1
|
Kabeya JK, Ngombe NK, Mutwale PK, Safari JB, Matlou GG, Krause RWM, Nkanga CI. Antimicrobial capping agents on silver nanoparticles made via green method using natural products from banana plant waste. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2025; 53:29-42. [PMID: 39920563 DOI: 10.1080/21691401.2025.2462335] [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: 08/23/2024] [Revised: 01/12/2025] [Accepted: 01/29/2025] [Indexed: 02/09/2025]
Abstract
Herein, we investigated the phytochemical composition and antibacterial activities of the organic layers from biosynthesized silver nanoparticles (AgNPs). AgNPs were synthesized using Musa paradisiaca and Musa sapientum extracts. UV-vis absorption in the 400-450 nm range indicated surface plasmonic resonance peak of AgNPs. Samples analyses using dynamic light scattering and transmission electron microscopy revealed the presence of particles within nanometric ranges, with sizes of 30-140 nm and 8-40 nm, respectively. Fourier transform infrared (FTIR) unveiled the presence of several organic functional groups on the surface of AgNPs, indicating the presence of phytochemicals from plant extracts. Thin layer chromatography (TLC) of the phytochemicals (capping agents) from AgNPs identified multiple groups of secondary metabolites. These phytochemical capping agents exhibited antibacterial activities against Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa, with minimum inhibitory concentrations ranging from 62.5 to 1000 µg/mL. Regardless of the bacterial species or plant parts (leaves or pseudo-stems), capping agents from M. sapientum nanoparticles displayed significantly enhanced antibacterial effectiveness compared to all other samples, including the raw plant extracts and biosynthesized capped and uncapped AgNPs. These results suggest the presence of antimicrobial phytochemicals on biosynthesized AgNPs, highlighting the promise of green nanoparticle synthesis as a valuable approach in bioprospecting antimicrobial agents.
Collapse
Affiliation(s)
- Jimmy K Kabeya
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa XI, Democratic Republic of the Congo
- Center of Chemico- and Bio-Medical Research (CCBR), Department of Chemistry, Faculty of Science, Rhodes University, Grahamstown, South Africa
- Centre d'Etudes des Substances Naturelles d'Origine Végétale (CESNOV), Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa XI, Democratic Republic of the Congo
| | - Nadège K Ngombe
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa XI, Democratic Republic of the Congo
- Centre d'Etudes des Substances Naturelles d'Origine Végétale (CESNOV), Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa XI, Democratic Republic of the Congo
| | - Paulin K Mutwale
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa XI, Democratic Republic of the Congo
- Centre d'Etudes des Substances Naturelles d'Origine Végétale (CESNOV), Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa XI, Democratic Republic of the Congo
| | - Justin B Safari
- Center of Chemico- and Bio-Medical Research (CCBR), Department of Chemistry, Faculty of Science, Rhodes University, Grahamstown, South Africa
- Department of Pharmacy, Faculty of Pharmaceutical Sciences and Public Health, Official University of Bukavu, Bukavu, Democratic Republic of the Congo
| | - Gauta Gold Matlou
- Electron Microscopy Unit, Sefako Makgatho Health Sciences University, Ga-Rankuwa, South Africa
| | - Rui W M Krause
- Center of Chemico- and Bio-Medical Research (CCBR), Department of Chemistry, Faculty of Science, Rhodes University, Grahamstown, South Africa
| | - Christian I Nkanga
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa XI, Democratic Republic of the Congo
| |
Collapse
|
2
|
Jain G, Chaurasia R, Kaur BP, Chowdhury OP, Roy H, Gupta RR, Biswas B, Chakrabarti S, Mukherjee M. Unleashing the antibacterial potential of ZIFs and their derivatives: mechanistic insights. J Mater Chem B 2025. [PMID: 39935286 DOI: 10.1039/d4tb02682a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Antibiotic resistance presents an alarming threat to global health, with bacterial infections now ranking among the leading causes of mortality. To address this escalating challenge, strategies such as antibiotic stewardship, development of antimicrobial therapies, and exploration of alternative treatment modalities are imperative. Metal-organic frameworks (MOFs), acclaimed for their outstanding biocompatibility and in vivo biodegradability, are promising avenues for the synthesis of novel antibiotic agents under mild conditions. Among these, zeolitic imidazolate frameworks (ZIFs), a remarkable subclass of MOFs, have emerged as potent antibacterial materials; the efficacy of which stems from their porous structure, metal ion content, and tunable functionalized groups. This could be further enhanced by incorporating or encapsulating metal ions, such as Cu, Fe, Ti, Ag, and others. This perspective aims to underscore the potential of ZIFs as antibacterial agents and their underlying mechanisms including the release of metal ions, generation of reactive oxygen species (ROS), disruption of bacterial cell walls, and synergistic interactions with other antibacterial agents. These attributes position ZIFs as promising candidates for advanced applications in combating bacterial infections. Furthermore, we propose a novel approach for synthesizing ZIFs and their derivatives, demonstrating exceptional antibacterial efficacy against Escherichia coli and Staphylococcus aureus. By highlighting the benefits of ZIFs and their derivatives as antibacterial agents, this perspective emphasizes their potential to address the critical challenge of antibiotic resistance.
Collapse
Affiliation(s)
- Geetika Jain
- Amity Institute of Nanotechnology, Amity University, Noida, UP 201313, India
- Molecular Science and Engineering Laboratory, Amity Institute of Click Chemistry Research & Studies, Amity University Noida, UP 201313, India.
| | - Radhika Chaurasia
- Molecular Science and Engineering Laboratory, Amity Institute of Click Chemistry Research & Studies, Amity University Noida, UP 201313, India.
| | - Bani Preet Kaur
- Molecular Science and Engineering Laboratory, Amity Institute of Click Chemistry Research & Studies, Amity University Noida, UP 201313, India.
| | | | - Hiranmay Roy
- Department of Chemistry, University of North Bengal, Darjeeling 734013, India
| | - Richa Rani Gupta
- Molecular Science and Engineering Laboratory, Amity Institute of Click Chemistry Research & Studies, Amity University Noida, UP 201313, India.
| | - Bhaskar Biswas
- Department of Chemistry, University of North Bengal, Darjeeling 734013, India
| | - Sandip Chakrabarti
- Molecular Science and Engineering Laboratory, Amity Institute of Click Chemistry Research & Studies, Amity University Noida, UP 201313, India.
| | - Monalisa Mukherjee
- Molecular Science and Engineering Laboratory, Amity Institute of Click Chemistry Research & Studies, Amity University Noida, UP 201313, India.
| |
Collapse
|
3
|
Soohoo AM, Aguilar RA, Cho H, Privalsky TM, Liu L, Nguyen KP, Walsh CT, Khosla C. New Insights into the Mechanism of Action of L-681,217, a Medicinally Promising Polyketide Inhibitor of Bacterial Protein Translation. Biochemistry 2024; 63:3336-3347. [PMID: 39576948 DOI: 10.1021/acs.biochem.4c00541] [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] [Indexed: 11/24/2024]
Abstract
An attractive strategy for combating antibacterial resistance involves the development of new antibiotics whose mechanisms differ from those of existing ones in the clinic. Elfamycin antibiotics, whose prototypes include kirromycin and aurodox, are illustrative examples based on their ability to target EF-Tu, an essential component for protein translation in bacteria. Our efforts to revisit this antibiotic class were enabled by two developments. First, we produced L-681,217, an understudied member of this polyketide family harboring a terminal carboxylic acid in place of a hydroxypyridone ring, and synthesized a biotinylated derivative with comparable activity to the natural product. Second, we established a sensitive cell-free protein synthesis (CFPS) assay in which superfolder green fluorescent protein (sfGFP) production was inhibited by L-681,217. Biotinyl-L-681,217 was used to drain the CFPS system of endogenous EF-Tu, allowing replenishment with orthologs to interrogate pathogen selectivity and propensity toward resistance. Comparative in vitro analysis of kirromycin and L-681,217 showed that, while both antibiotics are equipotent in CFPS assays, they interact distinctly with purified EF-Tu, a feature that presumably correlates with prior observations that kirromycin enhances GTP hydrolysis by EF-Tu whereas L-681,217 does not. Analysis of L-681,217 and kirromycin accumulation in selected mutant E. coli strains also revealed that antibiotic import and efflux contributed to resistance. The promise of L-681,217 as a medicinal lead was underscored by the observation that, unlike aurodox, this polyketide does not inhibit adenylosuccinate synthase.
Collapse
Affiliation(s)
- Alexander M Soohoo
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
- Sarafan ChEM-H, Stanford University, Stanford, California 94305, United States
| | - Rolin A Aguilar
- Sarafan ChEM-H, Stanford University, Stanford, California 94305, United States
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Heewon Cho
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Thomas M Privalsky
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Lin Liu
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Khanh P Nguyen
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Christopher T Walsh
- Sarafan ChEM-H, Stanford University, Stanford, California 94305, United States
| | - Chaitan Khosla
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
- Sarafan ChEM-H, Stanford University, Stanford, California 94305, United States
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
4
|
Chen S, Huang B, Tian J, Zhang W. Advancements of Porphyrin-Derived Nanomaterials for Antibacterial Photodynamic Therapy and Biofilm Eradication. Adv Healthc Mater 2024; 13:e2401211. [PMID: 39073000 DOI: 10.1002/adhm.202401211] [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: 04/01/2024] [Revised: 06/17/2024] [Indexed: 07/30/2024]
Abstract
The threat posed by antibiotic-resistant bacteria and the challenge of biofilm formation has highlighted the inadequacies of conventional antibacterial therapies, leading to increased interest in antibacterial photodynamic therapy (aPDT) in recent years. This approach offers advantages such as minimal invasiveness, low systemic toxicity, and notable effectiveness against drug-resistant bacterial strains. Porphyrins and their derivatives, known for their high molar extinction coefficients and singlet oxygen quantum yields, have emerged as crucial photosensitizers in aPDT. However, their practical application is hindered by challenges such as poor water solubility and aggregation-induced quenching. To address these limitations, extensive research has focused on the development of porphyrin-based nanomaterials for aPDT, enhancing the efficacy of photodynamic sterilization and broadening the range of antimicrobial activity. This review provides an overview of various porphyrin-based nanomaterials utilized in aPDT and biofilm eradication in recent years, including porphyrin-loaded inorganic nanoparticles, porphyrin-based polymer assemblies, supramolecular assemblies, metal-organic frameworks (MOFs), and covalent organic frameworks (COFs). Additionally, insights into the prospects of aPDT is offered, highlighting its potential for practical implementation.
Collapse
Affiliation(s)
- Suwen Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Baoxuan Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Jia Tian
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| |
Collapse
|
5
|
Gupta S, Luxami V, Paul K. Bacterial cell death to overcome drug resistance with multitargeting bis-naphthalimides as potent antibacterial agents against Enterococcus faecalis. J Mater Chem B 2024; 12:5645-5660. [PMID: 38747306 DOI: 10.1039/d3tb02804f] [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: 06/13/2024]
Abstract
The increasing frequency of drug-resistant pathogens poses serious health issues to humans around the globe, leading to the development of new antibacterial agents to conquer drug resistance and bacterial infections. In view of this, we have synthesized a series of bis-naphthalimides to respond to awful drug resistance. Bioactivity assay and structure-activity relationship disclosed that compounds 5d and 5o exhibit potent antibacterial activity against E. faecalis, outperforming the marketed antibiotics. These drug candidates not only inhibit the biofilm formation of E. faecalis but also display rapid bactericidal properties, thus delaying the development of drug resistance within 20 passages. To explore the mechanism of antibacterial activity against E. faecalis, biofunctional examination was carried out which unveiled that 5d and 5o effectively disrupt bacterial cell membranes, causing the leakage of cytoplasmic contents and metabolic activity loss. Concurrently, 5d and 5o effectively intercalate with DNA to block DNA replication, causing the build-up of excessive reactive oxygen species and inhibiting the glutathione activity, ultimately leading to oxidative damage of E. faecalis and cell death. In addition, these compounds readily bind with HSA with a high binding constant, indicating that these drug candidates could be easily delivered to the target site. The above finding manifested that these newly synthesized bis-naphthalimides with multitargeting antibacterial properties offer a new prospect to overcome drug resistance.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India.
| | - Vijay Luxami
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India.
| | - Kamaldeep Paul
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India.
| |
Collapse
|
6
|
Chedupaka R, Audipudi AV, Sangolkar AA, Mamidala S, Venkatesham P, Penta S, Vedula RR. Design, synthesis, molecular docking, and dynamic studies of novel thiazole derivatives incorporating benzimidazole moiety and assessment as antibacterial agents. Mol Divers 2024; 28:1565-1576. [PMID: 37490125 DOI: 10.1007/s11030-023-10675-x] [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: 02/20/2023] [Accepted: 06/15/2023] [Indexed: 07/26/2023]
Abstract
A general and sustainable approach for the synthesis of benzimidazole-thiazole compounds via an efficient, one-pot, domino, pseudo-four-component reaction using 5-amino-2-mercaptobenzimidazole, aralkyl halides, ammonium thiocyanate, and substituted α-bromo-acetophenones in glacial acetic acid at ambient temperature to give final compounds (4a-p) in good yields in shorter time. The spectral data of synthesized compounds were evaluated by analytical and spectral techniques (IR, 1H-NMR, 13C-NMR, and ESI-HRMS). Further, some of the synthesized compounds were screened for their in-vitro antibacterial activity studies using the agar well diffusion method against Gram-positive Streptococcus pneumoniae (2451) bacteria and Gram-negative Proteous mirabilis (2081) bacteria. Based on the MIC results, it was observed that the most active compounds 4b, 4e, 4f, and 4k show promising antibacterial activity with the zone of inhibition values of 2.85 cm 2.75 cm, 3.6 cm, and 3.3 cm against both Gram-negative and positive bacteria cell lines, respectively. Further, we have also insight into the molecular simulation studies, based on the binding results, compound 4i showed stable binding interactions with streptomycin drug with the active site of the gyrase protein (PDB ID: 1KIJ). The structure-activity relationship (SAR) studies of all the title scaffolds were also established. The antibacterial activity, molecular docking studies, and molecular dynamic simulations of the title compounds suggested that these are promising antibacterial active skeletons.
Collapse
Affiliation(s)
- Raju Chedupaka
- Department of Chemistry, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Amrutha V Audipudi
- Department of Botany and Microbiology, Acharya Nagarjuna University, Guntur, A.P., 522510, India
| | | | - Srikanth Mamidala
- Department of Chemistry, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Papisetti Venkatesham
- Department of Chemistry, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Santhosh Penta
- Department of Chemistry, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Rajeswar Rao Vedula
- Department of Chemistry, National Institute of Technology, Warangal, Telangana, 506004, India.
| |
Collapse
|
7
|
Esarev IV, Karge B, Zeng H, Lippmann P, Jones PG, Schrey H, Brönstrup M, Ott I. Silver Organometallics that are Highly Potent Thioredoxin and Glutathione Reductase Inhibitors: Exploring the Correlations of Solution Chemistry with the Strong Antibacterial Effects. ACS Infect Dis 2024; 10:1753-1766. [PMID: 38606463 PMCID: PMC11091889 DOI: 10.1021/acsinfecdis.4c00104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024]
Abstract
The antibacterial activity of silver species is well-established; however, their mechanism of action has not been adequately explored. Furthermore, issues of low-molecular silver compounds with cytotoxicity, stability, and solubility hamper their progress to drug leads. We have investigated silver N-heterocyclic carbene (NHC) halido complexes [(NHC)AgX, X = Cl, Br, and I] as a promising new type of antibacterial silver organometallics. Spectroscopic studies and conductometry established a higher stability for the complexes with iodide ligands, and nephelometry indicated that the complexes could be administered in solutions with physiological chloride levels. The complexes showed a broad spectrum of strong activity against pathogenic Gram-negative bacteria. However, there was no significant activity against Gram-positive strains. Further studies clarified that tryptone and yeast extract, as components of the culture media, were responsible for this lack of activity. The reduction of biofilm formation and a strong inhibition of both glutathione and thioredoxin reductases with IC50 values in the nanomolar range were confirmed for selected compounds. In addition to their improved physicochemical properties, the compounds with iodide ligands did not display cytotoxic effects, unlike the other silver complexes. In summary, silver NHC complexes with iodide secondary ligands represent a useful scaffold for nontoxic silver organometallics with improved physicochemical properties and a distinct mechanism of action that is based on inhibition of thioredoxin and glutathione reductases.
Collapse
Affiliation(s)
- Igor V. Esarev
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
| | - Bianka Karge
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Haoxuan Zeng
- Department
of Microbial Drugs, Helmholtz Centre for
Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig,
Inhoffenstraße 7, 38124 Braunschweig, Germany
- Institute
of Microbiology, Technische Universität
Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Petra Lippmann
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
| | - Peter G. Jones
- Institute
of Inorganic and Analytical Chemistry, Technische
Universität Braunschweig, Hagenring 30, 38106 Braunschweig, Germany
| | - Hedda Schrey
- Department
of Microbial Drugs, Helmholtz Centre for
Infection Research GmbH and German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig,
Inhoffenstraße 7, 38124 Braunschweig, Germany
- Institute
of Microbiology, Technische Universität
Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Mark Brönstrup
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Ingo Ott
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
| |
Collapse
|
8
|
Singh SB, Martin GE, McKittrick B, Crowther J, Fraenkel H, Lunn C, Bayne M, Perkins JB, Gullo V. History and Prospects of Drug Discovery and Development Collaboration between Industry and Academia. JOURNAL OF NATURAL PRODUCTS 2024; 87:1235-1245. [PMID: 38554098 DOI: 10.1021/acs.jnatprod.4c00081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2024]
Abstract
Research collaborations and licensing deals are critical for the discovery and development of life-saving drugs. This practice has been ongoing since the inception of the pharmaceutical industry. The current process of drug discovery and development is complex, regulated, and highly regimented, having evolved over time. Academia excels in the discovery of fundamental scientific concepts and biological processes, while industry excels in translational science and product development. Potential for collaboration exists at every step of the drug discovery and development continuum. This perspective walks through such collaborative activities, provides examples, and offers tips for potential collaborations.
Collapse
Affiliation(s)
- Sheo B Singh
- Charles A Dana Research Institute of Scientists Emeriti (RISE), Drew University, Madison, New Jersey 07054, United States
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Gary E Martin
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Brian McKittrick
- Charles A Dana Research Institute of Scientists Emeriti (RISE), Drew University, Madison, New Jersey 07054, United States
| | - Jonathan Crowther
- Charles A Dana Research Institute of Scientists Emeriti (RISE), Drew University, Madison, New Jersey 07054, United States
| | - Howard Fraenkel
- Charles A Dana Research Institute of Scientists Emeriti (RISE), Drew University, Madison, New Jersey 07054, United States
| | - Charles Lunn
- Charles A Dana Research Institute of Scientists Emeriti (RISE), Drew University, Madison, New Jersey 07054, United States
| | - Marvin Bayne
- Charles A Dana Research Institute of Scientists Emeriti (RISE), Drew University, Madison, New Jersey 07054, United States
| | - John B Perkins
- Charles A Dana Research Institute of Scientists Emeriti (RISE), Drew University, Madison, New Jersey 07054, United States
| | - Vincent Gullo
- Charles A Dana Research Institute of Scientists Emeriti (RISE), Drew University, Madison, New Jersey 07054, United States
| |
Collapse
|
9
|
Zhao X, Cao X, Qiu H, Liang W, Jiang Y, Wang Q, Wang W, Li C, Li Y, Han B, Tang K, Zhao L, Zhang X, Wang X, Liang H. Rational molecular design converting fascaplysin derivatives to potent broad-spectrum inhibitors against bacterial pathogens via targeting FtsZ. Eur J Med Chem 2024; 270:116347. [PMID: 38552428 DOI: 10.1016/j.ejmech.2024.116347] [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: 02/05/2024] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 04/21/2024]
Abstract
The filamentous temperature-sensitive mutant Z protein (FtsZ), a key player in bacterial cell division machinery, emerges as an attractive target to tackle the plight posed by the ever growing antibiotic resistance over the world. Therefore in this regard, agents with scaffold diversities and broad-spectrum antibacterial activity against Gram-positive and Gram-negative pathogens are highly needed. In this study, a new class of marine-derived fascaplysin derivatives has been designed and synthesized by Suzuki-Miyaura cross-coupling. Some compounds exhibited potent bactericidal activities against a panel of Gram-positive (MIC = 0.024-6.25 μg/mL) and Gram-negative (MIC = 1.56-12.5 μg/mL) bacteria including methicillin-resistant S. aureus (MRSA). They exerted their effects by dual action mechanism via disrupting the integrity of the bacterial cell membrane and targeting FtsZ protein. These compounds stimulated polymerization of FtsZ monomers and bundling of the polymers, and stabilized the resulting polymer network, thus leading to the dysfunction of FtsZ in cell division. In addition, these agents showed negligible hemolytic activity and low cytotoxicity to mammalian cells. The studies on docking and molecular dynamics simulations suggest that these inhibitors bind to the hydrophilic inter-domain cleft of FtsZ protein and the insights obtained in this study would facilitate the development of potential drugs with broad-spectrum bioactivities.
Collapse
Affiliation(s)
- Xing Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China; Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Xuanyu Cao
- Health Science Center, Ningbo University, Ningbo, 315211, China; Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China
| | - Hongda Qiu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Weida Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Yinli Jiang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Qiang Wang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Weile Wang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Chengxi Li
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Yang Li
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China
| | - Bowen Han
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China
| | - Keqi Tang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Lingling Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Xuan Zhang
- Health Science Center, Ningbo University, Ningbo, 315211, China; Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China.
| | - Xiao Wang
- Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Hongze Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
10
|
Peng X, Chen J, Gan Y, Yang L, Luo Y, Bu C, Huang Y, Chen X, Tan J, Yang YY, Yuan P, Ding X. Biofunctional lipid nanoparticles for precision treatment and prophylaxis of bacterial infections. SCIENCE ADVANCES 2024; 10:eadk9754. [PMID: 38578994 PMCID: PMC10997193 DOI: 10.1126/sciadv.adk9754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/04/2024] [Indexed: 04/07/2024]
Abstract
The lack of bacterial-targeting function in antibiotics and their prophylactic usage have caused overuse of antibiotics, which lead to antibiotic resistance and inevitable long-term toxicity. To overcome these issues, we develop neutrophil-bacterial hybrid cell membrane vesicle (HMV)-coated biofunctional lipid nanoparticles (LNP@HMVs), which are designed to transport antibiotics specifically to bacterial cells at the infection site for the effective treatment and prophylaxis of bacterial infection. The dual targeting ability of HMVs to inflammatory vascular endothelial cells and homologous Gram-negative bacterial cells results in targeted accumulation of LNP@HMVs in the site of infections. LNP@HMVs loaded with the antibiotic norfloxacin not only exhibit enhanced activity against planktonic bacteria and bacterial biofilms in vitro but also achieve potent therapeutic efficacy in treating both systemic infection and lung infection. Furthermore, LNP@HMVs trigger the activation of specific humoral and cellular immunity to prevent bacterial infection. Together, LNP@HMVs provide a promising strategy to effectively treat and prevent bacterial infection.
Collapse
Affiliation(s)
- Xinran Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Jiaoyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yingying Gan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Li Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yuanjing Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Changxin Bu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yi Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xinhai Chen
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518132, PR China
| | - Jeremy Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
- State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
11
|
Noto Guillen M, Li C, Rosener B, Mitchell A. Antibacterial activity of nonantibiotics is orthogonal to standard antibiotics. Science 2024; 384:93-100. [PMID: 38484036 DOI: 10.1126/science.adk7368] [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: 09/13/2023] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
Numerous nonantibiotic drugs have potent antibacterial activity and can adversely affect the human microbiome. The mechanistic underpinning of this toxicity remains largely unknown. We investigated the antibacterial activity of 200 drugs using genetic screens with thousands of barcoded Escherichia coli knockouts. We analyzed 2 million gene-drug interactions underlying drug-specific toxicity. Network-based analysis of drug-drug similarities revealed that antibiotics clustered into modules that are consistent with the mode of action of their established classes, whereas nonantibiotics remained unconnected. Half of the nonantibiotics clustered into separate modules, potentially revealing shared and unexploited targets for new antimicrobials. Analysis of efflux systems revealed that they widely affect antibiotics and nonantibiotics alike, suggesting that the impact of nonantibiotics on antibiotic cross-resistance should be investigated closely in vivo.
Collapse
Affiliation(s)
- Mariana Noto Guillen
- Department of Systems Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Carmen Li
- Department of Systems Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Brittany Rosener
- Department of Systems Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Amir Mitchell
- Department of Systems Biology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
12
|
Kasdekar N, Spieker MR, Crich D. Practical Synthesis from Streptomycin and Regioselective Partial Deprotections of (-)-(1 R,2 S,3 R,4 R,5 S,6 S)-1,3-Di(deamino)-1,3-diazido-2,5,6-tri- O-benzylstreptamine. J Org Chem 2024; 89:4225-4231. [PMID: 38427951 PMCID: PMC10949228 DOI: 10.1021/acs.joc.3c02922] [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: 12/20/2023] [Revised: 02/09/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
We describe the gram-scale synthesis of (-)-(1R,2S,3R,4R,5S,6S)-1,3-di(diamino)-1,3-diazido-2,5,6-tri-O-benzylstreptamine from streptomycin by (i) hydrolysis of the two streptomycin guanidine residues, (ii) reprotection of the amines as azides, (iii) protection of all alcohols as benzyl ethers, and (iv) glycosidic bond cleavage with HCl in methanol. Protocols for regioselective monodebenzylation and regioselective reduction of a single azide in the product are also described, providing four optically pure building blocks for exploitation in novel aminoglycoside synthesis.
Collapse
Affiliation(s)
- Niteshlal Kasdekar
- Department
of Pharmaceutical and Biomedical Sciences, University of Georgia, 250 West Green Street, Athens, Georgia 30602, United States
| | - Michael R. Spieker
- Department
of Pharmaceutical and Biomedical Sciences, University of Georgia, 250 West Green Street, Athens, Georgia 30602, United States
- Department
of Biochemistry and Molecular Biology, University
of Georgia, 120 East Green Street, Athens, Georgia 30602, United States
| | - David Crich
- Department
of Pharmaceutical and Biomedical Sciences, University of Georgia, 250 West Green Street, Athens, Georgia 30602, United States
- Department
of Chemistry, University of Georgia, 302 East Campus Road, Athens, Georgia 30602, United States
- Complex
Carbohydrate Research Center, University
of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| |
Collapse
|
13
|
Homayonia S, Ling CC. Epoxide-Mediated Trans-Thioglycosylation and Application to the Synthesis of Oligosaccharides Related to the Capsular Polysaccharides of C. jejuni HS:4. Chemistry 2024; 30:e202303753. [PMID: 38215247 DOI: 10.1002/chem.202303753] [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: 11/10/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/14/2024]
Abstract
The enzyme-resistant thioglycosides are highly valuable immunogens because of their enhanced metabolic stability. We report the first synthesis of a family of thiooligosaccharides related to the capsular polysaccharides (CPS) of Campylobacter jejuni HS:4 for potential use in conjugate vaccines. The native CPS structures of the pathogen consist of a challenging repeating disaccharide formed with β(1→4)-linked 6-deoxy-β-D-ido-heptopyranoside and N-acetyl-D-glucosamine; the rare 6-deoxy-ido-heptopyranosyl backbone and β-anomeric configuration of the former monosaccharide makes the synthesis of this family of antigens very challenging. So far, no synthesis of the thioanalogs of the CPS antigens have been reported. The unprecedented synthesis presented in this work is built on an elegant approach by using β-glycosylthiolate as a glycosyl donor to open the 2,3-epoxide functionality of pre-designed 6-deoxy-β-D-talo-heptopyranosides. Our results illustrated that this key trans-thioglycosylation can be designed in a modular and regio and stereo-selective manner. Built on the success of this novel approach, we succeeded the synthesis of a family of thiooligosaccharides including a thiohexasaccharide which is considered to be the desired antigen length and complexity for immunizations. We also report the first direct conversion of base-stable but acid-labile 2-trimethylsilylethyl glycosides to glycosyl-1-thioacetates in a one-pot manner.
Collapse
Affiliation(s)
- Saba Homayonia
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Chang-Chun Ling
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
14
|
Fernández-Pastor I, Ortiz-López FJ, Oves-Costales D, Martín J, Sánchez P, Melguizo Á, Reyes F, Weber T, Genilloud O. Dilarmycins A-C, Calcium-Dependent Lipopeptide Antibiotics with a Non-canonical Ca 2+-Binding Motif. Org Lett 2024; 26:1343-1347. [PMID: 38329455 DOI: 10.1021/acs.orglett.3c04195] [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: 02/09/2024]
Abstract
Genome analysis of strain Streptomyces sp. CA-278952 revealed a biosynthetic gene cluster encoding a putative lipopeptide with a sequence containing an Asp-Gly-Glu-Ala motif. We envisioned that this motif could mimic the canonical Asp-X-Asp-Gly sequence found in previously reported calcium-dependent lipopeptide antibiotics. Chemical investigation of the producing strain led to the discovery of three novel lipodepsipeptides, dilarmycins A-C. The calcium-dependent antibacterial activity of the new compounds was confirmed against the Gram-positive pathogens methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus.
Collapse
Affiliation(s)
- Ignacio Fernández-Pastor
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía. Avenida del Conocimiento, 34, 18016 Armilla, Granada, Spain
| | - Francisco Javier Ortiz-López
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía. Avenida del Conocimiento, 34, 18016 Armilla, Granada, Spain
| | - Daniel Oves-Costales
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía. Avenida del Conocimiento, 34, 18016 Armilla, Granada, Spain
| | - Jesús Martín
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía. Avenida del Conocimiento, 34, 18016 Armilla, Granada, Spain
| | - Pilar Sánchez
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía. Avenida del Conocimiento, 34, 18016 Armilla, Granada, Spain
| | - Ángeles Melguizo
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía. Avenida del Conocimiento, 34, 18016 Armilla, Granada, Spain
| | - Fernando Reyes
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía. Avenida del Conocimiento, 34, 18016 Armilla, Granada, Spain
| | - Tilmann Weber
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Søltoft Plads, Building 220, 2800 Kongens Lyngby, Denmark
| | - Olga Genilloud
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía. Avenida del Conocimiento, 34, 18016 Armilla, Granada, Spain
| |
Collapse
|
15
|
Barth SA, Preussger D, Pietschmann J, Feßler AT, Heller M, Herbst W, Schnee C, Schwarz S, Kloss F, Berens C, Menge C. In Vitro Antibacterial Activity of Microbial Natural Products against Bacterial Pathogens of Veterinary and Zoonotic Relevance. Antibiotics (Basel) 2024; 13:135. [PMID: 38391521 PMCID: PMC10886079 DOI: 10.3390/antibiotics13020135] [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: 12/07/2023] [Revised: 01/23/2024] [Accepted: 01/28/2024] [Indexed: 02/24/2024] Open
Abstract
Antimicrobial resistance (AMR) is considered one of the greatest threats to both human and animal health. Efforts to address AMR include implementing antimicrobial stewardship programs and introducing alternative treatment options. Nevertheless, effective treatment of infectious diseases caused by bacteria will still require the identification and development of new antimicrobial agents. Eight different natural products were tested for antimicrobial activity against seven pathogenic bacterial species (Brachyspira sp., Chlamydia sp., Clostridioides sp., Mannheimia sp., Mycobacterium sp., Mycoplasma sp., Pasteurella sp.). In a first pre-screening, most compounds (five out of eight) inhibited bacterial growth only at high concentrations, but three natural products (celastramycin A [CA], closthioamide [CT], maduranic acid [MA]) displayed activity at concentrations <2 µg/mL against Pasteurella sp. and two of them (CA and CT) also against Mannheimia sp. Those results were confirmed by testing a larger collection of isolates encompassing 64 Pasteurella and 56 Mannheimia field isolates originating from pigs or cattle, which yielded MIC90 values of 0.5, 0.5, and 2 µg/mL against Pasteurella and 0.5, 4, and >16 µg/mL against Mannheimia for CA, CT, and MA, respectively. CA, CT, and MA exhibited higher MIC50 and MIC90 values against Pasteurella isolates with a known AMR phenotype against commonly used therapeutic antimicrobial agents than against isolates with unknown AMR profiles. This study demonstrates the importance of whole-cell antibacterial screening of natural products to identify promising scaffolds with broad- or narrow-spectrum antimicrobial activity against important Gram-negative veterinary pathogens with zoonotic potential.
Collapse
Affiliation(s)
- Stefanie A Barth
- Friedrich-Loeffler-Institut-Federal Research Institute for Animal Health (FLI), Institute of Molecular Pathogenesis, 07743 Jena, Germany
| | - Daniel Preussger
- Friedrich-Loeffler-Institut-Federal Research Institute for Animal Health (FLI), Institute of Molecular Pathogenesis, 07743 Jena, Germany
| | - Jana Pietschmann
- Friedrich-Loeffler-Institut-Federal Research Institute for Animal Health (FLI), Institute of Molecular Pathogenesis, 07743 Jena, Germany
| | - Andrea T Feßler
- Institute of Microbiology and Epizootics, Freie Universität Berlin, 14163 Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, 14163 Berlin, Germany
| | - Martin Heller
- Friedrich-Loeffler-Institut-Federal Research Institute for Animal Health (FLI), Institute of Molecular Pathogenesis, 07743 Jena, Germany
| | - Werner Herbst
- Institute of Hygiene and Infectious Diseases of Animals, Justus-Liebig-University, 35392 Giessen, Germany
| | - Christiane Schnee
- Friedrich-Loeffler-Institut-Federal Research Institute for Animal Health (FLI), Institute of Molecular Pathogenesis, 07743 Jena, Germany
| | - Stefan Schwarz
- Institute of Microbiology and Epizootics, Freie Universität Berlin, 14163 Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, 14163 Berlin, Germany
| | - Florian Kloss
- Transfer Group Anti-Infectives, Leibniz Institute for Natural Product Research and Infection Biology, Leibniz-HKI, 07745 Jena, Germany
| | - Christian Berens
- Friedrich-Loeffler-Institut-Federal Research Institute for Animal Health (FLI), Institute of Molecular Pathogenesis, 07743 Jena, Germany
| | - Christian Menge
- Friedrich-Loeffler-Institut-Federal Research Institute for Animal Health (FLI), Institute of Molecular Pathogenesis, 07743 Jena, Germany
| |
Collapse
|
16
|
Mandal GH, Sathyamoorthi S. Sulfamate-Tethered Aza-Wacker Strategy for a Kasugamine Synthon. J Org Chem 2024; 89:793-797. [PMID: 38062940 PMCID: PMC10798055 DOI: 10.1021/acs.joc.3c02292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
We present our preparation of a kasugamine synthon, which proceeds in 14 steps from a literature epoxide. We expect that this kasugamine derivative can be used for the total syntheses of kasugamycin, minosaminomycin, and analogue antibiotics. A key step in the synthesis is our laboratory's sulfamate-tethered aza-Wacker cyclization.
Collapse
Affiliation(s)
- Gour Hari Mandal
- University of Kansas, Department of Medicinal Chemistry, Lawrence, KS, USA (66047)
| | - Shyam Sathyamoorthi
- University of Kansas, Department of Medicinal Chemistry, Lawrence, KS, USA (66047)
| |
Collapse
|
17
|
Theuretzbacher U, Blasco B, Duffey M, Piddock LJV. Unrealized targets in the discovery of antibiotics for Gram-negative bacterial infections. Nat Rev Drug Discov 2023; 22:957-975. [PMID: 37833553 DOI: 10.1038/s41573-023-00791-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 10/15/2023]
Abstract
Advances in areas that include genomics, systems biology, protein structure determination and artificial intelligence provide new opportunities for target-based antibacterial drug discovery. The selection of a 'good' new target for direct-acting antibacterial compounds is the first decision, for which multiple criteria must be explored, integrated and re-evaluated as drug discovery programmes progress. Criteria include essentiality of the target for bacterial survival, its conservation across different strains of the same species, bacterial species and growth conditions (which determines the spectrum of activity of a potential antibiotic) and the level of homology with human genes (which influences the potential for selective inhibition). Additionally, a bacterial target should have the potential to bind to drug-like molecules, and its subcellular location will govern the need for inhibitors to penetrate one or two bacterial membranes, which is a key challenge in targeting Gram-negative bacteria. The risk of the emergence of target-based drug resistance for drugs with single targets also requires consideration. This Review describes promising but as-yet-unrealized targets for antibacterial drugs against Gram-negative bacteria and examples of cognate inhibitors, and highlights lessons learned from past drug discovery programmes.
Collapse
Affiliation(s)
| | - Benjamin Blasco
- Global Antibiotic Research and Development Partnership (GARDP), Geneva, Switzerland
| | - Maëlle Duffey
- Global Antibiotic Research and Development Partnership (GARDP), Geneva, Switzerland
| | - Laura J V Piddock
- Global Antibiotic Research and Development Partnership (GARDP), Geneva, Switzerland.
| |
Collapse
|
18
|
Chen X, Li W, Li X, Li K, Zhang G, Hong W. Photodynamic Cationic Ultrasmall Copper Oxide Nanoparticles-Loaded Liposomes for Alleviation of MRSA Biofilms. Int J Nanomedicine 2023; 18:5441-5455. [PMID: 37753066 PMCID: PMC10519346 DOI: 10.2147/ijn.s426682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Introduction As we enter the post-antibiotic era, the rise of antibiotic-resistant pathogenic bacteria is becoming a serious threat to public health. This problem is further complicated by antibiotic-resistant biofilms, for which current treatment options are limited. Methods To tackle this challenge, we propose a novel approach that involves the use of photodynamic cationic pH-sensitive liposomes loaded with ultra-small copper oxide (Ce6@Lipo/UCONs) to effectively eliminate drug-resistant bacteria and eradicate biofilms while minimizing safety concerns and the risk of resistance development. Results Our study demonstrates that Ce6@Lipo/UCONs have minimal toxicity to mammalian cells and can significantly enhance the association affinity with methicillin-resistant Staphylococcus aureus (MRSA) as confirmed by fluorescent microscope and flow cytometry, thereby greatly improving the bactericidal effect against planktonic MRSA. The cationic nature of Ce6@Lipo/UCONs also enables them to penetrate MRSA biofilms and respond to the acidic microenvironment within the biofilm, effectively releasing the loaded UCONs. Our results indicate that Ce6@Lipo/UCONs could effectively eliminate biofilms under light irradiation conditions, as evidenced by both biomass analysis and scanning electron microscopy observations. In addition, significant antibacterial effects and abscess healing were observed in MRSA-infected mice treated with Ce6@Lipo/UCONs upon light irradiation, while good biocompatibility was achieved in vivo. Conclusion Taken together, our findings suggest that photodynamic cationic ultrasmall copper oxide nanoparticles-loaded liposomes are a highly promising nano platform for combating antibiotic-resistant microbial pathogens and biofilms. The effective biofilm penetration and synergistic effect between photodynamic inactivation and metal sterilization make them a valuable tool for overcoming the challenges posed by antibiotic resistance.
Collapse
Affiliation(s)
- Xiangjun Chen
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Wenting Li
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Xueling Li
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Keke Li
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Guilong Zhang
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| | - Wei Hong
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, People’s Republic of China
| |
Collapse
|
19
|
Panda S, Kumari S, Dixit M, Sharma NK. N-Salicyl-AA n-picolamide Foldameric Peptides Exhibit Quorum Sensing Inhibition of Pseudomonas aeruginosa (PA14). ACS OMEGA 2023; 8:30349-30358. [PMID: 37636905 PMCID: PMC10448664 DOI: 10.1021/acsomega.3c03404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023]
Abstract
An organic acid, salicylic acid, and its derivatives are constituents of various natural products possessing remarkable bioactivity. O-Acetyl salicylate (aspirin) is a well-known life-saving drug. Its peptide derivative salicylamide has also been explored in the designing of peptide-based therapeutic drugs. An organic base, picolylamine has been recently explored for designing diagnostic probes. However, both the acid and base have common features as metal chelating with coordinating metals. Thus, these scaffolds could be used for designing inhibitors of various metalloenzymes. Their characteristic properties encourage us to design peptides containing both scaffolds (salicylic acid and picolylamine) at opposite terminals. So far there is no report available on such conjugated peptides. This report describes the synthesis, conformational analysis, and biochemical assessment of rationally designed N-salicyl-AAn-picolamide peptides. Pleasantly, we have obtained the crystal structures of representative peptides that confirm their roles in conformational changes. Our biological assessment as quorum sensing inhibitors has revealed that their di/tripeptides inhibit quorum sensing of the pathogenic bacterium PA14 strain. Hence, these peptides have promising foldameric and therapeutic values.
Collapse
Affiliation(s)
- Subhashree
S. Panda
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO: Jatani, Bhubaneswar 752050, Odisha, India
- Homi
Bhabha National Institute, Training School
Complex, Anushaktinagar, Mumbai 400094, India
| | - Supriya Kumari
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO: Jatani, Bhubaneswar 752050, Odisha, India
- School
of biological Sciences, National Institute
of Science Education and Research (NISER), PO: Jatani, Bhubaneswar 752050, Odisha, India
| | - Manjusha Dixit
- School
of biological Sciences, National Institute
of Science Education and Research (NISER), PO: Jatani, Bhubaneswar 752050, Odisha, India
- Homi
Bhabha National Institute, Training School
Complex, Anushaktinagar, Mumbai 400094, India
| | - Nagendra K. Sharma
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO: Jatani, Bhubaneswar 752050, Odisha, India
- Homi
Bhabha National Institute, Training School
Complex, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
20
|
Fu J, Nakata Y, Itoh H, Panthee S, Hamamoto H, Sekimizu K, Inoue M. Molecular Editing Enhances Oxidation Resistance of Menaquinone-Targeting Antibiotics Lysocin E and WAP-8294A2. Chemistry 2023; 29:e202301224. [PMID: 37328428 DOI: 10.1002/chem.202301224] [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: 04/19/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 06/18/2023]
Abstract
Lysocin E (1 a) and WAP-8294A2 (2 a) are peptidic natural products with 37- and 40-membered macrocycles, respectively. Compounds 1 a and 2 a have potent antibacterial activities against Gram-positive bacteria and share a unique mode of action. The electron-rich indole ring of d-Trp-10 of 1 a and 2 a interacts with the electron-deficient benzoquinone ring of menaquinone, which is a co-enzyme in the bacterial respiratory chain. Formation of the electron-donor-acceptor complex causes membrane disruption, leading to cell death. Despite the promising activities of 1 a and 2 a, the susceptibility of Trp-10 to oxidative degradation potentially deters the development of these compounds as antibacterial drugs. To address this issue, we replaced the indole ring with more oxidation-resistant aromatics having a similar shape and electron-rich character. Specifically, analogues with benzofuran (1 b/2 b), benzothiophene (1 c/2 c), and 1-naphthalene (1 d/2 d) rings were designed, and chemically prepared by full solid-phase total syntheses. Antibacterial assays of the six analogues revealed similar activities of 1 d/2 d and markedly reduced activities of 1 b/2 b and 1 c/2 c compared with 1 a/2 a. Equipotent 1 d and 2 d both showed high resistance to oxidation by peroxyl radicals. Hence, the present study demonstrates a new molecular editing strategy for conferring oxidation stability on natural products with pharmacologically useful functions.
Collapse
Affiliation(s)
- Junhao Fu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yosuke Nakata
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroaki Itoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Suresh Panthee
- GenEndeavor LLC, 26219 Eden Landing Rd, Hayward, CA, 94545, USA
- Faculty of Pharma-Science, Teikyo University, 359 Otsuka, Hachioji, Tokyo, 192-0395, Japan
| | - Hiroshi Hamamoto
- Yamagata University Faculty of Medicine, 2-2-2 Iida-Nishi, Yamagata, Yamagata, 990-9585, Japan
| | - Kazuhisa Sekimizu
- Faculty of Pharma-Science, Teikyo University, 359 Otsuka, Hachioji, Tokyo, 192-0395, Japan
| | - Masayuki Inoue
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
21
|
Zeng P, Zhang P, Yi L, Wong KY, Chen S, Chan KF, Leung SSY. A novel ESKAPE-sensitive peptide with enhanced stability and its application in controlling multiple bacterial contaminations in chilled fresh pork. Food Chem 2023; 413:135647. [PMID: 36796263 DOI: 10.1016/j.foodchem.2023.135647] [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: 12/02/2022] [Revised: 02/02/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023]
Abstract
The co-existence of various pathogenic bacteria on the surface of pork products exacerbates difficulties in food safety control. Developing broad-spectrum and stable antibacterial agents that are not antibiotics is an unmet need. To address this issue, all l-arginine residues of a reported peptide (IIRR)4-NH2 (zp80) were substituted with the corresponding D enantiomers. This novel peptide (IIrr)4-NH2 (zp80r) was expected to maintain favourable bioactivity against ESKAPE strains and have enhanced proteolytic stability compared with zp80. In a series of experiments, zp80r maintained favourable bioactivities against starvation-induced persisters. Electron microscopy and fluorescent dye assays were used to verify the antibacterial mechanism of zp80r. Importantly, zp80r reduced bacterial colonies in chilled fresh pork contaminated with multiple bacterial species. This newly designed peptide is a potential antibacterial candidate to combat problematic foodborne pathogens during storage of pork.
Collapse
Affiliation(s)
- Ping Zeng
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Pengfei Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Lanhua Yi
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong; College of Food Science, Southwest University, Chongqing, PR China
| | - Kwok-Yin Wong
- State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Sheng Chen
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Kin-Fai Chan
- State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| | - Sharon Shui Yee Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
22
|
Labes A. Marine Resources Offer New Compounds and Strategies for the Treatment of Skin and Soft Tissue Infections. Mar Drugs 2023; 21:387. [PMID: 37504918 PMCID: PMC10381745 DOI: 10.3390/md21070387] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Bioprospecting of the marine environment for drug development has gained much attention in recent years owing to its massive chemical and biological diversity. Drugs for the treatment of skin and soft tissue infections have become part of the search, mainly with respect to enlarging the number of available antibiotics, with a special focus on multidrug-resistant Gram-positive bacteria, being the major causative agents in this field. Marine resources offer novel natural products with distinct biological activities of pharmaceutical importance, having the chance to provide new chemical scaffolds and new modes of action. New studies advance the field by proposing new strategies derived from an ecosystemic understanding for preventive activities against biofilms and new compounds suitable as disinfectants, which sustain the natural flora of the skin. Still, the development of new compounds is often stuck at the discovery level, as marine biotechnology also needs to overcome technological bottlenecks in drug development. This review summarizes its potential and shows these bottlenecks and new approaches.
Collapse
Affiliation(s)
- Antje Labes
- Department of Energy and Biotechnology, Flensburg University of Applied Sciences ZAiT, Kanzleistraße 91-93, D-24943 Flensburg, Germany
| |
Collapse
|
23
|
Li L. Accessing hidden microbial biosynthetic potential from underexplored sources for novel drug discovery. Biotechnol Adv 2023:108176. [PMID: 37211187 DOI: 10.1016/j.biotechadv.2023.108176] [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: 11/14/2022] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Microbial natural products and their structural analogues have widely used as pharmaceutical agents, especially for infectious diseases and cancer. Despite this success, new structural classes with innovative chemistry and modes of action are urgently needed to be developed to combat the growing antimicrobial resistance and other public health problems. The advances in next-generation sequencing technologies and powerful computational tools open up new opportunities to explore microbial biosynthetic potential from underexplored sources, with millions of secondary metabolites awaiting discovery. The review highlights challenges associated with discovery of new chemical entities, rich reservoirs provided by untapped taxa, ecological niches or host microbiomes, emerging synthetic biotechnologies to unearth the hidden microbial biosynthetic potential for novel drug discovery at scale and speed.
Collapse
Affiliation(s)
- Lei Li
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai 200240, China.
| |
Collapse
|
24
|
Bremner JB. An Update Review of Approaches to Multiple Action-Based Antibacterials. Antibiotics (Basel) 2023; 12:antibiotics12050865. [PMID: 37237768 DOI: 10.3390/antibiotics12050865] [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: 04/14/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Many approaches are being pursued to address the major global health challenge posed by the increasing resistance of pathogenic bacteria to antibacterial agents. One of the promising approaches being investigated includes the design and development of multiple action-based small-molecule antibacterials. Aspects of this broad area have been reviewed previously, and recent developments are addressed in this update review covering the literature mainly over the past three years. Considerations encompassing drug combinations, single-molecule hybrids and prodrugs are summarised in regard to the intentional design and development of multiple-action agents with a focus on potential triple or greater activities in bacteria. The hope for such single agents or combinations of single agents is that resistance development will be significantly hindered, and they may be useful in tackling bacterial disease caused by both resistant and non-resistant bacteria.
Collapse
Affiliation(s)
- John B Bremner
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
25
|
Zhuang QQ, Yang JL, Qiu HN, Huang KY, Yang Y, Peng HP, Deng HH, Jiang HQ, Chen W. Promoting the healing of methicillin-resistant Staphylococcus aureus-infected wound by a multi-target antimicrobial AIEgen of 6-Aza-2-thiothymine-decorated gold nanoclusters. Colloids Surf B Biointerfaces 2023; 226:113336. [PMID: 37167770 DOI: 10.1016/j.colsurfb.2023.113336] [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: 03/25/2023] [Revised: 04/24/2023] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
The use of conventional antibiotic therapies is in question owing to the emergence of drug-resistant pathogenic bacteria. Therefore, novel, highly efficient antibacterial agents to effectively overcome resistant bacteria are urgently needed. Accordingly, in this work, we described a novel class luminogen of 6-Aza-2-thiothymine-decorated gold nanoclusters (ATT-AuNCs) with aggregation-induced emission property that possessed potent antimicrobial activity against methicillin-resistant Staphylococcus aureus (MRSA). Scanning electron microscopy was performed to investigate the interactions between ATT-AuNCs and MRSA. In addition, ATT-AuNCs exhibited excellent ROS generation efficiency and could effectively ablate MRSA via their internalization to the cells. Finally, tandem mass tag-labeling proteome analysis was carried out to investigate the differential expression proteins in MRSA strains. The results suggested that ATT-AuNCs killed MRSA cells through altering the expression of multiple target proteins involved in DNA replication, aminoacyl-tRNA synthesis, peptidoglycan and arginine biosynthesis metabolism. Parallel reaction monitoring technique was further used for the validation of these proteome results. ATT-AuNCs could also be served as a wound-healing agent and accelerate the healing process. Overall, we proposed ATT-AuNCs could serve as a robust antimicrobial aggregation-induced emission luminogen (AIEgen) that shows the ability to alter the activities of multiple targets for the elimination of drug-resistant bacteria.
Collapse
Affiliation(s)
- Quan-Quan Zhuang
- Department of Pharmacy, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou 362000, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350004, China
| | - Jia-Lin Yang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350004, China
| | - Hui-Na Qiu
- Department of Laboratory Medicine, Quanzhou Infectious Disease Hospital, Quanzhou 362000, China
| | - Kai-Yuan Huang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350004, China
| | - Yu Yang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350004, China
| | - Hua-Ping Peng
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350004, China
| | - Hao-Hua Deng
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350004, China.
| | - Hui-Qiong Jiang
- Department of Cardiac Function Examination Room, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou 362000, China.
| | - Wei Chen
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350004, China.
| |
Collapse
|
26
|
Edalatmand A, McArthur AG. CARD*Shark: automated prioritization of literature curation for the Comprehensive Antibiotic Resistance Database. Database (Oxford) 2023; 2023:7133783. [PMID: 37079891 PMCID: PMC10118295 DOI: 10.1093/database/baad023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/01/2023] [Accepted: 03/27/2023] [Indexed: 04/22/2023]
Abstract
Scientific literature is published at a rate that makes manual data extraction a highly time-consuming task. The Comprehensive Antibiotic Resistance Database (CARD) utilizes literature to curate information on antimicrobial resistance genes and to enable time-efficient triage of publications we have developed a classification algorithm for identifying publications describing first reports of new resistance genes. Trained on publications contained in the CARD, CARD*Shark downloads, processes and identifies publications recently added to PubMed that should be reviewed by biocurators. With CARD*Shark, we can minimize the monthly scope of articles a biocurator reviews from hundreds of articles to a few dozen, drastically improving the speed of curation while ensuring no relevant publications are overlooked. Database URL http://card.mcmaster.ca.
Collapse
Affiliation(s)
- Arman Edalatmand
- David Braley Centre for Antibiotic Discovery, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| | - Andrew G McArthur
- David Braley Centre for Antibiotic Discovery, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
27
|
Gulia K, Hassan AHE, Lenhard JR, Farahat AA. Escaping ESKAPE resistance: in vitro and in silico studies of multifunctional carbamimidoyl-tethered indoles against antibiotic-resistant bacteria. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230020. [PMID: 37090961 PMCID: PMC10113819 DOI: 10.1098/rsos.230020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/24/2023] [Indexed: 05/03/2023]
Abstract
Combining the hybridization and repurposing strategies, six compounds from our in-house library and having a designed hybrid structure of MBX-1162, pentamidine and MMV688271 were repurposed as potential antibacterial agents. Among, compounds 1a and 1d elicited potential sub-µg ml-1 activity against the high-priority antibiotic-resistant Gram-positive members of ESKAPE bacteria as well as antibiotic-susceptible Gram-positive bacteria. Furthermore, they showed potential low µg ml-1 activity against the explored critical-priority antibiotic-resistant Gram-negative members of ESKAPE bacteria. In time-kill assay, compound 1a has effective 0.5 and 0.25 µg ml-1 antibacterial lethal concentrations against MRSA in exponential growth phase. In silico investigations predicted compounds 1a and 1d as inhibitors of the open conformation of undecaprenyl diphosphate synthase involved in bacterial isoprenoid synthesis. In addition, compounds 1a and 1d were predicted as inhibitors of NADPH-free but not NADPH-bound form of ketol-acid reductoisomerase and may also serve as potential B-DNA minor groove binders with possible differences in the molecular sequence recognition. Overall, compounds 1a and 1d are presented as multifunctional potential antibacterial agents for further development against high- and critical-priority Gram-positive and Gram-negative antibiotic-resistant ESKAPE bacterial pathogens as well as antibiotic-susceptible Gram-positive bacterial pathogens.
Collapse
Affiliation(s)
- Kanika Gulia
- Master of Pharmaceutical Sciences Program, California Northstate University, 9700 W Taron Dr., Elk Grove, CA 95757, USA
- College of Medicine, California Northstate University, 9700 W Taron Dr., Elk Grove, CA 95757, USA
| | - Ahmed H. E. Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Justin R. Lenhard
- Department of Clinical and Administrative Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| | - Abdelbasset A. Farahat
- Master of Pharmaceutical Sciences Program, California Northstate University, 9700 W Taron Dr., Elk Grove, CA 95757, USA
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
28
|
Li Y, Luo S, Wang H, Lai Y, Li D, Zhang Q, Huang H, Zhang P. Photoacidolysis-Mediated Iridium(III) Complex for Photoactive Antibacterial Therapy. J Med Chem 2023; 66:4840-4848. [PMID: 36966514 DOI: 10.1021/acs.jmedchem.2c02000] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2023]
Abstract
Photoactive antibacterial therapy is one of the novel therapeutic methods that has great application potential and prospects for curbing bacterial infections. In this work, a photoactivated iridium complex (Ir-Cl) is synthesized for photoactive antibacterial research. Ir-Cl exhibits photoacidolysis, which can generate H+ and be converted into a photolysis product Ir-OH under blue light irradiation. At the meantime, this process is accompanied by 1O2 generation. Notably, Ir-Cl can selectively permeate S. aureus and exhibit excellent photoactive antibacterial activity. Mechanism studies show that Ir-Cl can ablate bacterial membranes and biofilms under light irradiation. Metabolomics analysis proves that Ir-Cl with light exposure mainly disturbs some amino acids' degradation (e.g., valine, leucine, isoleucine, arginine) and pyrimidine metabolism, which indirectly causes the ablation of biofilms and ultimately produces irreversible damage to S. aureus. This work provides guidance for metal complexes in antibacterial application.
Collapse
Affiliation(s)
- Yue Li
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Shuangling Luo
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Haobing Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Yidan Lai
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Dan Li
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Qianling Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Huaiyi Huang
- School of Pharmaceutical Science (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Shenzhen 518107, China
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| |
Collapse
|
29
|
He J, Xiong Y, Mu H, Li P, Deng Y, Zou W, Zhao Q. Antibacterial Properties of Three-Dimensional Flower Cluster ZIF-L Modified by N-Doped Carbon Dots. CRYSTALS 2023; 13:564. [DOI: 10.3390/cryst13040564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
To overcome the problems of excessive ion release of inorganic antimicrobial agents and the biological toxicity of organic antimicrobial agents, metal organic framework (MOF) materials are attracting attention in the antimicrobial field due to their tunable structural properties and multifunctional applications. Most current studies are limited to zeolitic imidazolate framework-8 (ZIF-8), which has low antimicrobial efficiency by component release. Two-dimensional (2D) zeolitic imidazolate framework nanoleaf (ZIF-L) possesses better antimicrobial effect than ZIF-8 because of the physical destructionto bacteria by its blade tip. However, the in-situ synthesis method of two-dimensional ZIF-L, and the problem of leaf accumulation, limit the wider application of ZIF-L. In this paper, three-dimensional(3D) flower cluster-like ZIF-L (2–3 μm, +31.23 mv), with better antibacterial effects and a wider application range, was prepared by stirring without adding other reagents. To further improve the antibacterial performance of ZIF-L, nitrogen-doped carbon dots (NCDs) were electrostatically absorbed by ZIF-L to obtain NCDs@ZIF-L composites. The NCDs@ZIF-L composites showed over 95% and 85% antibacterial efficiency against E. coli and S. aureus, respectively, at a concentration of 0.25 mg/mL. In addition, polylactic acid (PLA) films mixed with ZIF-L and NCDs@ZIF-L composites with PLA showed good antimicrobial properties, indicating the applicability of ZIF-L and NCDs@ZIF-L composites for antibacterial materials. With a unique three-dimensional crystal shape and positive surface charge, ZIF-L and NCDs@ZIF-L composites exhibited excellent antibacterial properties, which provided a new perspective for the study of antimicrobial materials.
Collapse
Affiliation(s)
- Jing He
- School of Chemical Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Yuanyuan Xiong
- School of Chemical Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Huaixuan Mu
- School of Chemical Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Peini Li
- School of Chemical Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Yiqing Deng
- School of Chemical Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Wangcai Zou
- School of Chemical Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| | - Qiang Zhao
- School of Chemical Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu 610065, China
| |
Collapse
|
30
|
Cheng K, Wang H, Sun S, Wu M, Shen H, Chen K, Zhang Z, Li S, Lin H. Specific Chemiluminescence Imaging and Enhanced Photodynamic Therapy of Bacterial Infections by Hemin-Modified Carbon Dots. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2207868. [PMID: 36965080 DOI: 10.1002/smll.202207868] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/28/2023] [Indexed: 06/18/2023]
Abstract
Antibacterial photodynamic therapy (aPDT) is a promising antibiotics-alternative strategy for bacterial infectious diseases, which features broad-spectrum antibacterial activity with a low risk of inducing bacterial resistance. However, clinical applications of aPDT are still hindered by the hydrophobicity-caused inadequate photodynamic activity of conventional photosensitizers and the hypoxic microenvironment of bacterial infections. To address these problems, herein, a promising strategy is developed to achieve specific chemiluminescence (CL) imaging and enhanced PDT of bacterial infections using hemin-modified carbon dots (H-CDs). The H-CDs can be facilely prepared and exhibit favorable water solubility, augmented photodynamic activity, and unique peroxidase-mimicking capacity. Compared with the free CDs, the photodynamic efficacy of H-CDs is significantly augmented due to the increased electron-hole separation efficiency. Moreover, the peroxidase catalytic performance of H-CDs enables not only infection identification via bacterial infection microenvironment-responsive CL imaging but also oxygen self-supplied aPDT with hypoxia-relief-enhanced bacteria inactivation effects. Finally, the enhanced aPDT efficiencies of H-CDs are validated in both in vivo abscess and infected wound models. This work may provide an effective antibacterial platform for the selective imaging-guided treatment of bacterial infections.
Collapse
Affiliation(s)
- Ke Cheng
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| | - Henggang Wang
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| | - Shan Sun
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| | - Mingyu Wu
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| | - Hongzhe Shen
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| | - Ke Chen
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| | - Zhiyuan Zhang
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| | - Si Li
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| | - Hengwei Lin
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, P. R. China
| |
Collapse
|
31
|
Kaguchi R, Katsuyama A, Sato T, Takahashi S, Horiuchi M, Yokota SI, Ichikawa S. Discovery of Biologically Optimized Polymyxin Derivatives Facilitated by Peptide Scanning and In Situ Screening Chemistry. J Am Chem Soc 2023; 145:3665-3681. [PMID: 36708325 DOI: 10.1021/jacs.2c12971] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Peptides can be converted to highly active compounds by introducing appropriate substituents on the suitable amino acid residue. Although modifiable residues in peptides can be systematically identified by peptide scanning methodologies, there is no practical method for optimization at the "scanned" position. With the purpose of using derivatives not only for scanning but also as a starting point for further chemical functionalization, we herein report the "scanning and direct derivatization" strategy through chemoselective acylation of embedded threonine residues by a serine/threonine ligation (STL) with the help of in situ screening chemistry. We have applied this strategy to the optimization of the polymyxin antibiotics, which were selected as a model system to highlight the power of the rapid derivatization of active scanning derivatives. Using this approach, we explored the structure-activity relationships of the polymyxins and successfully prepared derivatives with activity against polymyxin-resistant bacteria and those with Pseudomonas aeruginosa selective antibacterial activity. This strategy opens up efficient structural exploration and further optimization of peptide sequences.
Collapse
Affiliation(s)
- Rintaro Kaguchi
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo060-0812, Japan
| | - Akira Katsuyama
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo060-0812, Japan.,Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo060-0812, Japan.,Global Station for Biosurfaces and Drug Discovery, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo060-0812, Japan
| | - Toyotaka Sato
- Laboratory of Veterinary Hygiene, School/Faculty of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo060-0818, Japan.,Graduate School of Infectious Diseases, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo060-0818, Japan.,One Health Research Center, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo060-0818, Japan
| | - Satoshi Takahashi
- Department of Infection Control and Laboratory Medicine, Sapporo Medical University School of Medicine, Minami-1, Nishi-16, Chuo-ku, Sapporo060-8543, Japan.,Division of Laboratory Medicine, Sapporo Medical University Hospital, Minami-1, Nishi-16, Chuo-ku, Sapporo060-8543, Japan
| | - Motohiro Horiuchi
- Laboratory of Veterinary Hygiene, School/Faculty of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo060-0818, Japan.,Graduate School of Infectious Diseases, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo060-0818, Japan.,One Health Research Center, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo060-0818, Japan
| | - Shin-Ichi Yokota
- Department of Microbiology, Sapporo Medical University School of Medicine, Minami-1, Nishi-17, Chuo-ku, Sapporo060-8556, Japan
| | - Satoshi Ichikawa
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo060-0812, Japan.,Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo060-0812, Japan.,Global Station for Biosurfaces and Drug Discovery, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo060-0812, Japan
| |
Collapse
|
32
|
Gao J, Hou H, Gao F. Current scenario of quinolone hybrids with potential antibacterial activity against ESKAPE pathogens. Eur J Med Chem 2023; 247:115026. [PMID: 36577217 DOI: 10.1016/j.ejmech.2022.115026] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
The ESKAPE (Escherichia coli/E. coli, Staphylococcus aureus/S. aureus, Klebsiella pneumonia/K. pneumoniae, Acinetobacter Baumannii/A. baumannii, Pseudomonas aeroginosa/P. aeroginosa and Enterobacter spp.) pathogens, which could escape or evade common therapies through diverse antimicrobial resistance mechanisms and biofilm formation, are deemed as highly virulent bacteria responsible for life-threatening diseases, calling for novel chemotherapeutics. Quinolones including 2-quinolones and 4-quinolones have occupied a propitious place in drug design and development due to their excellent pharmacological profiles. Quinolones especially fluoroquinolones could inhibit the synthesis of nucleic acid of ESKAPE pathogens, leading to the rupture of bacterial chromosome. However, the resistance of ESKAPE pathogens to quinolones develops rapidly and spreads widely. Accordingly, it has become increasingly urgent to enhance the potency of quinolones against both drug-susceptible and drug-resistant ESKAPE pathogens. Quinolone hybrids can bind with different drug targets simultaneously and have been considered as useful prototypes to circumvent drug resistance. The purpose of this review is to summarize the current scenario (2018-present) of quinolone hybrids with potential antibacterial activity against ESKAPE pathogens, together with the structure-activity relationships and mechanisms of action to facilitate further rational design of more effective candidates.
Collapse
Affiliation(s)
- Jingyue Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Haodong Hou
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
33
|
Lu L, Dong F, Chen X, Guo T, Qian J, Xu X, Liu Y, Ma L, Pang L, Chen R, Wang P, Tang X. Preparation and properties of an antimicrobial silane-modified polyether sealant. Polym Bull (Berl) 2023. [DOI: 10.1007/s00289-023-04701-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
34
|
Ani NI, Okolo KO, Offiah RO. Evaluation of antibacterial, antioxidant, and anti-inflammatory properties of GC/MS characterized methanol leaf extract of Terminalia superba (Combretaceae, Engl. & Diels). FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2023. [DOI: 10.1186/s43094-022-00455-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Abstract
Background
Terminalia superba is a well-known medicinal plant used in folk medicine for the management of various diseases and swelling. Validation of its efficacy in standardized scientific models is lacking. This gap needs to be filled as a way of enhancing modern drug discovery. The aim is to evaluate the antibacterial, antioxidant, and anti-inflammatory properties of T. superba in known and established models. Also, to establish and possibly correlate the established activity with the phytochemicals identified using GC/MS and qualitative methods.
Results
The result showed a dose-dependent percentage inhibition of DPPH, HO•, and Fe3+ reducing activity. The antibacterial activity showed dose-dependent significant (p < 0.05) inhibition against all the organisms used. The anti-inflammatory activity of METS was confirmed in the carrageenan model with significant (p < 0.05) inhibition of paw volume when compared to control while significantly decreasing (p < 0.05) weight of xylene-induced ear. For instance, after 6 h, there was a reduction of 42%, 33%, and 22% for diclofenac, 200 mg, and 100 mg, respectively, as against 4% in control. The significant (p < 0.05) increase in MDA was attenuated by the treatment with METS dose dependently. Phytochemical assay and GC/MS characterization showed that alkaloids, saponins, phenols, quinone, tannins, coumarins, proteins, flavonoids, and amino acids were dominant with fatty acids accounting for 53%. Others are esters (23%), organic compounds (12%), alkanes (9%), and carboxylic acids (3%).
Conclusions
T. superba possesses antioxidant, antibacterial, and anti-inflammatory properties which are believed to arise from the secondary metabolites observed in the GC–MS characterization.
Graphical Abstract
Collapse
|
35
|
Fan W, Han H, Lu Z, Huang Y, Zhang Y, Chen Y, Zhang X, Ji J, Yao K. ε-poly-L-lysine-modified polydopamine nanoparticles for targeted photothermal therapy of drug-resistant bacterial keratitis. Bioeng Transl Med 2023; 8:e10380. [PMID: 36684079 PMCID: PMC9842021 DOI: 10.1002/btm2.10380] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/16/2022] [Indexed: 01/25/2023] Open
Abstract
Bacterial keratitis can lead to intraocular infection and even blindness without prompt and potent treatments. Currently, clinical abuse of antibiotics encouraged the evolution of resistant bacteria. Conventional antibiotic eye drops based keratitis treatment has been heavily restricted due to the lack of bactericidal efficiency and easy induction of bacterial resistance. Hence, developing an effective treatment strategy for bacterial keratitis is of great significance. In this work, we investigated ε-poly-l-lysine (EPL)-modified polydopamine (PDA) nanoparticles (EPL@PDA NPs)-mediated antibacterial photothermal therapy (aPTT), to cope with methicillin-resistant Staphylococcus aureus (MRSA)-induced keratitis. The surface modification of cationic peptide EPL enables EPL@PDA NPs to specifically target negatively charged MRSA and induces local hyperthermia to kill the bacteria under low ambient temperature. Under near-infrared (NIR) irradiation, the sterilization efficiency of EPL@PDA NPs suspension for MRSA in vitro was up to 99.96%. The EPL@PDA-mediated aPTT presented potent antibacterial efficacy in treating MRSA-induced keratitis with little corneal epithelial cytotoxicity and good biocompatibility. In conclusion, the bacterial-targeting aPTT platform in this work provides a prospective method for the management of MRSA-induced refractory bacterial keratitis.
Collapse
Affiliation(s)
- Wenjie Fan
- Eye Center, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and EngineeringZhejiang UniversityHangzhouPeople's Republic of China
| | - Haijie Han
- Eye Center, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
| | - Zhouyu Lu
- Eye Center, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and EngineeringZhejiang UniversityHangzhouPeople's Republic of China
| | - Yin Zhang
- Eye Center, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
| | - Yaoyao Chen
- Eye Center, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
| | - Xiaobo Zhang
- Eye Center, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and EngineeringZhejiang UniversityHangzhouPeople's Republic of China
| | - Ke Yao
- Eye Center, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouPeople's Republic of China
| |
Collapse
|
36
|
Nagamalla S, Mague JT, Sathyamoorthi S. Progress towards the syntheses of Bactobolin A and C4- epi-Bactobolin A using a sulfamate-tethered aza-Wacker cyclization strategy. Tetrahedron 2022; 128:133112. [PMID: 37719878 PMCID: PMC10503945 DOI: 10.1016/j.tet.2022.133112] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We present a progress report towards Bactobolin A and C4-epi-Bactobolin A. Sulfamate-tethered aza-Wacker cyclization followed by a Tsuji-Wacker ketone synthesis furnishes a key tricyclic intermediate which we hypothesize can be elaborated into C4-epi-Bactobolin A. Epimerization of one of the stereocenters of this compound furnishes an intermediate which we hypothesize can be elaborated into Bactobolin A.
Collapse
Affiliation(s)
- Someshwar Nagamalla
- University of Kansas, Department of Medicinal Chemistry, Lawrence, KS, 66047, USA
| | - Joel T. Mague
- Tulane University, Department of Chemistry, New Orleans, LA, 70118, USA
| | - Shyam Sathyamoorthi
- University of Kansas, Department of Medicinal Chemistry, Lawrence, KS, 66047, USA
| |
Collapse
|
37
|
Cuervo L, Méndez C, Salas JA, Olano C, Malmierca MG. Volatile Compounds in Actinomycete Communities: A New Tool for Biosynthetic Gene Cluster Activation, Cooperative Growth Promotion, and Drug Discovery. Cells 2022; 11:3510. [PMID: 36359906 PMCID: PMC9655753 DOI: 10.3390/cells11213510] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 07/30/2023] Open
Abstract
The increasing appearance of multiresistant pathogens, as well as emerging diseases, has highlighted the need for new strategies to discover natural compounds that can be used as therapeutic alternatives, especially in the genus Streptomyces, which is one of the largest producers of bioactive metabolites. In recent years, the study of volatile compounds (VOCs) has raised interest because of the variety of their biological properties in addition to their involvement in cell communication. In this work, we analyze the implications of VOCs as mediating molecules capable of inducing the activation of biosynthetic pathways of bioactive compounds in surrounding Actinomycetes. For this purpose, several strains of Streptomyces were co-cultured in chamber devices that allowed VOC exchange while avoiding physical contact. In several of those strains, secondary metabolism was activated by VOCs emitted by companion strains, resulting in increased antibiotic production and synthesis of new VOCs. This study shows a novel strategy to exploit the metabolic potential of Actinomycetes as well as emphasizes the importance of studying the interactions between different microorganisms sharing the same ecological niche.
Collapse
Affiliation(s)
- Lorena Cuervo
- Functional Biology Department, University of Oviedo, 33006 Oviedo, Spain
- University Institute of Oncology of Asturias (I.U.O.P.A), University of Oviedo, 33006 Oviedo, Spain
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - Carmen Méndez
- Functional Biology Department, University of Oviedo, 33006 Oviedo, Spain
- University Institute of Oncology of Asturias (I.U.O.P.A), University of Oviedo, 33006 Oviedo, Spain
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - José A. Salas
- Functional Biology Department, University of Oviedo, 33006 Oviedo, Spain
- University Institute of Oncology of Asturias (I.U.O.P.A), University of Oviedo, 33006 Oviedo, Spain
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - Carlos Olano
- Functional Biology Department, University of Oviedo, 33006 Oviedo, Spain
- University Institute of Oncology of Asturias (I.U.O.P.A), University of Oviedo, 33006 Oviedo, Spain
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - Mónica G. Malmierca
- Functional Biology Department, University of Oviedo, 33006 Oviedo, Spain
- University Institute of Oncology of Asturias (I.U.O.P.A), University of Oviedo, 33006 Oviedo, Spain
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
38
|
Mota J, Bravo C, Santos C, Alves PC, Rijo P, Antunes AM, Grenho L, Helena Fernandes M, Alves MM, André V. Eco-friendly fabricated multibioactive Ca(II)-antibiotic coordination framework coating on zinc towards improved bone tissue regeneration. Colloids Surf B Biointerfaces 2022; 221:113008. [DOI: 10.1016/j.colsurfb.2022.113008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/24/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
|
39
|
Wang Y, Xu F, Nichols CB, Shi Y, Hellinga HW, Alspaugh JA, Distefano MD, Beese LS. Structure-Guided Discovery of Potent Antifungals that Prevent Ras Signaling by Inhibiting Protein Farnesyltransferase. J Med Chem 2022; 65:13753-13770. [PMID: 36218371 PMCID: PMC10755971 DOI: 10.1021/acs.jmedchem.2c00902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Infections by fungal pathogens are difficult to treat due to a paucity of antifungals and emerging resistances. Next-generation antifungals therefore are needed urgently. We have developed compounds that prevent farnesylation of Cryptoccoccus neoformans Ras protein by inhibiting protein farnesyltransferase with 3-4 nanomolar affinities. Farnesylation directs Ras to the cell membrane and is required for infectivity of this lethal pathogenic fungus. Our high-affinity compounds inhibit fungal growth with 3-6 micromolar minimum inhibitory concentrations (MICs), 4- to 8-fold better than Fluconazole, an antifungal commonly used in the clinic. Compounds bound with distinct inhibition mechanisms at two alternative, partially overlapping binding sites, accessed via different inhibitor conformations. We showed that antifungal potency depends critically on the selected inhibition mechanism because this determines the efficacy of an inhibitor at low in vivo levels of enzyme and farnesyl substrate. We elucidated how chemical modifications of the antifungals encode desired inhibitor conformation and concomitant inhibitory mechanism.
Collapse
Affiliation(s)
- You Wang
- Department of Biochemistry, Duke University School
of Medicine, Durham, North Carolina, USA 27710
| | - Feng Xu
- Department of Chemistry, University of Minnesota,
Minneapolis, Minnesota, USA 55455
| | - Connie B. Nichols
- Department of Medicine, Duke University School of
Medicine, Durham, North Carolina, USA 27710
- Department of Molecular Genetics and Microbiology,
Duke University School of Medicine, Durham, North Carolina, USA 27710
| | - Yuqian Shi
- Department of Biochemistry, Duke University School
of Medicine, Durham, North Carolina, USA 27710
| | - Homme W. Hellinga
- Department of Biochemistry, Duke University School
of Medicine, Durham, North Carolina, USA 27710
| | - J. Andrew Alspaugh
- Department of Medicine, Duke University School of
Medicine, Durham, North Carolina, USA 27710
- Department of Molecular Genetics and Microbiology,
Duke University School of Medicine, Durham, North Carolina, USA 27710
| | - Mark D. Distefano
- Department of Chemistry, University of Minnesota,
Minneapolis, Minnesota, USA 55455
| | - Lorena S. Beese
- Department of Biochemistry, Duke University School
of Medicine, Durham, North Carolina, USA 27710
| |
Collapse
|
40
|
Rawat V, Tiwari S, Khanna S, Gupta U, S N C S, Yadav DK, Kaul G, Akhir A, Saxena D, Matheshwaran S, Chopra S, Allimuthu D. Nitroisobenzofuranone, a small molecule inhibitor of multidrug-resistant Staphylococcus aureus, targets peptidoglycan biosynthesis. Chem Commun (Camb) 2022; 58:11669-11672. [PMID: 36172810 DOI: 10.1039/d2cc04102b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Antimicrobial resistance (AMR) is a global health concern. Targetting AMR, we present an in situ lactonization mechanism generating 4-nitroisobenzofuran-1(3H)-one (IITK2020), an exclusive S. aureus inhibitor at 2-4 μg mL-1 MIC including multidrug-resistant S. aureus clinical strains, that prevents peptidoglycan biosynthesis.
Collapse
Affiliation(s)
- Viral Rawat
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India.
| | - Sona Tiwari
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India.
| | - Shweta Khanna
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India.
| | - Umang Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Sridhar S N C
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India.
| | - Dharmendra K Yadav
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India.
| | - Grace Kaul
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India.,AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Abdul Akhir
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Deepanshi Saxena
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Saravanan Matheshwaran
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Sidharth Chopra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India.,AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Dharmaraja Allimuthu
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India.
| |
Collapse
|
41
|
Liu J, Li H, He Q, Chen K, Chen Y, Zhong R, Li H, Fang S, Liu S, Lin S. Design, synthesis, and biological evaluation of tetrahydroquinoline amphiphiles as membrane-targeting antimicrobials against pathogenic bacteria and fungi. Eur J Med Chem 2022; 243:114734. [PMID: 36088756 DOI: 10.1016/j.ejmech.2022.114734] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/15/2022]
Abstract
The rising prevalence of drug-resistant pathogens is one of the biggest threats to human health. The development of new antibiotics that can overcome drug resistance is in urgent need. Herein, we designed and synthesized a series of amphiphilic tetrahydroquinoline derivatives as small-molecule-based antimicrobial peptidomimetics. Two lead compounds 36 and 52 which contained the tetrahydroquinoline core, hydrophobic alkyl chains (n-nonyl or isoprenyl group), different spacer lengths (n = 4 or 8), and cationic guanidine moiety, showed poor hemolytic activity, low cytotoxicity, and potent broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria, as well as fungi. The further biological evaluation revealed that compounds 36 and 52 can kill bacteria and fungi rapidly via membrane-targeting action and avoid drug resistance development. More importantly, compounds 36 and 52 exhibited similarly potent in vivo antimicrobial activities in a murine corneal infection caused by Staphylococcus aureus ATCC29213 or Pseudomonas aeruginosa ATCC9027, as compared to vancomycin or gatifloxacin. These results suggest that compounds 36 and 52 have great potential as new broad-spectrum antimicrobial agents to combat microbial resistance.
Collapse
Affiliation(s)
- Jiayong Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hongxia Li
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Qile He
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kaiting Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yongzhi Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Rongcui Zhong
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haizhou Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shanfang Fang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shouping Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Shuimu Lin
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
42
|
Wang G, Hao C, Chen C, Kuang H, Xu C, Xu L. Six-Pointed Star Chiral Cobalt Superstructures with Strong Antibacterial Activity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2204219. [PMID: 36038354 DOI: 10.1002/smll.202204219] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/08/2022] [Indexed: 06/15/2023]
Abstract
Chiral inorganic nanomaterials have shown promise as a potential means of combating bacteria due to their high levels of biocompatibility, easy surface modification, and excellent optical properties. In this study, a diverse range of chiral hierarchical nanomaterials are prepared from Co2+ and L/D-Tartaric acid (Tar) ligands. By combining the ligands in different ratios, chiral Co superstructures (Co SS) are obtained with different morphologies, including chiral nanoflowers, chiral nanohanamaki, a chiral six-pointed star, a chiral fan shape, and a chiral fusiform shape. It is found that the chiral six-pointed star structures exhibit chiroptical activity across a broad range of wavelengths from 300 to 1300 nm and that the g-factor is as high as 0.033 with superparamagnetic properties. Under the action of electromagnetic fields, the chiral six-pointed star Co SS shows excellent killing ability against Gram-positive Staphylococcus aureus (ATCC 25923). Compared to L-Co SS, D-Co SS shows stronger levels of antibacterial ability. It is found that the levels of reactive oxygen species generated by D-Co SS are 1.59-fold higher than L-Co SS which is attributed to chiral-induced spin selectivity effects. These findings are of significance for the further development of chiral materials with antibacterial properties.
Collapse
Affiliation(s)
- Gaoyang Wang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Changlong Hao
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Chen Chen
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
| |
Collapse
|
43
|
Zhuang Z, Meng Z, Li J, Shen P, Dai J, Lou X, Xia F, Tang BZ, Zhao Z. Antibacterial Theranostic Agents with Negligible Living Cell Invasiveness: AIE-Active Cationic Amphiphiles Regulated by Alkyl Chain Engineering. ACS NANO 2022; 16:11912-11930. [PMID: 35917549 DOI: 10.1021/acsnano.2c01721] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
To address the threat of bacterial infection in the following post-antibiotic era, developing effective antibacterial approaches is of utmost urgency. Theranostic medicine integrating diagnosis and therapy is a promising protocol to fight against pathogenic bacteria. But numerous reported antibacterial theranostic materials are disclosed to be trapped in the excessive invasiveness to living mammal cells, leading to false positives and possible biosafety risks. Herein, a series of cationic pyridinium-substituted phosphindole oxide derivatives featuring aggregation-induced emission are designed, and alkyl chain engineering is conducted to finely tune their hydrophobicity and investigate their bioaffinity preference for living mammal cells and pathogenic bacteria. Most importantly, an efficient theranostic agent (PyBu-PIO) is acquired that is free from living cell invasiveness with negligible cytotoxicity and yet holds a good affinity for Gram-positive bacteria, including drug-resistant strains, with a superior inactivating effect. Externally applying PyBu-PIO onto Gram-positive bacteria-infected skin wounds can achieve creditable imaging effects and successfully accelerate the healing processes with reliable biosafety. This work proposes living cell invasiveness as a criterion for antibacterial theranostic materials and provides important enlightenment for the design of antibacterial theranostic materials.
Collapse
Affiliation(s)
- Zeyan Zhuang
- State Key Laboratory of Luminescent Materials and Devices, Key Laboratory of Luminescence from Molecular Aggregates of Guangdong Province, South China University of Technology, Guangzhou 510640, China
| | - Zijuan Meng
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Jianqing Li
- State Key Laboratory of Luminescent Materials and Devices, Key Laboratory of Luminescence from Molecular Aggregates of Guangdong Province, South China University of Technology, Guangzhou 510640, China
| | - Pingchuan Shen
- State Key Laboratory of Luminescent Materials and Devices, Key Laboratory of Luminescence from Molecular Aggregates of Guangdong Province, South China University of Technology, Guangzhou 510640, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaoding Lou
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Fan Xia
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Zujin Zhao
- State Key Laboratory of Luminescent Materials and Devices, Key Laboratory of Luminescence from Molecular Aggregates of Guangdong Province, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
44
|
Ma Y, Wei M, Wang X, Jiang L, Xiong Y, Cheng J, Tan Y, Liao X, Wang J. Synthesis and antibacterial against
S. aureus
of new ruthenium (II) polypyridine complexes containing pyrene groups. Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yuanyuan Ma
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Ming Wei
- Kangda College of Nanjing Medical University Lianyungang Jiangsu China
| | - Xuerong Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Li Jiang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Yanshi Xiong
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Jianxin Cheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Yanhui Tan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences Guangxi Normal University Guilin People’s Republic of China
| | - Xiangwen Liao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Jintao Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| |
Collapse
|
45
|
Aloe emodin-conjugated sulfonyl hydrazones as novel type of antibacterial modulators against S. aureus 25923 through multifaceted synergistic effects. Bioorg Chem 2022; 127:106035. [PMID: 35870413 DOI: 10.1016/j.bioorg.2022.106035] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 12/24/2022]
Abstract
Aloe emodin-conjugated sulfonyl hydrazones were designed and synthesized as novel type of antibacterial modulators. Aloe emodin benzenesulfonyl hydrazone 5a (AEBH-5a) was preponderant for the treatment of S. aureus 25923 (MIC = 0.5 μg/mL) over norfloxacin and presented high selectivity between bacterial membranes and mammalian membranes. Especially, AEBH-5a could eliminate the formed biofilms and relieve the development of S. aureus 25923 resistance. The antibacterial mechanism of AEBH-5a from extracellularity to intracellularity illustrated that AEBH-5a could destroy bacterial membrane integrity, leading to the leakage of protein and nucleic acid. Besides, AEBH-5a could not only interact with DNA and induce oxidative stress but also inhibit lactate dehydrogenase (LDH) activity as well as render metabolic inactivation. In silico ADME studies prediction of AEBH-5a revealed a favorable bioavailability score and prominent drug-likeness profile. This research showed that the multifaceted synergistic effect initiated by aloe emodin-conjugated sulfonyl hydrazones is a reasonable and effective tactic to combat menacing bacterial infections.
Collapse
|
46
|
Huang Y, Geng H, Wu Z, Sun L, Ji C, Grimes CA, Feng X, Cai Q. An Ag 2S@ZIF-Van nanosystem for NIR-II imaging of bacterial-induced inflammation and treatment of wound bacterial infection. Biomater Sci 2022; 10:3972-3980. [PMID: 35708482 DOI: 10.1039/d2bm00550f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bacterial diseases pose a serious threat to human health. Continued development of precise diagnostic methods and synergistic therapy techniques for combating bacteria are needed. Herein a hybrid nanosystem (Ag2S@ZIF-Van NS) was constructed by one-step self-assembly of Zn2+, vancomycin (Van) and Ag2S quantum dots (QDs). The nanosystem possesses excellent second near-infrared transparency window (NIR-II) fluorescence properties (∼1200 nm emission wavelength), good photothermal conversion properties, and biocompatibility. The material system enables precise, targeted NIR-II fluorescent imaging of bacterial inflammation in vivo as well as promoting anti-bacterial and wound healing effects.
Collapse
Affiliation(s)
- Yao Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, Hunan, China.
| | - Hongchao Geng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, Hunan, China.
| | - Zeming Wu
- Inner Mongolia Environmental Monitoring Center, Hohhot 010011, Inner Mongolia, China
| | - Leilei Sun
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, Hunan, China.
| | - Chenhui Ji
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, Hunan, China.
| | | | - Xinxin Feng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, Hunan, China.
| | - Qingyun Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, Hunan, China.
| |
Collapse
|
47
|
Isolation and Identification of Pathogenic Bacteria Causing Otitis Media in Misan Governorate. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2022. [DOI: 10.22207/jpam.16.2.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This study examined 150 ear swab samples from patients with otitis media who consulted at Al-Sadr Teaching Hospital from January to April 2021 in Misan, Iraq. The participants were aged 14–50 years, among which the infection rate was highest in participants aged 14–22 years and lowest in those aged 40–50 years. Subsequently, bacterial isolates were identified based on their morphology in various culture media and using biochemical tests. Six bacterial species were identified, namely Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli, Streptococcus pneumoniae, Klebsiella pneumoniae, and Staphylococcus epidermidis. Staphylococcus aureus had the highest infection rate (30%), whereas Staphylococcus epidermidis had the lowest infection rate (8.55%). When the sensitivity of each isolate to antibiotics was determined, Escherichia coli was the most sensitive to trimethoprim (TMP), whereas Pseudomonas aeruginosa was the most resistant to 75% of the tested antibiotics.
Collapse
|
48
|
Wang H, Niu M, Xue T, Ma L, Gu X, Wei G, li F, wang C. Development of Antibacterial Peptides with Efficient Antibacterial Activity, Low Toxicity, High Membrane Disruptive Activity and the Synergistic Antibacterial Effect. J Mater Chem B 2022; 10:1858-1874. [DOI: 10.1039/d1tb02852a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
It is urgent to develop new antimicrobial drugs to overcome bacterial resistance which is a serious threat to human health. Antimicrobial peptides (AMPs) which are ideal substitutes for traditional antibiotics...
Collapse
|