51
|
Chen C, Shi J, Wang D, Kong P, Wang Z, Liu Y. Antimicrobial peptides as promising antibiotic adjuvants to combat drug-resistant pathogens. Crit Rev Microbiol 2024; 50:267-284. [PMID: 36890767 DOI: 10.1080/1040841x.2023.2186215] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/19/2022] [Accepted: 10/26/2022] [Indexed: 03/10/2023]
Abstract
The widespread antimicrobial resistance (AMR) calls for the development of new antimicrobial strategies. Antibiotic adjuvant rescues antibiotic activity and increases the life span of the antibiotics, representing a more productive, timely, and cost-effective strategy in fighting drug-resistant pathogens. Antimicrobial peptides (AMPs) from synthetic and natural sources are considered new-generation antibacterial agents. Besides their direct antimicrobial activity, growing evidence shows that some AMPs effectively enhance the activity of conventional antibiotics. The combinations of AMPs and antibiotics display an improved therapeutic effect on antibiotic-resistant bacterial infections and minimize the emergence of resistance. In this review, we discuss the value of AMPs in the age of resistance, including modes of action, limiting evolutionary resistance, and their designing strategies. We summarise the recent advances in combining AMPs and antibiotics against antibiotic-resistant pathogens, as well as their synergistic mechanisms. Lastly, we highlight the challenges and opportunities associated with the use of AMPs as potential antibiotic adjuvants. This will shed new light on the deployment of synergistic combinations to address the AMR crisis.
Collapse
Affiliation(s)
- Chen Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jingru Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Dejuan Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Pan Kong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
52
|
Moreau F, Atamanyuk D, Blaukopf M, Barath M, Herczeg M, Xavier NM, Monbrun J, Airiau E, Henryon V, Leroy F, Floquet S, Bonnard D, Szabla R, Brown C, Junop MS, Kosma P, Gerusz V. Potentiating Activity of GmhA Inhibitors on Gram-Negative Bacteria. J Med Chem 2024; 67:6610-6623. [PMID: 38598312 PMCID: PMC11056994 DOI: 10.1021/acs.jmedchem.4c00037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/28/2024] [Accepted: 03/29/2024] [Indexed: 04/12/2024]
Abstract
Inhibition of the biosynthesis of bacterial heptoses opens novel perspectives for antimicrobial therapies. The enzyme GmhA responsible for the first committed biosynthetic step catalyzes the conversion of sedoheptulose 7-phosphate into d-glycero-d-manno-heptose 7-phosphate and harbors a Zn2+ ion in the active site. A series of phosphoryl- and phosphonyl-substituted derivatives featuring a hydroxamate moiety were designed and prepared from suitably protected ribose or hexose derivatives. High-resolution crystal structures of GmhA complexed to two N-formyl hydroxamate inhibitors confirmed the binding interactions to a central Zn2+ ion coordination site. Some of these compounds were found to be nanomolar inhibitors of GmhA. While devoid of HepG2 cytotoxicity and antibacterial activity of their own, they demonstrated in vitro lipopolysaccharide heptosylation inhibition in Enterobacteriaceae as well as the potentiation of erythromycin and rifampicin in a wild-type Escherichia coli strain. These inhibitors pave the way for a novel treatment of Gram-negative infections.
Collapse
Affiliation(s)
- François Moreau
- Mutabilis, 102 Avenue Gaston Roussel, Romainville 93230, France
| | | | - Markus Blaukopf
- Department
of Chemistry, University of Natural Resources
and Life Sciences, Muthgasse
18, Vienna A-1190, Austria
| | - Marek Barath
- Department
of Chemistry, University of Natural Resources
and Life Sciences, Muthgasse
18, Vienna A-1190, Austria
- Institute
of Chemistry, Center for Glycomics, Slovak
Academy of Sciences, Dúbravská cesta 9, Bratislava SK-845 38, Slovakia
| | - Mihály Herczeg
- Department
of Chemistry, University of Natural Resources
and Life Sciences, Muthgasse
18, Vienna A-1190, Austria
- Department
of Pharmaceutical Chemistry, University
of Debrecen, Debrecen 4032, Hungary
| | - Nuno M. Xavier
- Department
of Chemistry, University of Natural Resources
and Life Sciences, Muthgasse
18, Vienna A-1190, Austria
- Centro
de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Ed. C8, 5° Piso, Campo Grande, Lisboa 1749-016, Portugal
| | | | | | | | - Frédéric Leroy
- Carbosynth
Limited, 8&9 Old
Station Business Park, Compton, Berkshire RG20 6NE, U.K.
| | | | - Damien Bonnard
- Mutabilis, 102 Avenue Gaston Roussel, Romainville 93230, France
| | - Robert Szabla
- Department
of Biochemistry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Chris Brown
- Department
of Biochemistry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Murray S. Junop
- Department
of Biochemistry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Paul Kosma
- Department
of Chemistry, University of Natural Resources
and Life Sciences, Muthgasse
18, Vienna A-1190, Austria
| | - Vincent Gerusz
- Mutabilis, 102 Avenue Gaston Roussel, Romainville 93230, France
| |
Collapse
|
53
|
Caron AJ, Ali IJ, Delgado MJ, Johnson D, Reeks JM, Strzhemechny YM, McGillivray SM. Zinc oxide nanoparticles mediate bacterial toxicity in Mueller-Hinton Broth via Zn 2. Front Microbiol 2024; 15:1394078. [PMID: 38711974 PMCID: PMC11070567 DOI: 10.3389/fmicb.2024.1394078] [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: 02/29/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
As antibiotic resistance increases and antibiotic development dwindles, new antimicrobial agents are needed. Recent advances in nanoscale engineering have increased interest in metal oxide nanoparticles, particularly zinc oxide nanoparticles, as antimicrobial agents. Zinc oxide nanoparticles are promising due to their broad-spectrum antibacterial activity and low production cost. Despite many studies demonstrating the effectiveness of zinc oxide nanoparticles, the antibacterial mechanism is still unknown. Previous work has implicated the role of reactive oxygen species such as hydrogen peroxide, physical damage of the cell envelope, and/or release of toxic Zn2+ ions as possible mechanisms of action. To evaluate the role of these proposed methods, we assessed the susceptibility of S. aureus mutant strains, ΔkatA and ΔmprF, to zinc oxide nanoparticles of approximately 50 nm in size. These assays demonstrated that hydrogen peroxide and electrostatic interactions are not crucial for mediating zinc oxide nanoparticle toxicity. Instead, we found that Zn2+ accumulates in Mueller-Hinton Broth over time and that removal of Zn2+ through chelation reverses this toxicity. Furthermore, we found that the physical separation of zinc oxide nanoparticles and bacterial cells using a semi-permeable membrane still allows for growth inhibition. We concluded that soluble Zn2+ is the primary mechanism by which zinc oxide nanoparticles mediate toxicity in Mueller-Hinton Broth. Future work investigating how factors such as particle morphology (e.g., size, polarity, surface defects) and media contribute to Zn2+ dissolution could allow for the synthesis of zinc oxide nanoparticles that possess chemical and morphological properties best suited for antibacterial efficacy.
Collapse
Affiliation(s)
- Alexander J. Caron
- Department of Biology, Texas Christian University, Fort Worth, TX, United States
| | - Iman J. Ali
- Department of Biology, Texas Christian University, Fort Worth, TX, United States
| | - Michael J. Delgado
- Department of Biology, Texas Christian University, Fort Worth, TX, United States
| | - Dustin Johnson
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States
| | - John M. Reeks
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States
| | - Yuri M. Strzhemechny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States
| | | |
Collapse
|
54
|
Huang YJ, Zang YP, Peng LJ, Yang MH, Lin J, Chen WM. Cajaninstilbene acid derivatives conjugated with siderophores of 3-hydroxypyridin-4(1H)-ones as novel antibacterial agents against Gram-negative bacteria based on the Trojan horse strategy. Eur J Med Chem 2024; 269:116339. [PMID: 38537513 DOI: 10.1016/j.ejmech.2024.116339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/07/2024]
Abstract
The low permeability of the outer membrane of Gram-negative bacteria is a serious obstacle to the development of new antibiotics against them. Conjugation of antibiotic with siderophore based on the "Trojan horse strategy" is a promising strategy to overcome the outer membrane obstacle. In this study, series of antibacterial agents were designed and synthesized by conjugating the 3-hydroxypyridin-4(1H)-one based siderophores with cajaninstilbene acid (CSA) derivative 4 which shows good activity against Gram-positive bacteria by targeting their cell membranes but is ineffective against Gram-negative bacteria. Compared to the inactive parent compound 4, the conjugates 45c or 45d exhibits significant improvement in activity against Gram-negative bacteria, including Escherichia coli, Klebsiella pneumoniae and especially P. aeruginosa (minimum inhibitory concentrations, MICs = 7.8-31.25 μM). The antibacterial activity of the conjugates is attributed to the CSA derivative moiety, and the action mechanism is by disruption of bacterial cell membranes. Further studies on the uptake mechanisms showed that the bacterial siderophore-dependent iron transport system was involved in the uptake of the conjugates. In addition, the conjugates 45c and 45d showed a lower cytotoxic effects in vivo and in vitro and a positive therapeutic effect in the treatment of C. elegans infected by P. aeruginosa. Overall, our work describes a new class and a promising 3-hydroxypyridin-4(1H)-one-CSA derivative conjugates for further development as antibacterial agents against Gram-negative bacteria.
Collapse
Affiliation(s)
- Yong-Jun Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Yi-Peng Zang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Li-Jun Peng
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Ming-Han Yang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Jing Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| | - Wei-Min Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| |
Collapse
|
55
|
Cook GD, Stasulli NM. Employing synthetic biology to expand antibiotic discovery. SLAS Technol 2024; 29:100120. [PMID: 38340893 DOI: 10.1016/j.slast.2024.100120] [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: 06/16/2023] [Revised: 01/04/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Antimicrobial-resistant (AMR) bacterial pathogens are a continually growing threat as our methods for combating these infections continue to be overcome by the evolution of resistance mechanisms. Recent therapeutic methods have not staved off the concern of AMR infections, so continued research focuses on new ways of identifying small molecules to treat AMR pathogens. While chemical modification of existing antibiotics is possible, there has been rapid development of resistance by pathogens that were initially susceptible to these compounds. Synthetic biology is becoming a key strategy in trying to predict and induce novel, natural antibiotics. Advances in cloning and mutagenesis techniques applied through a synthetic biology lens can help characterize the native regulation of antibiotic biosynthetic gene clusters (BGCs) to identify potential modifications leading to more potent antibiotic activity. Additionally, many cryptic antibiotic BGCs are derived from non-ribosomal peptide synthase (NRPS) and polyketide synthase (PKS) biosynthetic pathways; complex, clustered genetic sequences that give rise to amino acid-derived natural products. Synthetic biology can be applied to modify and metabolically engineer these enzyme-based systems to promote rapid and sustainable production of natural products and their variants. This review will focus on recent advances related to synthetic biology as applied to genetic pathway characterization and identification of antibiotics from naturally occurring BGCs. Specifically, we will summarize recent efforts to characterize BGCs via general genomic mutagenesis, endogenous gene expression, and heterologous gene expression.
Collapse
Affiliation(s)
- Greta D Cook
- Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Rd, Dodds Hall 316, West Haven 06516 USA
| | - Nikolas M Stasulli
- Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Rd, Dodds Hall 316, West Haven 06516 USA.
| |
Collapse
|
56
|
Tchinda AM, Nanfack ARD, Tamokou JDD, Matsuete-Takongmo G, Tsopmo A, Ali MS, Tene M. Echinograciolide, a new antibacterial nor-triterpenoid and other constituents from Echinops gracilis O. Hoffm. (Asteraceae). Nat Prod Res 2024; 38:1108-1119. [PMID: 36214723 DOI: 10.1080/14786419.2022.2132497] [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: 05/11/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 10/17/2022]
Abstract
Antibacterial resistance is a serious threat against humankind and the search for new therapeutics is needed. This study aims to investigate the antibacterial activity of extracts and compounds from Echinops gracilis O. Hoffm. Standard chromatographic and spectroscopic methods were used to isolate and characterize compounds (1-15) from the methanol extract. The extract, chromatographic fractions and compounds 1-3, 8, 11, 13 and 14 were subjected to in vitro antibacterial assays against Staphylococcus aureus ATCC25923, Salmonella Typhi ATCC6539, Klebsiella pneumoniae 22, and Salmonella Typhi 68, using broth micro-dilution method. As results, a new nor-triterpenoid (1) and fourteen known compounds (2-15) were characterized. The extract and fractions displayed moderate (128 ≤ MIC ≤ 512 μg/mL) and significant (MIC 64 μg/mL) antibacterial activities. Compounds 1 and 14 showed the best anti-staphylococcal and anti-salmonella activity (MIC 16 µg/mL), respectively. These results partially justified the antimicrobial uses of E. gracilis in traditional medicine.
Collapse
Affiliation(s)
- Arnaud Mbe Tchinda
- Natural Products Chemistry Research Unit, Department of Chemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Arno R Donfack Nanfack
- Natural Products Chemistry Research Unit, Department of Chemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
- International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Jean-De-Dieu Tamokou
- Research Unit of Microbiology and Antimicrobial Substances, Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Germaine Matsuete-Takongmo
- Research Unit of Microbiology and Antimicrobial Substances, Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Apollinaire Tsopmo
- Food Science Program, Department of Chemistry, Carleton University, Ottawa, Canada
| | - Muhammad Shaiq Ali
- International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Mathieu Tene
- Natural Products Chemistry Research Unit, Department of Chemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| |
Collapse
|
57
|
Mohamed-Ezzat RA, Elgemeie GH. Novel synthesis of new triazine sulfonamides with antitumor, anti-microbial and anti-SARS-CoV-2 activities. BMC Chem 2024; 18:58. [PMID: 38532431 DOI: 10.1186/s13065-024-01164-9] [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: 01/14/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Novel approach for synthesizing triazine sulfonamide derivatives is accomplished via reacting the sulfaguanidine derivatives with N-cyanodithioiminocarbonate. Further reaction of the novel triazine sulfonamide analogues with various secondary amines and anilines generated various substituted triazine sulfonamide analogues of promising broad-spectrum activities including anti-microbial, anti-tumor, and anti-viral properties. The in vitro anti-proliferative activities of most of the novel compounds were evaluated on the NCI-60 cell line panel. The antifungal and antibacterial activities of the compounds were also estimated. The anti-viral activity against SARS CoV-2 virus was performed using MTT cytotoxicity assay to evaluate the half-maximal cytotoxic concentration (CC50) and inhibitory concentration 50 (IC50) of a representative compound from the novel triazine sulfonamide category. Compound 3a demonstrated potent antiviral activity against SARS-CoV-2 with IC50 = 2.378 µM as compared to the activity of the antiviral drug remdesivir (IC50 = 10.11 µM). Our results indicate that, upon optimization, these new triazine sulfonamides could potentially serve as novel antiviral drugs.
Collapse
Affiliation(s)
- Reham A Mohamed-Ezzat
- Chemistry of Natural & Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Center, Cairo, Egypt
| | - Galal H Elgemeie
- Department of Chemistry, Faculty of Science, Helwan University, Helwan, Cairo, Egypt.
| |
Collapse
|
58
|
Chen S, Qin S, Li R, Qu Y, Ampomah-Wireko M, Nininahazwe L, Wang M, Gao C, Zhang E. Design, synthesis and antibacterial evaluation of low toxicity amphiphilic-cephalosporin derivatives. Eur J Med Chem 2024; 268:116293. [PMID: 38447461 DOI: 10.1016/j.ejmech.2024.116293] [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: 11/27/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Global public health is facing a serious problem as a result of the rise in antibiotic resistance and the decline in the discovery of new antibiotics. In this study, two series of amphiphilic-cephalosporins were designed and synthesized, several of which showed good antibacterial activity against both Gram-positive and Gram-negative bacteria. Structure-activity relationships indicated that the length of the hydrophobic alkyl chain significantly affects the antibacterial activity against Gram-negative bacteria. The best compound 2d showed high activity against drug-susceptible Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA) with MICs of 0.5 and 2-4 μg/mL, respectively. Furthermore, 2d remained active in complex mammalian body fluids and had a longer post-antibiotic effect (PAE) than vancomycin. Mechanism studies indicated that compound 2d lacks membrane-damaging properties and can target penicillin-binding proteins to disrupt bacterial cell wall structure, inhibit the metabolic activity and induce the accumulation of reactive oxygen species (ROS) in bacteria. Compound 2d showed minimal drug resistance and was nontoxic to HUVEC and HBZY-1 cells with CC50 > 128 μg/mL. These findings suggest that 2d is a promising drug candidate for treating bacterial infections.
Collapse
Affiliation(s)
- Shengcong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Ruirui Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Ye Qu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Lauraine Nininahazwe
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Meng Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Chen Gao
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - En Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China; Pingyuan Laboratory (Zhengzhou University), PR China.
| |
Collapse
|
59
|
Wang L, Huang X, Cao X, Zhou F, Liu B, Wei S, Liu X, Yang X, Yin S. Confining the Growth of AgNPs onto Epigallocatechin Gallate-Decorated Zein Nanoparticles for Constructing Potent Protein-Based Antibacterial Nanocomposites. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4928-4938. [PMID: 38393975 DOI: 10.1021/acs.jafc.3c08029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Sliver nanoparticles (AgNPs) have attracted tremendous interest as an alternative to commercially available antibiotics due to their low microbial resistance and broad-spectrum antimicrobial activity. However, AgNPs are highly reactive and unstable and are susceptible to fast oxidation. Synthesizing stable and efficient AgNPs using green chemistry principles remains a major challenge. To address this issue, we establish a facile route to form AgNP-doped zein nanoparticle core-satellite superstructures with ultralow minimum bactericidal concentration (MBC). In brief, polyphenol surface-functionalization of zein nanoparticles was performed, and the epigallocatechin gallate (EGCG) layer on zein nanoparticles served as a reducing-cum-stabilizing agent. We used EGCG-decorated zein nanoparticles (ZE) as a template to direct the nucleation and growth of AgNPs to develop metallized hybrid nanoparticles (ZE-Ag). The highly monodispersed core-satellite nanoparticles (∼150 nm) decorated with ∼4.9 nm AgNPs were synthesized successfully. The spatial restriction of EGCG by zein nanoparticles confined the nucleation and growth of AgNPs only on the surface of the particles, which prevented the formation of entangled clusters of polyphenols and AgNPs and concomitantly inhibited the coalescence and oxidation of AgNPs. Thus, this strategy improved the effective specific surface area of AgNPs, and as a result, ZE-Ag efficiently killed the indicator bacteria, Escherichia coli (E. coli) and Methicillin-resistant Staphylococcus aureus(MRSA) after 20 min of incubation, with MBCs of 2 and 4 μg/mL, respectively. This situation indicated that as-prepared core-satellite nanoparticles possessed potent short-term sterilization capability. Moreover, the simulated wound infection model also confirmed the promising application of ZE-Ag as an efficient antimicrobial composite. This work provides new insights into the synthesis and emerging application of AgNPs in food preservation, packaging, biomedicine, and catalysis.
Collapse
Affiliation(s)
- Like Wang
- Research and Development Centre of Food Proteins, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products Safety, South China University of Technology, Guangzhou 510640, P. R. China
| | - Xiaonan Huang
- Research and Development Centre of Food Proteins, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products Safety, South China University of Technology, Guangzhou 510640, P. R. China
| | - Xiaoxuan Cao
- Research and Development Centre of Food Proteins, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products Safety, South China University of Technology, Guangzhou 510640, P. R. China
| | - Fuzhen Zhou
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Bo Liu
- Research and Development Centre of Food Proteins, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products Safety, South China University of Technology, Guangzhou 510640, P. R. China
| | - Shuheng Wei
- Research and Development Centre of Food Proteins, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products Safety, South China University of Technology, Guangzhou 510640, P. R. China
| | - Xia Liu
- Research and Development Centre of Food Proteins, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products Safety, South China University of Technology, Guangzhou 510640, P. R. China
| | - Xiaoquan Yang
- Research and Development Centre of Food Proteins, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products Safety, South China University of Technology, Guangzhou 510640, P. R. China
| | - Shouwei Yin
- Research and Development Centre of Food Proteins, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products Safety, South China University of Technology, Guangzhou 510640, P. R. China
| |
Collapse
|
60
|
Shah KN, Shah PN, Agobe FO, Lovato K, Gao H, Ogun O, Hoffman C, Yabe-Gill M, Chen Q, Sweatt J, Chirra B, Muñoz-Medina R, Farmer DE, Kürti L, Cannon CL. Antimicrobial activity of a natural compound and analogs against multi-drug-resistant Gram-positive pathogens. Microbiol Spectr 2024; 12:e0151522. [PMID: 38289721 PMCID: PMC10913730 DOI: 10.1128/spectrum.01515-22] [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: 05/09/2022] [Accepted: 11/06/2023] [Indexed: 02/01/2024] Open
Abstract
The increasing prevalence of methicillin-resistant Staphylococcus aureus (MRSA) has sparked global concern due to the dwindling availability of effective antibiotics. To increase our treatment options, researchers have investigated naturally occurring antimicrobial compounds and have identified MC21-A (C58), which has potent antimicrobial activity against MRSA. Recently, we have devised total synthesis schemes for C58 and its chloro-analog, C59. Here, we report that both compounds eradicate 90% of the 39 MRSA isolates tested [MIC90 and minimum bactericidal concentration (MBC90)] at lower or comparable concentrations compared to several standard-of-care (SoC) antimicrobials including daptomycin, vancomycin, and linezolid. Furthermore, a stable, water-soluble sodium salt of C59, C59Na, demonstrates antimicrobial activity comparable to C59. C59, unlike vancomycin, kills stationary-phase MRSA in a dose-dependent manner and completely eradicates MRSA biofilms. In contrast to vancomycin, exposing MRSA to sub-MIC concentrations of C59 does not result in the emergence of spontaneous resistance. Similarly, in a multi-step study, C59 demonstrates a low propensity of resistance acquisition when compared to SoC antimicrobials, such as linezolid and clindamycin. Our findings suggest C58, C59, and C59Na are non-toxic to mammalian cells at concentrations that exert antimicrobial activity; the lethal dose at median cell viability (LD50) is at least fivefold higher than the MBC90 in the two mammalian cell lines tested. A morphological examination of the effects of C59 on a MRSA isolate suggests the inhibition of the cell division process as a mechanism of action. Our results demonstrate the potential of this naturally occurring compound and its analogs as non-toxic next-generation antimicrobials to combat MRSA infections. IMPORTANCE The rapid emergence of methicillin-resistant Staphylococcus aureus (MRSA) isolates has precipitated a critical need for novel antibiotics. We have developed a one-pot synthesis method for naturally occurring compounds such as MC21-A (C58) and its chloro-analog, C59. Our findings demonstrate that these compounds kill MRSA isolates at lower or comparable concentrations to standard-of-care (SoC) antimicrobials. C59 eradicates MRSA cells in biofilms, which are notoriously difficult to treat with SoC antibiotics. Additionally, the lack of resistance development observed with C59 treatment is a significant advantage when compared to currently available antibiotics. Furthermore, these compounds are non-toxic to mammalian cell lines at effective concentrations. Our findings indicate the potential of these compounds to treat MRSA infections and underscore the importance of exploring natural products for novel antibiotics. Further investigation will be essential to fully realize the therapeutic potential of these next-generation antimicrobials to address the critical issue of antimicrobial resistance.
Collapse
Affiliation(s)
- Kush N. Shah
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Parth N. Shah
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Francesca O. Agobe
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Kaitlyn Lovato
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Hongyin Gao
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Oluwadara Ogun
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Cason Hoffman
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Marium Yabe-Gill
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Qingquan Chen
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Jordan Sweatt
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Bhagath Chirra
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Ricardo Muñoz-Medina
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Delaney E. Farmer
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - László Kürti
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Carolyn L. Cannon
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| |
Collapse
|
61
|
Mourino N, Varela-Lema L, Santiago-Pérez MI, Braun JM, Rey-Brandariz J, Candal-Pedreira C, Pérez-Ríos M. Antibiotic consumption in the first months of life: A cross-sectional study. An Pediatr (Barc) 2024; 100:164-172. [PMID: 38355328 DOI: 10.1016/j.anpede.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 09/20/2023] [Indexed: 02/16/2024] Open
Abstract
INTRODUCTION The factors and patterns associated with antibiotic consumption in infants are unclear. Our aim was to assess the cumulative incidence of antibiotic consumption from birth to 16 months and identify factors associated with antibiotic consumption among infants aged 4-16 months. MATERIAL AND METHODS We conducted a cross-sectional study in 2016 in a sample of 18 882 women from Galicia, Spain, who had given birth to a live child between September 1, 2015 and August 31, 2016. We calculated the cumulative incidence of antibiotic consumption based on maternal reports regarding the infant's consumption from birth to 14 months obtained through interviews; we did not estimate consumption at ages 15 and 16 months due to the small sample size. To assess which factors were associated with antibiotic consumption, we carried out a nested case-control study matching cases and controls for birth month on a 1:1 ratio. RESULTS The cumulative incidence of antibiotic consumption among infants aged 0-14 months increased from 7.5% to 66.0%. The case-control study included data for 1852 cases and 1852 controls. Daycare attendance (OR: 3.8 [95% CI: 3.2-4.6]), having older siblings (OR: 1.8 [95% CI: 1.6-2.1]), health care visits to private clinics (OR: 1.6 [95% CI: 1.4-2.0]), and passive smoking (OR: 1.3 [95% CI: 1.1-1.6]) were associated with an increased probability of antibiotic consumption. Maternal age between 30-39 years or 40 years and over at the time of birth was associated with a decreased probability of antibiotic consumption (OR: 0.8 [95% CI, 0.7-1.0] and OR: 0.6 [95% CI: 0.5-0.8], respectively). CONCLUSIONS Some of the factors associated with antibiotic consumption in infants are modifiable and should be considered in the development of public health measures aimed at reducing antibiotic consumption.
Collapse
Affiliation(s)
- Nerea Mourino
- Área de Medicina Preventiva y Salud Pública, Universidade de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Leonor Varela-Lema
- Área de Medicina Preventiva y Salud Pública, Universidade de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Instituto para el Desarrollo e Integración de la Sanidad (IDIS), Santiago de Compostela, A Coruña, Spain.
| | - María Isolina Santiago-Pérez
- Sección de Epidemiología, Dirección General de Salud Pública de Galicia, Santiago de Compostela, A Coruña, Spain
| | - Joseph M Braun
- Departamento de Epidemiología, Brown University, Providence, RI, United States
| | - Julia Rey-Brandariz
- Área de Medicina Preventiva y Salud Pública, Universidade de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Instituto para el Desarrollo e Integración de la Sanidad (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Cristina Candal-Pedreira
- Área de Medicina Preventiva y Salud Pública, Universidade de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Instituto para el Desarrollo e Integración de la Sanidad (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Mónica Pérez-Ríos
- Área de Medicina Preventiva y Salud Pública, Universidade de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Instituto para el Desarrollo e Integración de la Sanidad (IDIS), Santiago de Compostela, A Coruña, Spain
| |
Collapse
|
62
|
Li LJ, Xu F, Xu JX, Yan Y, Su JQ, Zhu YG, Li H. Spatiotemporal Changes of Antibiotic Resistance, Potential Pathogens, and Health Risk in Kindergarten Dust. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:3919-3930. [PMID: 38353611 DOI: 10.1021/acs.est.3c07935] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
The microorganisms present in kindergartens are extremely important for children's health during their three-year preschool education. To assess the risk of outdoor dust in kindergartens, the antibiotic resistome and potential pathogens were investigated in dust samples collected from 59 kindergartens in Xiamen, southeast China in both the winter and summer. Both high-throughput quantitative PCR and metagenome analysis revealed a higher richness and abundance of antibiotic resistance genes (ARGs) in winter (P < 0.05). Besides, the bloom of ARGs and potential pathogens was evident in the urban kindergartens. The co-occurrence patterns among ARGs, mobile genetic elements (MGEs), and potential pathogens suggested some bacterial pathogens were potential hosts of ARGs and MGEs. We found a large number of high-risk ARGs in the dust; the richness and abundance of high-risk ARGs were higher in winter and urban kindergartens compared to in summer and peri-urban kindergartens, respectively. The results of the co-occurrence patterns and high-risk ARGs jointly reveal that urbanization will significantly increase the threat of urban dust to human beings and their risks will be higher in winter. This study unveils the close association between ARGs/mobile ARGs and potential pathogens and emphasizes that we should pay more attention to the health risks induced by their combination.
Collapse
Affiliation(s)
- Li-Juan Li
- Key Laboratory of Urban Environment and Health, Ningbo Observation and Research Station, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Fei Xu
- Key Laboratory of Urban Environment and Health, Ningbo Observation and Research Station, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Jian-Xin Xu
- Department of Environmental and Resource Engineering, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Yu Yan
- Department of Environmental Science and Engineering, Huaqiao University, Xiamen 361021, China
| | - Jian-Qiang Su
- Key Laboratory of Urban Environment and Health, Ningbo Observation and Research Station, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Yong-Guan Zhu
- Key Laboratory of Urban Environment and Health, Ningbo Observation and Research Station, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
- State Key Laboratory of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Hu Li
- Key Laboratory of Urban Environment and Health, Ningbo Observation and Research Station, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
63
|
Yan Q, Wang F, Zhou B, Lin X. Hybrid 2D/3D-quantitative structure-activity relationship studies on the bioactivities and molecular mechanism of antibacterial peptides. Amino Acids 2024; 56:16. [PMID: 38358574 PMCID: PMC10869384 DOI: 10.1007/s00726-024-03381-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
Antimicrobial peptide (AMP) is the polypeptide, which protects the organism avoiding attack from pathogenic bacteria. Studies have shown that there were some antimicrobial peptides with molecular action mechanism involved in crossing the cell membrane without inducing severe membrane collapse, then interacting with cytoplasmic target-nucleic acid, and exerting antibacterial activity by interfacing the transmission of genetic information of pathogenic microorganisms. However, the relationship between the antibacterial activities and peptide structures was still unclear. Therefore, in the present work, a series of AMPs with a sequence of 20 amino acids was extracted from DBAASP database, then, quantitative structure-activity relationship (QSAR) methods were conducted on these peptides. In addition, novel antimicrobial peptides with stronger antimicrobial activities were designed according to the information originated from the constructed models. Hence, the outcome of this study would lay a solid foundation for the in-silico design and exploration of novel antibacterial peptides with improved activity activities.
Collapse
Affiliation(s)
- Qingguo Yan
- School of Life Science, Linyi University, Linyi, 276000, China
| | - Fangfang Wang
- School of Life Science, Linyi University, Linyi, 276000, China.
| | - Bo Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, College of Basic Medical, Guizhou Medical University, Guizhou, 550004, China
| | - Xiangna Lin
- School of Life Science, Linyi University, Linyi, 276000, China
| |
Collapse
|
64
|
Guo W, Yang Z, Wang K, Li W, Zhao Y, Yang Y, Chang W, Gong Z, Liu Z, Chen Y, Li Q. Discovery of Unique Bis-Substituted Aromatic Amide Derivatives as Novel Highly Potent Antibiotics for Combating Methicillin-Resistant Staphylococcus aureus (MRSA). J Med Chem 2024; 67:2129-2151. [PMID: 38289145 DOI: 10.1021/acs.jmedchem.3c02064] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Due to the increasing antibiotic resistance, developing novel antimicrobials to fight infections caused by resistant bacteria is imperative. Herein, a series of novel bis-substituted aromatic amides were designed and synthesized through modifying the hit compound 1, and their antimicrobial activities were evaluated. Among them, compound 4t, as the most potent lead, exhibited excellent antimicrobial activities against Gram-positive bacteria, including clinical methicillin-resistant Staphylococcus aureus (MRSA) isolates, while keeping weak hemolytic and mammalian cytotoxic activities. Furthermore, compound 4t displayed rapid bactericidal capabilities, low tendency to produce resistance, and favorable capacities to destroy bacterial biofilms. Further explorations indicated that compound 4t induces bacterial death by binding to cardiolipin (CL) on the bacterial membrane, disrupting the cell membrane, and facilitating the accumulation of reactive oxygen species (ROS). Additionally, compound 4t showed remarkable anti-MRSA activity in vivo, demonstrating compound 4t could be developed as a potential candidate to combat MRSA infections.
Collapse
Affiliation(s)
- Weikai Guo
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Zhengfan Yang
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Kexiao Wang
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Wenyu Li
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Yanyang Zhao
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Yuqing Yang
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Wenjing Chang
- Henan Provincial Center for Disease Control and Prevention, Zhengzhou 450003, China
| | - Zhen Gong
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhou Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yihua Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qiming Li
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| |
Collapse
|
65
|
Weigert Muñoz A, Zhao W, Sieber SA. Monitoring host-pathogen interactions using chemical proteomics. RSC Chem Biol 2024; 5:73-89. [PMID: 38333198 PMCID: PMC10849124 DOI: 10.1039/d3cb00135k] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/09/2023] [Indexed: 02/10/2024] Open
Abstract
With the rapid emergence and the dissemination of microbial resistance to conventional chemotherapy, the shortage of novel antimicrobial drugs has raised a global health threat. As molecular interactions between microbial pathogens and their mammalian hosts are crucial to establish virulence, pathogenicity, and infectivity, a detailed understanding of these interactions has the potential to reveal novel therapeutic targets and treatment strategies. Bidirectional molecular communication between microbes and eukaryotes is essential for both pathogenic and commensal organisms to colonise their host. In particular, several devastating pathogens exploit host signalling to adjust the expression of energetically costly virulent behaviours. Chemical proteomics has emerged as a powerful tool to interrogate the protein interaction partners of small molecules and has been successfully applied to advance host-pathogen communication studies. Here, we present recent significant progress made by this approach and provide a perspective for future studies.
Collapse
Affiliation(s)
- Angela Weigert Muñoz
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 D-85748 Garching Germany
| | - Weining Zhao
- College of Pharmacy, Shenzhen Technology University Shenzhen 518118 China
| | - Stephan A Sieber
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 D-85748 Garching Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Germany
| |
Collapse
|
66
|
Chery-Karschney L, Patrapuvich R, Mudeppa DG, Kokkonda S, Chakrabarti R, Sriwichai P, O'Connor RM, Rathod PK, White J. Tartrolon E, a secondary metabolite of a marine symbiotic bacterium, is a potent inhibitor of asexual and sexual Plasmodium falciparum. Antimicrob Agents Chemother 2024; 68:e0068423. [PMID: 38193705 PMCID: PMC10848769 DOI: 10.1128/aac.00684-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/10/2023] [Indexed: 01/10/2024] Open
Abstract
Due to the spread of resistance to front-line artemisinin derivatives worldwide, there is a need for new antimalarials. Tartrolon E (TrtE), a secondary metabolite of a symbiotic bacterium of marine bivalve mollusks, is a promising antimalarial because it inhibits the growth of sexual and asexual blood stages of Plasmodium falciparum at sub-nanomolar levels. The potency of TrtE warrants further investigation into its mechanism of action, cytotoxicity, and ease with which parasites may evolve resistance to it.
Collapse
Affiliation(s)
| | - Rapatbhorn Patrapuvich
- Drug Research Unit for Malaria, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Sreekanth Kokkonda
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Rimi Chakrabarti
- Department of Chemistry, University of Washington, Seattle, Washington, USA
- Department of Medicine, Goa Medical College and Hospital, Bambolim, Goa, India
| | - Patchara Sriwichai
- Department of Medical Entomology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Roberta M. O'Connor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - John White
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| |
Collapse
|
67
|
Couturier C, Ronzon Q, Lattanzi G, Lingard I, Coyne S, Cazals V, Dubarry N, Yvon S, Leroi-Geissler C, Gracia OR, Teague J, Sordello S, Corbett D, Bauch C, Monlong C, Payne L, Taillier T, Fuchs H, Broenstrup M, Harrison PH, Moynié L, Lakshminarayanan A, Gianga TM, Hussain R, Naismith JH, Mourez M, Bacqué E, Björkling F, Sabuco JF, Franzyk H. Studies of antibacterial activity (in vitro and in vivo) and mode of action for des-acyl tridecaptins (DATs). Eur J Med Chem 2024; 265:116097. [PMID: 38157595 DOI: 10.1016/j.ejmech.2023.116097] [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: 11/09/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Tridecaptins comprise a class of linear cationic lipopeptides with an N-terminal fatty acyl moiety. These 13-mer antimicrobial peptides consist of a combination of d- and l-amino acids, conferring increased proteolytic stability. Intriguingly, they are biosynthesized by non-ribosomal peptide synthetases in the same bacterial species that also produce the cyclic polymyxins displaying similar fatty acid tails. Previously, the des-acyl analog of TriA1 (termed H-TriA1) was found to possess very weak antibacterial activity, albeit it potentiated the effect of several antibiotics. In the present study, two series of des-acyl tridecaptins were explored with the aim of improving the direct antibacterial effect. At the same time, overall physico-chemical properties were modulated by amino acid substitution(s) to diminish the risk of undesired levels of hemolysis and to avoid an impairment of mammalian cell viability, since these properties are typically associated with highly hydrophobic cationic peptides. Microbiology and biophysics tools were used to determine bacterial uptake, while circular dichroism and isothermal calorimetry were used to probe the mode of action. Several analogs had improved antibacterial activity (as compared to that of H-TriA1) against Enterobacteriaceae. Optimization enabled identification of the lead compound 29 that showed a good ADMET profile as well as in vivo efficacy in a variety of mouse models of infection.
Collapse
Affiliation(s)
- Cédric Couturier
- Evotec, 1541, Avenue Marcel Mérieux, 69280, Marcy L'Etoile, France.
| | - Quentin Ronzon
- Evotec, 1541, Avenue Marcel Mérieux, 69280, Marcy L'Etoile, France
| | - Giulia Lattanzi
- Evotec-Aptuit (Verona) Srl, Via Alessandro Fleming 4, 37135, Verona, Italy
| | - Iain Lingard
- Evotec-Aptuit (Verona) Srl, Via Alessandro Fleming 4, 37135, Verona, Italy
| | | | | | | | | | | | | | - Joanne Teague
- Evotec, No. 23F, Mereside, Alderley Park, Cheshire, SK10 4TG, United Kingdom
| | | | - David Corbett
- Evotec, No. 23F, Mereside, Alderley Park, Cheshire, SK10 4TG, United Kingdom
| | - Caroline Bauch
- Evotec-Cyprotex, No. 24, Mereside, Alderley Park, Cheshire, SK10 4TG, United Kingdom
| | | | - Lloyd Payne
- Evotec, No. 23F, Mereside, Alderley Park, Cheshire, SK10 4TG, United Kingdom
| | | | - Hazel Fuchs
- Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Mark Broenstrup
- Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Peter H Harrison
- Division of Structural Biology, Wellcome Trust Centre of Human Genomics, 7 Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Lucile Moynié
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Didcot, OX11 0QS, United Kingdom
| | - Abirami Lakshminarayanan
- Division of Structural Biology, Wellcome Trust Centre of Human Genomics, 7 Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Tiberiu-Marius Gianga
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0DE, United Kingdom
| | - Rohanah Hussain
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0DE, United Kingdom
| | - James H Naismith
- Division of Structural Biology, Wellcome Trust Centre of Human Genomics, 7 Roosevelt Drive, Oxford, OX3 7BN, United Kingdom; Rosalind Franklin Institute, Harwell Science and Innovation Campus, Didcot, OX11 0QS, United Kingdom
| | | | - Eric Bacqué
- Evotec, 1541, Avenue Marcel Mérieux, 69280, Marcy L'Etoile, France
| | - Fredrik Björkling
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Denmark
| | | | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Denmark
| |
Collapse
|
68
|
Zhao X, Verma R, Sridhara MB, Sharath Kumar KS. Fluorinated azoles as effective weapons in fight against methicillin-resistance staphylococcus aureus (MRSA) and its SAR studies. Bioorg Chem 2024; 143:106975. [PMID: 37992426 DOI: 10.1016/j.bioorg.2023.106975] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/22/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023]
Abstract
The rapid spread of Methicillin-resistant Staphylococcus aureus (MRSA) and its difficult-to-treat skin and filmsy diseases are making MRSA a threat to human life. The most dangerous feature is the fast emergence of MRSA resistance to all recognized antibiotics, including vancomycin. The creation of novel, effective, and non-toxic drug candidates to combat MRSA isolates is urgently required. Fluorine containing small molecules have taken a centre stage in the field of drug development. Over the last 50 years, there have been a growing number of fluorinated compounds that have been approved since the clinical usage of fluorinated corticosteroids in the 1950 s and fluoroquinolones in the 1980 s. Due to its advantages in terms of potency and ADME (absorption, distribution, metabolism, and excretion), fluoro-pharmaceuticals have been regarded as a potent and useful tool in the rational drug design method. The flexible bioactive fluorinated azoles are ideal candidates for the development of new antibiotics. This review summarizes the decade developments of fluorinated azole derivatives with a wide antibacterial activity against diverged MRSA strains. In specific, we correlated the efficacy of structurally varied fluorinated azole analogues including thiazole, benzimidazole, oxadiazole and pyrazole against MRSA and discussed different angles of structure-activity relationship (SAR).
Collapse
Affiliation(s)
- Xuanming Zhao
- Energy Engineering College, Yulin University, Yulin City-719000, P. R. China
| | - Rameshwari Verma
- School of New Energy, Yulin University, Yulin 719000, Shaanxi, P. R. China
| | - M B Sridhara
- Department of Chemistry, Rani Channamma University, Vidyasangama, Belagavi 591156, India
| | | |
Collapse
|
69
|
Lavekar AG, Thakare R, Saima, Equbal D, Chopra S, Sinha AK. Indole-based aryl sulfides target the cell wall of Staphylococcus aureus without detectable resistance. Drug Dev Res 2024; 85:e22123. [PMID: 37840429 DOI: 10.1002/ddr.22123] [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: 07/10/2023] [Revised: 09/05/2023] [Accepted: 10/02/2023] [Indexed: 10/17/2023]
Abstract
Sulfur-containing classes of the scaffold "Arylthioindoles" have been evaluated for antibacterial activity; they demonstrated excellent potency against methicillin-resistant Staphylococcus aureus (MRSA) as well as against vancomycin-resistant strains and a panel of clinical isolates of resistant strains. In this study, we have elucidated the mechanism of action of lead compounds, wherein they target the cell wall of S. aureus. Further, S. aureus failed to develop resistance against two lead compounds tested in a serial passage experiment in the presence of the compounds over a period of 40 days. Both the compounds demonstrated comparable in vivo efficacy with vancomycin in a neutropenic mice thigh infection model. The results of these antibacterial activities emphasize the excellent potential of thioethers for developing novel antibiotics and may fill in as a target for the adjustment of accessible molecules to develop new powerful antibacterial agents with fewer side effects.
Collapse
Affiliation(s)
- Aditya G Lavekar
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Ritesh Thakare
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Department of Cell and Cancer Biology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Saima
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- School of Advanced Chemical Sciences, Solan, Himachal Pradesh, India
| | - Danish Equbal
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Sidharth Chopra
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Arun K Sinha
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
- Ranchi University, Ranchi, Jharkhand, India
| |
Collapse
|
70
|
Yue C, Yuan Z, Xu G, Guan XN, Wei B, Yao H, Yang CG, Zhang T. Structure-Guided Design, Synthesis, and Antivirulence Assessment of Covalent Staphylococcus aureus Sortase A Inhibitors. J Med Chem 2024; 67:1127-1146. [PMID: 38170998 DOI: 10.1021/acs.jmedchem.3c01615] [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: 01/05/2024]
Abstract
Sortase A (SrtA) is a membrane-associated cysteine transpeptidase required for bacterial virulence regulation and anchors surface proteins to cell wall, thereby assisting biofilm formation. SrtA is targeted in antivirulence treatments against Gram-positive bacterial infections. However, the development of potent small-molecule SrtA inhibitors is constrained owing to the limited understanding of the mode of action of inhibitors in the SrtA binding pocket. Herein, we designed and synthesized a novel class of covalent SrtA inhibitors based on the binding mode detailed in the X-ray crystal structure of the ML346/Streptococcus pyogenes SrtA complex. ML346 analog Y40 exhibited 2-fold increased inhibitory activity on Staphylococcus aureus SrtA and showed superior inhibitory effects on biofilm formation in vitro. Y40 protected Galleria mellonella larvae fromS. aureusinfections in vivo while minimally attenuating staphylococcal growth in vitro. Our study indicates that the covalent SrtA inhibitor Y40 is an antivirulence agent that is effective againstS. aureusinfections.
Collapse
Affiliation(s)
- Chuan Yue
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ziqi Yuan
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guobin Xu
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiang-Na Guan
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bingyan Wei
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Hequan Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Cai-Guang Yang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Tao Zhang
- State Key Laboratory of Drug Research, Centre for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
71
|
Olcay B, Ozdemir GD, Ozdemir MA, Ercan UK, Guren O, Karaman O. Prediction of the synergistic effect of antimicrobial peptides and antimicrobial agents via supervised machine learning. BMC Biomed Eng 2024; 6:1. [PMID: 38233957 DOI: 10.1186/s42490-024-00075-z] [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: 03/24/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Infectious diseases not only cause severe health problems but also burden the healthcare system. Therefore, the effective treatment of those diseases is crucial. Both conventional approaches, such as antimicrobial agents, and novel approaches, like antimicrobial peptides (AMPs), are used to treat infections. However, due to the drawbacks of current approaches, new solutions are still being investigated. One recent approach is the use of AMPs and antimicrobial agents in combination, but determining synergism is with a huge variety of AMPs time-consuming and requires multiple experimental studies. Machine learning (ML) algorithms are widely used to predict biological outcomes, particularly in the field of AMPs, but no previous research reported on predicting the synergistic effects of AMPs and antimicrobial agents. RESULTS Several supervised ML models were implemented to accurately predict the synergistic effect of AMPs and antimicrobial agents. The results demonstrated that the hyperparameter-optimized Light Gradient Boosted Machine Classifier (oLGBMC) yielded the best test accuracy of 76.92% for predicting the synergistic effect. Besides, the feature importance analysis reveals that the target microbial species, the minimum inhibitory concentrations (MICs) of the AMP and the antimicrobial agents, and the used antimicrobial agent were the most important features for the prediction of synergistic effect, which aligns with recent experimental studies in the literature. CONCLUSION This study reveals that ML algorithms can predict the synergistic activity of two different antimicrobial agents without the need for complex and time-consuming experimental procedures. The implications support that the ML models may not only reduce the experimental cost but also provide validation of experimental procedures.
Collapse
Affiliation(s)
- Basak Olcay
- Department of Biomedical Engineering, Graduate School of Natural and Applied Sciences, Izmir Katip Celebi University, Izmir, Turkey
| | - Gizem D Ozdemir
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Izmir, Turkey
| | - Mehmet A Ozdemir
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Izmir, Turkey.
| | - Utku K Ercan
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Izmir, Turkey
| | - Onan Guren
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Izmir, Turkey
| | - Ozan Karaman
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Izmir, Turkey
| |
Collapse
|
72
|
Chen T, Yao J, Quan K, Xu J, Hang X, Tong Q, Liu G, Luo P, Zeng L, Feng G, Bi H. Repurposing a human acetyl-CoA carboxylase inhibitor firsocostat to treat fungal candidiasis alone and in combination. Antimicrob Agents Chemother 2024; 68:e0113123. [PMID: 38018962 PMCID: PMC10777831 DOI: 10.1128/aac.01131-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/15/2023] [Indexed: 11/30/2023] Open
Abstract
Opportunistic fungal infections, particularly caused by Candida albicans, remain a common cause of high morbidity and mortality in immunocompromised patients. The escalating prevalence of antifungal drug resistance necessitates the immediate exploration of alternative treatment strategies to combat these life-threatening fungal diseases. In this study, we investigated the antifungal efficacy of firsocostat, a human acetyl-CoA carboxylase (ACC) inhibitor, against C. albicans. Firsocostat alone displayed moderate antifungal activity, while combining it with voriconazole, itraconazole, or amphotericin B exhibited synergistic effects across almost all drug-sensitive and drug-resistant C. albicans strains tested. These observed synergies were further validated in two mouse models of oropharyngeal and systemic candidiasis, where the combination therapies demonstrated superior fungicidal effects compared to monotherapy. Moreover, firsocostat was shown to directly bind to C. albicans ACC and inhibit its enzymatic activity. Sequencing spontaneous firsocostat-resistant mutants revealed mutations mapping to C. albicans ACC, confirming that firsocostat has retained its target in C. albicans. Overall, our findings suggest that repurposing firsocostat, either alone or in combination with other antifungal agents, holds promising potential in the development of antifungal drugs and the treatment of candidiasis.
Collapse
Affiliation(s)
- Tianyu Chen
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, Jiangsu, China
| | - Jing Yao
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Keao Quan
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingchen Xu
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xudong Hang
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Tong
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Genyan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peipei Luo
- Department of Gastroenterology, Wujin People’s Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu, China
| | - Liping Zeng
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ganzhu Feng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongkai Bi
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
73
|
Han S, Shin R, Ryu SH, Unno T, Hur HG, Shin H. A Potential Indicator Gene, tetM, to Assess Contamination by Antibiotic Resistance Genes in Greenhouses in South Korea. Microbes Environ 2024; 39:ME24053. [PMID: 39756985 PMCID: PMC11821766 DOI: 10.1264/jsme2.me24053] [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: 06/23/2024] [Accepted: 09/07/2024] [Indexed: 01/07/2025] Open
Abstract
Antibiotic resistance genes (ARGs) have been emerging as a concerning threat to both environment and public health. The continuous input of manure, irrigation water, and fertilizers increases the abundance of ARGs in agricultural environments. However, current risk assessments have focused on clinical settings, which are not applicable to environmental settings. Therefore, we herein aimed to identify and assess indicator genes to reduce the time and effort required for ARG surveillance. A nationwide ana-lysis of 322 ARGs and 58 mobile genetic elements (MGEs) was performed on 42 greenhouse and 19 control soil samples. The chemical properties and pH of soil were also investigated to characterize differences between greenhouse and control soil samples. The results obtained showed that the abundance of ARGS was significantly higher and ion concentrations were higher in greenhouse samples than in control samples. These results indicate that agricultural activities increased the abundance of ARGs. Furthermore, the abundance of core genes was significantly higher in greenhouse samples than in control samples, and the chemical characteristics of soil significantly differed between these samples. Among the discriminatory genes selected, tetM was identified as an ARG surveillance indicator gene based on its clinical relevance, prevalence in the soil resistome, and relationship with mobile genetic elements. The present results will contribute to the continuous and rapid surveillance of antibiotic resistance dissemination and proliferation in greenhouses in South Korea.
Collapse
Affiliation(s)
- Seunggyun Han
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Raan Shin
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Song-Hee Ryu
- Residual Agrochemical Assessment Division, National Institute of Agricultural Sciences, Wanju-gun, South Korea
| | - Tatsuya Unno
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Republic of Korea
| | - Hor-Gil Hur
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hanseob Shin
- Center for Health Effects of Environmental Contamination, University of Iowa, W195 Chemistry Building, University of Iowa, Iowa city, Iowa, United States
- State Hygienic Laboratory, University of Iowa, Coralville, Iowa, United States
| |
Collapse
|
74
|
Nenni M, Karahuseyin S. Medicinal Plants, Secondary Metabolites, and Their Antiallergic Activities. BIOTECHNOLOGY OF MEDICINAL PLANTS WITH ANTIALLERGY PROPERTIES 2024:37-126. [DOI: 10.1007/978-981-97-1467-4_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
75
|
Renard S, Versluys S, Taillier T, Dubarry N, Leroi-Geissler C, Rey A, Cornaire E, Sordello S, Carry JCB, Angouillant-Boniface O, Gouyon T, Thompson F, Lebourg G, Certal V, Balazs L, Arranz E, Doerflinger G, Bretin F, Gervat V, Brohan E, Kraft V, Boulenc X, Ducelier C, Bacqué E, Couturier C. Optimization of the Antibacterial Spectrum and the Developability Profile of the Novel-Class Natural Product Corramycin. J Med Chem 2023; 66:16869-16887. [PMID: 38088830 DOI: 10.1021/acs.jmedchem.3c01564] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Corramycin 1 is a novel zwitterionic antibacterial peptide isolated from a culture of the myxobacterium Corallococcus coralloides. Though Corramycin displayed a narrow spectrum and modest MICs against sensitive bacteria, its ADMET and physchem profile as well as its high tolerability in mice along with an outstanding in vivo efficacy in an Escherichia coli septicemia mouse model were promising and prompted us to embark on an optimization program aiming at enlarging the spectrum and at increasing the antibacterial activities by modulating membrane permeability. Scanning the peptidic moiety by the Ala-scan strategy followed by key stabilization and introduction of groups such as a primary amine or siderophore allowed us to enlarge the spectrum and increase the overall developability profile. The optimized Corramycin 28 showed an improved mouse IV PK and a broader spectrum with high potency against key Gram-negative bacteria that translated into excellent efficacy in several in vivo mouse infection models.
Collapse
Affiliation(s)
| | | | - Thomas Taillier
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| | | | | | - Astrid Rey
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| | - Emilie Cornaire
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| | | | | | | | - Thierry Gouyon
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | | | - Gilles Lebourg
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Victor Certal
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Laszlo Balazs
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Esther Arranz
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | | | | | - Vincent Gervat
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Eric Brohan
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Volker Kraft
- Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, Frankfurt am Main 65926, Germany
| | | | - Cécile Ducelier
- Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin 91385, France
| | - Eric Bacqué
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| | - Cédric Couturier
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| |
Collapse
|
76
|
Hofkens N, Gestels Z, Abdellati S, De Baetselier I, Gabant P, Martin A, Kenyon C, Manoharan-Basil SS. Microbisporicin (NAI-107) protects Galleria mellonella from infection with Neisseria gonorrhoeae. Microbiol Spectr 2023; 11:e0282523. [PMID: 37823634 PMCID: PMC10715042 DOI: 10.1128/spectrum.02825-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE We screened 66 bacteriocins to see if they exhibited anti-gonococcal activity. We found 12 bacteriocins with anti-gonococcal effects, and 4 bacteriocins showed higher anti-gonococcal activity. Three bacteriocins, lacticin Z, lacticin Q, and Garvicin KS (ABC), showed in vitro anti-gonococcal activity but no in vivo inhibitory effects against the Neisseria gonorrhoeae (WHO-P) isolate. On the other hand, NAI-107 showed in vivo anti-gonococcal activity. The findings suggest that NAI-107 is a promising alternative to treat gonorrhea infections.
Collapse
Affiliation(s)
- Nele Hofkens
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Zina Gestels
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Said Abdellati
- Clinical Reference Laboratory, Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Irith De Baetselier
- Clinical Reference Laboratory, Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | | | | | - Christopher Kenyon
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | | |
Collapse
|
77
|
Duan S, Su H, Xu W, Hu X, Xu Y, Cao Y, Wen G. Concentrations, distribution, and key influencing factors of antibiotic resistance genes and bacterial community in water and reared fish tissues in a typical tilapia farm in South China. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART. B, PESTICIDES, FOOD CONTAMINANTS, AND AGRICULTURAL WASTES 2023; 59:21-35. [PMID: 38009809 DOI: 10.1080/03601234.2023.2284617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Although previous studies have investigated the occurrence of antibiotic resistance genes (ARGs) in aquaculture, few have monitored the concentrations and propagation of ARGs in biological tissues or investigated the key factors influencing their spread in aquaculture. This study investigated the concentration, propagation, and distribution of ARGs and bacterial communities in water sources, pond water, and tilapia tissues, and their key influencing factors, in a typical tilapia farm. ErmF, sul1, and sul2 were the dominant ARGs with high concentrations. The total concentrations of ARGs (TCAs) in tilapia tissues decreased in the following order: stomach > scales > intestine > gills (P < 0.05). Redundancy analysis and multiple linear regression revealed that suspended solids (SS) and chemical oxygen demand (COD) were positively correlated with the dominant ARGs ermF sul2, and the TCAs (P < 0.05); additionally, Chloroflexi and Bacteroidetes in tilapia aquaculture water were positively correlated with the dominant ARGs ermF and sul2, as well as the TCAs (P < 0.05). This study suggests that SS and COD were the key factors driving the distribution and spread of ARGs in tilapia aquaculture water. Additionally, Chloroflexi and Bacteroidetes were the key bacterial flora affecting the propagation of ARGs in tilapia aquaculture systems.
Collapse
Affiliation(s)
- Sijia Duan
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan, China
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Beijing, China
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Haochang Su
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan, China
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Beijing, China
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| | - Wujie Xu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Beijing, China
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| | - Xiaojuan Hu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Beijing, China
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| | - Yu Xu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Beijing, China
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| | - Yucheng Cao
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Beijing, China
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| | - Guoliang Wen
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan, China
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Beijing, China
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| |
Collapse
|
78
|
Tang L, Zhou S, Li F, Sun L, Lu H. Ozone Micronano-bubble-Enhanced Selective Degradation of Oxytetracycline from Production Wastewater: The Overlooked Singlet Oxygen Oxidation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:18550-18562. [PMID: 36474357 DOI: 10.1021/acs.est.2c06008] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The efficient and selective removal of refractory antibiotics from high-strength antibiotic production wastewater is crucial but remains a substantial challenge. In this study, a novel ozone micronano-bubble (MNB)-enhanced treatment system was constructed for antibiotic production wastewater treatment. Compared with conventional ozone, ozone MNBs exhibit excellent treatment efficiency for oxytetracycline (OTC) degradation and toxicity decrease. Notably, this study identifies the overlooked singlet oxygen (1O2) for the first time as a crucial active species in the ozone MNB system through probe and electron paramagnetic resonance methods. Subsequently, the oxidation mechanisms of OTC by ozone MNBs are systematically investigated. Owing to the high reactivity of OTC toward 1O2, ozone MNBs enhance the selective and anti-interference performance of OTC degradation in raw OTC production wastewater with complex matrixes. This study provides insights into the mechanism of ozone MNB-enhanced pollutant degradation and a new perspective for the efficient treatment of high-concentration industrial wastewater using ozone MNBs. In addition, this study presents a promising technology with scientific guidance for the treatment of antibiotic production wastewater.
Collapse
Affiliation(s)
- Lan Tang
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou510275, China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou510275, China
| | - Sining Zhou
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou510275, China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou510275, China
| | - Fan Li
- Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Institute of Environmental Research at Greater Bay, Guangzhou University, Guangzhou510006, China
| | - Lianpeng Sun
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou510275, China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou510275, China
| | - Hui Lu
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou510275, China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou510275, China
| |
Collapse
|
79
|
Muteeb G, Rehman MT, Shahwan M, Aatif M. Origin of Antibiotics and Antibiotic Resistance, and Their Impacts on Drug Development: A Narrative Review. Pharmaceuticals (Basel) 2023; 16:1615. [PMID: 38004480 PMCID: PMC10675245 DOI: 10.3390/ph16111615] [Citation(s) in RCA: 138] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Antibiotics have revolutionized medicine, saving countless lives since their discovery in the early 20th century. However, the origin of antibiotics is now overshadowed by the alarming rise in antibiotic resistance. This global crisis stems from the relentless adaptability of microorganisms, driven by misuse and overuse of antibiotics. This article explores the origin of antibiotics and the subsequent emergence of antibiotic resistance. It delves into the mechanisms employed by bacteria to develop resistance, highlighting the dire consequences of drug resistance, including compromised patient care, increased mortality rates, and escalating healthcare costs. The article elucidates the latest strategies against drug-resistant microorganisms, encompassing innovative approaches such as phage therapy, CRISPR-Cas9 technology, and the exploration of natural compounds. Moreover, it examines the profound impact of antibiotic resistance on drug development, rendering the pursuit of new antibiotics economically challenging. The limitations and challenges in developing novel antibiotics are discussed, along with hurdles in the regulatory process that hinder progress in this critical field. Proposals for modifying the regulatory process to facilitate antibiotic development are presented. The withdrawal of major pharmaceutical firms from antibiotic research is examined, along with potential strategies to re-engage their interest. The article also outlines initiatives to overcome economic challenges and incentivize antibiotic development, emphasizing international collaborations and partnerships. Finally, the article sheds light on government-led initiatives against antibiotic resistance, with a specific focus on the Middle East. It discusses the proactive measures taken by governments in the region, such as Saudi Arabia and the United Arab Emirates, to combat this global threat. In the face of antibiotic resistance, a multifaceted approach is imperative. This article provides valuable insights into the complex landscape of antibiotic development, regulatory challenges, and collaborative efforts required to ensure a future where antibiotics remain effective tools in safeguarding public health.
Collapse
Affiliation(s)
- Ghazala Muteeb
- Department of Nursing, College of Applied Medical Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Md Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11437, Saudi Arabia;
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates;
| | - Moayad Shahwan
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates;
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates
| | - Mohammad Aatif
- Department of Public Health, College of Applied Medical Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| |
Collapse
|
80
|
Colilla M, Vallet-Regí M. Organically Modified Mesoporous Silica Nanoparticles against Bacterial Resistance. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2023; 35:8788-8805. [PMID: 38027542 PMCID: PMC10653088 DOI: 10.1021/acs.chemmater.3c02192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/22/2023] [Indexed: 12/01/2023]
Abstract
Bacterial antimicrobial resistance is posed to become a major hazard to global health in the 21st century. An aggravating issue is the stalled antibiotic research pipeline, which requires the development of new therapeutic strategies to combat antibiotic-resistant infections. Nanotechnology has entered into this scenario bringing up the opportunity to use nanocarriers capable of transporting and delivering antimicrobials to the target site, overcoming bacterial resistant barriers. Among them, mesoporous silica nanoparticles (MSNs) are receiving growing attention due to their unique features, including large drug loading capacity, biocompatibility, tunable pore sizes and volumes, and functionalizable silanol-rich surface. This perspective article outlines the recent research advances in the design and development of organically modified MSNs to fight bacterial infections. First, a brief introduction to the different mechanisms of bacterial resistance is presented. Then, we review the recent scientific approaches to engineer multifunctional MSNs conceived as an assembly of inorganic and organic building blocks, against bacterial resistance. These elements include specific ligands to target planktonic bacteria, intracellular bacteria, or bacterial biofilm; stimuli-responsive entities to prevent antimicrobial cargo release before arriving at the target; imaging agents for diagnosis; additional constituents for synergistic combination antimicrobial therapies; and aims to improve the therapeutic outcomes. Finally, this manuscript addresses the current challenges and future perspectives on this hot research area.
Collapse
Affiliation(s)
- Montserrat Colilla
- Departamento
de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación
Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - María Vallet-Regí
- Departamento
de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación
Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
81
|
Kim MJ, Kang D, Lee G, Kim K, Kim J, Shin JH, Lee S. Interplays between cyanobacterial blooms and antibiotic resistance genes. ENVIRONMENT INTERNATIONAL 2023; 181:108268. [PMID: 37897871 DOI: 10.1016/j.envint.2023.108268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/04/2023] [Accepted: 10/13/2023] [Indexed: 10/30/2023]
Abstract
Cyanobacterial harmful algal blooms (cyanoHABs), which are a form of microbial dysbiosis in freshwater environments, are an emerging environmental and public health concern. Additionally, the freshwater environment serves as a reservoir of antibiotic resistance genes (ARGs), which pose a risk of transmission during microbial dysbiosis, such as cyanoHABs. However, the interactions between potential synergistic pollutants, cyanoHABs, and ARGs remain poorly understood. During cyanoHABs, Microcystis and high microcystin levels were dominant in all the nine regions of the river sampled. The resistome, mobilome, and microbiome were interrelated and linked to the physicochemical properties of freshwater. Planktothrix and Pseudanabaena competed with Actinobacteriota and Proteobacteria during cyanoHABs. Forty two ARG carriers were identified, most of which belonged to Actinobacteriota and Proteobacteria. ARG carriers showed a strong correlation with ARGs density, which decreased with the severity of cyanoHAB. Although ARGs decreased due to a reduction of ARG carriers during cyanoHABs, mobile gene elements (MGEs) and virulence factors (VFs) genes increased. We explored the relationship between cyanoHABs and ARGs for potential synergistic interaction. Our findings demonstrated that cyanobacteria compete with freshwater commensal bacteria such as Actinobacteriota and Proteobacteria, which carry ARGs in freshwater, resulting in a reduction of ARGs levels. Moreover, cyanoHABs generate biotic and abiotic stress in the freshwater microbiome, which may lead to an increase in MGEs and VFs. Exploration of the intricate interplays between microbiome, resistome, mobilome, and pathobiome during cyanoHABs not only revealed that the mechanisms underlying the dynamics of microbial dysbiosis but also emphasizes the need to prioritize the prevention of microbial dysbiosis in the risk management of ARGs.
Collapse
Affiliation(s)
- Min-Ji Kim
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea.
| | - Dayun Kang
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea.
| | - GyuDae Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea.
| | - Kyeongnam Kim
- Institute of Quality and Safety Evaluation of Agricultural Products, Kyungpook National University, Daegu 41566, Republic of Korea.
| | - Jinnam Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea.
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea; NGS Core Facility, Kyungpook National University, Daegu 41566, Republic of Korea.
| | - Seungjun Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
82
|
Liu P, Jiang Y, Jiao L, Luo Y, Wang X, Yang T. Strategies for the Discovery of Oxazolidinone Antibacterial Agents: Development and Future Perspectives. J Med Chem 2023; 66:13860-13873. [PMID: 37807849 DOI: 10.1021/acs.jmedchem.3c01040] [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: 10/10/2023]
Abstract
Oxazolidinones represent a significant class of synthetic bacterial protein synthesis inhibitors that are primarily effective against Gram-positive bacteria. The commercial success of linezolid, the first FDA-approved oxazolidinone antibiotic, has motivated researchers to develop more potent oxazolidinones by employing various drug development strategies to fight against antimicrobial resistance, some of which have shown promising results. Thus, this Perspective aims to discuss the strategies employed in constructing oxazolidinone-based antibacterial agents and summarize recent advances in discovering oxazolidinone antibiotics to provide valuable insights for potentially developing next-generation oxazolidinone antibacterial agents or other pharmaceuticals.
Collapse
Affiliation(s)
- Pingxian Liu
- Center of Infectious Diseases and Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunhan Jiang
- Center of Infectious Diseases and Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Jiao
- Center of Infectious Diseases and Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaodong Wang
- Department of Breast Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yang
- Center of Infectious Diseases and Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
83
|
Wohlgemuth R. Synthesis of Metabolites and Metabolite-like Compounds Using Biocatalytic Systems. Metabolites 2023; 13:1097. [PMID: 37887422 PMCID: PMC10608848 DOI: 10.3390/metabo13101097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 10/28/2023] Open
Abstract
Methodologies for the synthesis and purification of metabolites, which have been developed following their discovery, analysis, and structural identification, have been involved in numerous life science milestones. The renewed focus on the small molecule domain of biological cells has also created an increasing awareness of the rising gap between the metabolites identified and the metabolites which have been prepared as pure compounds. The design and engineering of resource-efficient and straightforward synthetic methodologies for the production of the diverse and numerous metabolites and metabolite-like compounds have attracted much interest. The variety of metabolic pathways in biological cells provides a wonderful blueprint for designing simplified and resource-efficient synthetic routes to desired metabolites. Therefore, biocatalytic systems have become key enabling tools for the synthesis of an increasing number of metabolites, which can then be utilized as standards, enzyme substrates, inhibitors, or other products, or for the discovery of novel biological functions.
Collapse
Affiliation(s)
- Roland Wohlgemuth
- MITR, Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego Street 116, 90-924 Lodz, Poland;
- Swiss Coordination Committee Biotechnology (SKB), 8021 Zurich, Switzerland
- European Society of Applied Biocatalysis (ESAB), 1000 Brussels, Belgium
| |
Collapse
|
84
|
Choi SR, Narayanasamy P. In Vitro and In Vivo Antimicrobial Activity of an Oxidative Stress-Mediated Bicyclic Menaquinone Biosynthesis Inhibitor against MRSA. ACS Infect Dis 2023; 9:2016-2024. [PMID: 37655755 DOI: 10.1021/acsinfecdis.3c00319] [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: 09/02/2023]
Abstract
Menaquinone (MK) is an essential component in the oxidative phosphorylation pathway of Gram-positive bacteria. Drugs targeting enzymes involved in MK biosynthesis can prevent electron transfer, which leads to ATP starvation and thereby death of microorganisms. Previously, we reported a series of MenA inhibitors and demonstrated their antimicrobial activity against Gram-positive bacteria, including Methicillin-resistant Staphylococcus aureus (MRSA) and mycobacteria. These inhibitors were developed by mimicking demethylmenaquinone, a product of MenA enzymatic reaction in MK biosynthesis. In this study, compound NM4, MK biosynthesis inhibitor, inhibited the formation of MRSA biofilm and it was screened against 1952 transposon mutants to elucidate mechanisms of action; however, no resistant mutants were found. Also, compound NM4 induced the production of reactive oxygen species (ROS) by blocking electron transfer in the oxidative phosphorylation pathway as observed by MRSA growth recovery using various ROS scavengers. An oxygen consumption assay also showed that NM4 blocks the oxygen consumption by MRSA, but the addition of menaquinone (MK) restores growth of MRSA. The NM4-treated MRSA induced the expression of catalase by more than 25%, as quantified by the native gel. A pulmonary murine model exhibited that NM4 significantly reduced bacterial lung load in mice without toxicity. An NM4-resistant USA300 strain was developed to attempt to identify the targets participating in the mechanism of resistance. Our results support that respiration and oxidative phosphorylation are potential targets for developing antimicrobial agents against MRSA. Altogether, our findings suggest the potential use of MK biosynthesis inhibitors as an effective antimicrobial agent against MRSA.
Collapse
Affiliation(s)
- Seoung-Ryoung Choi
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Prabagaran Narayanasamy
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
85
|
Caramiello AM, Bellucci MC, Ottaviano E, Ancona S, Borghi E, Volonterio A. Synthesis of amphiphilic hydantoin-based universal peptidomimetics as antibiotic agents. Org Biomol Chem 2023; 21:7702-7706. [PMID: 37698587 DOI: 10.1039/d3ob01247f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Three model hydantoin-based universal peptidomimetics were designed and synthetized. Their preferred amphiphilic β-turn conformation was assessed using molecular modeling and NMR experiments, and their antibacterial activity was tested against Gram-positive and Gram-negative bacteria strains, which demonstrated that these compounds could be a captivating class of antibiotics to fight emergent drug resistance.
Collapse
Affiliation(s)
- Alessio M Caramiello
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, 20131 Milano, Italy.
| | - Maria Cristina Bellucci
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, via Celoria 2, 20131 Milano, Italy
| | - Emerenziana Ottaviano
- Department of Health Sciences, Università degli Studi di Milano, via Di Rudinì 8, 20142, Milan, Italy
| | - Silvia Ancona
- Department of Health Sciences, Università degli Studi di Milano, via Di Rudinì 8, 20142, Milan, Italy
| | - Elisa Borghi
- Department of Health Sciences, Università degli Studi di Milano, via Di Rudinì 8, 20142, Milan, Italy
| | - Alessandro Volonterio
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, 20131 Milano, Italy.
| |
Collapse
|
86
|
Khunti P, Chantakorn K, Tantibhadrasapa A, Htoo HH, Thiennimitr P, Nonejuie P, Chaikeeratisak V. A novel coli myophage and antibiotics synergistically inhibit the growth of the uropathogenic E. coli strain CFT073 in stoichiometric niches. Microbiol Spectr 2023; 11:e0088923. [PMID: 37732769 PMCID: PMC10580823 DOI: 10.1128/spectrum.00889-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/20/2023] [Indexed: 09/22/2023] Open
Abstract
Urinary tract infections are widespread bacterial infections affecting millions of people annually, with Escherichia coli being the most prevalent. Although phage therapy has recently gained interest as a promising alternative therapy for antibiotic-resistant bacteria, several studies have raised concerns regarding the evolution of phage resistance, making the therapy ineffective. In this study, we discover a novel coli myophage designated as Killian that targets E. coli strains, including the uropathogenic E. coli (UPEC) strain CFT073. It requires at least 20 minutes for 90% of its particles to adsorb to the host cells, undergoes subcellular activities for replication for 30 minutes, and eventually lyses the cells with a burst size of about 139 particles per cell. Additionally, Killian can withstand a wide variety of temperatures (4-50°C) and pHs (4-10). Genome analysis reveals that Killian's genome consists of 169,905 base pairs with 35.5% GC content, encoding 276 open reading frames; of these, 209 are functionally annotated with no undesirable genes detected, highlighting its potential as an antibiotic alternative against UPEC. However, after an 8-hour phage treatment at high multiplicities of infection, bacterial density continuously increases, indicating an onset of bacterial growth revival. Thus, the combination study between the phage and three different antibiotics, including amikacin, ciprofloxacin, and piperacillin, was performed and showed that certain pairs of phage and antibiotics exhibited synergistic interactions in suppressing the bacterial growth revival. These findings suggest that Killian-antibiotic combinations are effective in inhibiting the growth of UPEC. IMPORTANCE Phage therapy has recently been in the spotlight as a viable alternative therapy for bacterial infections. However, several studies have raised concerns about the emergence of phage resistance that occurs during treatment, making the therapy not much effective. Here, we present the discovery of a novel E. coli myophage that, by itself, can effectively kill the uropathogenic E. coli, but the emergence of bacterial growth revival was detected during the treatment. Phage and antibiotics are then combined to improve the efficiency of the phage in suppressing the bacterial re-growth. This research would pave the way for the future development of phage-antibiotic cocktails for the sustainable use of phages for therapeutic purposes.
Collapse
Affiliation(s)
- Patiphan Khunti
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | | | | | - Htut Htut Htoo
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Parameth Thiennimitr
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center of Microbial Diversity and Sustainable Utilization, Chiang Mai University, Chiang Mai, Thailand
| | - Poochit Nonejuie
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | | |
Collapse
|
87
|
Al-Mijalli SH, Jeddi M, El Hachlafi N, M. Abdallah E, Assaggaf H, Qasem A, S. Rajab B, Lee LH, Bouyahya A, Goh KW, Ming LC, Mrabti HN. Combination of sweet orange, lentisk and lemon eucalyptus essential oils: Optimization of a new complete antimicrobial formulation using a mixture design methodology. Heliyon 2023; 9:e19814. [PMID: 37809691 PMCID: PMC10559161 DOI: 10.1016/j.heliyon.2023.e19814] [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: 05/25/2023] [Revised: 09/01/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Sweet orange (Citrus × sinensis (L.) Osbeck), lentisk (Pistacia lentiscus L.) and lemon eucalyptus (Eucalyptus citriodora Hook) are medicinal plants known by its culinary virtues. Their volatile oils have demonstrated promising antimicrobial activity against a panel of microbial strains, including those implicated in food deterioration. In this exploratory investigation, we aimed to determine the antimicrobial formulation of sweet orange, lentisk and lemon eucalyptus essential oils (EOs) using the simplex-centroid mixture design approach coupled with a broth microdilution method. EOs were first extracted by hydrodistillation, and then their phytochemical profile was characterized using Gas chromatography-mass spectrometry (GC-MS). GC-MS analysis identified d-limonene (14.27%), careen-3 (14.11%), β-myrcene (12.53%) as main components of lentisk EOs, while lemon eucalyptus was dominated by citronellal (39.40%), β-citronellol (16.39%) and 1,8-cineole (9.22%). For sweet orange EOs, d-limonene (87.22%) was the principal compound. The three EOs exhibited promising antimicrobial potential against various microorganisms. Lemon eucalyptus and sweet orange EO showed high activity against most tested microorganisms, while lentisk EO exerted important effect against some microbes but only moderate activity against others. The optimization formulations of antimicrobial potential showed interesting synergistic effects between three EOs. The best combinations predicted on C. albicans, S. aureus, E. coli, S. enterica and B. cereus correspond to 44%/55%/0%, 54%/16%/28%, 43%/22%/33%, 45%/17%/36% and 36%/30%/32% of Citrus sinensis, Pistacia lentiscus and Eucalyptus citriodora EOs, respectively. These findings suggest that the combination of EOs could be used as natural food preservatives and antimicrobial agents. However, further studies are needed to determine the mechanisms of action and efficacy of these EOs against different microorganisms.
Collapse
Affiliation(s)
- Samiah Hamad Al-Mijalli
- Department of Biology, College of Sciences, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Mohamed Jeddi
- Laboratory of Microbial Biotechnology and Bioactive Molecules, Faculty of Sciences and Technologies Faculty, Sidi Mohamed Ben Abdellah University, P.O. Box 2202, Imouzzer Road, Fez, Morocco
| | - Naoufal El Hachlafi
- Laboratory of Microbial Biotechnology and Bioactive Molecules, Faculty of Sciences and Technologies Faculty, Sidi Mohamed Ben Abdellah University, P.O. Box 2202, Imouzzer Road, Fez, Morocco
| | - Emad M. Abdallah
- Department of Science Laboratories, College of Science and Arts, Qassim University, Ar Rass 51921, Saudi Arabia
| | - Hamza Assaggaf
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ahmed Qasem
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Bodour S. Rajab
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Learn-Han Lee
- Sunway Microbiomics Centre, School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Sunway City, Malaysia
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, 10106, Morocco
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, 71800 Nilai, Malaysia
| | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia
| | - Hanae Naceiri Mrabti
- Laboratory of Pharmacology and Toxicology, Bio Pharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
- High Institute of Nursing Professions and Health Techniques of Casablanca, Casablanca, Morocco
| |
Collapse
|
88
|
Wan J, Ma N, Yuan H. Recent advances in the direct cloning of large natural product biosynthetic gene clusters. ENGINEERING MICROBIOLOGY 2023; 3:100085. [PMID: 39628928 PMCID: PMC11611023 DOI: 10.1016/j.engmic.2023.100085] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 12/06/2024]
Abstract
Large-scale genome-mining analyses have revealed that microbes potentially harbor a huge reservoir of uncharacterized natural product (NP) biosynthetic gene clusters (BGCs), and this has spurred a renaissance of novel drug discovery. However, the majority of these BGCs are often poorly or not at all expressed in their native hosts under laboratory conditions, and thus are regarded as silent/orphan BGCs. Currently, connecting silent BGCs to their corresponding NPs quickly and on a large scale is particularly challenging because of the lack of universal strategies and enabling technologies. Generally, the heterologous host-based genome mining strategy is believed to be a suitable alternative to the native host-based approach for prioritization of the vast and ever-increasing number of uncharacterized BGCs. In the last ten years, a variety of methods have been reported for the direct cloning of BGCs of interest, which is the first and rate-limiting step in the heterologous expression strategy. Essentially, each method requires that the following three issues be resolved: 1) how to prepare genomic DNA; 2) how to digest the bilateral boundaries for release of the target BGC; and 3) how to assemble the BGC and the capture vector. Here, we summarize recent reports regarding how to directly capture a BGC of interest and briefly discuss the advantages and disadvantages of each method, with an emphasis on the notion that direct cloning is very beneficial for accelerating genome mining research and large-scale drug discovery.
Collapse
Affiliation(s)
- Jiaying Wan
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China
| | - Nan Ma
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China
| | - Hua Yuan
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China
| |
Collapse
|
89
|
Fang Y, Liu X, Guo H, Zhang Y, Wu H, Zhou X, Chen X, Qin H, Gao H, Liu Y. AIE Bioconjugates for Accurate Identification and In Vivo Targeted Treatment of Bacterial Infection Based on Bioorthogonal Reaction. Adv Healthc Mater 2023; 12:e2300044. [PMID: 37368932 DOI: 10.1002/adhm.202300044] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/17/2023] [Accepted: 06/26/2023] [Indexed: 06/29/2023]
Abstract
Targeted killing multidrug-resistant bacteria with high efficiency is urgently needed for the treatment of infection with minimal collateral damage. Herein, a new near-infrared (NIR) fluorescence nanoprobe is designed and synthesized with aggregation-induced emission (AIE) features, which also is excellent reactive oxygen species (ROS) generator. The as-prepared AIE nanoparticles (NPs) present outstanding sterilizing rate on methicillin-resistant Staphylococcus aureus (MRSA) and kanamycin-resistant Escherichia coli (KREC). Meanwhile, considering the differences in the surface structure of animal cells and bacteria, a non-invasive image-guided strategy for precise treatment of bacterial infection has been successfully implemented based on bioorthogonal reaction which can perform and control unnatural chemical reactions inside living organisms. The AIE NPs are thus specifically trapped on the bacterial surface while not on the normal cells, realizing real-time tracking of the infected site distribution in vivo and guiding photodynamic therapy (PDT) for eliminating bacteria in inflammation region. That significantly improves the accuracy and sterilization rate of bacterial-infected wounds with negligible side effects. The investigation developed a potential antibacterial agent and also provides an instructive way for targeting treatment based on bioorthogonal reaction.
Collapse
Affiliation(s)
- Yuan Fang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin Key Laboratory of Food Quality and Health, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Xin Liu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin Key Laboratory of Food Quality and Health, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Hanqiong Guo
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin Key Laboratory of Food Quality and Health, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yujie Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin Key Laboratory of Food Quality and Health, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Haotian Wu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin Key Laboratory of Food Quality and Health, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Xiao Zhou
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin Key Laboratory of Food Quality and Health, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Xiying Chen
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin Key Laboratory of Food Quality and Health, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Haijuan Qin
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin Key Laboratory of Food Quality and Health, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Heqi Gao
- College of Physics and Optoelectronic Engineering, College of Materials Science and Engineering, Center for AIE Research, Shenzhen University, Shenzhen, 518060, China
| | - Yaqing Liu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin Key Laboratory of Food Quality and Health, Tianjin University of Science and Technology, Tianjin, 300457, China
| |
Collapse
|
90
|
Tsai MJ, Zambrano RA, Susas JL, Silva L, Takahashi MK. Identifying Antisense Oligonucleotides to Disrupt Small RNA Regulated Antibiotic Resistance via a Cell-Free Transcription-Translation Platform. ACS Synth Biol 2023; 12:2245-2251. [PMID: 37540186 PMCID: PMC10443041 DOI: 10.1021/acssynbio.3c00245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Indexed: 08/05/2023]
Abstract
Bacterial small RNAs (sRNAs) regulate many important physiological processes in cells, including antibiotic resistance and virulence genes, through base-pairing interactions with mRNAs. Antisense oligonucleotides (ASOs) have great potential as therapeutics against bacterial pathogens by targeting sRNAs such as MicF, which regulates outer membrane protein OmpF expression and limits the permeability of antibiotics. Here we devised a cell-free transcription-translation (TX-TL) assay to identify ASO designs that sufficiently sequester MicF. ASOs were then ordered as peptide nucleic acids conjugated to cell-penetrating peptides (CPP-PNA) to allow for effective delivery into bacteria. Subsequent minimum inhibitory concentration (MIC) assays demonstrated that simultaneously targeting the regions of MicF responsible for sequestering the start codon and the Shine-Dalgarno sequence of ompF with two different CPP-PNAs synergistically reduced the MIC for a set of antibiotics. This investigation offers a TX-TL-based approach to identify novel therapeutic candidates to combat intrinsic sRNA-mediated antibiotic resistance mechanisms.
Collapse
Affiliation(s)
- Min Jen Tsai
- Department
of Biology, California State University
Northridge, Northridge, California 91330, United States
| | - Raphael Angelo
I. Zambrano
- Department
of Biology, California State University
Northridge, Northridge, California 91330, United States
- Department
of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912, United States
| | - Jeremiah Lyn Susas
- Department
of Biology, California State University
Northridge, Northridge, California 91330, United States
| | - Lizette Silva
- Department
of Biology, California State University
Northridge, Northridge, California 91330, United States
| | - Melissa K. Takahashi
- Department
of Biology, California State University
Northridge, Northridge, California 91330, United States
| |
Collapse
|
91
|
Naheed S, Din IU, Qamar MU, Rasool N, Ahmad M, Bilal M, Khalid A, Ahmad G, Al-Hussain SA, Zaki MEA. Synthesis, Anti-Bacterial and Molecular Docking Studies of Arylated Butyl 2-Bromoisonicotinate Against Clinical Isolates of ESBL-Producing Escherichia coli ST405 and Methicillin-Resistant Staphylococcus aureus. Infect Drug Resist 2023; 16:5295-5308. [PMID: 37601564 PMCID: PMC10438430 DOI: 10.2147/idr.s407891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/12/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Global public health concerns include the emergence and spread of methicillin-resistant Staphylococcus aureus (MRSA) and extended-spectrum beta-lactamase Escherichia coli (ESBL-E. coli). These pathogens cause infections that are difficult to treat, which can have fatal outcomes and require lengthy hospital stays. As a result, we created butyl 2-bromoisonicotinate and tested its antibacterial effectiveness against the ESBL-E. coli ST 405 and MRSA pathogens. Natural product discovery is complemented by synthetic compound synthesis because of the latter's potential for superior characteristics, target specificity, scalability, intellectual advantages, and chemical diversity. Because of this, the potential for discovering new medicinal compounds is increased, and the constraints placed on natural sources are overcome. Natural items are tough to obtain since they are hard to isolate and synthesize. Therefore, modern science is actively searching for small molecules as therapeutic agents by applying sustainable techniques that can be commercialized. Methods Two patients' blood samples were taken, and the BACTEC/Alert system was used to process them. On blood and MacConkey agar, the positive samples were subcultured and incubated aerobically at 37 °C. Using the VITEK 2 compact system, the isolates were subjected to isolate identification and MIC. MLST of the ESBL-E. coli was performed by PCR. Additionally, Fischer esterification was used to create butyl 2-bromoisonicotinate in excellent yields. A commercially available palladium catalyst was then used to arylate the compound, resulting in medium to good yields of arylated butyl 2-bromoisonicotinates. Using the agar well diffusion assay and the micro-broth dilution method, we assessed the in-vitro activities of the synthesized molecules (3, 5a-h) against clinically isolated ESBL-E. coli ST405, and MRSA. A molecular operating environment was used to carry out in silico validation of the synthesized compounds' binding to the active site and to evaluate the stability of their molecular interactions with the target E. coli 2Y2T protein. Results MRSA and ESBL-producing E. coli were identified as the two clinical isolates. While MRSA was also resistant to beta-lactam drugs and least resistant to vancomycin, ESBL-producing E. coli belonged to ST405 and was resistant to cephalosporins and sensitive to carbapenems. Good yields of the desired compounds were produced by our effective and economical synthesis. By using a micro-broth dilution assay, the Molecules (3, 5a, and 5d) were most effective against both resistant strains. The Molecules (3, 5a, 5b, and 5d) also displayed good binding energies. Conclusion The butyl 2-bromoisonicotinate displayed antibacterial efficacy against ESBL-producing E. coli ST405 and MRSA strains. After the in-vivo trial, this substance might offer an alternative therapeutic option.
Collapse
Affiliation(s)
- Shazia Naheed
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Irum Umar Din
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Muhammad Usman Qamar
- Institute of Microbiology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Nasir Rasool
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Matloob Ahmad
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Muhammad Bilal
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Aqsa Khalid
- School of Interdisciplinary Engineering & Science (SINES), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Gulraiz Ahmad
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Sami A Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University, Riyad, 11623, Saudi Arabia
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University, Riyad, 11623, Saudi Arabia
| |
Collapse
|
92
|
Kimishima A, Sakai K, Honsho M, Matsui H, Wasuwanich P, Watanabe Y, Iwatsuki M, Sunazuka T, Arima N, Abe K, Hanaki H, Asami Y. A combination strategy of a semisynthetic macrolide, 5-O-mycaminosyltylonolide with polymyxin B nonapeptide for multi-drug resistance P. aeruginosa. J Antibiot (Tokyo) 2023; 76:499-501. [PMID: 37208456 DOI: 10.1038/s41429-023-00628-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023]
Abstract
The emergence and spread of antimicrobial resistant pathogens continue to threaten our ability to combat several infections. Among them, Pseudomonas aeruginosa (P. aeruginosa) poses a major threat to human health. P. aeruginosa has intrinsic resistance to many antibiotics due to the impermeability of its outer membrane and a resistance-nodulation-cell division type multidrug efflux pump system. Therefore, only limited therapeutic drugs are effective against the pathogen. To address this problem, we have recently discovered an overlooked anti- P. aeruginosa compound, 5-O-mycaminosyltylonolide (OMT) from the Ōmura Natural Compound library using an efflux pump deletion P. aeruginosa mutant strain, YM64. In this report, we aim to demonstrate the promising potential of OMT for as a novel anti- P. aeruginosa compound and performed combination assays of OMT with polymyxin B nonapeptide, an example of a permeabilizing agent, against multi-drug resistant P. aeruginosa clinical isolates.
Collapse
Affiliation(s)
- Aoi Kimishima
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Kazunari Sakai
- Research Management Department, Kowa Company LTD., 4-13-3 Nihonbashi-honcho, Chuo-ku, Tokyo, 103-8433, Japan
| | - Masako Honsho
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Hidehito Matsui
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Paul Wasuwanich
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Yoshihiro Watanabe
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Masato Iwatsuki
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Toshiaki Sunazuka
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
| | - Naoaki Arima
- Research Management Department, Kowa Company LTD., 4-13-3 Nihonbashi-honcho, Chuo-ku, Tokyo, 103-8433, Japan
| | - Kazutoyo Abe
- Research Management Department, Kowa Company LTD., 4-13-3 Nihonbashi-honcho, Chuo-ku, Tokyo, 103-8433, Japan
| | - Hideaki Hanaki
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan
- Research Management Department, Kowa Company LTD., 4-13-3 Nihonbashi-honcho, Chuo-ku, Tokyo, 103-8433, Japan
| | - Yukihiro Asami
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
- Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, 108-8641, Japan.
| |
Collapse
|
93
|
MacNair CR, Tsai CN, Rutherford ST, Tan MW. Returning to Nature for the Next Generation of Antimicrobial Therapeutics. Antibiotics (Basel) 2023; 12:1267. [PMID: 37627687 PMCID: PMC10451936 DOI: 10.3390/antibiotics12081267] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
Antibiotics found in and inspired by nature are life-saving cures for bacterial infections and have enabled modern medicine. However, the rise in resistance necessitates the discovery and development of novel antibiotics and alternative treatment strategies to prevent the return to a pre-antibiotic era. Once again, nature can serve as a source for new therapies in the form of natural product antibiotics and microbiota-based therapies. Screening of soil bacteria, particularly actinomycetes, identified most of the antibiotics used in the clinic today, but the rediscovery of existing molecules prompted a shift away from natural product discovery. Next-generation sequencing technologies and bioinformatics advances have revealed the untapped metabolic potential harbored within the genomes of environmental microbes. In this review, we first highlight current strategies for mining this untapped chemical space, including approaches to activate silent biosynthetic gene clusters and in situ culturing methods. Next, we describe how using live microbes in microbiota-based therapies can simultaneously leverage many of the diverse antimicrobial mechanisms found in nature to treat disease and the impressive efficacy of fecal microbiome transplantation and bacterial consortia on infection. Nature-provided antibiotics are some of the most important drugs in human history, and new technologies and approaches show that nature will continue to offer valuable inspiration for the next generation of antibacterial therapeutics.
Collapse
Affiliation(s)
- Craig R. MacNair
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA 94080, USA;
| | - Caressa N. Tsai
- School of Law, University of California, Berkeley, Berkeley, CA 94704, USA;
| | - Steven T. Rutherford
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA 94080, USA;
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA 94080, USA;
| |
Collapse
|
94
|
Singh S, Verma T, Khamari B, Bulagonda EP, Nandi D, Umapathy S. Antimicrobial Resistance Studies Using Raman Spectroscopy on Clinically Relevant Bacterial Strains. Anal Chem 2023. [PMID: 37463121 DOI: 10.1021/acs.analchem.3c01453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
There has been a steep rise in the emergence of antibiotic-resistant bacteria in the past few years. A timely diagnosis can help in initiating appropriate antibiotic therapy. However, conventional techniques for diagnosing antibiotic resistance are time-consuming and labor-intensive. Therefore, we investigated the potential of Raman spectroscopy as a rapid surveillance technology for tracking the emergence of antibiotic resistance. In this study, we used Raman spectroscopy to differentiate clinical isolates of antibiotic-resistant and -sensitive bacteria of Escherichia coli, Acinetobacter baumannii, and Enterobacter species. The spectra were collected with or without exposure to various antibiotics (ciprofloxacin, gentamicin, meropenem, and nitrofurantoin), each having a distinct mechanism of action. Ciprofloxacin- and meropenem-treated sensitive strains showed a decrease in the intensity of Raman bands associated with DNA (667, 724, 785, 1378, 1480, and 1575 cm-1) and proteins (640 and 1662 cm-1), coupled with an increase in the intensity of lipid bands (891, 960, and 1445 cm-1). Gentamicin- and nitrofurantoin-treated sensitive strains showed an increase in the intensity of nucleic acid bands (668, 724, 780, 810, 1378, 1480, and 1575 cm-1) while a decrease in the intensity of protein bands (640, 1003, 1606, and 1662 cm-1) and the lipid band (1445 cm-1). The Raman spectral changes observed in the antibiotic-resistant strains were opposite to that of antibiotic-sensitive strains. The Raman spectral data correlated well with the antimicrobial susceptibility test results. The Raman spectral dataset was used for partial least-squares (PLS) analysis to validate the biomarkers obtained from the univariate analysis. Overall, this study showcases the potential of Raman spectroscopy for detecting antibiotic-resistant and -sensitive bacteria.
Collapse
Affiliation(s)
- Saumya Singh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Taru Verma
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Balaram Khamari
- Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi 515134, Andhra Pradesh, India
| | - Eswarappa Pradeep Bulagonda
- Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi 515134, Andhra Pradesh, India
| | - Dipankar Nandi
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, Karnataka, India
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Siva Umapathy
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, Karnataka, India
- Department of Instrumentation and Applied Physics, Indian Institute of Science, Bangalore 560012, Karnataka, India
| |
Collapse
|
95
|
Jia Y, Chen W, Tang R, Zhang J, Liu X, Dong R, Hu F, Jiang X. Multi-armed antibiotics for Gram-positive bacteria. Cell Host Microbe 2023; 31:1101-1110.e5. [PMID: 37442098 DOI: 10.1016/j.chom.2023.06.008] [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: 01/17/2023] [Revised: 04/03/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023]
Abstract
Antibiotic resistance is a serious threat to public health. Here, we propose a multi-armed chemical scaffold (MACS) for antibiotic screening, which refers to multi-armed molecules (MAMs) consisting of a core unit and three or four arms, neither of which is active for pathogens. Based on a structure-activity relationship study of MAMs, we discover a class of multi-armed antibiotics (MAAs) with a core similar to ethylene (E), carbon atom (C), benzene (B), nitrogen atom (N), and triazine (T) and three or four 4-phenylbenzoic acid (PBA) arms, or a B core and three 4-vinylbenzoic acid (VBA) or 4-ethynylbenzoic acid (EBA) arms. They can selectively interact with Gram-positive bacteria and inhibit cell wall assembly by targeting the lipid carriers of cell wall biosynthesis. MAAs have excellent antibacterial activities against Gram-positive bacteria, including clinical multi-drug-resistant (MDR) isolates. Our study provides a chemical scaffold and identifies eight antibacterial lead compounds for the development of antibiotics.
Collapse
Affiliation(s)
- Yuexiao Jia
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China; Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Wenwen Chen
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, P.R. China
| | - Rongbing Tang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China
| | - Jiangjiang Zhang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China
| | - Xiaoyan Liu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China
| | - Ruihua Dong
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China
| | - Fupin Hu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xingyu Jiang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd., Nanshan District, Shenzhen, Guangdong 518055, P.R. China.
| |
Collapse
|
96
|
Morais DC, Fontes ML, Oliveira AB, Gabbai-Armelin PR, Ferrisse TM, De Oliveira LFC, Brighenti FL, Barud HS, De Sousa FB. Combining Polymer and Cyclodextrin Strategy for Drug Release of Sulfadiazine from Electrospun Fibers. Pharmaceutics 2023; 15:1890. [PMID: 37514076 PMCID: PMC10386385 DOI: 10.3390/pharmaceutics15071890] [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: 06/07/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
This study reports the fabrication of polymeric matrices through electrospinning using polymethyl methacrylate (PMMA) and poly(lactic-co-glycolic acid) (PLGA), biocompatible polymers commonly used in medical systems. These polymers were combined with an antibacterial drug, sulfadiazine sodium salt (SDS) or its supramolecular system formed with hydroxypropyl-β-cyclodextrin (HPβ/CD) at 1:1 molar ratio, aiming to assemble a transdermal drug delivery system. The formation of fibers was confirmed by scanning electron microscopy (SEM), and the fibers' surface properties were analyzed using contact angle and water vapor permeability techniques. Drug release tests and cell viability assays were performed to evaluate the potential toxicity of the material. SEM images demonstrated that the obtained fibers had nanoscale- and micrometer-scale diameters in PLGA and PMMA systems, respectively. The contact angle analyses indicated that, even in the presence of hydrophilic molecules (SDS and HPβCD), PMMA fibers exhibited hydrophobic characteristics, while PLGA fibers exhibited hydrophilic surface properties. These data were also confirmed by water vapor permeability analysis. The drug release profiles demonstrated a greater release of SDS in the PLGA system. Moreover, the presence of HPβCD improved the drug release in both polymeric systems and the cell viability in the PMMA SDS/HPβCD system. In terms of antibacterial activity, all membranes yielded positive outcomes; nevertheless, the PLGA SDS/HPβCD membrane exhibited the most remarkable results, with the lowest microbial load values. Additionally, the pseudo wound healing analysis demonstrated that the PLGA SDS/HPβCD fiber exhibited results similar to the control group. Consequently, these findings exemplify the substantial potential of the obtained materials for use in wound healing applications.
Collapse
Affiliation(s)
- Diego C. Morais
- Laboratório de Sistemas Poliméricos e Supramoleculares (LSPS), Instituto de Física e Química, Universidade Federal de Itajubá (UNIFEI), Itajubá 37500-903, MG, Brazil;
| | - Marina L. Fontes
- Laboratório de Biopolímeros e Biomateriais, Universidade de Araraquara (UNIARA), Araraquara 14801-340, SP, Brazil; (M.L.F.); (H.S.B.)
| | - Analú B. Oliveira
- Department of Morphology and Pediatric Dentistry, School of Dentistry, São Paulo State University (UNESP), Araraquara 14801-903, SP, Brazil; (A.B.O.); (P.R.G.-A.); (F.L.B.)
| | - Paulo R. Gabbai-Armelin
- Department of Morphology and Pediatric Dentistry, School of Dentistry, São Paulo State University (UNESP), Araraquara 14801-903, SP, Brazil; (A.B.O.); (P.R.G.-A.); (F.L.B.)
| | - Túlio M. Ferrisse
- Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara 14801-903, SP, Brazil;
| | - Luiz F. C. De Oliveira
- Núcleo de Espectroscopia E Estrutura Molecular—Departamento de Química—ICE, Universidade Federal de Juiz de Fora (UFJF), Juiz de Fora 36036-900, MG, Brazil
| | - Fernanda Lourenção Brighenti
- Department of Morphology and Pediatric Dentistry, School of Dentistry, São Paulo State University (UNESP), Araraquara 14801-903, SP, Brazil; (A.B.O.); (P.R.G.-A.); (F.L.B.)
| | - Hernane S. Barud
- Laboratório de Biopolímeros e Biomateriais, Universidade de Araraquara (UNIARA), Araraquara 14801-340, SP, Brazil; (M.L.F.); (H.S.B.)
| | - Frederico B. De Sousa
- Laboratório de Sistemas Poliméricos e Supramoleculares (LSPS), Instituto de Física e Química, Universidade Federal de Itajubá (UNIFEI), Itajubá 37500-903, MG, Brazil;
| |
Collapse
|
97
|
Wang X, Zhou N, Wang B. Bacterial synthetic biology: tools for novel drug discovery. Expert Opin Drug Discov 2023; 18:1087-1097. [PMID: 37482696 DOI: 10.1080/17460441.2023.2239704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
INTRODUCTION Bacterial synthetic biology has provided powerful tools to revolutionize the drug discovery process. These tools can be harnessed to generate bacterial novel pharmaceutical compounds with enhanced bioactivity and selectivity or to create genetically modified microorganisms as living drugs. AREAS COVERED This review provides a current overview of the state-of-the-art in bacterial synthetic biology tools for novel drug discovery. The authors discuss the application of these tools including bioinformatic tools, CRISPR tools, engineered bacterial transcriptional regulators, and synthetic biosensors for novel drug discovery. Additionally, the authors present the recent progress on reprogramming bacteriophages as living drugs to fight against antibiotic-resistant pathogens. EXPERT OPINION The field of using bacterial synthetic biology tools for drug discovery is rapidly advancing. However, challenges remain in developing reliable and robust methods to engineer bacteria. Further advancements in synthetic biology hold promise to speed up drug discovery, facilitating the development of novel therapeutics against various diseases.
Collapse
Affiliation(s)
- Xiyan Wang
- College of Chemical and Biological Engineering & ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Nan Zhou
- College of Chemical and Biological Engineering & ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Baojun Wang
- College of Chemical and Biological Engineering & ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- Research Center of Biological Computation, Zhejiang Laboratory, Hangzhou, China
| |
Collapse
|
98
|
Park Y, Taguchi A, Baidin V, Kahne D, Walker S. A Time-Resolved FRET Assay Identifies a Small Molecule that Inhibits the Essential Bacterial Cell Wall Polymerase FtsW. Angew Chem Int Ed Engl 2023; 62:e202301522. [PMID: 37099323 PMCID: PMC10330507 DOI: 10.1002/anie.202301522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/10/2023] [Accepted: 04/26/2023] [Indexed: 04/27/2023]
Abstract
The peptidoglycan cell wall is essential for bacterial survival. To form the cell wall, peptidoglycan glycosyltransferases (PGTs) polymerize Lipid II to make glycan strands and then those strands are crosslinked by transpeptidases (TPs). Recently, the SEDS (for shape, elongation, division, and sporulation) proteins were identified as a new class of PGTs. The SEDS protein FtsW, which produces septal peptidoglycan during cell division, is an attractive target for novel antibiotics because it is essential in virtually all bacteria. Here, we developed a time-resolved Förster resonance energy transfer (TR-FRET) assay to monitor PGT activity and screened a Staphylococcus aureus lethal compound library for FtsW inhibitors. We identified a compound that inhibits S. aureus FtsW in vitro. Using a non-polymerizable Lipid II derivative, we showed that this compound competes with Lipid II for binding to FtsW. The assays described here will be useful for discovering and characterizing other PGT inhibitors.
Collapse
Affiliation(s)
- Youngseon Park
- Department of Microbiology, Harvard Medical School, 4 Blackfan Circle, Boston, Massachusetts 02115, United States
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St., Cambridge, Massachusetts 02138, United States
| | - Atsushi Taguchi
- Department of Microbiology, Harvard Medical School, 4 Blackfan Circle, Boston, Massachusetts 02115, United States
- (Current location) SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Vadim Baidin
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St., Cambridge, Massachusetts 02138, United States
| | - Daniel Kahne
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St., Cambridge, Massachusetts 02138, United States
| | - Suzanne Walker
- Department of Microbiology, Harvard Medical School, 4 Blackfan Circle, Boston, Massachusetts 02115, United States
| |
Collapse
|
99
|
Stojković D, Petrović J, Carević T, Soković M, Liaras K. Synthetic and Semisynthetic Compounds as Antibacterials Targeting Virulence Traits in Resistant Strains: A Narrative Updated Review. Antibiotics (Basel) 2023; 12:963. [PMID: 37370282 PMCID: PMC10295040 DOI: 10.3390/antibiotics12060963] [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: 05/10/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
This narrative review paper provides an up-to-date overview of the potential of novel synthetic and semisynthetic compounds as antibacterials that target virulence traits in resistant strains. The review focused on research conducted in the last five years and investigated a range of compounds including azoles, indoles, thiophenes, glycopeptides, pleuromutilin derivatives, lactone derivatives, and chalcones. The emergence and spread of antibiotic-resistant bacterial strains is a growing public health concern, and new approaches are urgently needed to combat this threat. One promising approach is to target virulence factors, which are essential for bacterial survival and pathogenesis, but not for bacterial growth. By targeting virulence factors, it may be possible to reduce the severity of bacterial infections without promoting the development of resistance. We discuss the mechanisms of action of the various compounds investigated and their potential as antibacterials. The review highlights the potential of targeting virulence factors as a promising strategy to combat antibiotic resistance and suggests that further research is needed to identify new compounds and optimize their efficacy. The findings of this review suggest that novel synthetic and semisynthetic compounds that target virulence factors have great potential as antibacterials in the fight against antibiotic resistance.
Collapse
Affiliation(s)
- Dejan Stojković
- Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (D.S.); (J.P.); (T.C.); (M.S.)
| | - Jovana Petrović
- Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (D.S.); (J.P.); (T.C.); (M.S.)
| | - Tamara Carević
- Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (D.S.); (J.P.); (T.C.); (M.S.)
| | - Marina Soković
- Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (D.S.); (J.P.); (T.C.); (M.S.)
| | - Konstantinos Liaras
- Department of Life and Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417 Nicosia, Cyprus
| |
Collapse
|
100
|
Liu S, Xu Q, Lou S, Tu J, Yin W, Li X, Jin Y, Radnaeva LD, Nikitina E, Makhinov AN, Araruna JT, Fedorova IV. Spatiotemporal distributions of sulfonamide and tetracycline resistance genes and microbial communities in the coastal areas of the Yangtze River Estuary. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 259:115025. [PMID: 37216861 DOI: 10.1016/j.ecoenv.2023.115025] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023]
Abstract
In this paper, water and sediments were sampled at eight monitoring stations in the coastal areas of the Yangtze River Estuary in summer and autumn 2021. Two sulfonamide resistance genes (sul1 and sul2), six tetracycline resistance genes (tetM, tetC, tetX, tetA, tetO, and tetQ), one integrase gene (intI1), 16 S rRNA genes, and microbial communities were examined and analyzed. Most resistance genes showed relatively higher abundance in summer and lower abundance in autumn. One-way analysis of variance (ANOVA) showed significant seasonal variation of some ARGs (7 ARGs in water and 6 ARGs in sediment). River runoff and WWTPs are proven to be the major sources of resistance genes along the Yangtze River Estuary. Significant and positive correlations between intI1 and other ARGs were found in water samples (P < 0.05), implying that intI1 may influence the spread and propagation of resistance genes in aquatic environments. Proteobacteria was the dominant phylum along the Yangtze River Estuary, with an average proportion of 41.7%. Redundancy analysis indicated that the ARGs were greatly affected by temperature, dissolved oxygen, and pH in estuarine environments. Network analysis showed that Proteobacteria and Cyanobacteria were the potential host phyla for ARGs in the coastal areas of the Yangtze River Estuary.
Collapse
Affiliation(s)
- Shuguang Liu
- Department of Hydraulic Engineering, College of Civil Engineering, Tongji University, Shanghai, China; Key Laboratory of Yangtze River Water Environment, Ministry of Education, Tongji University, Shanghai, China
| | - Qiuhong Xu
- Department of Hydraulic Engineering, College of Civil Engineering, Tongji University, Shanghai, China
| | - Sha Lou
- Department of Hydraulic Engineering, College of Civil Engineering, Tongji University, Shanghai, China; Key Laboratory of Yangtze River Water Environment, Ministry of Education, Tongji University, Shanghai, China.
| | - Junbiao Tu
- State Key Laboratory of Marine Geology, Tongji University, Shanghai, China
| | - Wenjun Yin
- State Key Laboratory of Pollution Control and Resources Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - Xin Li
- Department of Hydraulic Engineering, College of Civil Engineering, Tongji University, Shanghai, China
| | - Yuchen Jin
- Department of Hydraulic Engineering, College of Civil Engineering, Tongji University, Shanghai, China
| | - Larisa Dorzhievna Radnaeva
- Laboratory of Chemistry of Natural Systems, Baikal Institute of Nature Management of Siberian Branch of the Russian Academy of Sciences, Republic of Buryatia, Russian Republic, Russia
| | - Elena Nikitina
- Laboratory of Chemistry of Natural Systems, Baikal Institute of Nature Management of Siberian Branch of the Russian Academy of Sciences, Republic of Buryatia, Russian Republic, Russia
| | | | | | - Irina Viktorovna Fedorova
- Institute of Earth Sciences, Saint Petersburg State University, 7-9 Universitetskaya Embankment, St Petersburg, Russia
| |
Collapse
|