1
|
Ellward GL, Binda ME, Dzurny DI, Bucher MJ, Dees WR, Czyż DM. A Screen of Traditional Chinese Medicinal Plant Extracts Reveals 17 Species with Antimicrobial Properties. Antibiotics (Basel) 2024; 13:1220. [PMID: 39766610 PMCID: PMC11726858 DOI: 10.3390/antibiotics13121220] [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: 10/06/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 01/04/2025] Open
Abstract
Background/Objectives: Antimicrobial resistance (AMR) is a growing threat that undermines the effectiveness of global healthcare. The Centers for Disease Control and Prevention and the World Health Organization have identified numerous microbial organisms, particularly members of the ESKAPEE pathogens, as critical threats to global health and economic security. Many clinical isolates of these pathogens have become completely resistant to current antibiotics, making treatment nearly impossible. Herbal remedies, such as those found in Traditional Chinese Medicine (TCM), have been practiced for thousands of years and successfully used to treat a wide range of ailments, including infectious diseases. Surprisingly, despite this extensive knowledge of folk medicine, no plant-derived antibacterial drugs are currently approved for clinical use. As such, the objective of this study is to evaluate the antimicrobial properties of extracts derived from TCM plants. Methods: This study explores a comprehensive library comprising 664 extracts from 132 distinct TCM plant species for antimicrobial properties against gram-negative (Escherichia coli) and gram-positive (Micrococcus luteus) bacteria using liquid and solid in vitro assays. Results: Intriguingly, our results reveal 17 plant species with potent antimicrobial properties effective primarily against gram-positive organisms, including Streptococcus aureus and epidermidis. A literature search revealed that nearly 100 purified compounds from the identified TCM plants were previously isolated and confirmed for their antimicrobial properties, collectively inhibiting 45 different bacterial species. Conclusions: Our results indicate that phytobiotics from the identified plants could serve as potential candidates for novel antimicrobials.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel M. Czyż
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, FL 32611, USA; (G.L.E.); (D.I.D.)
| |
Collapse
|
2
|
Mooney E, Twamley B, Cooke G, Caraher E, Tacke M, Kelleher F, Creaven BS. Heteroleptic Coumarin-Based Silver(I) Complexes: Possible New Antimicrobial Agents. Molecules 2024; 29:5917. [PMID: 39770006 PMCID: PMC11678713 DOI: 10.3390/molecules29245917] [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: 11/11/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 01/04/2025] Open
Abstract
Heteroleptic coumarin-based silver(I) complexes with improved solubility profiles were synthesised using either triphenylphosphine or an N-heterocyclic carbene as adduct ligands, and were fully characterised using IR and NMR spectroscopy, elemental analysis, and, where possible, X-ray crystallography. The triphenylphosphine adducts formed well-resolved structures, where the oxyacetate ligands asymmetrically chelated the silver(I) ion in a bidentate chelating mode, and the silver(I) ion was also bound to two triphenylphosphine ligands. The solubility profile and photostability of the adducts were considerably improved compared to those of previously isolated simple coumarin silver(I) complexes. Analysis of the coumarin N-heterocyclic carbene(NHC) silver(I) adduct indicated that it likely formed as a complex aggregate species with an overall stoichiometry of 1:1:1 coumarin:Ag(I):NHC. The Kirby Bauer assay and broth microdilution assays were used to assess the silver(I) complexes' and adducts' antimicrobial activity against pathogenic strains of Pseudomonas aeruginosa, Escherichia coli, and MRSA. Interestingly, the formation of more soluble complexes did not increase the activity of the silver(I) complexes and, in effect, made them less effective antimicrobial agents, particularly against Escherichia coli and Pseudomonas aeruginosa, although they retained their activity against MRSA.
Collapse
Affiliation(s)
- Erika Mooney
- School of Chemical and BioPharmaceutical Sciences, Technological University Dublin, TU Dublin, Tallaght Campus, D24 FKT9 Dublin, Ireland; (E.M.); (G.C.)
- Centre for AMR and One Health Research, Technological University Dublin, TU Dublin, Tallaght Campus, D24 FKT9 Dublin, Ireland
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Gordon Cooke
- School of Chemical and BioPharmaceutical Sciences, Technological University Dublin, TU Dublin, Tallaght Campus, D24 FKT9 Dublin, Ireland; (E.M.); (G.C.)
- Centre for AMR and One Health Research, Technological University Dublin, TU Dublin, Tallaght Campus, D24 FKT9 Dublin, Ireland
| | - Emma Caraher
- School of Chemical and BioPharmaceutical Sciences, Technological University Dublin, TU Dublin, Tallaght Campus, D24 FKT9 Dublin, Ireland; (E.M.); (G.C.)
- Centre for AMR and One Health Research, Technological University Dublin, TU Dublin, Tallaght Campus, D24 FKT9 Dublin, Ireland
| | - Matthias Tacke
- UCD School of Chemistry, Science Centre South, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland;
| | - Fintan Kelleher
- School of Chemical and BioPharmaceutical Sciences, Technological University Dublin, TU Dublin, Tallaght Campus, D24 FKT9 Dublin, Ireland; (E.M.); (G.C.)
- Centre for AMR and One Health Research, Technological University Dublin, TU Dublin, Tallaght Campus, D24 FKT9 Dublin, Ireland
| | - Bernadette S. Creaven
- Centre for AMR and One Health Research, Technological University Dublin, TU Dublin, Tallaght Campus, D24 FKT9 Dublin, Ireland
- School of Chemical and BioPharmaceutical Sciences, Technological University Dublin, Central Quad Building, Grangegorman, D07 ADY7 Dublin, Ireland
| |
Collapse
|
3
|
Azlisham NA, Rahman FS, Mahmood Z, Mohamad D, Johari Y, Al-Batayneh OB. Comparative analysis of hydrazinyl coumarin derivative incorporation in resin-modified and conventional glass ionomer cement. J Taibah Univ Med Sci 2024; 19:1119-1129. [PMID: 39758353 PMCID: PMC11699269 DOI: 10.1016/j.jtumed.2024.11.003] [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: 07/19/2024] [Revised: 10/09/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025] Open
Abstract
Objective The study aimed to conduct a comparative analysis of the effects of incorporating hydrazinyl coumarin derivative (HCD) in resin-modified (RMGIC) and conventional glass ionomer cement (cGIC) on their release profiles and antibacterial properties. Method Resin-modified GIC, Fuji II LC (F2) and high-fluoride cGIC, Fuji VII (F7) were used as controls. HCD was synthesized in-house, incorporated into both RMGIC and cGICs at 1 % and 2 % weight percentages (w/w), and chemically analyzed using Fourier transform infrared (FTIR) spectroscopy. Then, the F2 containing HCD (GIC-HCD F2) and F7 containing HCD (GIC-HCD F7) were evaluated for HCD and fluoride release profiles using UV Visible spectrophotometer and pH/ISE benchtop fluoridemeter, respectively. The antibacterial properties were assessed against Streptococcus sanguinis using the agar well diffusion method and measurement of bacterial growth turbidity, followed by the observation of the bacterial morphology using scanning electron microscope. The data were statistically analyzed using one-way ANOVA and Bonferroni post-hoc tests. Results The FTIR spectra confirmed the presence of HCD in the GIC-HCD matrices. HCD was successfully released from both GIC-HCD F2 and GIC-HCD F7 matrices at both weight percentages. Higher fluoride release and inhibitory zones were observed compared to the control groups, with GIC-HCD F2 having a more significant effect than GIC-HCD F7. Additionally, the incorporation of HCD slowed down the growth of Streptococcus sanguinis and showed remarkable changes in bacterial shape specifically on GIC-HCD F2. Conclusion The incorporation of HCD into both RMGIC and cGIC improved fluoride release and enhanced the antibacterial activities, with a more significant effect observed in RMGIC compared to cGIC.
Collapse
Affiliation(s)
- Nor A.F. Azlisham
- Unit of Biomaterials, School of Dental Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Fatimah S.A. Rahman
- Unit of Biomaterials, School of Dental Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Zuliani Mahmood
- Unit of Paediatric Dentistry, School of Dental Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Dasmawati Mohamad
- Unit of Biomaterials, School of Dental Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Yanti Johari
- Unit of Prosthodontics, School of Dental Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Ola B. Al-Batayneh
- Department of Preventive Dentistry, Faculty of Dentistry, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
4
|
Mitsiou VPM, Antonaki AMN, Douka MD, Litinas KE. An Overview on the Synthesis of Lamellarins and Related Compounds with Biological Interest. Molecules 2024; 29:4032. [PMID: 39274880 PMCID: PMC11396623 DOI: 10.3390/molecules29174032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
Lamellarins are natural products with a [3,4]-fused pyrrolocoumarin skeleton possessing interesting biological properties. More than 70 members have been isolated from diverse marine organisms, such as sponges, ascidians, mollusks, and tunicates. There is a continuous interest in the synthesis of these compounds. In this review, the synthetic strategies for the synthesis of the title compounds are presented along with their biological properties. Three routes are followed for the synthesis of lamellarins. Initially, pyrrole derivatives are the starting or intermediate compounds, and then they are fused to isoquinoline or a coumarin moiety. Second, isoquinoline is the starting compound fused to an indole moiety. In the last route, coumarins are the starting compounds, which are fused to a pyrrole moiety and an isoquinoline scaffold. The synthesis of isolamellarins, azacoumestans, isoazacoumestans, and analogues is also described. The above synthesis is achieved via metal-catalyzed cross-coupling, [3 + 2] cycloaddition, substitution, and lactonization reactions. The title compounds exhibit cytotoxic, multidrug resistance (MDR), topoisomerase I-targeted antitumor, anti-HIV, antiproliferative, anti-neurodegenerative disease, and anti-inflammatory activities.
Collapse
Affiliation(s)
- Vasiliki-Panagiota M Mitsiou
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Anastasia-Maria N Antonaki
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Matina D Douka
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos E Litinas
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
5
|
Jung HS, Park YJ, Gu BH, Han G, Ji W, Hwang SM, Kim M. Coumarin derivatives ameliorate the intestinal inflammation and pathogenic gut microbiome changes in the model of infectious colitis through antibacterial activity. Front Cell Infect Microbiol 2024; 14:1362773. [PMID: 39081865 PMCID: PMC11287663 DOI: 10.3389/fcimb.2024.1362773] [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: 12/29/2023] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Coumarin, a phenolic compound, is a secondary metabolite produced by plants such as Tanga and Lime. Coumarin derivatives were prepared via Pechmann condensation. In this study, we performed in vitro and in vivo experiments to determine the antimicrobial and gut immune-regulatory functions of coumarin derivatives. For the in vitro antimicrobial activity assay, coumarin derivatives C1 and C2 were selected based on their pathogen-killing activity against various pathogenic microbes. We further demonstrated that the selected coumarin derivatives disrupted bacterial cell membranes. Next, we examined the regulatory function of the coumarin derivatives in gut inflammation using an infectious colitis model. In an in vivo infectious colitis model, administration of selected C1 coumarin derivatives reduced pathogen loads, the number of inflammatory immune cells (Th1 cells and Th17 cells), and inflammatory cytokine levels (IL-6 and IL-1b) in the intestinal tissue after pathogen infection. In addition, we found that the administration of C1 coumarin derivatives minimized abnormal gut microbiome shift-driven pathogen infection. Potential pathogenic gut microbes, such as Enterobacteriaceae and Staphylococcaceae, were increased by pathogen infection. However, this pathogenic microbial expansion was minimized and beneficial bacteria, such as Ligilactobacillus and Limosilactobacillus, increased with C1 coumarin derivative treatment. Functional gene enrichment assessment revealed that the relative abundance of genes associated with lipid and nucleotide metabolism was reduced by pathogen infection; however, this phenomenon was not observed in C1 coumarin derivative-treated animals. Collectively, our data suggest that C1 coumarin derivative is effective antibacterial agents that minimize pathogen-induced gut inflammation and abnormal gut microbiome modulation through their antibacterial activity.
Collapse
Affiliation(s)
- Hui-su Jung
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea
| | - Yei Ju Park
- R & D Center, EyeGene, Goyang, Republic of Korea
| | - Bon-Hee Gu
- Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| | - Goeun Han
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea
- Future Earth Research Institute, PNU JYS Science Academy, Pusan National University, Busan, Republic of Korea
| | - Woonhak Ji
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea
| | - Su mi Hwang
- Department of Biomedical Laboratory Science, College of Health and Medical Science, Sangji University, Wonju, Republic of Korea
| | - Myunghoo Kim
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea
- Future Earth Research Institute, PNU JYS Science Academy, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
6
|
Kapidou E, Litinas KE. An Overview of the Synthesis of 3,4-Fused Pyrrolocoumarins of Biological Interest. Molecules 2024; 29:2748. [PMID: 38930816 PMCID: PMC11206682 DOI: 10.3390/molecules29122748] [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: 05/15/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
3,4-Fused pyrrolocoumarins, synthetically prepared or naturally occurring, possess interesting biological properties. In this review, the synthetic strategies for the synthesis of the title compounds are presented along with their biological activities. Two routes are followed for that synthesis. In one, the pyrrole ring is formed from coumarin derivatives, such as aminocoumarins or other coumarins. In the other approach, the pyranone moiety is built from an existing pyrrole derivative or through the simultaneous formation of coumarin and pyrrole frameworks. The above syntheses are achieved via 1,3-dipolar cycloaddition reactions, Michael reaction, aza-Claisen rearrangement reactions, multi-component reactions (MCR), as well as metal-catalyzed reactions. Pyrrolocoumarins present cytotoxic, antifungal, antibacterial, α-glucosidase inhibition, antioxidant, lipoxygenase (LOX) inhibition, and fluorescent activities, as well as benzodiazepine receptor ability.
Collapse
Affiliation(s)
| | - Konstantinos E. Litinas
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
7
|
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
|
8
|
Abdel-Halim MS, El-Ganiny AM, Mansour B, Yahya G, Latif HKAE, Askoura M. Phenotypic, molecular, and in silico characterization of coumarin as carbapenemase inhibitor to fight carbapenem-resistant Klebsiella pneumoniae. BMC Microbiol 2024; 24:67. [PMID: 38413891 PMCID: PMC10898048 DOI: 10.1186/s12866-024-03214-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Carbapenems represent the first line treatment of serious infections caused by drug-resistant Klebsiella pneumoniae. Carbapenem-resistant K. pneumoniae (CRKP) is one of the urgent threats to human health worldwide. The current study aims to evaluate the carbapenemase inhibitory potential of coumarin and to test its ability to restore meropenem activity against CRKP. Disk diffusion method was used to test the antimicrobial susceptibility of K. pneumoniae clinical isolates to various antibiotics. Carbapenemase genes (NDM-1, VIM-2, and OXA-9) were detected using PCR. The effect of sub-MIC of coumarin on CRKP isolates was performed using combined disk assay, enzyme inhibition assay, and checkerboard assay. In addition, qRT-PCR was used to estimate the coumarin effect on expression of carbapenemase genes. Molecular docking was used to confirm the interaction between coumarin and binding sites within three carbapenemases. RESULTS K. pneumoniae clinical isolates were found to be multi-drug resistant and showed high resistance to meropenem. All bacterial isolates harbor at least one carbapenemase-encoding gene. Coumarin significantly inhibited carbapenemases in the crude periplasmic extract of CRKP. The checkerboard assay indicated that coumarin-meropenem combination was synergistic exhibiting a fractional inhibitory concentration index ≤ 0.5. In addition, qRT-PCR results revealed that coumarin significantly decreased carbapenemase-genes expression. Molecular docking revealed that the binding energies of coumarin to NDM1, VIM-2, OXA-48 and OXA-9 showed a free binding energy of -7.8757, -7.1532, -6.2064 and - 7.4331 Kcal/mol, respectively. CONCLUSION Coumarin rendered CRKP sensitive to meropenem as evidenced by its inhibitory action on hydrolytic activity and expression of carbapenemases. The current findings suggest that coumarin could be a possible solution to overcome carbapenems resistance in CRKP.
Collapse
Affiliation(s)
- Mahmoud Saad Abdel-Halim
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Amira M El-Ganiny
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Basem Mansour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| | - Galal Yahya
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Hemat K Abd El Latif
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Momen Askoura
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
9
|
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
|
10
|
Khwaza V, Aderibigbe BA. Antifungal Activities of Natural Products and Their Hybrid Molecules. Pharmaceutics 2023; 15:2673. [PMID: 38140014 PMCID: PMC10747321 DOI: 10.3390/pharmaceutics15122673] [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: 10/26/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
The increasing cases of drug resistance and high toxicity associated with the currently used antifungal agents are a worldwide public health concern. There is an urgent need to develop new antifungal drugs with unique target mechanisms. Plant-based compounds, such as carvacrol, eugenol, coumarin, cinnamaldehyde, curcumin, thymol, etc., have been explored for the development of promising antifungal agents due to their diverse biological activities, lack of toxicity, and availability. However, researchers around the world are unable to fully utilize the potential of natural products due to limitations, such as their poor bioavailability and aqueous solubility. The development of hybrid molecules containing natural products is a promising synthetic approach to overcome these limitations and control microbes' capability to develop resistance. Based on the potential advantages of hybrid compounds containing natural products to improve antifungal activity, there have been different reported synthesized hybrid compounds. This paper reviews different literature to report the potential antifungal activities of hybrid compounds containing natural products.
Collapse
Affiliation(s)
- Vuyolwethu Khwaza
- Department of Chemistry, University of Fort Hare, Alice Campus, Alice 5700, Eastern Cape, South Africa
| | - Blessing A. Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Alice 5700, Eastern Cape, South Africa
| |
Collapse
|
11
|
Jiang Y, Hou J, Liu C, Zhao C, Xu Y, Song W, Shu Z, Wang B. Inhibitory Effect of Salicin on Staphylococcus aureus Coagulase. ChemMedChem 2023; 18:e202300302. [PMID: 37755368 DOI: 10.1002/cmdc.202300302] [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: 06/11/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 09/28/2023]
Abstract
The massive use of antibiotics has resulted in an alarming increase in antibiotic resistance in Staphylococcus aureus (S. aureus). This study aimed to identify the inhibitory effect of salicin on S. aureus. Coagulase (Coa) activity was assessed using in vitro Coa assays and Western blot, thermal shift assay (TSA), fluorescence quenching and molecular docking experiments were conducted to verify the interaction between salicin and Coa. An in vivo mouse pneumonia model demonstrated that salicin can reduce the virulence of S. aureus. In vitro Coa assays elucidated that salicin directly inhibited Coa activity. The Western blot and TSA results suggested that salicin did not block the expression of Coa but affected the thermal stability of the protein by binding to Coa. The fluorescence quenching, molecular docking and molecular dynamics assays have found that the most promising binding site between salicin and Coa was GLN-97. The pneumonia model of mice infected with S. aureus revealed that salicin could not only reduce the content of lung bacteria in mice but also prolong their survival. Salicin was identified as a novel anti-infective candidate compound with the potential to target Coa and inhibit its activity by binding to it, which would facilitate the development of roadmaps for future research.
Collapse
Affiliation(s)
- Yijing Jiang
- Changchun University of Chinese Medicine, No.1035 Boshuo Road Jingyue National High-tech Industrial Development Zone, Changchun, 130117, China
| | - Juan Hou
- Changchun University of Chinese Medicine, No.1035 Boshuo Road Jingyue National High-tech Industrial Development Zone, Changchun, 130117, China
| | - Chang Liu
- Changchun University of Chinese Medicine, No.1035 Boshuo Road Jingyue National High-tech Industrial Development Zone, Changchun, 130117, China
| | - Chunhui Zhao
- Changchun University of Chinese Medicine, No.1035 Boshuo Road Jingyue National High-tech Industrial Development Zone, Changchun, 130117, China
| | - Yangming Xu
- Changchun University of Chinese Medicine, No.1035 Boshuo Road Jingyue National High-tech Industrial Development Zone, Changchun, 130117, China
| | - Wu Song
- Changchun University of Chinese Medicine, No.1035 Boshuo Road Jingyue National High-tech Industrial Development Zone, Changchun, 130117, China
| | - Zunhua Shu
- The Third Affiliated Hospital to Changchun University of Chinese Medicine, No.1643, Jingyue Street Nanguan District, Changchun, 130118, China
| | - Bingmei Wang
- Changchun University of Chinese Medicine, No.1035 Boshuo Road Jingyue National High-tech Industrial Development Zone, Changchun, 130117, China
| |
Collapse
|
12
|
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
|