda
- Department of Microbiology, University of Granada, Campus Fuentenueva s/n, 18071 Granada, Spain
| | - B Alpha-Bazin
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 30200 Bagnols sur Cèze, France
| | - C Berthomieu
- CEA, CNRS, Aix-Marseille Université, BIAM, IPM, 13108 Saint-Paul-lez-Durance, France
| | - J Armengaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 30200 Bagnols sur Cèze, France
| | - M L Merroun
- Department of Microbiology, University of Granada, Campus Fuentenueva s/n, 18071 Granada, Spain
| |
Collapse
|
29
|
Thamilselvan G, Sarveswari HB, Vasudevan S, Stanley A, Shanmugam K, Vairaprakash P, Solomon AP. Development of an Antibiotic Resistance Breaker to Resensitize Drug-Resistant Staphylococcus aureus: In Silico and In Vitro Approach. Front Cell Infect Microbiol 2021; 11:700198. [PMID: 34485178 PMCID: PMC8415528 DOI: 10.3389/fcimb.2021.700198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022] Open
Abstract
Efflux pumps are one of the predominant microbial resistant mechanisms leading to the development of multidrug resistance. In Staphylococcus aureus, overexpression of NorA protein enables the efflux of antibiotics belonging to the class of fluoroquinolones and, thus, makes S. aureus resistant. Hence, NorA efflux pumps are being extensively exploited as the potential drug target to evade bacterial resistance and resensitize bacteria to the existing antibiotics. Although several molecules are reported to inhibit NorA efflux pump effectively, boronic acid derivatives were shown to have promising NorA efflux pump inhibition. In this regard, the current study exploits 6-(3-phenylpropoxy)pyridine-3-boronic acid to further improve the activity and reduce cytotoxicity using the bioisostere approach, a classical medicinal chemistry concept. Using the SWISS-Bioisostere online tool, from the parent compound, 42 compounds were obtained upon the replacement of the boronic acid. The 42 compounds were docked with modeled NorA protein, and key molecular interactions of the prominent compounds were assessed. The top hit compounds were further analyzed for their drug-like properties using ADMET studies. The identified potent lead, 5-nitro-2-(3-phenylpropoxy)pyridine (5-NPPP), was synthesized, and in vitro efficacy studies have been proven to show enhanced efflux inhibition, thus acting as a potent antibiotic breaker to resensitize S. aureus without elucidating any cytotoxic effect to the host Hep-G2 cell lines.
Collapse
Affiliation(s)
- Gopalakrishnan Thamilselvan
- Quorum Sensing Laboratory, Centre of Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Hema Bhagavathi Sarveswari
- Quorum Sensing Laboratory, Centre of Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Sahana Vasudevan
- Quorum Sensing Laboratory, Centre of Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Alex Stanley
- Department of Bioinformatics, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Karthi Shanmugam
- Quorum Sensing Laboratory, Centre of Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India.,Department of Bioinformatics, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Pothiappan Vairaprakash
- Department of Chemistry, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre of Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
30
|
Dwivedi GR, Rai R, Pratap R, Singh K, Pati S, Sahu SN, Kant R, Darokar MP, Yadav DK. Drug resistance reversal potential of multifunctional thieno[3,2-c]pyran via potentiation of antibiotics in MDR P. aeruginosa. Biomed Pharmacother 2021; 142:112084. [PMID: 34449308 DOI: 10.1016/j.biopha.2021.112084] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022] Open
Abstract
We explored the antibacterial potential (alone and combination) against multidrug resistant (MDR) Pseudomonas aeruginosa isolates KG-P2 using synthesized thieno[3,2-c]pyran-2-ones in combination with different antibiotics. Out of 14 compounds, two compounds (3g and 3l) abridged the MIC of tetracycline (TET) by 16 folds. Compounds was killing the KG-P2 cells, in time dependent manner, lengthened post-antibiotic effect (PAE) of TET and found decreased the mutant prevention concentration (MPC) of TET. In ethidium bromide efflux experiment, two compounds repressed the drug transporter (efflux pumps) which is further supported by molecular docking of these compounds with efflux complex MexAB-OprM. In another study, these compounds inhibited the synthesis of biofilm.
Collapse
Affiliation(s)
- Gaurav Raj Dwivedi
- Microbiology Department, ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur 273013, India.
| | - Reeta Rai
- Department of Biochemistry, AIIMS Ansari Nagar, New Delhi 110029, India
| | - Ramendra Pratap
- Department of Chemistry, North campus University of Delhi, Delhi 110007, India.
| | - Khusbu Singh
- Microbiology Department, ICMR-Regional Medical Research Centre, Bhubaneshwar 751023, Odisha, India
| | - Sanghamitra Pati
- Microbiology Department, ICMR-Regional Medical Research Centre, Bhubaneshwar 751023, Odisha, India
| | - Satya Narayan Sahu
- Government College Balrampur, Balrampur-Ramanujganj, Chhattisgarh 497119, India
| | - Rajni Kant
- Microbiology Department, ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur 273013, India
| | - Mahendra P Darokar
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, ̥Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow 226015, India
| | - Dharmendra K Yadav
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, 191 Hambakmoeiro, Yeonsu-gu, Incheon 21924, Republic of Korea.
| |
Collapse
|
31
|
Agreles MAA, Cavalcanti IDL, Cavalcanti IMF. The Role of Essential Oils in the Inhibition of Efflux Pumps and Reversion of Bacterial Resistance to Antimicrobials. Curr Microbiol 2021; 78:3609-3619. [PMID: 34432112 DOI: 10.1007/s00284-021-02635-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 08/17/2021] [Indexed: 01/09/2023]
Abstract
Due to the deaths from infections caused by multidrug-resistant microorganisms worldwide, the World Health Organization considers antibiotic resistance to be a critical global public health problem. Bacterial resistance mechanisms are diverse and can be acquired through the overexpression of transmembrane proteins that are called efflux pumps, which act by expelling drugs from the intracellular environment, thereby preventing their action and contributing to the severity of infections. Efflux pumps are one of the main mechanisms of bacterial resistance, and it is important to identify new molecules that are capable of inhibiting the action of efflux pumps and circumvent the problem of resistance linked to the expression of these transmembrane proteins. The plants are promising candidates for obtaining biologically active substances, such as essential oils, with antimicrobial activity and inhibitors of efflux pumps, which can help in the resensitization of bacterial strains resistant to antibiotics. Therefore, this review aims to present the recently reported inhibitory activity of essential oils against bacterial pathogens that produce efflux pumps.
Collapse
Affiliation(s)
- Maria Anndressa Alves Agreles
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, CEP: 50670-901, Brazil
| | - Iago Dillion Lima Cavalcanti
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, CEP: 50670-901, Brazil
| | - Isabella Macário Ferro Cavalcanti
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, CEP: 50670-901, Brazil.
- Laboratory of Microbiology and Immunology, Academic Center of Vitória (CAV), Federal University of Pernambuco (UFPE), Rua do Alto do Reservatório s/n, Bela Vista, Vitória de Santo Antão, Pernambuco, CEP: 55608-680, Brazil.
| |
Collapse
|
32
|
Steenhuis M, Corona F, ten Hagen-Jongman CM, Vollmer W, Lambin D, Selhorst P, Klaassen H, Versele M, Chaltin P, Luirink J. Combining Cell Envelope Stress Reporter Assays in a Screening Approach to Identify BAM Complex Inhibitors. ACS Infect Dis 2021; 7:2250-2263. [PMID: 34125508 PMCID: PMC8369490 DOI: 10.1021/acsinfecdis.0c00728] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Indexed: 12/11/2022]
Abstract
The development of new antibiotics is particularly problematic in Gram-negative bacteria due to the presence of the outer membrane (OM), which serves as a permeability barrier. Recently, the β-barrel assembly machine (BAM), located in the OM and responsible for β-barrel type OM protein (OMP) assembly, has been validated as a novel target for antibiotics. Here, we identified potential BAM complex inhibitors using a screening approach that reports on cell envelope σE and Rcs stress in Escherichia coli. Screening a library consisting of 316 953 compounds yielded five compounds that induced σE and Rcs stress responses, while not inducing the intracellular heat-shock response. Two of the five compounds (compounds 2 and 14) showed the characteristics of known BAM complex inhibitors: synergy with OMP biogenesis mutants, decrease in the abundance of various OMPs, and loss of OM integrity. Importantly, compound 2 also inhibited BAM-dependent OMP folding in an in vitro refolding assay using purified BAM complex reconstituted in proteoliposomes.
Collapse
Affiliation(s)
- Maurice Steenhuis
- Department
of Molecular Microbiology, Amsterdam Institute
of Molecular and Life Sciences (AIMMS), Vrije Universiteit, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands
| | - Federico Corona
- Centre
for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle
upon Tyne NE2 4HH, United
Kingdom
| | - Corinne M. ten Hagen-Jongman
- Department
of Molecular Microbiology, Amsterdam Institute
of Molecular and Life Sciences (AIMMS), Vrije Universiteit, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands
| | - Waldemar Vollmer
- Centre
for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle
upon Tyne NE2 4HH, United
Kingdom
| | - Dominique Lambin
- Centre
for Innovation and Stimulation of Drug Discovery (CISTIM), Gaston Geenslaan 2, B-3001 Leuven, Belgium
| | - Philippe Selhorst
- Centre
for Innovation and Stimulation of Drug Discovery (CISTIM), Gaston Geenslaan 2, B-3001 Leuven, Belgium
| | - Hugo Klaassen
- Centre
for Innovation and Stimulation of Drug Discovery (CISTIM), Gaston Geenslaan 2, B-3001 Leuven, Belgium
| | - Matthias Versele
- Centre
for Innovation and Stimulation of Drug Discovery (CISTIM), Gaston Geenslaan 2, B-3001 Leuven, Belgium
| | - Patrick Chaltin
- Center
for Drug Design and Development (CD3), KU
Leuven R&D, Waaistraat 6, B-3000 Leuven, Belgium
| | - Joen Luirink
- Department
of Molecular Microbiology, Amsterdam Institute
of Molecular and Life Sciences (AIMMS), Vrije Universiteit, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
33
|
Rossi I, Bettini R, Buttini F. Resistant Tuberculosis: the Latest Advancements of Second-line Antibiotic Inhalation Products. Curr Pharm Des 2021; 27:1436-1452. [PMID: 33480336 DOI: 10.2174/1381612827666210122143214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 11/22/2022]
Abstract
Drug-resistant tuberculosis (TB) can be considered the man-made result of interrupted, erratic or inadequate TB therapy. As reported in WHO data, resistant Mycobacterium tuberculosis (Mtb) strains continue to constitute a public health crisis. Mtb is naturally able to survive host defence mechanisms and to resist most antibiotics currently available. Prolonged treatment regimens using the available first-line drugs give rise to poor patient compliance and a rapid evolution of strains resistant to rifampicin only or to both rifampicin and isoniazid (multi drug-resistant, MDR-TB). The accumulation of mutations may give rise to extensively drug-resistant strains (XDR-TB), i.e. strains with resistance also to fluoroquinolones and to the injectable aminoglycoside, which represent the second-line drugs. Direct lung delivery of anti-tubercular drugs, as an adjunct to conventional routes, provides high concentrations within the lungs, which are the intended target site of drug delivery, representing an interesting strategy to prevent or reduce the development of drug-resistant strains. The purpose of this paper is to describe and critically analyse the most recent and advanced results in the formulation development of WHO second-line drug inhalation products, with particular focus on dry powder formulation. Although some of these formulations have been developed for other lung infectious diseases (Pseudomonas aeruginosa, nontuberculous mycobacteria), they could be valuable to treat MDR-TB and XDR-TB.
Collapse
Affiliation(s)
- Irene Rossi
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Ruggero Bettini
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Francesca Buttini
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| |
Collapse
|
34
|
da Silva PB, Araújo VHS, Fonseca-Santos B, Solcia MC, Ribeiro CM, da Silva IC, Alves RC, Pironi AM, Silva ACL, Victorelli FD, Fernandes MA, Ferreira PS, da Silva GH, Pavan FR, Chorilli M. Highlights Regarding the Use of Metallic Nanoparticles against Pathogens Considered a Priority by the World Health Organization. Curr Med Chem 2021; 28:1906-1956. [PMID: 32400324 DOI: 10.2174/0929867327666200513080719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/11/2020] [Accepted: 03/20/2020] [Indexed: 11/22/2022]
Abstract
The indiscriminate use of antibiotics has facilitated the growing resistance of bacteria, and this has become a serious public health problem worldwide. Several microorganisms are still resistant to multiple antibiotics and are particularly dangerous in the hospital and nursing home environment, and to patients whose care requires devices, such as ventilators and intravenous catheters. A list of twelve pathogenic genera, which especially included bacteria that were not affected by different antibiotics, was released by the World Health Organization (WHO) in 2017, and the research and development of new antibiotics against these genera has been considered a priority. The nanotechnology is a tool that offers an effective platform for altering the physicalchemical properties of different materials, thereby enabling the development of several biomedical applications. Owing to their large surface area and high reactivity, metallic particles on the nanometric scale have remarkable physical, chemical, and biological properties. Nanoparticles with sizes between 1 and 100 nm have several applications, mainly as new antimicrobial agents for the control of microorganisms. In the present review, more than 200 reports of various metallic nanoparticles, especially those containing copper, gold, platinum, silver, titanium, and zinc were analyzed with regard to their anti-bacterial activity. However, of these 200 studies, only 42 reported about trials conducted against the resistant bacteria considered a priority by the WHO. All studies are in the initial stage, and none are in the clinical phase of research.
Collapse
Affiliation(s)
- Patricia Bento da Silva
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| | | | - Bruno Fonseca-Santos
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| | - Mariana Cristina Solcia
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| | | | | | - Renata Carolina Alves
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| | - Andressa Maria Pironi
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| | | | | | - Mariza Aires Fernandes
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| | - Paula Scanavez Ferreira
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| | - Gilmar Hanck da Silva
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| | - Fernando Rogério Pavan
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| | - Marlus Chorilli
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara-SP, Brazil
| |
Collapse
|
35
|
Chemical synthesis, molecular docking and MepA efflux pump inhibitory effect by 1,8-naphthyridines sulfonamides. Eur J Pharm Sci 2021; 160:105753. [PMID: 33581258 DOI: 10.1016/j.ejps.2021.105753] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/27/2021] [Accepted: 02/08/2021] [Indexed: 11/20/2022]
Abstract
This study aimed to evaluate the antibacterial activity and to verify, in silico and in vitro, the inhibition of efflux mechanisms using a series of synthesized 1,8-naphthyridines sulfonamides against Staphylococcus aureus strains carrying MepA efflux pumps. The chemical synthesis occurred through the thermolysis of the Meldrum's acid adduct. The sulfonamide derivatives were obtained by the sulfonylation of 2-amino-5‑chloro-1,8-naphthyridine with commercial benzenesulfonyl chloride. Antibacterial activity was assessed by the broth microdilution test. Efflux pump inhibitory capacity was evaluated in silico by molecular docking and in vitro by analyzing synergistic effects on ciprofloxacin and ethidium bromide (EtBr) and by EtBr fluorescence emission assays. The following 1,8-naphthyridines were synthesized: 4-methyl-N-(5‑chloro-1,8-naphthyridin-2-yl)-benzenesulfonamide (Compound 10a); 2,5-dichloro-N-(5‑chloro-1,8-naphthyridin-2-yl)-benzenesulfonamide (Compound 10b); 4-fluoro-N-(5‑chloro-1,8-naphthyridin-2-yl)-benzenesulfonamide (Compound 10c); 2,3,4-trifluoro-N-(5‑chloro-1,8-naphthyridin-2-yl)-benzenesulfonamide (Compound 10d); 3-trifluoromethyl-N-(5‑chloro-1,8-naphthyridin-2-yl)-benzenesulfonamide (Compound 10e); 4‑bromo-2,5-difluoro-N-(5‑chloro-1,8-naphthyridin-2-yl)-benzenesulfonamide (Compound 10f). The 1,8-naphthyridines derivatives associated with sulfonamides did not show antibacterial activity. However, they showed a favorable pharmacokinetic profile with possible MepA efflux pump inhibitory action, demonstrated in molecular docking. In addition to the promising results in reducing the concentration of intracellular EtBr. 1,8-naphthyridines act as putative agents in the inhibitory action of the MepA efflux pump.
Collapse
|
36
|
Langendonk RF, Neill DR, Fothergill JL. The Building Blocks of Antimicrobial Resistance in Pseudomonas aeruginosa: Implications for Current Resistance-Breaking Therapies. Front Cell Infect Microbiol 2021; 11:665759. [PMID: 33937104 PMCID: PMC8085337 DOI: 10.3389/fcimb.2021.665759] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
P. aeruginosa is classified as a priority one pathogen by the World Health Organisation, and new drugs are urgently needed, due to the emergence of multidrug-resistant (MDR) strains. Antimicrobial-resistant nosocomial pathogens such as P. aeruginosa pose unwavering and increasing threats. Antimicrobial stewardship has been a challenge during the COVID-19 pandemic, with a majority of those hospitalized with SARS-CoV2 infection given antibiotics as a safeguard against secondary bacterial infection. This increased usage, along with increased handling of sanitizers and disinfectants globally, may further accelerate the development and spread of cross-resistance to antibiotics. In addition, P. aeruginosa is the primary causative agent of morbidity and mortality in people with the life-shortening genetic disease cystic fibrosis (CF). Prolonged periods of selective pressure, associated with extended antibiotic treatment and the actions of host immune effectors, results in widespread adaptive and acquired resistance in P. aeruginosa found colonizing the lungs of people with CF. This review discusses the arsenal of resistance mechanisms utilized by P. aeruginosa, how these operate under high-stress environments such as the CF lung and how their interconnectedness can result in resistance to multiple antibiotic classes. Intrinsic, adaptive and acquired resistance mechanisms will be described, with a focus on how each layer of resistance can serve as a building block, contributing to multi-tiered resistance to antimicrobial activity. Recent progress in the development of anti-resistance adjuvant therapies, targeting one or more of these building blocks, should lead to novel strategies for combatting multidrug resistant P. aeruginosa. Anti-resistance adjuvant therapy holds great promise, not least because resistance against such therapeutics is predicted to be rare. The non-bactericidal nature of anti-resistance adjuvants reduce the selective pressures that drive resistance. Anti-resistance adjuvant therapy may also be advantageous in facilitating efficacious use of traditional antimicrobials, through enhanced penetration of the antibiotic into the bacterial cell. Promising anti-resistance adjuvant therapeutics and targets will be described, and key remaining challenges highlighted. As antimicrobial stewardship becomes more challenging in an era of emerging and re-emerging infectious diseases and global conflict, innovation in antibiotic adjuvant therapy can play an important role in extending the shelf-life of our existing antimicrobial therapeutic agents.
Collapse
Affiliation(s)
- R. Frèdi Langendonk
- Institute of Infection, Veterinary and Ecological Science, University of Liverpool, Liverpool, United Kingdom
| | | | | |
Collapse
|
37
|
Pinel-Cabello M, Jroundi F, López-Fernández M, Geffers R, Jarek M, Jauregui R, Link A, Vílchez-Vargas R, Merroun ML. Multisystem combined uranium resistance mechanisms and bioremediation potential of Stenotrophomonas bentonitica BII-R7: Transcriptomics and microscopic study. JOURNAL OF HAZARDOUS MATERIALS 2021; 403:123858. [PMID: 33264934 DOI: 10.1016/j.jhazmat.2020.123858] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/18/2020] [Accepted: 08/24/2020] [Indexed: 06/12/2023]
Abstract
The potential use of microorganisms in the bioremediation of U pollution has been extensively described. However, a lack of knowledge on molecular resistance mechanisms has become a challenge for the use of these technologies. We reported on the transcriptomic and microscopic response of Stenotrophomonas bentonitica BII-R7 exposed to 100 and 250 μM of U. Results showed that exposure to 100 μM displayed up-regulation of 185 and 148 genes during the lag and exponential phases, respectively, whereas 143 and 194 were down-regulated, out of 3786 genes (>1.5-fold change). Exposure to 250 μM of U showed up-regulation of 68 genes and down-regulation of 290 during the lag phase. Genes involved in cell wall and membrane protein synthesis, efflux systems and phosphatases were up-regulated under all conditions tested. Microscopic observations evidenced the formation of U-phosphate minerals at membrane and extracellular levels. Thus, a biphasic process is likely to occur: the increased cell wall would promote the biosorption of U to the cell surface and its precipitation as U-phosphate minerals enhanced by phosphatases. Transport systems would prevent U accumulation in the cytoplasm. These findings contribute to an understanding of how microbes cope with U toxicity, thus allowing for the development of efficient bioremediation strategies.
Collapse
Affiliation(s)
- M Pinel-Cabello
- Department of Microbiology, University of Granada, Campus Fuentenueva s/n, 18071, Granada, Spain.
| | - F Jroundi
- Department of Microbiology, University of Granada, Campus Fuentenueva s/n, 18071, Granada, Spain
| | - M López-Fernández
- Institute of Resource Ecology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - R Geffers
- Genome Analytics, Helmholtz Centre for Infection Research (HZI), 38124, Braunschweig, Germany
| | - M Jarek
- Genome Analytics, Helmholtz Centre for Infection Research (HZI), 38124, Braunschweig, Germany
| | - R Jauregui
- AgResearch Grasslands Research Centre, Tennent drive, Palmerston North, New Zealand
| | - A Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, University of Magdeburg, Leipziger Str. 44.39120, Magdeburg, Germany
| | - R Vílchez-Vargas
- Department of Gastroenterology, Hepatology and Infectious Diseases, University of Magdeburg, Leipziger Str. 44.39120, Magdeburg, Germany
| | - M L Merroun
- Department of Microbiology, University of Granada, Campus Fuentenueva s/n, 18071, Granada, Spain
| |
Collapse
|
38
|
Non-steroidal anti-inflammatory drugs, plant extracts, and characterized microparticles to modulate antimicrobial resistance of epidemic mecA positive S. aureus of dairy origin. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-020-01628-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
39
|
Oliveira-Tintino CDDM, Muniz DF, Barbosa CRDS, Pereira RLS, Begnini IM, Rebelo RA, Silva LED, Mireski SL, Nasato MC, Krautler MIL, Pereira PS, Costa JGMD, Rodrigues FFG, Teixeira AMR, Ribeiro-Filho J, Tintino SR, de Menezes IRA, Coutinho HDM, Silva TGD. The 1,8-naphthyridines sulfonamides are NorA efflux pump inhibitors. J Glob Antimicrob Resist 2020; 24:233-240. [PMID: 33385589 DOI: 10.1016/j.jgar.2020.11.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 10/24/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Efflux pumps are transmembrane proteins associated with bacterial resistance mechanisms. Bacteria use these proteins to actively transport antibiotics to the extracellular medium, preventing the pharmacological action of these drugs. This study aimed to evaluate in vitro the antibacterial activity of 1,8-naphthyridines sulfonamides, as well as their ability to inhibit efflux systems of Staphylococcus aureus strains expressing different levels of the NorA efflux pump. METHODS The broth microdilution test was performed to assess antibacterial activity. Efflux pump inhibition was evaluated in silico by molecular docking and in vitro by fluorometric tests, and the minimum inhibitory concentration (MIC) was determined. The MIC was determined in the association between 1,8-naphthyridine and norfloxacin or ethidium bromide. RESULTS The 1,8-naphthyridines did not show direct antibacterial activity. However, they effectively reduced the MIC of multidrug-resistant bacteria by associating with norfloxacin and ethidium bromide, in addition to increasing the fluorescence emission. In silico analysis addressing the binding between NorA and 1,8-naphthyridines suggests that hydrogen bonds and hydrophilic interactions represent the interactions with the most favourable binding energy, corroborating the experimental data. CONCLUSION Our data suggest that 1,8-naphthyridines sulfonamides inhibit bacterial resistance through molecular mechanisms associated with inhibition of the NorA efflux pump in S. aureus strains.
Collapse
Affiliation(s)
| | - Débora Feitosa Muniz
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | | | - Raimundo Luiz Silva Pereira
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | - Iêda Maria Begnini
- Department of Chemistry, Regional University of Blumenau, FURB, Itoupava Seca, 89030-903, Blumenau, SC, Brazil
| | - Ricardo Andrade Rebelo
- Department of Chemistry, Regional University of Blumenau, FURB, Itoupava Seca, 89030-903, Blumenau, SC, Brazil
| | - Luiz Everson da Silva
- Postgraduate Program in Sustainable Territorial Development, Coastal Sector, Federal University of Paraná, Curitiba, PR, Brazil
| | - Sandro Lucio Mireski
- Department of Chemistry, Regional University of Blumenau, FURB, Itoupava Seca, 89030-903, Blumenau, SC, Brazil
| | - Michele Caroline Nasato
- Department of Chemistry, Regional University of Blumenau, FURB, Itoupava Seca, 89030-903, Blumenau, SC, Brazil
| | | | - Pedro Silvino Pereira
- Laboratory of Pharmatoxicological Prospecting of Bioactive Products, Department of Antibiotics, Federal University of Pernambuco, UFPE, Recife, PE, Brazil
| | - José Galberto Martins da Costa
- Laboratory of Natural Products, Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | | | - Alexandre Magno Rodrigues Teixeira
- Laboratory of simulations and molecular spectroscopy, Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | - Jaime Ribeiro-Filho
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, IGM-Fiocruz, Salvador, BA, Brazil
| | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | - Irwin Rose Alencar de Menezes
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | - Henrique Douglas Melo Coutinho
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, URCA, Crato, CE, Brazil
| | - Teresinha Gonçalves da Silva
- Laboratory of Pharmatoxicological Prospecting of Bioactive Products, Department of Antibiotics, Federal University of Pernambuco, UFPE, Recife, PE, Brazil
| |
Collapse
|
40
|
Anti-staphylococcal activity and mode of action of thioridazine photoproducts. Sci Rep 2020; 10:18043. [PMID: 33093568 PMCID: PMC7582912 DOI: 10.1038/s41598-020-74752-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
Antibiotic resistance became an increasing risk for population health threatening our ability to fight infectious diseases. The objective of this study was to evaluate the activity of laser irradiated thioridazine (TZ) against clinically-relevant bacteria in view to fight antibiotic resistance. TZ in ultrapure water solutions was irradiated (1–240 min) with 266 nm pulsed laser radiation. Irradiated solutions were characterized by UV–Vis and FTIR absorption spectroscopy, thin layer chromatography, laser-induced fluorescence, and dynamic surface tension measurements. Molecular docking studies were made to evaluate the molecular mechanisms of photoproducts action against Staphylococcus aureus and MRSA. More general, solutions were evaluated for their antimicrobial and efflux inhibitory activity against a panel of bacteria of clinical relevance. We observed an enhanced antimicrobial activity of TZ photoproducts against Gram-positive bacteria. This was higher than ciprofloxacin effects for methicillin- and ciprofloxacin-resistant Staphylococcus aureus. Molecular docking showed the Penicillin-binding proteins PBP3 and PBP2a inhibition by sulforidazine as a possible mechanism of action against Staphylococcus aureus and MRSA strains, respectively. Irradiated TZ reveals possible advantages in the treatment of infectious diseases produced by antibiotic-resistant Gram-positive bacteria. TZ repurposing and its photoproducts, obtained by laser irradiation, show accelerated and low-costs of development if compared to chemical synthesis.
Collapse
|
41
|
Liu JL, Yao J, Zhu X, Zhou DL, Duran R, Mihucz VG, Bashir S, Hudson-Edwards KA. Metagenomic exploration of multi-resistance genes linked to microbial attributes in active nonferrous metal(loid) tailings. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 273:115667. [PMID: 33497944 DOI: 10.1016/j.envpol.2020.115667] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 06/12/2023]
Abstract
Mine tailings sites are considered as a continuous source of discharged metal(loid)s and residual organic flotation reagents. They are extremely toxic environments representing unique ecological niches for microbial communities. Mine tailings as a source of multi-resistance genes have been poorly investigated. Metagenomic analysis for four active nonferrous metal(loid) tailings sites with different environmental parameters was conducted. The abundance of Thiobacillus, able to tolerate acidity and showing iron- and sulfur/sulfide oxidation capacities, was significantly different (p < 0.05) between acid and neutral tailings sites. Correlation analyses showed that Zn, Pb, TP, Cd, and Cu were the main drivers influencing the bacterial compositions. Multi-metal resistance genes (MRGs) and antibiotic resistance genes (ARGs), such as baca and copA, were found to be co-selected by high concentrations of metal(loid)s tailings. The main contributors to different distributions of MRGs were Thiobacillus and Nocardioides genus, while genera with low abundance (<0.1%) were the main contributors for ARGs. Functional metabolic pathways related to Fe-S metabolism, polycyclic aromatic hydrocarbons (PAHs) degradation and acid stress were largely from Altererythrobacter, Lysobacter, and Thiobacillus, respectively. Such information provides new insights on active tailings with highly toxic contaminants. Short-term metal(loid) exposure of microorganism in active nonferrous metal(loid) tailings contribute to the co-occurrence of ARGs and MRGs, and aggravation of tailings acidification. Our results recommend that the management of microorganisms involved in acid tolerance and metal/antibiotic resistance is of key importance for in-suit treatment of the continuous discharge of tailings with multiple metal(loid) contaminants into impoundments.
Collapse
Affiliation(s)
- Jian-Li Liu
- School of Water Resources and Environment and Research Center of Environmental Science and Engineering, Sino-Hungarian Joint Laboratory of Environmental Science and Health, China University of Geosciences (Beijing), 29 Xueyuan Road, Haidian District, 100083, Beijing, China
| | - Jun Yao
- School of Water Resources and Environment and Research Center of Environmental Science and Engineering, Sino-Hungarian Joint Laboratory of Environmental Science and Health, China University of Geosciences (Beijing), 29 Xueyuan Road, Haidian District, 100083, Beijing, China.
| | - Xiaozhe Zhu
- School of Water Resources and Environment and Research Center of Environmental Science and Engineering, Sino-Hungarian Joint Laboratory of Environmental Science and Health, China University of Geosciences (Beijing), 29 Xueyuan Road, Haidian District, 100083, Beijing, China
| | - De-Liang Zhou
- Beijing Zhongdianyida Technology Co., Ltd, Beijing, 100190, China
| | - Robert Duran
- School of Water Resources and Environment and Research Center of Environmental Science and Engineering, Sino-Hungarian Joint Laboratory of Environmental Science and Health, China University of Geosciences (Beijing), 29 Xueyuan Road, Haidian District, 100083, Beijing, China; Equipe Environnement et Microbiologie, MELODY Group, Université de Pau et des Pays de L'Adour/E2S UPPA, IPREM UMR CNRS 5254, BP 1155, 64013, Pau Cedex, France
| | - Victor G Mihucz
- Sino-Hungarian Joint Research Laboratory for Environmental Sciences and Health, Eötvös Loránd University, H-1117 Budapest, Pázmány Péter Stny. 1/A, Hungary
| | - Safdar Bashir
- Sub-campus Depalpur, University of Agriculture Faisalabad, Okara 56130, Pakistan
| | - Karen A Hudson-Edwards
- Environment & Sustainability Institute and Camborne School of Mines, University of Exeter, Penryn, Cornwall TR10 9DF, UK
| |
Collapse
|
42
|
Impey RE, Hawkins DA, Sutton JM, Soares da Costa TP. Overcoming Intrinsic and Acquired Resistance Mechanisms Associated with the Cell Wall of Gram-Negative Bacteria. Antibiotics (Basel) 2020; 9:E623. [PMID: 32961699 PMCID: PMC7558195 DOI: 10.3390/antibiotics9090623] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/19/2022] Open
Abstract
The global increase in multi-drug-resistant bacteria is severely impacting our ability to effectively treat common infections. For Gram-negative bacteria, their intrinsic and acquired resistance mechanisms are heightened by their unique cell wall structure. The cell wall, while being a target of some antibiotics, represents a barrier due to the inability of most antibacterial compounds to traverse and reach their intended target. This means that its composition and resulting mechanisms of resistance must be considered when developing new therapies. Here, we discuss potential antibiotic targets within the most well-characterised resistance mechanisms associated with the cell wall in Gram-negative bacteria, including the outer membrane structure, porins and efflux pumps. We also provide a timely update on the current progress of inhibitor development in these areas. Such compounds could represent new avenues for drug discovery as well as adjuvant therapy to help us overcome antibiotic resistance.
Collapse
Affiliation(s)
- Rachael E. Impey
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (R.E.I.); (D.A.H.)
| | - Daniel A. Hawkins
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (R.E.I.); (D.A.H.)
| | - J. Mark Sutton
- National Infection Service, Research and Development Institute, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK;
| | - Tatiana P. Soares da Costa
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (R.E.I.); (D.A.H.)
| |
Collapse
|
43
|
AlMatar M, Albarri O, Makky EA, Köksal F. Efflux pump inhibitors: new updates. Pharmacol Rep 2020; 73:1-16. [PMID: 32946075 DOI: 10.1007/s43440-020-00160-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/17/2022]
Abstract
The discovery of antibiotics ought to have ended the issue of bacterial infections, but this was not the case as it has led to the evolution of various mechanisms of bacterial resistance against various antibiotics. The efflux pump remains one of the mechanisms through which organisms develop resistance against antibiotics; this is because organisms can extrude most of the clinically relevant antibiotics from the interior cell environment to the exterior environment via the efflux pumps. Efflux pumps are thought to contribute significantly to biofilm formation as highlighted by various studies. Therefore, the inhibition of these efflux pumps can be a potential way of improving the activity of antibiotics, particularly now that the discovery of novel antibiotics is becoming tedious. Efflux pump inhibitors (EPIs) are molecules that can inhibit efflux pumps; they have been considered potential therapeutic agents for rejuvenating the activity of antibiotics that have already lost their activity against bacteria. However, studies are yet to determine the specific substrates for such pumps; the effect of altered efflux activity of these pumps on biofilm formation is still being investigated. A clear knowledge of the involvement of efflux pumps in biofilm development could aid in developing new agents that can interfere with their function and help to prevent biofilms formation; thereby, improving the outcome of treatment strategies. This review focuses on the novel update of EPIs and discusses the evidence of the roles of efflux pumps in biofilm formation; the potential approaches towards overcoming the increasing problem of biofilm-based infections are also discussed.
Collapse
Affiliation(s)
- Manaf AlMatar
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang (UMP), 26300, Gambang, Kuantan, Malaysia.
| | - Osman Albarri
- Department of Biotechnology, Institute of Natural and Applied Sciences (Fen Bilimleri Enstitüsü), Çukurova University, Adana, Turkey
| | - Essam A Makky
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang (UMP), 26300, Gambang, Kuantan, Malaysia.
| | - Fatih Köksal
- Department of Medical Microbiology, Faculty of Medicine, Çukurova University, Adana, Turkey
| |
Collapse
|
44
|
Sutera V, Hennebique A, Lopez F, Fernandez N, Schneider D, Maurin M. Genomic trajectories to fluoroquinolone resistance in Francisella tularensis subsp. holarctica live vaccine strain. Int J Antimicrob Agents 2020; 56:106153. [PMID: 32911069 DOI: 10.1016/j.ijantimicag.2020.106153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 07/04/2020] [Accepted: 08/29/2020] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Fluoroquinolone (FQ)-resistant mutants were previously selected from the live vaccine strain (LVS) of Francisella tularensis (F. tularensis) subsp. holarctica. This study further characterised all genetic changes that occurred in these mutants during the evolutionary trajectory toward high-level FQ resistance, and their potential impact on F. tularensis antibiotic resistance and intracellular fitness. METHODS The whole genomes of FQ-resistant mutants were determined and compared with those of their parental strain. All detected mutations were evaluated for their potential impact on FQ resistance and intracellular multiplication of F. tularensis. RESULTS As compared with the parental LVS genome, 28 mutations were found in the derived FQ-resistant mutants. These mutations involved all genes encoding type II topoisomerases (i.e. gyrA, gyrB, parC, and parE). Interestingly, some of them were not previously associated with FQ resistance, warranting further characterisation. Mutations associated with FQ resistance were also found in other genes, including three encoding proteins involved in transport processes. Most of the detected mutations did not alter multiplication of the corresponding mutants in J774 cells. In contrast, all mutations at locus FTL_0439 encoding FupA/B, a membrane protein involved in iron transport, were associated with FQ resistance and fitness loss. CONCLUSION FQ resistance in F. tularensis is complex and may involve single or combined mutations in genes encoding type II topoisomerases, transport systems and FupA/B. In vivo studies are now required to assess the potential role of these mutations in FQ treatment failures.
Collapse
Affiliation(s)
- Vivien Sutera
- Centre National de Référence Francisella tularensis, Laboratoire de Bactériologie, Institut de Biologie et de Pathologie, CHU Grenoble Alpes, Grenoble, France; Laboratoire Techniques de l'Ingénierie Médicale et de la Complexité Informatique - Mathématiques et Applications (TIMC-IMAG), Univ. Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Grenoble, France
| | - Aurélie Hennebique
- Centre National de Référence Francisella tularensis, Laboratoire de Bactériologie, Institut de Biologie et de Pathologie, CHU Grenoble Alpes, Grenoble, France; Laboratoire Techniques de l'Ingénierie Médicale et de la Complexité Informatique - Mathématiques et Applications (TIMC-IMAG), Univ. Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Grenoble, France
| | - Fabrice Lopez
- Technological Advances for Genomics and Clinics (TAGC), Univ. Aix-Marseille II, Marseille, France; Transcriptomic and Genomic Marseille-Luminy (TGML), IBiSA platform, Marseille, France
| | - Nicolas Fernandez
- Technological Advances for Genomics and Clinics (TAGC), Univ. Aix-Marseille II, Marseille, France; Transcriptomic and Genomic Marseille-Luminy (TGML), IBiSA platform, Marseille, France
| | - Dominique Schneider
- Laboratoire Techniques de l'Ingénierie Médicale et de la Complexité Informatique - Mathématiques et Applications (TIMC-IMAG), Univ. Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Grenoble, France
| | - Max Maurin
- Centre National de Référence Francisella tularensis, Laboratoire de Bactériologie, Institut de Biologie et de Pathologie, CHU Grenoble Alpes, Grenoble, France; Laboratoire Techniques de l'Ingénierie Médicale et de la Complexité Informatique - Mathématiques et Applications (TIMC-IMAG), Univ. Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Grenoble, France.
| |
Collapse
|
45
|
Ermakova AY, Beletsky AV, Mardanov AV, Petrova MA, Ravin NV, Rakitin AL. A Novel Plasmid pALWVS1.4 from Acinetobacter lwoffii Strain VS15, Carrying the Chloramphenicol Resistance Gene. Microbiology (Reading) 2020. [DOI: 10.1134/s0026261720050070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
46
|
Yewale PP, Lokhande KB, Sridhar A, Vaishnav M, Khan FA, Mandal A, Swamy KV, Jass J, Nawani N. Molecular profiling of multidrug-resistant river water isolates: insights into resistance mechanism and potential inhibitors. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:27279-27292. [PMID: 31236860 DOI: 10.1007/s11356-019-05738-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/10/2019] [Indexed: 06/09/2023]
Abstract
Polluted waters are an important reservoir for antibiotic resistance genes and multidrug-resistant bacteria. This report describes the microbial community, antibiotic resistance genes, and the genetic profile of extended spectrum β-lactamase strains isolated from rivers at, Pune, India. ESBL-producing bacteria isolated from diverse river water catchments running through Pune City were characterized for their antibiotic resistance. The microbial community and types of genes which confer antibiotic resistance were identified followed by the isolation of antibiotic-resistant bacteria on selective media and their genome analysis. Four representative isolates were sequenced using next generation sequencing for genomic analysis. They were identified as Pseudomonas aeruginosa, Escherichia coli, and two isolates were Enterobacter cloacae. The genes associated with the multidrug efflux pumps, such as tolC, macA, macB, adeL, and rosB, were detected in the isolates. As MacAB-TolC is an ABC type efflux pump responsible for conferring resistance in bacteria to several antibiotics, potential efflux pump inhibitors were identified by molecular docking. The homology model of their MacB protein with that from Escherichia coli K12 demonstrated structural changes in different motifs of MacB. Molecular docking of reported efflux pump inhibitors revealed the highest binding affinity of compound MC207-110 against MacB. It also details the potential efflux pump inhibitors that can serve as possible drug targets in drug development and discovery.
Collapse
Affiliation(s)
- Priti Prabhakar Yewale
- Microbial Diversity Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Pune, 411 033, India
| | - Kiran Bharat Lokhande
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Pune, 411 033, India
| | - Aishwarya Sridhar
- Microbial Diversity Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Pune, 411 033, India
| | - Monika Vaishnav
- Microbial Diversity Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Pune, 411 033, India
| | - Faisal Ahmad Khan
- The Life Science Centre-Biology, School of Science and Technology, Örebro University, 705 10, Örebro, Sweden
| | - Abul Mandal
- Systems Biology Research Center, School of Biosciences, University of Skӧvde, Skӧvde, Sweden
| | - Kakumani Venkateswara Swamy
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Pune, 411 033, India
| | - Jana Jass
- The Life Science Centre-Biology, School of Science and Technology, Örebro University, 705 10, Örebro, Sweden.
| | - Neelu Nawani
- Microbial Diversity Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Pune, 411 033, India.
| |
Collapse
|
47
|
Ledger L, Eidt J, Cai HY. Identification of Antimicrobial Resistance-Associated Genes through Whole Genome Sequencing of Mycoplasma bovis Isolates with Different Antimicrobial Resistances. Pathogens 2020; 9:pathogens9070588. [PMID: 32707642 PMCID: PMC7400188 DOI: 10.3390/pathogens9070588] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 01/08/2023] Open
Abstract
Antimicrobial resistance (AMR) in Mycoplasma bovis has been previously associated with topoisomerase and ribosomal gene mutations rather than specific resistance-conferring genes. Using whole genome sequencing (WGS) to identify potential new AMR mechanisms for M. bovis, it was found that a 2019 clinical isolate with high MIC (2019-043682) for fluoroquinolones, macrolides, lincosamides, pleuromutilins and tetracyclines had a new core genome multilocus sequencing (cgMLST) type (ST10-like) and 91% sequence similarity to the published genome of M. bovis PG45. Closely related to PG45, a 1982 isolate (1982-M6152) shared the same cgMLST type (ST17), 97.2% sequence similarity and low MIC results. Known and potential AMR- associated genetic events were identified through multiple sequence alignment of the three genomes. Isolate 2019-043682 had 507 genes with non-synonymous mutations (NSMs) and 67 genes disrupted. Isolate 1982-M6152 had 81 NSMs and 20 disruptions. Using functional roles and known mechanisms of antimicrobials, a 55 gene subset was assessed for AMR potential. Seventeen were previously identified from other bacteria as sites of AMR mutation, 38 shared similar functions to them, and 11 contained gene-disrupting mutations. This study indicated that M. bovis may obtain high AMR characteristics by mutating or disrupting other functional genes, in addition to topoisomerases and ribosomal genes.
Collapse
|
48
|
BacEffluxPred: A two-tier system to predict and categorize bacterial efflux mediated antibiotic resistance proteins. Sci Rep 2020; 10:9287. [PMID: 32518231 PMCID: PMC7283322 DOI: 10.1038/s41598-020-65981-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 05/12/2020] [Indexed: 11/08/2022] Open
Abstract
Efflux proteins are transport proteins, which are involved in transporting different substrates from the cell to the external environment, including antibiotics. The efflux mechanism and efflux pumps are a major reason underlying emerging rampant antibiotic resistance (AR) in microbes. To reduce the resources required and time of identification, characterization and classification of bacterial efflux proteins, we have developed a fast and accurate support vector machine based two-tier prediction system, BacEffluxPred, which can predict bacterial efflux proteins responsible for AR and identify their corresponding families. A leave-one-out cross-validation also called jackknife procedure was used for performance evaluation. The accuracy to discriminate bacterial AR efflux from non-AR efflux was obtained as 85.81% (at tier-I) while accuracies for prediction of efflux pump families like ABC, MFS, RND and MATE family were found 92.13%, 85.39%, 91.01% and 99.44%, respectively (at tier-II). Benchmarking on an independent dataset also showed that BacEffluxPred had comparable accuracy for prediction of bacterial AR efflux pumps and their families. This is the first in-silico tool for predicting bacterial AR efflux proteins and their families and is freely available as both web-server and standalone versions at http://proteininformatics.org/mkumar/baceffluxpred/.
Collapse
|
49
|
Abstract
Efflux is an important mechanism in Gram-negative bacteria conferring multidrug resistance. Inhibition of efflux is an encouraging strategy to restore the antibacterial activity of antibiotics. Chlorpromazine and amitriptyline have been shown to behave as efflux inhibitors. However, their mode of action is poorly understood. Exposure of Salmonella enterica serovar Typhimurium and Escherichia coli to chlorpromazine selected for mutations within genes encoding RamR and MarR, regulators of the multidrug tripartite efflux pump AcrAB-TolC. Further experiments with S. Typhimurium containing AcrB D408A (a nonfunctional efflux pump) and chlorpromazine or amitriptyline resulted in the reversion of the mutant acrB allele to the wild type. Together, this suggests these drugs are AcrB efflux substrates. Subsequent docking studies with AcrB from S. Typhimurium and E. coli, followed by molecular dynamics simulations and free energy calculations showed that chlorpromazine and amitriptyline bind at the hydrophobic trap, a preferred binding site for substrates and inhibitors within the distal binding pocket of AcrB. Based on these simulations, we suggest that chlorpromazine and amitriptyline inhibit AcrB-mediated efflux by interfering with substrate binding. Our findings provide evidence that these drugs are substrates and inhibitors of AcrB, yielding molecular details of their mechanism of action and informing drug discovery of new efflux inhibitors.IMPORTANCE Efflux pumps of the resistance nodulation-cell division (RND) superfamily are major contributors to multidrug resistance for most of the Gram-negative ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) pathogens. The development of inhibitors of these pumps would be highly desirable; however, several issues have thus far hindered all efforts at designing new efflux inhibitory compounds devoid of adverse effects. An alternative route to de novo design relies on the use of marketed drugs, for which side effects on human health have been already assessed. In this work, we provide experimental evidence that the antipsychotic drugs chlorpromazine and amitriptyline are inhibitors of the AcrB transporter, the engine of the major RND efflux pumps in Escherichia coli and Salmonella enterica serovar Typhimurium. Furthermore, in silico calculations have provided a molecular-level picture of the inhibition mechanism, allowing rationalization of experimental data and paving the way for similar studies with other classes of marketed compounds.
Collapse
|
50
|
Cruz RMD, Zelli R, Benhsain S, Cruz RMD, Siqueira‐Júnior JP, Décout J, Mingeot‐Leclercq M, Mendonça‐Junior FJB. Synthesis and Evaluation of 2‐Aminothiophene Derivatives as
Staphylococcus aureus
Efflux Pump Inhibitors. ChemMedChem 2020; 15:716-725. [DOI: 10.1002/cmdc.201900688] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/17/2020] [Indexed: 11/10/2022]
Affiliation(s)
- Rayssa M. D. Cruz
- Department of Biological Sciences State University of Paraiba Laboratory of Synthesis and Drug Delivery João Pessoa PB Brazil
- Department of Pharmaceutical Sciences Federal University of Paraiba Post-Graduation Program in Natural and Synthetic Bioactive Products João Pessoa PB Brazil
- Département de Pharmacochimie Moléculaire University Grenoble Alpes CNRS Grenoble France
- Department of Pharmacologie Cellulaire et Moléculaire Université Catholique de Louvain, Louvain Drug Research Institute Brussels Belgium
| | - Renaud Zelli
- Département de Pharmacochimie Moléculaire University Grenoble Alpes CNRS Grenoble France
| | - Sarah Benhsain
- Department of Pharmacologie Cellulaire et Moléculaire Université Catholique de Louvain, Louvain Drug Research Institute Brussels Belgium
| | - Ryldene M. D. Cruz
- Department of Pharmaceutical Sciences Federal University of Paraiba Post-Graduation Program in Natural and Synthetic Bioactive Products João Pessoa PB Brazil
| | - José P. Siqueira‐Júnior
- Department of Molecular Biology Federal University of Paraiba Laboratory of Microorganism Genetics João Pessoa/PB Brazil
| | - Jean‐Luc Décout
- Département de Pharmacochimie Moléculaire University Grenoble Alpes CNRS Grenoble France
| | - Marie‐Paule Mingeot‐Leclercq
- Department of Pharmacologie Cellulaire et Moléculaire Université Catholique de Louvain, Louvain Drug Research Institute Brussels Belgium
| | - Francisco J. B. Mendonça‐Junior
- Department of Biological Sciences State University of Paraiba Laboratory of Synthesis and Drug Delivery João Pessoa PB Brazil
- Department of Pharmaceutical Sciences Federal University of Paraiba Post-Graduation Program in Natural and Synthetic Bioactive Products João Pessoa PB Brazil
| |
Collapse
|