1
|
Menezes Dantas DD, Macêdo NS, Sousa Silveira ZD, Santos Barbosa CRD, Muniz DF, Bezerra AH, Sousa JTD, Alencar GG, Morais Oliveira-Tintino CDD, Tintino SR, da Rocha MN, Marinho ES, Marinho MM, Dos Santos HS, Melo Coutinho HD, Cunha FABD. Naringenin as potentiator of norfloxacin efficacy through inhibition of the NorA efflux pump in Staphylococcus aureus. Microb Pathog 2025; 203:107504. [PMID: 40154849 DOI: 10.1016/j.micpath.2025.107504] [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: 10/07/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Bacterial resistance is a major challenge in the treatment of Staphylococcus aureus infections, with efflux mechanisms highlighted as reducing the efficacy of antibiotics. In this study, we investigated the potential of naringenin, a natural flavonoid, as an antibacterial agent and efflux pump inhibitor in S. aureus strains 1199 and 1199B. The studies used minimum inhibitory concentration (MIC) assays, ethidium bromide (EtBr) fluorescence emission enhancement assays, cell membrane permeability assays, and in silico molecular docking and ADME prediction assays. Naringenin showed no relevant antibacterial activity (MIC ≥1024 μg/mL). However, it potentiated the effect of norfloxacin and EtBr, reducing their MICs and increasing the fluorescence emission of EtBr, suggesting a possible inhibition of the NorA efflux pump. Bacterial membrane permeability was not significantly affected. Molecular docking assays indicated that naringenin interacts with the chlorpromazine binding site and has more favorable affinity energy than the chlorpromazine-NorA complex. ADME prediction showed favorable physicochemical properties, good oral absorption, metabolic stability and central nervous system safety. Therefore, naringenin demonstrates the potential to reverse the efficacy of norfloxacin in S. aureus by associating with efflux inhibition through effective interactions with the NorA protein, suggesting its therapeutic potential against bacterial resistance.
Collapse
Affiliation(s)
- Debora de Menezes Dantas
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil.
| | - Nair Silva Macêdo
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Zildene de Sousa Silveira
- Graduate Program in Biological Sciences- PPGCB, Federal University of Pernambuco - UFPE, Recife, Pernambuco, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Cristina Rodrigues Dos Santos Barbosa
- Postdoctoral Intern at the Semiarid Bioprospecting Laboratory (LABSEMA), Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Débora Feitosa Muniz
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Antônio Henrique Bezerra
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Josivânia Teixeira de Sousa
- Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Gabriel Gonçalves Alencar
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil
| | - Cícera Datiane de Morais Oliveira-Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil; Postdoctoral Intern at the Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil; Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Matheus Nunes da Rocha
- Ceará State University, Postgraduate Program in Natural Sciences, Laboratory of Chemistry of Natural Products, Fortaleza, CE, Brazil
| | - Emmanuel Silva Marinho
- Ceará State University, Postgraduate Program in Natural Sciences, Laboratory of Chemistry of Natural Products, Fortaleza, CE, Brazil
| | | | - Hélcio Silva Dos Santos
- Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Henrique Douglas Melo Coutinho
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil; Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Francisco Assis Bezerra da Cunha
- Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil; Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| |
Collapse
|
2
|
Wu Z, Lei X, Zhang Y, Wu S, Hou Z, Ma K, Pei H, Shang F, Xue T. The membrane protein DtpT plays an important role in biofilm formation and stress resistance in foodborne Staphylococcus aureus RMSA49. Food Res Int 2025; 208:116249. [PMID: 40263806 DOI: 10.1016/j.foodres.2025.116249] [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: 12/17/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
Staphylococcus aureus has been a major contributor to the contamination of dairy products and preserved foods due to its capacity for biofilm formation and strong resistance to environmental stress. The membrane transport protein di-and tripeptides transporter (DtpT) is the primary transporter of di- and tripeptides in S. aureus, yet its impact on biofilm formation and stress resistance in S. aureus has not been previously reported. Our study focused on the foodborne S. aureus strain RMSA49, revealing that mutation of the dtpT resulted in diminished biofilm formation ability and reduced tolerance to environmental stress (high temperature, dryness, oxidative stress, and salt stress). These findings highlight the significance of DtpT in both biofilm formation and response to environmental stress in foodborne S. aureus. Our study represents the first report demonstrating the crucial role of DtpT in biofilm formation and environmental tolerance in S. aureus, providing new avenues for future research on this protein while also identifying potential target genes for further investigation into S. aureus tolerance mechanisms during food processing and control of biofilm formation.
Collapse
Affiliation(s)
- Ziheng Wu
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xiaolu Lei
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yunying Zhang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Siyao Wu
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zhiyuan Hou
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Kai Ma
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Hao Pei
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| | - Fei Shang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| | - Ting Xue
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
3
|
de Sousa Silveira Z, Macêdo NS, Menezes Dantas DD, Rodrigues Dos Santos Barbosa C, Muniz DF, Morais Oliveira-Tintino CDD, Relison Tintino S, Alencar GG, Marinho ES, Rocha MND, Marinho MM, Santos HSD, Coutinho HDM, Cunha FABD, Silva MVD. Evaluation of the antibacterial and inhibitory activity of the NorA and TetK efflux pumps of Staphylococcus aureus by p-coumaric acid. Microb Pathog 2025; 200:107318. [PMID: 39848298 DOI: 10.1016/j.micpath.2025.107318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/25/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
The NorA and TetK efflux pumps mediate resistance to fluoroquinolone and tetracycline antibiotics by actively extruding these compounds and reducing their intracellular concentrations. Consequently, intense research has focused on inhibiting these efflux mechanisms using antimicrobial agents derived from natural or synthetic sources. This study used Fourier transform infrared spectroscopy (ATR-FTIR) to analyze the various functional groups present in p-coumaric acid. We also investigated the antibacterial activity of p-coumaric acid on strains of Staphylococcus aureus carrying the NorA and TetK efflux pumps, as well as the compound's ability to increase the fluorescence of ethidium bromide (EtBr) and Sytox Green. In addition, the interactions of this compound with NorA were analyzed using molecular docking, and its pharmacokinetic properties were evaluated using ADMET (Absorption, Distribution, Metabolism, Excretion and Toxicity) modeling. The results revealed that p-coumaric acid exhibited no direct antibacterial activity against the tested Staphylococcus aureus strains. However, significant reductions in the minimum inhibitory concentrations (MICs) of norfloxacin and EtBr, both used as NorA substrates, were observed when combined with p-coumaric acid. It was also observed that p-coumaric acid increased the fluorescence emission of EtBr and Sytox Green in strains 1199 and 1199B, suggesting the inhibition of the efflux mechanism and enhanced membrane permeabilization in S. aureus. The in silico analysis demonstrated that p-coumaric acid exhibits a favorable binding energy with NorA, comparable to that of chlorpromazine. These results position p-coumaric acid as a promising antibiotic adjuvant and efflux inhibitor in strains harboring NorA.
Collapse
Affiliation(s)
- Zildene de Sousa Silveira
- Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), 50670-901, Recife, PE, Brazil.
| | - Nair Silva Macêdo
- Postgraduate Program in Biological Chemistry (PPQB), Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil.
| | - Débora de Menezes Dantas
- Postgraduate Program in Biological Chemistry (PPQB), Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil.
| | | | - Débora Feitosa Muniz
- Postgraduate Program in Biological Chemistry (PPQB), Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil
| | | | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Cariri Regional University (URCA), Crato, CE, Brazil.
| | - Gabriel Gonçalves Alencar
- Laboratory of Microbiology and Molecular Biology (LMBM), Cariri Regional University (URCA), Crato, CE, Brazil.
| | - Emmanuel Silva Marinho
- State University of Ceará, Postgraduate Program in Natural Sciences, Natural Resources Bioprospecting and Monitoring Laboratory (LBMRN), Fortaleza, Ceará, Brazil.
| | - Matheus Nunes da Rocha
- State University of Ceará, Postgraduate Program in Natural Sciences, Natural Resources Bioprospecting and Monitoring Laboratory (LBMRN), Fortaleza, Ceará, Brazil.
| | - Marcia Machado Marinho
- State University of Ceará, Postgraduate Program in Natural Sciences, Natural Resources Bioprospecting and Monitoring Laboratory (LBMRN), Fortaleza, Ceará, Brazil.
| | - Hélcio Silva Dos Santos
- Northeast Biotechnology Network (RENORBIO-Nucleadora UECE), State University of Acaraú Valley (UVA), Sobral, CE, Brazil.
| | - Henrique Douglas Melo Coutinho
- Postgraduate Program in Biological Chemistry (PPQB), Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil.
| | | | - Márcia Vanusa da Silva
- Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), 50670-901, Recife, PE, Brazil.
| |
Collapse
|
4
|
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
|
5
|
Salikin NH, Keong LC, Azemin WA, Philip N, Yusuf N, Daud SA, Rashid SA. Combating multidrug-resistant (MDR) Staphylococcus aureus infection using terpene and its derivative. World J Microbiol Biotechnol 2024; 40:402. [PMID: 39627623 DOI: 10.1007/s11274-024-04190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024]
Abstract
Multidrug-resistant (MDR) Staphylococcus aureus represents a major global health issue resulting in a wide range of debilitating infections and fatalities. The slow progression of new antibiotics, limited choices for treatment, and scarcity of new drug approvals create immense obstacles in new drug line development. S. aureus poses a significant public health risk, due to the emergence of methicillin-resistant (MRSA) and vancomycin-resistant strains (VRSA), necessitating novel antibiotics for effective control management. Current studies are delving into the terpenes' potential as an antimicrobial agent, indicating positive prospects as promising substitutes or complementary to conventional antibiotics. Concurrent reactions of terpenes with conventional antibiotics create synergistic effects that significantly enhance antibiotic efficacy. Accumulated evidence has shown that while efflux pump (e.g., NorA, TetK, and MepA) is revealed as an essential defense of S. aureus against antibiotics, terpene and its derivative act as its potent inhibitor, suggesting the promising potential of terpenes in combating those infectious pathogens. Furthermore, pronounced cell membrane disruptive activity and antibiofilm properties by terpenes have been exerted, signifying their significance as promising prevention against microbial pathogenesis and antimicrobial resistance. This review provides an overview of the potential of terpenes and their derivatives in combating S. aureus infections, highlighting their potential mechanisms of action (MOA), synergistic effects with conventional antibiotics, and challenges in clinical translation. The unique properties of terpenes offer an opportunity for their use in developing an exceptional defense strategy against antibiotic-resistant S. aureus.
Collapse
Affiliation(s)
- Nor Hawani Salikin
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Lee Chee Keong
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Wan-Atirah Azemin
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Noraini Philip
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Nurhaida Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Syiah Kuala University, Aceh, Indonesia
| | - Siti Aisyah Daud
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Syarifah Ab Rashid
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia.
| |
Collapse
|
6
|
Zhang L, Liu Y, Liu S, Wang T, Ouyang F, Pei Z, Ren Y, Shuai Q. NIR-intervened thermally accelerated urease-propelled MOF nanosubmarine for antibiotic-free antibacterial inhibition via single-wavelength synergistic PDT/PTT. Int J Biol Macromol 2024; 282:137367. [PMID: 39515705 DOI: 10.1016/j.ijbiomac.2024.137367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
The increasing resistance of bacteria to antibiotics poses a serious threat to global human health. Herein, we have developed a thermal-accelerated biomacromolecular urease-driven MOF-based nanomotor (ZIF-8@PDA@ICG@Ur) mediated via NIR-intervened antimicrobial therapy. In this system, we have attempted for the first time to introduce an easy-to-operate light combination therapy strategy (only one light source is required) into an enzyme-driven MOF motor system to achieve antibiotic-free antibacterial therapy. The purpose of this research is to utilize the increased thermal energy from the photothermal effect to enhance the activity of urease, boost the driving force, which further increases the contact efficiency of the nanocarrier with the bacteria to enhance the bacterial killing power. In 50 mM urea solution, the rate of nanomotor movement was increased by about 1.24 times under NIR light irradiation compared to that without light. In addition, the synergistic effect of enhanced autonomous movement, NIR-controlled on-demand phototherapy and zinc ions release achieved 99.99 % antibacterial activity without antibiotics. The developed NIR-intervened design in this manuscript endows urease-based nanomotors with attractive propulsion feature and contributes to a novel strategy for against diseases caused by bacterial infections.
Collapse
Affiliation(s)
- Li Zhang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yu Liu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Shupeng Liu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Tao Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Feng Ouyang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhichao Pei
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Yixia Ren
- College of Chemistry and Chemical Engineering, Yan'an University, Yan'an 716000, PR China.
| | - Qi Shuai
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
7
|
Benzenine D, Daoud I, Aissaoui N, Kibou Z, Seijas JA, Vázquez-Tato MP, Ziani-Cherif C, Belarbi L, Choukchou-Braham N. Design, Synthesis, Molecular Docking, and ADME-Tox Investigations of Imidazo[1,2-a]Pyrimidines Derivatives as Antimicrobial Agents. Molecules 2024; 29:5058. [PMID: 39519699 PMCID: PMC11547262 DOI: 10.3390/molecules29215058] [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: 08/20/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
A convenient and effective synthesis of imidazo[1,2-a]pyrimidine derivatives has been developed under microwave irradiations using Al2O3 as a catalyst in solvent-free conditions. The functionalized imidazo[1,2-a]pyrimidine derivatives are useful in biochemistry and medical science. In our investigation, the antimicrobial activity of the synthesized compounds was evaluated against 13 microorganisms, including 6 Gram-positive bacteria, 4 Gram-negative bacteria, and 3 pathogenic fungi. Bioactivity tests revealed that the majority of the compounds exhibited good antimicrobial activity. Finally, molecular docking simulations and ADME-T predictions were performed, showing that the most active compounds have good binding modes with microbial targets and promising pharmacokinetic safety profiles.
Collapse
Affiliation(s)
- Djamila Benzenine
- Laboratoire de Catalyse et Synthèse en Chimie Organique, Faculté des Sciences, Université de Tlemcen, BP 119, Tlemcen 13000, Algeria; (D.B.)
- Faculté des Sciences et de la Technologie, Université de Ain Témouchent, BP 284, Ain Témouchent 46000, Algeria
| | - Ismail Daoud
- Department of Matter Sciences, University Mohamed Khider, BP 145 RP, Biskra 07000, Algeria;
- Laboratory of Natural and Bioactive Substances, University of Abou-Bakr Belkaid, BP 119, Tlemcen 13000, Algeria
| | - Nadia Aissaoui
- Laboratory of the Sustainable Management of Natural Resources in Arid and Semi-Arid Areas, Institute of Sciences, University Center of Naama, BP 66, Naama 45000, Algeria;
| | - Zahira Kibou
- Laboratoire de Catalyse et Synthèse en Chimie Organique, Faculté des Sciences, Université de Tlemcen, BP 119, Tlemcen 13000, Algeria; (D.B.)
- Faculté des Sciences et de la Technologie, Université de Ain Témouchent, BP 284, Ain Témouchent 46000, Algeria
| | - Julio A. Seijas
- Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Santiago de Compostela, Avda. Alfonso X El Sabio s/n, 27002 Lugo, Spain;
| | - M. Pilar Vázquez-Tato
- Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Santiago de Compostela, Avda. Alfonso X El Sabio s/n, 27002 Lugo, Spain;
| | - Chewki Ziani-Cherif
- Laboratoire de Catalyse et Synthèse en Chimie Organique, Faculté des Sciences, Université de Tlemcen, BP 119, Tlemcen 13000, Algeria; (D.B.)
| | - Lahcen Belarbi
- Faculté des Sciences et de la Technologie, Université de Ain Témouchent, BP 284, Ain Témouchent 46000, Algeria
- Laboratorie de Chimie Appliquée, Université de Ain Témouchent, BP 284, Ain Témouchent 46000, Algeria
| | - Noureddine Choukchou-Braham
- Laboratoire de Catalyse et Synthèse en Chimie Organique, Faculté des Sciences, Université de Tlemcen, BP 119, Tlemcen 13000, Algeria; (D.B.)
| |
Collapse
|
8
|
Judan Cruz KG, Takumi O, Bongulto KA, Gandalera EE, Kagia N, Watanabe K. Natural compound-induced downregulation of antimicrobial resistance and biofilm-linked genes in wastewater Aeromonas species. Front Cell Infect Microbiol 2024; 14:1456700. [PMID: 39469451 PMCID: PMC11513397 DOI: 10.3389/fcimb.2024.1456700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024] Open
Abstract
Addressing the global antimicrobial resistance (AMR) crisis requires a multifaceted innovative approach to mitigate impacts on public health, healthcare and economic systems. In the complex evolution of AMR, biofilms and the acquisition of antimicrobial resistance genes (ARGs) play a pivotal role. Aeromonas is a major AMR player that often forms biofilm, harbors ARGs and is frequently detected in wastewater. Existing wastewater treatment plants (WWTPs) do not have the capacity to totally eliminate antimicrobial-resistant bacteria favoring the evolution of ARGs in wastewater. Besides facilitating the emergence of AMR, biofilms contribute significantly to biofouling process within the activated sludge of WWTP bioreactors. This paper presents the inhibition of biofilm formation, the expression of biofilm-linked genes and ARGs by phytochemicals andrographolide, docosanol, lanosterol, quercetin, rutin and thymohydroquinone. Aeromonas species were isolated and purified from activated sludge samples. The ARGs were detected in the isolated Aeromonas species through PCR. Aeromonas biofilms were quantified following the application of biocompounds through the microtiter plate assay. qPCR analyses of related genes were done for confirmation. Findings showed that the natural compounds inhibited the formation of biofilms and reduced the expression of genes linked to biofilm production as well as ARGs in wastewater Aeromonas. This indicates the efficacy of these compounds in targeting and controlling both ARGs and biofilm formation, highlighting their potential as innovative solutions for combating antimicrobial resistance and biofouling.
Collapse
Affiliation(s)
- Khristina G. Judan Cruz
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime, Japan
- Department of Biological Sciences, College of Science, Central Luzon State University, Science City of Muñoz, Nueva Ecija, Philippines
| | - Okamoto Takumi
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime, Japan
| | - Kenneth A. Bongulto
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime, Japan
| | - Emmanuel E. Gandalera
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime, Japan
- Department of Biological Sciences, College of Science, Central Luzon State University, Science City of Muñoz, Nueva Ecija, Philippines
| | - Ngure Kagia
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime, Japan
| | - Kozo Watanabe
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, Ehime, Japan
| |
Collapse
|
9
|
Brito GS, Dutra RP, Fernandes Pereira AL, Ferreira AGN, Neto MS, Holanda CA, Fidelis QC. Nanoemulsions of essential oils against multi-resistant microorganisms: An integrative review. Microb Pathog 2024; 195:106837. [PMID: 39103128 DOI: 10.1016/j.micpath.2024.106837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024]
Abstract
Microbial resistance to drugs continues to be a global public health issue that demands substantial investment in research and development of new antimicrobial agents. Essential oils (EO) have demonstrated satisfactory and safe antimicrobial action, being used in pharmaceutical, cosmetic, and food formulations. In order to improve solubility, availability, and biological action, EO have been converted into nanoemulsions (NE). This review identified scientific evidence corroborating the antimicrobial action of nanoemulsions of essential oils (NEEO) against antibiotic-resistant pathogens. Using integrative review methodology, eleven scientific articles evaluating the antibacterial or antifungal assessment of NEEO were selected. The synthesis of evidence indicates that NEEO are effective in combating multidrug-resistant microorganisms and in the formation of their biofilms. Factors such as NE droplet size, chemical composition of essential oils, and the association of NE with antibiotics are discussed. Furthermore, NEEO showed satisfactory results in vitro and in vivo evaluations against resistant clinical isolates, making them promising for the development of new antimicrobial and antivirulence drugs.
Collapse
Affiliation(s)
- Gabriel Sousa Brito
- Program in Health and Technology, Imperatriz Science Center, Federal University of Maranhão, Imperatriz, 65915-240, Brazil
| | - Richard Pereira Dutra
- Program in Health and Technology, Imperatriz Science Center, Federal University of Maranhão, Imperatriz, 65915-240, Brazil
| | - Ana Lúcia Fernandes Pereira
- Program in Health and Technology, Imperatriz Science Center, Federal University of Maranhão, Imperatriz, 65915-240, Brazil
| | | | - Marcelino Santos Neto
- Program in Health and Technology, Imperatriz Science Center, Federal University of Maranhão, Imperatriz, 65915-240, Brazil
| | - Carlos Alexandre Holanda
- Program in Health and Technology, Imperatriz Science Center, Federal University of Maranhão, Imperatriz, 65915-240, Brazil
| | - Queli Cristina Fidelis
- Program in Health and Technology, Imperatriz Science Center, Federal University of Maranhão, Imperatriz, 65915-240, Brazil.
| |
Collapse
|
10
|
La Monica G, Bono A, Alamia F, Lauria A, Martorana A. Bioisosteric heterocyclic analogues of natural bioactive flavonoids by scaffold-hopping approaches: State-of-the-art and perspectives in medicinal chemistry. Bioorg Med Chem 2024; 109:117791. [PMID: 38870715 DOI: 10.1016/j.bmc.2024.117791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/13/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
The flavonoid family is a set of well-known bioactive natural molecules, with a wide range of potential therapeutic applications. Despite the promising results obtained in preliminary in vitro/vivo studies, their pharmacokinetic and pharmacodynamic profiles are severely compromised by chemical instability. To address this issue, the scaffold-hopping approach is a promising strategy for the structural optimization of natural leads to discover more potent analogues. In this scenario, this Perspective provides a critical analysis on how the replacement of the chromon-4-one flavonoid core with other bioisosteric nitrogen/sulphur heterocycles might affect the chemical, pharmaceutical and biological properties of the resulting new chemical entities. The investigated derivatives were classified on the basis of their biological activity and potential therapeutic indications. For each session, the target(s), the specific mechanism of action, if available, and the key pharmacophoric moieties were highlighted, as revealed by X-ray crystal structures and in silico structure-based studies. Biological activity data, in vitro/vivo studies, were examined: a particular focus was given on the improvements observed with the new heterocyclic analogues compared to the natural flavonoids. This overview of the scaffold-hopping advantages in flavonoid compounds is of great interest to the medicinal chemistry community to better exploit the vast potential of these natural molecules and to identify new bioactive molecules.
Collapse
Affiliation(s)
- Gabriele La Monica
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy
| | - Alessia Bono
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy
| | - Federica Alamia
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy
| | - Antonino Lauria
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy
| | - Annamaria Martorana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, University of Palermo, Viale delle Scienze, Ed. 17, I-90128 Palermo, Italy.
| |
Collapse
|
11
|
Martin ALAR, Pereira RLS, Rocha JE, Farias PAM, Freitas TS, Caldas FRDL, Figueredo FG, Sampaio NFL, Oliveira-Tintino CDDM, Tintino SR, da Hora GCA, Lima MCP, de Menezes IRA, Carvalho DT, Coutinho HDM, Fonteles MMF. Unlocking bacterial defense: Exploring the potent inhibition of NorA efflux pump by coumarin derivatives in Staphylococcus aureus. Microb Pathog 2024; 190:106608. [PMID: 38503396 DOI: 10.1016/j.micpath.2024.106608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/27/2024] [Accepted: 03/03/2024] [Indexed: 03/21/2024]
Abstract
The occurrence of bacterial resistance has been increasing, compromising the treatment of various infections. The high virulence of Staphylococcus aureus allows for the maintenance of the infectious process, causing many deaths and hospitalizations. The MepA and NorA efflux pumps are transporter proteins responsible for expelling antimicrobial agents such as fluoroquinolones from the bacterial cell. Coumarins are phenolic compounds that have been studied for their diverse biological actions, including against bacteria. A pharmacokinetic in silico characterization of compounds C10, C11, C13, and C14 was carried out according to the principles of Lipinski's Rule of Five, in addition to searching for similarity in ChemBL and subsequent search for publications in CAS SciFinder. All compounds were evaluated for their in vitro antibacterial and modulatory activity against standard and multidrug-resistant Gram-positive and Gram-negative strains. The effect of coumarins C9, C10, C11, C13, and C14 as efflux pump inhibitors in Staphylococcus aureus strains was evaluated using the microdilution method (MepA or NorA) and fluorimetry (NorA). The behavior of coumarins regarding the efflux pump was determined from their interaction properties with the membrane and coumarin-protein using molecular docking and molecular dynamics simulations. Only the isolated coumarin compound C13 showed antibacterial activity against standard strains of Staphylococcus aureus and Escherichia coli. However, the other tested coumarins showed modulatory capacity for fluoroquinolone and aminoglycoside antibacterials. Compounds C10, C13, and C14 were effective in reducing the MIC of both antibiotics for both multidrug-resistant strains, while C11 potentiated the effect of norfloxacin and gentamicin for Gram-positive and Gram-negative bacteria and only norfloxacin for Gram-negative. Only coumarin C14 produced synergistic effects when associated with ciprofloxacin in MepA-carrying strains. All tested coumarins have the ability to inhibit the NorA efflux pump present in Staphylococcus aureus, both in reducing the MIC and inducing increased ethidium bromide fluorescence emission in fluorimetry. The findings of this study offer an atomistic perspective on the potential of coumarins as active inhibitors of the NorA pump, highlighting their specific mode of action mainly targeting protein inhibition. In molecular docking, it was observed that coumarins are capable of interacting with various amino acid residues of the NorA pump. The simulation showed that coumarin C10 can cross the bilayer; however, the other coumarins interacted with the membrane but were unable to cross it. Coumarins demonstrated their potentiating role in the effect of norfloxacin through a dual mechanism: efflux pump inhibition through direct interaction with the protein (C9, C10, C11, and C13) and increased interaction with the membrane (C10 and C13). In the context of pharmacokinetic prediction studies, the studied structures have a suitable chemical profile for possible oral use. We suggest that coumarin derivatives may be an interesting alternative in the future for the treatment of resistant bacterial infections, with the possibility of a synergistic effect with other antibacterials, although further studies are needed to characterize their therapeutic effects and toxicity.
Collapse
Affiliation(s)
- Ana Luíza A R Martin
- Department of Physiology and Pharmacology, Federal University of Ceará - UFC, 60430-160, Fortaleza, Brazil; Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil; School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil
| | | | - Janaína Esmeraldo Rocha
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | - Pablo A M Farias
- School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil; CECAPE College, 63024-015, Juazeiro do Norte, Brazil
| | - Thiago S Freitas
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | | | - Fernando G Figueredo
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil; School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil
| | - Nadghia Figueiredo Leite Sampaio
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil; School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil
| | | | - Saulo Relison Tintino
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | | | | | - Irwin Rose A de Menezes
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | - Diogo T Carvalho
- School of Pharmacy, Federal University of Alfenas - UNIFAL, 37130-001, Alfenas, Brazil
| | - Henrique D M Coutinho
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil.
| | - Marta M F Fonteles
- Department of Physiology and Pharmacology, Federal University of Ceará - UFC, 60430-160, Fortaleza, Brazil
| |
Collapse
|
12
|
Adhikrao PA, Motiram GM, Kumar G. Tackling Nontuberculous Mycobacteria by Repurposable Drugs and Potential Leads from Natural Products. Curr Top Med Chem 2024; 24:1291-1326. [PMID: 38288807 DOI: 10.2174/0115680266276938240108060247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 07/25/2024]
Abstract
Nontuberculous Mycobacteria (NTM) refer to bacteria other than all Mycobacterium species that do not cause tuberculosis or leprosy, excluding the species of the Mycobacterium tuberculosis complex, M. leprae and M. lepromatosis. NTM are ubiquitous and present in soils and natural waters. NTM can survive in a wide range of environmental conditions. The direct inoculum of the NTM from water or other materials is most likely a source of infections. NTMs are responsible for several illnesses, including pulmonary alveolar proteinosis, cystic fibrosis, bronchiectasis, chronic obstructive pneumoconiosis, and pulmonary disease. Recent reports suggest that NTM species have become insensitive to sterilizing agents, antiseptics, and disinfectants. The efficacy of existing anti-NTM regimens is diminishing and has been compromised due to drug resistance. New and recurring cases of multidrug-resistant NTM strains are increasing. Thus, there is an urgent need for ant-NTM regimens with novel modes of action. This review sheds light on the mode of antimicrobial resistance in the NTM species. Then, we discussed the repurposable drugs (antibiotics) that have shown new indications (activity against NTM strains) that could be developed for treating NTM infections. Also, we have summarised recently identified natural leads acting against NTM, which have the potential for treating NTM-associated infections.
Collapse
Affiliation(s)
- Patil Amruta Adhikrao
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| | - Gudle Mayuri Motiram
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| | - Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| |
Collapse
|
13
|
de Araújo-Neto JB, Oliveira-Tintino CDDM, de Araújo GA, Alves DS, Ribeiro FR, Brancaglion GA, Carvalho DT, Lima CMG, Mohammed Ali HSH, Rather IA, Wani MY, Emran TB, Coutinho HDM, Balbino VDQ, Tintino SR. 3-Substituted Coumarins Inhibit NorA and MepA Efflux Pumps of Staphylococcus aureus. Antibiotics (Basel) 2023; 12:1739. [PMID: 38136773 PMCID: PMC10741188 DOI: 10.3390/antibiotics12121739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Coumarins are compounds with scientifically proven antibacterial properties, and modifications to the chemical structure are known to improve their effects. This information is even more relevant with the unbridled advances of antibiotic resistance, where Staphylococcus aureus and its efflux pumps play a prominent role. The study's objective was to evaluate the potential of synthetic coumarins with different substitutions in the C-3 position as possible inhibitors of the NorA and MepA efflux pumps of S. aureus. For this evaluation, the following steps took place: (i) the determination of the minimum inhibitory concentration (MIC); (ii) the association of coumarins with fluoroquinolones and ethidium bromide (EtBr); (iii) the assessment of the effect on EtBr fluorescence emission; (iv) molecular docking; and (v) an analysis of the effect on membrane permeability. Coumarins reduced the MICs of fluoroquinolones and EtBr between 50% and 87.5%. Coumarin C1 increased EtBr fluorescence emission between 20 and 40% by reinforcing the evidence of efflux inhibition. The molecular docking results demonstrated that coumarins have an affinity with efflux pumps and establish mainly hydrogen bonds and hydrophobic interactions. Furthermore, C1 did not change the permeability of the membrane. Therefore, we conclude that these 3-substituted coumarins act as inhibitors of the NorA and MepA efflux pumps of S. aureus.
Collapse
Affiliation(s)
- José B. de Araújo-Neto
- Postgraduate Program in Biological Sciences, Biosciences Center, Federal University of Pernambuco, Recife 50740-570, PE, Brazil; (J.B.d.A.-N.); (V.d.Q.B.)
| | - Cícera D. de M. Oliveira-Tintino
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, Crato 63105-000, CE, Brazil; (C.D.d.M.O.-T.); (G.A.d.A.); (D.S.A.); (S.R.T.)
| | - Gildênia A. de Araújo
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, Crato 63105-000, CE, Brazil; (C.D.d.M.O.-T.); (G.A.d.A.); (D.S.A.); (S.R.T.)
| | - Daniel S. Alves
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, Crato 63105-000, CE, Brazil; (C.D.d.M.O.-T.); (G.A.d.A.); (D.S.A.); (S.R.T.)
| | - Fernanda R. Ribeiro
- Pharmaceutical Chemistry Research Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil; (F.R.R.); (G.A.B.); (D.T.C.)
| | - Guilherme A. Brancaglion
- Pharmaceutical Chemistry Research Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil; (F.R.R.); (G.A.B.); (D.T.C.)
| | - Diogo T. Carvalho
- Pharmaceutical Chemistry Research Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil; (F.R.R.); (G.A.B.); (D.T.C.)
| | | | - Hani S. H. Mohammed Ali
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (H.S.H.M.A.); (I.A.R.)
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (H.S.H.M.A.); (I.A.R.)
| | - Mohmmad Y. Wani
- Department of Chemistry, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia;
| | - Talha B. Emran
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA;
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Henrique D. M. Coutinho
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, Crato 63105-000, CE, Brazil; (C.D.d.M.O.-T.); (G.A.d.A.); (D.S.A.); (S.R.T.)
| | - Valdir de Q. Balbino
- Postgraduate Program in Biological Sciences, Biosciences Center, Federal University of Pernambuco, Recife 50740-570, PE, Brazil; (J.B.d.A.-N.); (V.d.Q.B.)
| | - Saulo R. Tintino
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, Crato 63105-000, CE, Brazil; (C.D.d.M.O.-T.); (G.A.d.A.); (D.S.A.); (S.R.T.)
| |
Collapse
|
14
|
Patel MB, Spikes H, Bailey RS, Connell T, Gill H, Gokel MR, Harris R, Meisel JW, Negin S, Yin SA, Gokel GW. Antimicrobial and Adjuvant Potencies of Di- n-alkyl Substituted Diazalariat Ethers. Antibiotics (Basel) 2023; 12:1513. [PMID: 37887214 PMCID: PMC10603992 DOI: 10.3390/antibiotics12101513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
Lariat ethers are macrocyclic polyethers-crown ethers-to which sidearms are appended. 4,13-Diaza-18-crown-6 having twin alkyl chains at the nitrogens show biological activity. They exhibit antibiotic activity, but when co-administered at with an FDA-approved antibiotic, the latter's potency is often strongly enhanced. Potency enhancements and resistance reversals have been documented in vitro for a range of Gram-negative and Gram-positive bacteria with a variety of antimicrobials. Strains of E. coli and Staphylococcus aureus having resistance to a range of drugs have been studied and the potency enhancements (checkerboards) are reported here. Drugs included in the present study are ampicillin, cefepime, chlortetracycline, ciprofloxacin, doxycycline, kanamycin, minocycline, norfloxacin, oxycycline, penicillin G, and tetracycline. Enhancements of norfloxacin potency against S. aureus 1199B of up to 128-fold were observed. The properties of these lariat ethers have been studied to determine solubility, their membrane penetration, cytotoxicity and mammalian cell survival, and their effect on bacterial efflux pumps. It is shown that in some cases, the lariat ethers have complex antimicrobials with considerable selectivity. Based on these observations, including 1:1 complexation between lariat ethers and antimicrobials and the cytotoxicity of the MeI salts showing a separation index of 32-fold, they hold significant potential for further development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - George W. Gokel
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, MO 63121, USA
| |
Collapse
|
15
|
Xie P, Gao Y, Wu C, Li X, Yang Y. The inhibitory mechanism of echinacoside against Staphylococcus aureus Ser/Thr phosphatase Stp1 by virtual screening and molecular modeling. J Mol Model 2023; 29:320. [PMID: 37725157 DOI: 10.1007/s00894-023-05723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/09/2023] [Indexed: 09/21/2023]
Abstract
CONTEXT Stp1 is a new potential target closely related to the pathogenicity of Staphylococcus aureus (S. aureus). In this study, effective Stp1 inhibitors were screened via virtual screening and enzyme activity experiments, and the inhibition mechanism was analyzed using molecular dynamics simulation. METHODS AutoDock Vina 4.0 software was used for virtual screening. The molecular structures of Stp1 and ligands were obtained from the RCSB Protein Data Bank and Zinc database, respectively. The molecular dynamics simulation used the Gromacs 4.5.5 software package with the Amberff99sb force field and TIP3P water model. AutoDock Tools was used to add polar hydrogen atoms to Stp1 and distribute part of the charge generated by Kollman's combined atoms. The binding free energies were calculated using the Amber 10 package. RESULTS The theoretical calculation results are consistent with the experimental results. We found that echinacoside (ECH) substantially inhibits the hydrolytic activity of Stp1. ECH competes with the substrate by binding to the active center of Stp1, resulting in a decrease in Stp1 activity. In addition, Met39, Gly41, Asp120, Asn162, and Ile163 were identified to play key roles in the binding of Stp1 to ECH. The benzene ring of ECH also plays an important role in complex binding. These findings provide a robust foundation for the development of innovative anti-infection drugs.
Collapse
Affiliation(s)
- Peng Xie
- Faculty of Food Science and Technology, Suzhou Polytechnic Institute of Agriculture, Suzhou, 215008, China
| | - Yue Gao
- Faculty of Food Science and Technology, Suzhou Polytechnic Institute of Agriculture, Suzhou, 215008, China
| | - Chenqi Wu
- Faculty of Food Science and Technology, Suzhou Polytechnic Institute of Agriculture, Suzhou, 215008, China
| | - Xuenan Li
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, Jilin, China
| | - Yanan Yang
- Faculty of Food Science and Technology, Suzhou Polytechnic Institute of Agriculture, Suzhou, 215008, China.
| |
Collapse
|
16
|
De Gaetano GV, Lentini G, Famà A, Coppolino F, Beninati C. Antimicrobial Resistance: Two-Component Regulatory Systems and Multidrug Efflux Pumps. Antibiotics (Basel) 2023; 12:965. [PMID: 37370284 DOI: 10.3390/antibiotics12060965] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The number of multidrug-resistant bacteria is rapidly spreading worldwide. Among the various mechanisms determining resistance to antimicrobial agents, multidrug efflux pumps play a noteworthy role because they export extraneous and noxious substrates from the inside to the outside environment of the bacterial cell contributing to multidrug resistance (MDR) and, consequently, to the failure of anti-infective therapies. The expression of multidrug efflux pumps can be under the control of transcriptional regulators and two-component systems (TCS). TCS are a major mechanism by which microorganisms sense and reply to external and/or intramembrane stimuli by coordinating the expression of genes involved not only in pathogenic pathways but also in antibiotic resistance. In this review, we describe the influence of TCS on multidrug efflux pump expression and activity in some Gram-negative and Gram-positive bacteria. Taking into account the strict correlation between TCS and multidrug efflux pumps, the development of drugs targeting TCS, alone or together with already discovered efflux pump inhibitors, may represent a beneficial strategy to contribute to the fight against growing antibiotic resistance.
Collapse
Affiliation(s)
| | - Germana Lentini
- Department of Human Pathology, University of Messina, 98124 Messina, Italy
| | - Agata Famà
- Department of Human Pathology, University of Messina, 98124 Messina, Italy
| | - Francesco Coppolino
- Department of Biomedical, Dental and Imaging Sciences, University of Messina, 98124 Messina, Italy
| | - Concetta Beninati
- Department of Human Pathology, University of Messina, 98124 Messina, Italy
- Scylla Biotech Srl, 98124 Messina, Italy
| |
Collapse
|
17
|
Varela MF, Stephen J, Bharti D, Lekshmi M, Kumar S. Inhibition of Multidrug Efflux Pumps Belonging to the Major Facilitator Superfamily in Bacterial Pathogens. Biomedicines 2023; 11:1448. [PMID: 37239119 PMCID: PMC10216197 DOI: 10.3390/biomedicines11051448] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Bacterial pathogens resistant to multiple structurally distinct antimicrobial agents are causative agents of infectious disease, and they thus constitute a serious concern for public health. Of the various bacterial mechanisms for antimicrobial resistance, active efflux is a well-known system that extrudes clinically relevant antimicrobial agents, rendering specific pathogens recalcitrant to the growth-inhibitory effects of multiple drugs. In particular, multidrug efflux pump members of the major facilitator superfamily constitute central resistance systems in bacterial pathogens. This review article addresses the recent efforts to modulate these antimicrobial efflux transporters from a molecular perspective. Such investigations can potentially restore the clinical efficacy of infectious disease chemotherapy.
Collapse
Affiliation(s)
- Manuel F. Varela
- Department of Biology, Eastern New Mexico University, Station 33, Portales, NM 88130, USA
| | - Jerusha Stephen
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India; (J.S.); (D.B.); (M.L.); (S.K.)
| | - Deeksha Bharti
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India; (J.S.); (D.B.); (M.L.); (S.K.)
| | - Manjusha Lekshmi
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India; (J.S.); (D.B.); (M.L.); (S.K.)
| | - Sanath Kumar
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India; (J.S.); (D.B.); (M.L.); (S.K.)
| |
Collapse
|