1
|
Dziadas M, Pachura N, Duda-Madej A, Garbicz M, Gębarowski T, Dominguez-Martin A, Rowińska-Żyrek M. Chloramphenicol glycoside derivative: A way to overcome its antimicrobial resistance and toxicity. Carbohydr Res 2025; 550:109387. [PMID: 39862554 DOI: 10.1016/j.carres.2025.109387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/30/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Triggered by the urgent need to tackle the global crisis of multidrug-resistant bacterial infections, in this work, we present a way to overcome chloramphenicol resistance by introducing modifications based on the glycosylation of its hydroxyl groups. The synthesized derivatives demonstrate complete resistance to the action of recombinant chloramphenicol acetyltransferase (CAT) from Escherichia coli and efficacy against methicillin-resistant Staphylococcus aureus (MRSA), Escherichia coli ESBL, and Pseudomonas aeruginosa ATCC 27853. Glycosylation gives chloramphenicol an additional advantage - the stable glycosidic form is less toxic to human dermal fibroblasts and has significantly better water solubility than non-glycosylated chloramphenicol. Using a specific glycosidase, chloramphenicol can be almost immediately released from the stable prodrug at the site of polybacterial infections.
Collapse
Affiliation(s)
- Mariusz Dziadas
- Faculty of Chemistry, University of Wrocław, Wrocław, 50-383, Poland.
| | - Natalia Pachura
- Department of Food Chemistry and Biocatalysis, Wrocław University of Environmental and Life Sciences, Wrocław, 50-375, Poland
| | - Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Wrocław, 50-368, Poland
| | - Mateusz Garbicz
- Faculty of Chemistry, University of Wrocław, Wrocław, 50-383, Poland
| | - Tomasz Gębarowski
- Department of Biostructure and Animal Physiology, Wrocław University of Environmental and Life Sciences, Wrocław, 51-631, Poland
| | - Alicia Dominguez-Martin
- Department of Inorganic Chemistry, Faculty of Pharmacy, University of Granada, Granada, E-18071, Spain
| | | |
Collapse
|
2
|
Rathod NV, Mishra S. Synthesis and Biological Evaluation of Bile Acid-Triclosan Conjugates: A Study on Antibacterial, Antibiofilm, and Molecular Docking. Bioconjug Chem 2025; 36:276-290. [PMID: 39841879 DOI: 10.1021/acs.bioconjchem.4c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
This work describes the synthesis, characterization, and antibacterial properties of four bile acid-triclosan conjugates. The in vitro antibacterial activity of synthetic bile acid-triclosan conjugates was investigated against a panel of Gram-positive and Gram-negative bacteria. Conjugates 3 and 4 show high activity against Escherichia coli (ATCC25922), with IC50 values of 2.94 ± 0.7 and 1.51 ± 0.05 μM, respectively. Conjugate 4 demonstrated 9 times the activity of triclosan (6.77 μM) and 18 times the potency of kanamycin, a well-known antibiotic. Compound 3 showed higher potential activity against all evaluated strains, including Bacillus megaterium (IC50: 3.05 ± 0.02), Bacillus amyloquefaciens (IC50: 8.79 ± 0.01), Serratia marcescens (IC50: 6.77 ± 0.4), and E. coli (IC50: 1.51 ± 0.05 μM). These findings indicate that it has broad-spectrum antibacterial activity. Bile acid-triclosan conjugates prevent biofilms by up to 99% at low doses (conjugates 4; 4.16 ± 0.8 μM), compared to triclosan. Conjugate 5 was most potent against B. amyloquefaciens (IC50 = 5.23 ± 0.2 μM), while conjugate 4 was most effective against B. megaterium (IC50 = 4.16 ± 0.8 μM) in biofilm formation. These conjugates inhibit biofilm formation by limiting the extracellular polymeric substance generation. The in vitro antibacterial study revealed that bile acid-triclosan conjugates were more effective than the parent molecule triclosan at inhibiting bacterial growth and biofilm formation against both Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
- Neha V Rathod
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar 382426, India
| | - Satyendra Mishra
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar 382426, India
| |
Collapse
|
3
|
Al-Sawarees DK, Darwish RM, Abu-Zurayk R, Masri MA. Assessing silver nanoparticle and antimicrobial combinations for antibacterial activity and biofilm prevention on surgical sutures. J Appl Microbiol 2024; 135:lxae063. [PMID: 38471695 DOI: 10.1093/jambio/lxae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/28/2024] [Accepted: 03/11/2024] [Indexed: 03/14/2024]
Abstract
AIMS To evaluate the effect of silver nanoparticles alone and in combination with Triclosan, and trans-cinnamaldehyde against Staphylococcus aureus and Escherichia coli biofilms on sutures to improve patients' outcomes. METHODS AND RESULTS Silver nanoparticles were prepared by chemical method and characterized by UV-visible spectrophotometer and dynamic light scattering. The minimum inhibitory concentration was assessed by the Microdilution assay. The antibiofilm activity was determined using crystal violet assay. A checkerboard assay using the fractional inhibitory concentration index and time-kill curve was used to investigate the synergistic effect of silver nanoparticle combinations. The hemolytic activity was determined using an erythrocyte hemolytic assay. Our results revealed that silver nanoparticles, Triclosan, and trans-cinnamaldehyde (TCA) inhibited S.aureus and E.coli biofilms. Silver nanoparticles with TCA showed a synergistic effect (FICI values 0.35 and 0.45 against S. aureus and E. coli biofilms, respectively), and silver nanoparticles with Triclosan showed complete inhibition of S. aureus biofilm. The hemolytic activity was <2.50% for the combinations.
Collapse
Affiliation(s)
- Diana K Al-Sawarees
- Department of Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| | - Rula M Darwish
- Department of Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| | - Rund Abu-Zurayk
- Hamdi Mango Center for Scientific Research, The University of Jordan, Amman 11942, Jordan
| | - Mahmoud Al Masri
- King Hussain Cancer center, Surgery Department, Amman 11941, Jordan
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
4
|
Breijyeh Z, Karaman R. Design and Synthesis of Novel Antimicrobial Agents. Antibiotics (Basel) 2023; 12:628. [PMID: 36978495 PMCID: PMC10045396 DOI: 10.3390/antibiotics12030628] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The necessity for the discovery of innovative antimicrobials to treat life-threatening diseases has increased as multidrug-resistant bacteria has spread. Due to antibiotics' availability over the counter in many nations, antibiotic resistance is linked to overuse, abuse, and misuse of these drugs. The World Health Organization (WHO) recognized 12 families of bacteria that present the greatest harm to human health, where options of antibiotic therapy are extremely limited. Therefore, this paper reviews possible new ways for the development of novel classes of antibiotics for which there is no pre-existing resistance in human bacterial pathogens. By utilizing research and technology such as nanotechnology and computational methods (such as in silico and Fragment-based drug design (FBDD)), there has been an improvement in antimicrobial actions and selectivity with target sites. Moreover, there are antibiotic alternatives, such as antimicrobial peptides, essential oils, anti-Quorum sensing agents, darobactins, vitamin B6, bacteriophages, odilorhabdins, 18β-glycyrrhetinic acid, and cannabinoids. Additionally, drug repurposing (such as with ticagrelor, mitomycin C, auranofin, pentamidine, and zidovudine) and synthesis of novel antibacterial agents (including lactones, piperidinol, sugar-based bactericides, isoxazole, carbazole, pyrimidine, and pyrazole derivatives) represent novel approaches to treating infectious diseases. Nonetheless, prodrugs (e.g., siderophores) have recently shown to be an excellent platform to design a new generation of antimicrobial agents with better efficacy against multidrug-resistant bacteria. Ultimately, to combat resistant bacteria and to stop the spread of resistant illnesses, regulations and public education regarding the use of antibiotics in hospitals and the agricultural sector should be combined with research and technological advancements.
Collapse
Affiliation(s)
- Zeinab Breijyeh
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem P.O. Box 20002, Palestine
| | - Rafik Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem P.O. Box 20002, Palestine
- Department of Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| |
Collapse
|
5
|
Design, Synthesis, In Silico and POM Studies for the Identification of the Pharmacophore Sites of Benzylidene Derivatives. Molecules 2023; 28:molecules28062613. [PMID: 36985587 PMCID: PMC10053039 DOI: 10.3390/molecules28062613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Due to the uneven distribution of glycosidase enzyme expression across bacteria and fungi, glycoside derivatives of antimicrobial compounds provide prospective and promising antimicrobial materials. Therefore, herein, we report the synthesis and characterization of six novel methyl 4,6-O-benzylidene-α-d-glucopyranoside (MBG) derivatives (2–7). The structures were ascertained using spectroscopic techniques and elemental analyses. Antimicrobial tests (zone of inhibition, MIC and MBC) were carried out to determine their ability to inhibit the growth of different Gram-positive, Gram-negative bacteria and fungi. The highest antibacterial activity was recorded with compounds 4, 5, 6 and 7. The compounds with the most significant antifungal efficacy were 4, 5, 6 and 7. Based on the prediction of activity spectra for substances (PASS), compounds 4 and 7 have promising antimicrobial capacity. Molecular docking studies focused on fungal and bacterial proteins where derivatives 3 and 6 exhibited strong binding affinities. The molecular dynamics study revealed that the complexes formed by these derivatives with the proteins L,D-transpeptidase Ykud and endoglucanase from Aspergillus niger remained stable, both over time and in physiological conditions. Structure–activity relationships, including in vitro and in silico results, revealed that the acyl chains [lauroyl-(CH3(CH2)10CO-), cinnamoyl-(C6H5CH=CHCO-)], in combination with sugar, were found to have the most potential against human and fungal pathogens. Synthetic, antimicrobial and pharmacokinetic studies revealed that MBG derivatives have good potential for antimicrobial activity, developing a therapeutic target for bacteria and fungi. Furthermore, the Petra/Osiris/Molinspiration (POM) study clearly indicated the presence of an important (O1δ−----O2δ−) antifungal pharmacophore site. This site can also be explored as a potential antiviral moiety.
Collapse
|
6
|
Yang Y, Ding J, Zhu X, Tian Z, Zhu S. Triclosan to Improve the Antimicrobial Performance of Universal Adhesives. Polymers (Basel) 2023; 15:polym15020304. [PMID: 36679185 PMCID: PMC9863416 DOI: 10.3390/polym15020304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
To solve the proble ms of composite restoration failure caused by secondary caries, this study reports a light curable antibacterial triclosan derivative (TCS-IH), which was synthesized and added to the existing commercial universal adhesive to achieve a long-term antibacterial effect The effect of mixing different mass percentages of TCS-IH on the bond strength of dentin was also investigated.TCS-IH was synthesized by solution polymerization and characterized by nuclear magnetic resonance hydrogen spectroscopy (1H NMR) and Fourier transform infrared (FTIR) spectroscopy. Two commercial universal adhesives, Single Bond Universal and All Bond Universal, were selected and used as the control group, and universal adhesives with different mass percentages (1 wt%, 3 wt%, 5 wt% and 7 wt%) of TCS-IH were used as the experimental group. The antibacterial properties were analysed by means of colony count experiments, biofilm formation detection, plotting of growth curves, biofilm metabolic activity detection, insoluble extracellular polysaccharide measurements and observations by confocal laser scanning microscopy and scanning electron microscopy (SEM). The effect of adhesives on biofilm formation, metabolism, extracellular matrix production, distribution of live and dead bacteria, and bacterial morphology of Streptococcus mutans (S. mutans) was analysed. The mechanical properties were evaluated by the degree of conversion and microtensile bonding strength under different conditions. Its biosafety was tested. We found that the addition of TCS-IH significantly improved the antibacterial performance of the universal adhesive, with the 5 wt% and 7 wt% groups showing the best antibacterial effect and effectively inhibiting the formation of biofilm. In addition, the adhesive strength test results showed that there was no statistical difference (p < 0.05) in the microtensile bond strength measured under various factors in all experimental groups except for the 7 wt% group in the self-etch bonding mode, and all of them had good biosafety. In summary, the 5 wt% group of antibacterial monomer TCS-IH was selected as the optimum addition to the universal adhesive to ensure the antimicrobial properties of the universal adhesive and the stability and durability of the adhesive interface. This study provides a reference for the clinical application of adhesives with antimicrobial activity to improve the stability and durability of adhesive restorations.
Collapse
|
7
|
Hutchins M, Bovill RA, Stephens PJ, Brazier JA, Osborn HMI. Glycosides of Nadifloxacin-Synthesis and Antibacterial Activities against Methicillin-Resistant Staphylococcus aureus. Molecules 2022; 27:1504. [PMID: 35268604 PMCID: PMC8912027 DOI: 10.3390/molecules27051504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
The increase in the number of bacteria that are resistant to multiple antibiotics poses a serious clinical problem that threatens the health of humans worldwide. Nadifloxacin (1) is a highly potent antibacterial agent with broad-spectrum activity. However, its poor aqueous solubility has limited its use to topical applications. To increase its solubility, it was glycosylated herein to form a range of trans-linked (3a-e) and cis-linked (7a,b) glycosides, each of which was prepared and purified to afford single anomers. The seven glycoside derivatives (3a-e, 7a,b) were examined for potency against eight strains of S. aureus, four of which were methicillin-resistant. Although less potent than free nadifloxacin (1), the α-L-arabinofuransoside (3a) was effective against all strains that were tested (minimum inhibitory concentrations of 1-8 μg/mL compared to 0.1-0.25 μg/mL for nadifloxacin), demonstrating the potential of this glycoside as an antibacterial agent. Estimation of Log P as well as observations made during preparation of these compounds reveal that the solubilities of the glycosides were greatly improved compared with nadifloxacin (1), raising the prospect of its use in oral applications.
Collapse
Affiliation(s)
- Mark Hutchins
- ThermoFisher Scientific, Wade Road, Basingstoke RG24 8PW, Hampshire, UK
| | - Richard A. Bovill
- ThermoFisher Scientific, Wade Road, Basingstoke RG24 8PW, Hampshire, UK
| | - Peter J. Stephens
- ThermoFisher Scientific, Wade Road, Basingstoke RG24 8PW, Hampshire, UK
| | - John A. Brazier
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, Berkshire, UK
| | - Helen M. I. Osborn
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, Berkshire, UK
| |
Collapse
|
8
|
Holt BA, Tuttle M, Xu Y, Su M, Røise JJ, Wang X, Murthy N, Kwong GA. Dimensionless parameter predicts bacterial prodrug success. Mol Syst Biol 2022; 18:e10495. [PMID: 35005851 PMCID: PMC8744131 DOI: 10.15252/msb.202110495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022] Open
Abstract
Understanding mechanisms of antibiotic failure is foundational to combating the growing threat of multidrug-resistant bacteria. Prodrugs-which are converted into a pharmacologically active compound after administration-represent a growing class of therapeutics for treating bacterial infections but are understudied in the context of antibiotic failure. We hypothesize that strategies that rely on pathogen-specific pathways for prodrug conversion are susceptible to competing rates of prodrug activation and bacterial replication, which could lead to treatment escape and failure. Here, we construct a mathematical model of prodrug kinetics to predict rate-dependent conditions under which bacteria escape prodrug treatment. From this model, we derive a dimensionless parameter we call the Bacterial Advantage Heuristic (BAH) that predicts the transition between prodrug escape and successful treatment across a range of time scales (1-104 h), bacterial carrying capacities (5 × 104 -105 CFU/µl), and Michaelis constants (KM = 0.747-7.47 mM). To verify these predictions in vitro, we use two models of bacteria-prodrug competition: (i) an antimicrobial peptide hairpin that is enzymatically activated by bacterial surface proteases and (ii) a thiomaltose-conjugated trimethoprim that is internalized by bacterial maltodextrin transporters and hydrolyzed by free thiols. We observe that prodrug failure occurs at BAH values above the same critical threshold predicted by the model. Furthermore, we demonstrate two examples of how failing prodrugs can be rescued by decreasing the BAH below the critical threshold via (i) substrate design and (ii) nutrient control. We envision such dimensionless parameters serving as supportive pharmacokinetic quantities that guide the design and administration of prodrug therapeutics.
Collapse
Affiliation(s)
- Brandon Alexander Holt
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Tech College of Engineering and Emory School of MedicineAtlantaGAUSA
| | - McKenzie Tuttle
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Tech College of Engineering and Emory School of MedicineAtlantaGAUSA
| | - Yilin Xu
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Tech College of Engineering and Emory School of MedicineAtlantaGAUSA
| | - Melanie Su
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Tech College of Engineering and Emory School of MedicineAtlantaGAUSA
| | - Joachim J Røise
- Department of BioengineeringInnovative Genomics InstituteUniversity of CaliforniaBerkeleyCAUSA
| | - Xioajian Wang
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech UniversityNanjingChina
| | - Niren Murthy
- Department of BioengineeringInnovative Genomics InstituteUniversity of CaliforniaBerkeleyCAUSA
| | - Gabriel A Kwong
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Tech College of Engineering and Emory School of MedicineAtlantaGAUSA
- Parker H. Petit Institute of Bioengineering and BioscienceAtlantaGAUSA
- Institute for Electronics and NanotechnologyGeorgia TechAtlantaGAUSA
- Integrated Cancer Research CenterGeorgia TechAtlantaGAUSA
- Georgia ImmunoEngineering ConsortiumGeorgia Tech and Emory UniversityAtlantaGAUSA
- Emory School of MedicineAtlantaGAUSA
- Emory Winship Cancer InstituteAtlantaGAUSA
| |
Collapse
|
9
|
Lin JH, Shih YH, Huang CH, Lai MF, Lee SA, Shiu BC, Lou CW. Evaluations of Electrostatic Filtration Efficiency and Antibacterial Efficacy of Antibacterial Electret Polypropylene Filters: Effects of Using Low Molecular Antibacterial Agent as Additive. Polymers (Basel) 2021; 13:polym13193303. [PMID: 34641119 PMCID: PMC8512406 DOI: 10.3390/polym13193303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/25/2022] Open
Abstract
In recent years, air filtration has been gaining much attention, and now people are much more concerned about antibacterial filters due to the spreading of COVID-19. The electret polypropylene (PP) nonwoven fabrics possess excellent filtration efficiency but a limited antibacterial effect against S. aureus and E. coli, and therefore triclosan is used in this study. Serving as an antibacterial agent, triclosan with a low molecular weight is an effective additive for the test results, indicating that the presence of triclosan strengthens the antibacterial effects of the filters. In addition, triclosan also strengthens the PP’s crystallinity, which in turn betters the filtration efficiency of the filters concurrently. Demonstrating powerful filtration and antibacterial performances, the antibacterial electret PP filters are highly qualified for filter applications.
Collapse
Affiliation(s)
- Jia-Horng Lin
- College of Material and Chemical Engineering, Minjiang University, Fuzhou 350108, China; (J.-H.L.); (B.-C.S.)
- Laboratory of Fiber Application and Manufacturing, Department of Fiber and Composite Materials, Feng Chia University, Taichung 40724, Taiwan; (Y.-H.S.); (M.-F.L.)
- School of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
- Advanced Medical Care and Protection Technology Research Center, College of Textile and Clothing, Qingdao University, Qingdao 266071, China
| | - Ying-Huei Shih
- Laboratory of Fiber Application and Manufacturing, Department of Fiber and Composite Materials, Feng Chia University, Taichung 40724, Taiwan; (Y.-H.S.); (M.-F.L.)
| | - Chen-Hung Huang
- Department of Aerospace and Systems Engineering, Feng Chia University, Taichung 40724, Taiwan
- Correspondence: (C.-H.H.); (C.-W.L.)
| | - Mei-Feng Lai
- Laboratory of Fiber Application and Manufacturing, Department of Fiber and Composite Materials, Feng Chia University, Taichung 40724, Taiwan; (Y.-H.S.); (M.-F.L.)
| | - Shu-An Lee
- Department of Environmental Engineering and Science, Feng Chia University, Taichung 40724, Taiwan;
| | - Bing-Chiuan Shiu
- College of Material and Chemical Engineering, Minjiang University, Fuzhou 350108, China; (J.-H.L.); (B.-C.S.)
| | - Ching-Wen Lou
- Advanced Medical Care and Protection Technology Research Center, College of Textile and Clothing, Qingdao University, Qingdao 266071, China
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Fujian Key Laboratory of Novel Functional Fibers and Materials, Minjiang University, Fuzhou 350108, China
- Correspondence: (C.-H.H.); (C.-W.L.)
| |
Collapse
|
10
|
Dwivedi GR, Rai R, Pratap R, Singh K, Pati S, Sahu SN, Kant R, Darokar MP, Yadav DK. Drug resistance reversal potential of multifunctional thieno[3,2-c]pyran via potentiation of antibiotics in MDR P. aeruginosa. Biomed Pharmacother 2021; 142:112084. [PMID: 34449308 DOI: 10.1016/j.biopha.2021.112084] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022] Open
Abstract
We explored the antibacterial potential (alone and combination) against multidrug resistant (MDR) Pseudomonas aeruginosa isolates KG-P2 using synthesized thieno[3,2-c]pyran-2-ones in combination with different antibiotics. Out of 14 compounds, two compounds (3g and 3l) abridged the MIC of tetracycline (TET) by 16 folds. Compounds was killing the KG-P2 cells, in time dependent manner, lengthened post-antibiotic effect (PAE) of TET and found decreased the mutant prevention concentration (MPC) of TET. In ethidium bromide efflux experiment, two compounds repressed the drug transporter (efflux pumps) which is further supported by molecular docking of these compounds with efflux complex MexAB-OprM. In another study, these compounds inhibited the synthesis of biofilm.
Collapse
Affiliation(s)
- Gaurav Raj Dwivedi
- Microbiology Department, ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur 273013, India.
| | - Reeta Rai
- Department of Biochemistry, AIIMS Ansari Nagar, New Delhi 110029, India
| | - Ramendra Pratap
- Department of Chemistry, North campus University of Delhi, Delhi 110007, India.
| | - Khusbu Singh
- Microbiology Department, ICMR-Regional Medical Research Centre, Bhubaneshwar 751023, Odisha, India
| | - Sanghamitra Pati
- Microbiology Department, ICMR-Regional Medical Research Centre, Bhubaneshwar 751023, Odisha, India
| | - Satya Narayan Sahu
- Government College Balrampur, Balrampur-Ramanujganj, Chhattisgarh 497119, India
| | - Rajni Kant
- Microbiology Department, ICMR-Regional Medical Research Centre, BRD Medical College Campus, Gorakhpur 273013, India
| | - Mahendra P Darokar
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, ̥Near Kukrail Picnic Spot, P.O. CIMAP, Lucknow 226015, India
| | - Dharmendra K Yadav
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, 191 Hambakmoeiro, Yeonsu-gu, Incheon 21924, Republic of Korea.
| |
Collapse
|
11
|
Wu L, Li X, Li P, Pan L, Ji Z, Feng Y, Shi C. Bioabsorbable flexible elastomer of
PTMC‐b‐PEG‐b‐PTMC
copolymer as intestinal anastomosis scaffold. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Lei Wu
- School of Chemical Engineering and Technology Tianjin University Tianjin China
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| | - Xujian Li
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| | - Pengpeng Li
- School of Biomedical Engineering Wenzhou Medical University Wenzhou Zhejiang China
| | - Luqi Pan
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| | - Zhixiao Ji
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| | - Yakai Feng
- School of Chemical Engineering and Technology Tianjin University Tianjin China
- Key Laboratory of Systems Bioengineering (Ministry of Education) Tianjin University Tianjin China
| | - Changcan Shi
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| |
Collapse
|
12
|
Meiers J, Zahorska E, Röhrig T, Hauck D, Wagner S, Titz A. Directing Drugs to Bugs: Antibiotic-Carbohydrate Conjugates Targeting Biofilm-Associated Lectins of Pseudomonas aeruginosa. J Med Chem 2020; 63:11707-11724. [PMID: 32924479 PMCID: PMC7586336 DOI: 10.1021/acs.jmedchem.0c00856] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic infections by Pseudomonas aeruginosa are characterized by biofilm formation, which effectively enhances resistance toward antibiotics. Biofilm-specific antibiotic delivery could locally increase drug concentration to break antimicrobial resistance and reduce the drug's peripheral side effects. Two extracellular P. aeruginosa lectins, LecA and LecB, are essential structural components for biofilm formation and thus render a possible anchor for biofilm-targeted drug delivery. The standard-of-care drug ciprofloxacin suffers from severe systemic side effects and was therefore chosen for this approach. We synthesized several ciprofloxacin-carbohydrate conjugates and established a structure-activity relationship. Conjugation of ciprofloxacin to lectin probes enabled biofilm accumulation in vitro, reduced the antibiotic's cytotoxicity, but also reduced its antibiotic activity against planktonic cells due to a reduced cell permeability and on target activity. This work defines the starting point for new biofilm/lectin-targeted drugs to modulate antibiotic properties and ultimately break antimicrobial resistance.
Collapse
Affiliation(s)
- Joscha Meiers
- Chemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, D-38124 Braunschweig, Germany.,Department of Pharmacy and Department of Chemistry, Saarland University, D-66123 Saarbrücken, Germany
| | - Eva Zahorska
- Chemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, D-38124 Braunschweig, Germany.,Department of Pharmacy and Department of Chemistry, Saarland University, D-66123 Saarbrücken, Germany
| | - Teresa Röhrig
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, D-38124 Braunschweig, Germany.,Drug Design and Optimization (DDOP), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany
| | - Dirk Hauck
- Chemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, D-38124 Braunschweig, Germany
| | - Stefanie Wagner
- Chemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, D-38124 Braunschweig, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, D-38124 Braunschweig, Germany.,Department of Pharmacy and Department of Chemistry, Saarland University, D-66123 Saarbrücken, Germany
| |
Collapse
|
13
|
Lima CSAD, Varca GHC, Costa SMD, Ferraz HG, Santos ACDS, Lopes PS, Costa SAD. Development of natural polymeric microcapsules for antimicrobial drug delivery: triclosan loaded chitosan and alginate-based microcapsules. Drug Dev Ind Pharm 2020; 46:1477-1486. [PMID: 32783646 DOI: 10.1080/03639045.2020.1809445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The goal of this work was the development of natural polymeric microcapsules for antimicrobial drug delivery - triclosan loaded alginate and chitosan-based microcapsules for potential coating applications in substrates such as textiles or plastics. Microcapsules containing 2.5% (w/w) or 3% (w/w) triclosan in both core and matrix were synthesized and evaluated by Fourier-transform infrared spectroscopy, scanning electron microscopy, confocal microscopy, differential scanning calorimetry, thermogravimetry, and antimicrobial activity. The microcapsules produced featured spherical and mostly irregularly-shaped surfaces composed by an alginate core in a chitosan outer matrix, as revealed by confocal microscopy, and antimicrobial activity against S. aureus and E. coli with inhibition halos up to 60 mm and 25 mm respectively, granted by a triclosan loading of 61.66%. The thermal analysis suggested that the polymers protected the active substance from temperature-induced degradation. In conclusion, these microcapsules may be applied toward antimicrobial functionalization of plastics, textiles and other materials.
Collapse
Affiliation(s)
| | | | | | - Humberto Gomes Ferraz
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Patrícia Santos Lopes
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | | |
Collapse
|
14
|
Yang L, Zhang C, Huang F, Liu J, Zhang Y, Yang C, Ren C, Chu L, Liu B, Liu J. Triclosan-based supramolecular hydrogels as nanoantibiotics for enhanced antibacterial activity. J Control Release 2020; 324:354-365. [PMID: 32454121 DOI: 10.1016/j.jconrel.2020.05.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/17/2020] [Accepted: 05/21/2020] [Indexed: 12/20/2022]
Abstract
With the emergence of drug-resistant bacteria, conventional antibiotics are becoming increasingly ineffective for the treatment of bacterial infections. Nanomaterial-modified antibiotics, denoted as "nanoantibiotics", can usually circumvent most of the shortcomings of conventional antibiotics, thus improving antibacterial activities. Here, we developed triclosan-based supramolecular hydrogel nanoantibiotics by conjugating small molecule antibiotic triclosan (TCS) to self-assembling peptides. The resultant nanoantibiotics presented many beneficial characteristics: (i) a stable three-dimensional nanofiber structure; (ii) increased TCS solubility by 850-fold; (iii) acid-responsive TCS release; (iv) favorable biocompatibility. In consequence, the nanoantibiotics showed potent in vitro broad-spectrum antibacterial activities against both Gram-positive and Gram-negative bacteria based on the cooperative effect of antibiotic TCS and the nanostructure-induced bacterial membrane disruption. Furthermore, the TCS-based supramolecular hydrogel nanoantibiotics exhibited enhanced antibacterial activities with low side effects, according to the in vivo antibacterial evaluation at the macro and micro level. Therefore, the simple and effective hydrogel nanoantibiotics developed here hold great potential for the treatment of intractable bacterial infections.
Collapse
Affiliation(s)
- Lijun Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300192, PR China
| | - Congrou Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300192, PR China
| | - Fan Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300192, PR China
| | - Jinjian Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300192, PR China
| | - Yumin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300192, PR China
| | - Cuihong Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300192, PR China
| | - Chunhua Ren
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300192, PR China
| | - Liping Chu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300192, PR China.
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
15
|
Antibacterial Prodrugs to Overcome Bacterial Resistance. Molecules 2020; 25:molecules25071543. [PMID: 32231026 PMCID: PMC7180472 DOI: 10.3390/molecules25071543] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/17/2022] Open
Abstract
Bacterial resistance to present antibiotics is emerging at a high pace that makes the development of new treatments a must. At the same time, the development of novel antibiotics for resistant bacteria is a slow-paced process. Amid the massive need for new drug treatments to combat resistance, time and effort preserving approaches, like the prodrug approach, are most needed. Prodrugs are pharmacologically inactive entities of active drugs that undergo biotransformation before eliciting their pharmacological effects. A prodrug strategy can be used to revive drugs discarded due to a lack of appropriate pharmacokinetic and drug-like properties, or high host toxicity. A special advantage of the use of the prodrug approach in the era of bacterial resistance is targeting resistant bacteria by developing prodrugs that require bacterium-specific enzymes to release the active drug. In this article, we review the up-to-date implementation of prodrugs to develop medications that are active against drug-resistant bacteria.
Collapse
|
16
|
Insights into the complexation of glucose-6-phosphate (G6P) with V(III), Ru(III), Au(III), and Se(IV) ions in binary solvent system. J Mol Liq 2019. [DOI: 10.1016/j.molliq.2019.111999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Microorganisms control and quality improvement of stewed pork with carrots using ZnO nanoparticels combined with radio frequency pasteurization. FOOD BIOSCI 2019. [DOI: 10.1016/j.fbio.2019.100487] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|