1
|
Dong HM, Chen JX, Cai YX, Tian LX, Yang ZC. Compounds Derived from 5-Fluoropyridine and Benzo[b]thiophene: Killing Mycobacterium tuberculosis and Reducing its Virulence. Chem Biodivers 2024; 21:e202401191. [PMID: 39058423 DOI: 10.1002/cbdv.202401191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 07/28/2024]
Abstract
The rise of drug-resistant Mycobacterium tuberculosis (Mtb) has extended the duration of tuberculosis (TB) treatment and reduced the likelihood of cure. One strategy to combat this issue is the development of inhibitors targeting the virulence factors of bacterial pathogens. Mtb' catalase (KatG) is crucial for its detoxification mechanisms and also serves as a significant virulence factor for the bacterium. In this study, twelve derivatives synthesized from 5-fluoropyridine and benzo[b]thiophene demonstrated antimycobacterial efficacy with minimum inhibitory concentrations (MICs) varying between 0.5 and 32 μg/mL. Compound 2, 1-(benzo[b]thiophen-2-ylmethylene) thiosemicarbazide, emerged as the most potent candidate. It effectively inhibited Mtb KatG, enhanced the production of reactive oxygen species (ROS) in Mtb, and achieved Mtb killing within 96 hours at a concentration of 2 μg/mL (4×MIC). Molecular docking simulations revealed that compound 2 binds tightly to the active site of Mtb-KatG with a docking score of 114, indicating that it may serve as a potent inhibitor of Mtb-KatG. The rabbit skin tuberculosis model was employed to assess the virulence of Mtb. Animal study results indicated that the granulomas induced by Mtb after treatment with compound 2 were reduced in size, exhibited a lower bacterial load, and the bacteria were no longer aggregated, in contrast to those caused by untreated Mtb. Hence, compound 2 can be regarded as a molecule capable of neutralizing the virulence factors of Mtb. This research offers insights into the design of anti-Mtb molecules with novel mechanisms of action.
Collapse
Affiliation(s)
- Hong-Mei Dong
- College of Pharmacy, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
| | - Jun-Xian Chen
- College of Pharmacy, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
| | - Yu-Xiang Cai
- College of Pharmacy, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
| | - Li-Xia Tian
- College of Pharmacy, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
| | - Zai-Chang Yang
- College of Pharmacy, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Science Road, Guiyang, 550014, China
| |
Collapse
|
2
|
Butler MS, Vollmer W, Goodall ECA, Capon RJ, Henderson IR, Blaskovich MAT. A Review of Antibacterial Candidates with New Modes of Action. ACS Infect Dis 2024; 10:3440-3474. [PMID: 39018341 PMCID: PMC11474978 DOI: 10.1021/acsinfecdis.4c00218] [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: 03/17/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 07/19/2024]
Abstract
There is a lack of new antibiotics to combat drug-resistant bacterial infections that increasingly threaten global health. The current pipeline of clinical-stage antimicrobials is primarily populated by "new and improved" versions of existing antibiotic classes, supplemented by several novel chemical scaffolds that act on traditional targets. The lack of fresh chemotypes acting on previously unexploited targets (the "holy grail" for new antimicrobials due to their scarcity) is particularly unfortunate as these offer the greatest opportunity for innovative breakthroughs to overcome existing resistance. In recognition of their potential, this review focuses on this subset of high value antibiotics, providing chemical structures where available. This review focuses on candidates that have progressed to clinical trials, as well as selected examples of promising pioneering approaches in advanced stages of development, in order to stimulate additional research aimed at combating drug-resistant infections.
Collapse
Affiliation(s)
- Mark S. Butler
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Waldemar Vollmer
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Emily C. A. Goodall
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Robert J. Capon
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Ian R. Henderson
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Mark A. T. Blaskovich
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
3
|
Leslie K, Berry SS, Miller GJ, Mahon CS. Sugar-Coated: Can Multivalent Glycoconjugates Improve upon Nature's Design? J Am Chem Soc 2024; 146:27215-27232. [PMID: 39340450 PMCID: PMC11467903 DOI: 10.1021/jacs.4c08818] [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: 06/30/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Multivalent interactions between receptors and glycans play an important role in many different biological processes, including pathogen infection, self-recognition, and the immune response. The growth in the number of tools and techniques toward the assembly of multivalent glycoconjugates means it is possible to create synthetic systems that more and more closely resemble the diversity and complexity we observe in nature. In this Perspective we present the background to the recognition and binding enabled by multivalent interactions in nature, and discuss the strategies used to construct synthetic glycoconjugate equivalents. We highlight key discoveries and the current state of the art in their applications to glycan arrays, vaccines, and other therapeutic and diagnostic tools, with an outlook toward some areas we believe are of most interest for future work in this area.
Collapse
Affiliation(s)
- Kathryn
G. Leslie
- Department
of Chemistry, Durham University, Durham DH1 3LE, United Kingdom
| | - Sian S. Berry
- Centre
for Glycoscience and School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
| | - Gavin J. Miller
- Centre
for Glycoscience and School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
| | - Clare S. Mahon
- Department
of Chemistry, Durham University, Durham DH1 3LE, United Kingdom
| |
Collapse
|
4
|
Borho J, Kögel M, Eckert A, Barth H. Repurposing FDA-approved disulfiram for targeted inhibition of diphtheria toxin and the binary protein toxins of Clostridium botulinum and Bacillus anthracis. Front Pharmacol 2024; 15:1455696. [PMID: 39346565 PMCID: PMC11427369 DOI: 10.3389/fphar.2024.1455696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024] Open
Abstract
Many bacteria act pathogenic by the release of AB-type protein toxins that efficiently enter human or animal cells and act as enzymes in their cytosol. This leads to disturbed cell functions and the clinical symptoms characteristic for the individual toxin. Therefore, molecules that directly target and neutralize these toxins provide promising novel therapeutic options. Here, we found that the FDA-approved drug disulfiram (DSF), used for decades to treat alcohol abuse, protects cells from intoxication with diphtheria toxin (DT) from Corynebacterium diphtheria, the causative agent of diphtheria, lethal toxin (LT) from Bacillus anthracis, which contributes to anthrax, and C2 enterotoxin from Clostridium botulinum when applied in concentrations lower than those found in plasma of patients receiving standard DSF treatment for alcoholism (up to 20 µM). Moreover, this inhibitory effect is increased by copper, a known enhancer of DSF activity. LT and C2 are binary toxins, consisting of two non-linked proteins, an enzyme (A) and a separate binding/transport (B) subunit. To act cytotoxic, their proteolytically activated B subunits PA63 and C2IIa, respectively, form barrel-shaped heptamers that bind to their cellular receptors and form complexes with their respective A subunits LF and C2I. The toxin complexes are internalized via receptor-mediated endocytosis and in acidified endosomes, PA63 and C2IIa form pores in endosomal membranes, which facilitate translocation of LF and C2I into the cytosol, where they act cytotoxic. In DT, A and B subunits are located within one protein, but DT also forms pores in endosomes that facilitate translocation of the A subunit. If cell binding, membrane translocation, or substrate modification is inhibited, cells are protected from intoxication. Our results implicate that DSF neither affects cellular binding nor the catalytic activity of the investigated toxins to a relevant extend, but interferes with the toxin pore-mediated translocation of the A subunits of DT, LT and C2 toxin, as demonstrated by membrane-translocation assays and toxin pore conductivity experiments in the presence or absence of DSF. Since toxin translocation across intracellular membranes represents a central step during cellular uptake of many bacterial toxins, DSF might neutralize a broad spectrum of medically relevant toxins.
Collapse
Affiliation(s)
| | | | | | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
5
|
Manoharan D, Wang LC, Chen YC, Li WP, Yeh CS. Catalytic Nanoparticles in Biomedical Applications: Exploiting Advanced Nanozymes for Therapeutics and Diagnostics. Adv Healthc Mater 2024; 13:e2400746. [PMID: 38683107 DOI: 10.1002/adhm.202400746] [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: 02/26/2024] [Revised: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Catalytic nanoparticles (CNPs) as heterogeneous catalyst reveals superior activity due to their physio-chemical features, such as high surface-to-volume ratio and unique optical, electric, and magnetic properties. The CNPs, based on their physio-chemical nature, can either increase the reactive oxygen species (ROS) level for tumor and antibacterial therapy or eliminate the ROS for cytoprotection, anti-inflammation, and anti-aging. In addition, the catalytic activity of nanozymes can specifically trigger a specific reaction accompanied by the optical feature change, presenting the feasibility of biosensor and bioimaging applications. Undoubtedly, CNPs play a pivotal role in pushing the evolution of technologies in medical and clinical fields, and advanced strategies and nanomaterials rely on the input of chemical experts to develop. Herein, a systematic and comprehensive review of the challenges and recent development of CNPs for biomedical applications is presented from the viewpoint of advanced nanomaterial with unique catalytic activity and additional functions. Furthermore, the biosafety issue of applying biodegradable and non-biodegradable nanozymes and future perspectives are critically discussed to guide a promising direction in developing span-new nanozymes and more intelligent strategies for overcoming the current clinical limitations.
Collapse
Affiliation(s)
- Divinah Manoharan
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Liu-Chun Wang
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Ying-Chi Chen
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wei-Peng Li
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chen-Sheng Yeh
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
6
|
Sun S, Chen X. Mechanism-guided strategies for combating antibiotic resistance. World J Microbiol Biotechnol 2024; 40:295. [PMID: 39122871 DOI: 10.1007/s11274-024-04106-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Bacterial antibiotic resistance has been recognized as a global threat to public health. It challenges the antibiotics currently used in clinical practice and causes severe and often fatal infectious diseases. Fighting against antibiotic-resistant bacteria (ARB) is growing more urgent. While understanding the molecular mechanisms that underlie resistance is a prerequisite, several major mechanisms have been previously proposed including bacterial efflux systems, reduced cell membrane permeability, antibiotic inactivation by enzymes, target modification, and target protection. In this context, this review presents a panel of promising and potential strategies to combat antibiotic resistance/resistant bacteria. Different types of direct-acting and indirect resistance breakers, such as efflux pump inhibitors, antibiotic adjuvants, and oxidative treatments are discussed. In addition, the emerging multi-omics approaches for rapid resistance identification and promising alternatives to existing antibiotics are highlighted. Overall, this review suggests that continued effort and investment in research are required to develop new antibiotics and alternatives to existing antibiotics and translate them into environmental and clinical applications.
Collapse
Affiliation(s)
- Shengwei Sun
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China.
| | - Xueyingzi Chen
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| |
Collapse
|
7
|
Vargas-Lizarazo AY, Ali MA, Mazumder NA, Kohli GM, Zaborska M, Sons T, Garnett M, Senanayake IM, Goodson BM, Vargas-Muñiz JM, Pond A, Jensik PJ, Olson ME, Hamilton-Brehm SD, Kohli P. Electrically polarized nanoscale surfaces generate reactive oxygenated and chlorinated species for deactivation of microorganisms. SCIENCE ADVANCES 2024; 10:eado5555. [PMID: 39093965 PMCID: PMC11636998 DOI: 10.1126/sciadv.ado5555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024]
Abstract
Because of the decreasing supply of new antibiotics, recent outbreaks of infectious diseases, and the emergence of antibiotic-resistant microorganisms, it is imperative to develop new effective strategies for deactivating a broad spectrum of microorganisms and viruses. We have implemented electrically polarized nanoscale metallic (ENM) coatings that deactivate a wide range of microorganisms including Gram-negative and Gram-positive bacteria with greater than 6-log reduction in less than 10 minutes of treatment. The electrically polarized devices were also effective in deactivating lentivirus and Candida albicans. The key to the high deactivation effectiveness of ENM devices is electrochemical production of micromolar cuprous ions, which mediated reduction of oxygen to hydrogen peroxide. Formation of highly damaging species, hydroxyl radicals and hypochlorous acid, from hydrogen peroxide contributed to antimicrobial properties of the ENM devices. The electric polarization of nanoscale coatings represents an unconventional tool for deactivating a broad spectrum of microorganisms through in situ production of reactive oxygenated and chlorinated species.
Collapse
Affiliation(s)
- Annie Y. Vargas-Lizarazo
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - M. Aswad Ali
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - Nehal A. Mazumder
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | | | - Miroslava Zaborska
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - Tyler Sons
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA
| | - Michelle Garnett
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA
| | - Ishani M. Senanayake
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - Boyd M. Goodson
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - José M. Vargas-Muñiz
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA
| | - Amber Pond
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA
| | - Philip J. Jensik
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA
| | - Michael E. Olson
- Department of Medical Microbiology, Immunology and Cell Biology, School of Medicine, Southern Illinois University, Springfield, IL 62702, USA
| | | | - Punit Kohli
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
- Integrated Microscopy and Graphics Expertise (IMAGE) Center, Southern Illinois University, Carbondale, IL 62901, USA
| |
Collapse
|
8
|
Stefanetti V, Passamonti F, Rampacci E. Antimicrobial Strategies Proposed for the Treatment of S. pseudintermedius and Other Dermato-Pathogenic Staphylococcus spp. in Companion Animals: A Narrative Review. Vet Sci 2024; 11:311. [PMID: 39057995 PMCID: PMC11281426 DOI: 10.3390/vetsci11070311] [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: 05/31/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The treatment of dermato-pathogenic Staphylococcus spp., particularly Staphylococcus pseudintermedius, in companion animals presents significant challenges due to rising antimicrobial resistance. This review explores innovative strategies to combat these infections. We examined novel antimicrobials and the repurposing of existing drugs to enhance their efficacy against resistant strains. Additionally, we evaluate the potential of natural products, nanomaterials, and skin antiseptics as alternative treatments. The review also investigates the use of antimicrobial peptides and bacteriophages, highlighting their targeted action against staphylococcal pathogens. Furthermore, the role of adjuvants in antibiotic treatments, such as antimicrobial resistance breakers, is discussed, emphasizing their ability to enhance therapeutic outcomes. Our analysis underscores the importance of a multifaceted approach in developing effective antimicrobial strategies for companion animals, aiming to mitigate resistance and improve clinical management of staphylococcal skin infections.
Collapse
Affiliation(s)
- Valentina Stefanetti
- Department of Human Science and Promotion of Quality Life, San Raffaele Telematic University, 00166 Rome, Italy;
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy;
| | - Fabrizio Passamonti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy;
| | - Elisa Rampacci
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy;
| |
Collapse
|
9
|
de Souza CM, Silvério de Oliveira W, Fleitas Martínez O, Dos Santos Neto NA, Buccini DF, Nieto Marín V, de Faria Júnior C, Rocha Maximiano M, Soller Ramada MH, Franco OL. Evaluating virulence features of Acinetobacter baumannii resistant to polymyxin B. Lett Appl Microbiol 2024; 77:ovae061. [PMID: 38942450 DOI: 10.1093/lambio/ovae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/17/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024]
Abstract
The increasing resistance to polymyxins in Acinetobacter baumannii has made it even more urgent to develop new treatments. Anti-virulence compounds have been researched as a new solution. Here, we evaluated the modification of virulence features of A. baumannii after acquiring resistance to polymyxin B. The results showed lineages attaining unstable resistance to polymyxin B, except for Ab7 (A. baumannii polymyxin B resistant lineage), which showed stable resistance without an associated fitness cost. Analysis of virulence by a murine sepsis model indicated diminished virulence in Ab7 (A. baumannii polymyxin B resistant lineage) compared with Ab0 (A. baumannii polymyxin B susceptible lineage). Similarly, downregulation of virulence genes was observed by qPCR at 1 and 3 h of growth. However, an increase in bauE, abaI, and pgAB expression was observed after 6 h of growth. Comparison analysis of Ab0, Ab7, and Pseudomonas aeruginosa suggested no biofilm formation by Ab7. In general, although a decrease in virulence was observed in Ab7 when compared with Ab0, some virulence feature that enables infection could be maintained. In light of this, virulence genes bauE, abaI, and pgAB showed a potential relevance in the maintenance of virulence in polymyxin B-resistant strains, making them promising anti-virulence targets.
Collapse
Affiliation(s)
- Camila Maurmann de Souza
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-160, Brazil
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande CEP 79.117-900, Brazil
| | - Warley Silvério de Oliveira
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-160, Brazil
| | - Osmel Fleitas Martínez
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-160, Brazil
| | | | - Danieli Fernanda Buccini
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande CEP 79.117-900, Brazil
| | - Valentina Nieto Marín
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande CEP 79.117-900, Brazil
| | - Célio de Faria Júnior
- Microbiology Department, Laboratório Central de Saúde Pública LACEN, Brasília 70830-010, Brazil
| | - Mariana Rocha Maximiano
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-160, Brazil
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande CEP 79.117-900, Brazil
| | - Marcelo Henrique Soller Ramada
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-160, Brazil
| | - Octávio Luiz Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília 70790-160, Brazil
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande CEP 79.117-900, Brazil
| |
Collapse
|
10
|
Zhang J, Gao L, Lin H, Liang Y, You M, Ding L, Feng F, Yang B, Liu Y. Discovery of Antibacterial Compounds against Xanthomonas citri subsp. citri from a Marine Fungus Aspergillus terreus SCSIO 41202 and the Mode of Action. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12596-12606. [PMID: 38771666 DOI: 10.1021/acs.jafc.4c02769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Citrus canker, caused by Xanthomonas citri subsp. citri (Xcc), is a severe citrus disease. Currently, copper-containing pesticides are widely used to manage this disease, posing high risks to the environment and human health. This study reports the discovery of naturally occurring anti-Xcc compounds from a deep-sea fungus, Aspergillus terreus SCSIO 41202, and the possible mode of action. The ethyl acetate extract of A. terreus was subjected to bioassay-guided isolation, resulting in the discovery of eight anti-Xcc compounds (1-8) with minimum inhibitory concentrations (MICs) ranging from 0.078 to 0.625 mg/mL. The chemical structures of these eight metabolites were determined by integrative analysis of various spectroscopic data. Among these compounds, Asperporonin A (1) and Asperporonin B (2) were identified as novel compounds with a very unusual structural skeleton. The electronic circular dichroism was used to determine the absolute configurations of 1 and 2 through quantum chemical calculation. A bioconversion pathway involving pinacol rearrangement was proposed to produce the unusual compounds (1-2). Compound 6 exhibited an excellent anti-Xcc effect with a MIC value of 0.078 mg/mL, which was significantly more potent than the positive control CuSO4 (MIC = 0.3125 mg/mL). Compound 6 inhibited cell growth by disrupting biofilm formation, destroying the cell membrane, and inducing the accumulation of reactive oxygen species. In vivo tests indicated that compound 6 is highly effective in controlling citrus canker disease. These results indicate that compounds 1-8, especially 6, have the potential as lead compounds for the development of new, environmentally friendly, and efficient anti-Xcc pesticides.
Collapse
Affiliation(s)
- Jun Zhang
- National Engineering Research Center of Navel Orange, Gannan Normal University, Ganzhou 341000, China
| | - Liangliang Gao
- National Engineering Research Center of Navel Orange, Gannan Normal University, Ganzhou 341000, China
| | - Huiting Lin
- National Engineering Research Center of Navel Orange, Gannan Normal University, Ganzhou 341000, China
| | - Yan Liang
- National Engineering Research Center of Navel Orange, Gannan Normal University, Ganzhou 341000, China
| | - Mingnan You
- National Engineering Research Center of Navel Orange, Gannan Normal University, Ganzhou 341000, China
| | - Lijian Ding
- Department of Marine Pharmacy, Ningbo University, Ningbo 315211, China
| | - Fangjian Feng
- Department of Marine Pharmacy, Ningbo University, Ningbo 315211, China
| | - Bin Yang
- Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510000, P. R. China
| | - Yonghong Liu
- Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510000, P. R. China
| |
Collapse
|
11
|
Zhao J, Wang Z, Yang M, Guo J, Gao Z, Song P, Song YY. Pore-Forming Toxin-Driven Recovery of Peroxidase-Mimicking Activity in Biomass Channels for Label-Free Electrochemical Bacteria Sensing. Anal Chem 2024; 96:7661-7668. [PMID: 38687969 DOI: 10.1021/acs.analchem.4c00589] [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: 05/02/2024]
Abstract
The development of sensitive, selective, and rapid methods to detect bacteria in complex media is essential to ensuring human health. Virulence factors, particularly pore-forming toxins (PFTs) secreted by pathogenic bacteria, play a crucial role in bacterial diseases and serve as indicators of disease severity. In this study, a nanochannel-based label-free electrochemical sensing platform was developed for the detection of specific pathogenic bacteria based on their secreted PFTs. In this design, wood substrate channels were functionalized with a Fe-based metal-organic framework (FeMOF) and then protected with a layer of phosphatidylcholine (PC)-based phospholipid membrane (PM) that serves as a peroxidase mimetic and a channel gatekeeper, respectively. Using Staphylococcus aureus (S. aureus) as the model bacteria, the PC-specific PFTs secreted by S. aureus perforate the PM layer. Now exposed to the FeMOF, uncharged 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonate) (ABTS) molecules in the electrolyte undergo oxidation to cationic products (ABTS•+). The measured transmembrane ionic current indicates the presence of S. aureus and methicillin-resistant S. aureus (MRSA) with a low detection limit of 3 cfu mL-1. Besides excellent specificity, this sensing approach exhibits satisfactory performance for the detection of target bacteria in the complex media of food.
Collapse
Affiliation(s)
- Junjian Zhao
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| | - Zirui Wang
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| | - Mei Yang
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| | - Junli Guo
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
- Foshan Graduate School of Innovation, Northeastern University, Foshan 528311, China
| | - Zhida Gao
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| | - Pei Song
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Yan-Yan Song
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| |
Collapse
|
12
|
Sajeevan A, Ramamurthy T, Solomon AP. Vibrio cholerae virulence and its suppression through the quorum-sensing system. Crit Rev Microbiol 2024:1-22. [PMID: 38441045 DOI: 10.1080/1040841x.2024.2320823] [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: 07/30/2023] [Accepted: 02/10/2024] [Indexed: 03/06/2024]
Abstract
Vibrio cholerae is a cholera-causing pathogen known to instigate severe contagious diarrhea that affects millions globally. Survival of vibrios depend on a combination of multicellular responses and adapt to changes that prevail in the environment. This process is achieved through a strong communication at the cellular level, the process has been recognized as quorum sensing (QS). The severity of infection is highly dependent on the QS of vibrios in the gut milieu. The quorum may exist in a low/high cell density (LCD/HCD) state to exert a positive or negative response to control the regulatory pathogenic networks. The impact of this regulation reflects on the transition of pathogenic V. cholerae from the environment to infect humans and cause outbreaks or epidemics of cholera. In this context, the review portrays various regulatory processes and associated virulent pathways, which maneuver and control LCD and HCD states for their survival in the host. Although several treatment options are existing, promotion of therapeutics by exploiting the virulence network may potentiate ineffective antibiotics to manage cholera. In addition, this approach is also useful in resource-limited settings, where the accessibility to antibiotics or conventional therapeutic options is limited.
Collapse
Affiliation(s)
- Anusree Sajeevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Thandavarayan Ramamurthy
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Disease, Kolkata, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
13
|
Gao Y, Wang H, Niu X. A hydrogen-bonded curdlan-chitosan/polyvinyl alcohol edible dual functional hydrogel bandage against MRSA promotes wound healing. Int J Biol Macromol 2024; 259:129351. [PMID: 38216019 DOI: 10.1016/j.ijbiomac.2024.129351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/23/2023] [Accepted: 01/07/2024] [Indexed: 01/14/2024]
Abstract
The most prevalent complication arising from skin injuries is bacterial infection, where pathogenic bacteria proliferate significantly at the wound site, leading to subsequent complications like septic shock and sepsis. Although antibiotics presently effectively manage wound infections caused by common bacteria, the escalating prevalence of antibiotic-resistant strains necessitates urgent novel approaches for addressing such infections. Here, we present CS9P1-RA, a dual functional hydrogel dressing, based on polyvinyl alcohol (PVA) matrix crosslinked through hydrogen bonding. CS9P1-RA combines chitosan (CS), a food-derived antibacterial agent, with the natural compound rosmarinic acid (RA) to specifically target skin injuries caused by MRSA. Computational and molecular biology assays illustrate RA's ability to selectively inhibit the activity of Staphylococcus aureus (S. aureus) serine/threonine phosphatase (Stp1), reducing the S. aureus pathogenicity. CS9P1-RA showcases exceptional antibacterial efficacy (MIC = 1 mg/mL) and demonstrates potency in reducing virulence (IC50 = 7.424 μM on Stp1). Notably, it effectively curbs bacterial growth and accelerates wound healing in the mice model, thereby fulfilling the practical requirements for clinical applications. Moreover, the mechanical properties of CS9P1-RA ensure user comfort during treatment. This work introduces a fresh design paradigm for dressing materials, offering a promising solution for treating skin injuries inflicted by antibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Yawen Gao
- College of Food Science and Engineering, Jilin University, Changchun 130062, PR China
| | - Hongsu Wang
- College of Food Science and Engineering, Jilin University, Changchun 130062, PR China
| | - Xiaodi Niu
- College of Food Science and Engineering, Jilin University, Changchun 130062, PR China.
| |
Collapse
|
14
|
Lu L, Wang J, Wang C, Zhu J, Wang H, Liao L, Zhao Y, Wang X, Yang C, He Z, Li M. Plant-derived virulence arresting drugs as novel antimicrobial agents: Discovery, perspective, and challenges in clinical use. Phytother Res 2024; 38:727-754. [PMID: 38014754 DOI: 10.1002/ptr.8072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/23/2023] [Accepted: 11/05/2023] [Indexed: 11/29/2023]
Abstract
Antimicrobial resistance (AMR) emerges as a severe crisis to public health and requires global action. The occurrence of bacterial pathogens with multi-drug resistance appeals to exploring alternative therapeutic strategies. Antivirulence treatment has been a positive substitute in seeking to circumvent AMR, which aims to target virulence factors directly to combat bacterial infections. Accumulated evidence suggests that plant-derived natural products, which have been utilized to treat infectious diseases for centuries, can be abundant sources for screening potential virulence-arresting drugs (VADs) to develop advanced therapeutics for infectious diseases. This review sums up some virulence factors and their actions in various species of bacteria, as well as recent advances pertaining to plant-derived natural products as VAD candidates. Furthermore, we also discuss natural VAD-related clinical trials and patents, the perspective of VAD-based advanced therapeutics for infectious diseases and critical challenges hampering clinical use of VADs, and genomics-guided identification for VAD therapeutic. These newly discovered natural VADs will be encouraging and optimistic candidates that may sustainably combat AMR.
Collapse
Affiliation(s)
- Lan Lu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Jingya Wang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Chongrui Wang
- Faculty of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, P.R. China
| | - Jie Zhu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Hongping Wang
- Safety Evaluation Center, Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), Chengdu, Sichuan, P.R. China
| | - Li Liao
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Yuting Zhao
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Xiaobo Wang
- Department of Hepatobiliary Surgery, Langzhong People's Hospital, Langzhong, Sichuan, P.R. China
| | - Chen Yang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Zhengyou He
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| |
Collapse
|
15
|
Su P, Zhang Y, Jiang C, Zhang T, Bao B, Wang L. Ag +-Adsorbing Semiconducting Polymer Nanosponge for Smart Local Treatment of Wound Infection. ACS APPLIED BIO MATERIALS 2023; 6:4421-4429. [PMID: 37755335 DOI: 10.1021/acsabm.3c00616] [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] [Indexed: 09/28/2023]
Abstract
Nanoplatform combined with photothermal therapy (PTT) and silver nanoparticles have been widely used to combat bacterial infections. However, the development of environmentally benign antibacterial nanoplatforms with controllable and long-term antibacterial activity is still challenging. Herein, we synthesized an Ag+-adsorbing organic semiconducting polymeric nanosponge (PDPP3T NPe@Ag+) to realize Ag+ enhanced photothermal anti-infective therapy. Furthermore, the PDPP3T NPe@Ag+ sponge can also spatiotemporally release silver ions in a pH/NIR light-responsive manner for controllable and long-term antimicrobial therapy. Owing to good biocompatibility and controlled release of silver ions, PDPP3T NPe@Ag+ can effectively kill bacteria in vitro and promote wound healing in vivo. We expect that this antimicrobial platform could be utilized as a robust antibacterial agent for infective therapy.
Collapse
Affiliation(s)
- Peng Su
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Yueyue Zhang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Cheng Jiang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tao Zhang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Biqing Bao
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lianhui Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| |
Collapse
|
16
|
Wang J, Song Y, Huang Z, Lin W, Yu G, Xiong Y, Jiang G, Tan Y, Wang J, Liao X. Coupling a Virulence-Targeting Moiety with Ru-Based AMP Mimics Efficiently Improved Its Anti-Infective Potency and Therapeutic Index. J Med Chem 2023; 66:13304-13318. [PMID: 37704628 DOI: 10.1021/acs.jmedchem.3c01282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
The surge of antibiotic resistance in Staphylococcus aureus calls for novel drugs that attack new targets. Developing antimicrobial peptides (AMPs) or antivirulence agents (AvAs) is a promising strategy to tackle this challenge. However, AMPs, which kill bacteria by disrupting cell membranes, suffer from low stability and high synthesis cost, while AvAs, which inhibit toxin secretion, have relatively poor bactericidal activity. Here, to address their respective shortcomings, we combined these two different antibacterial activities on the same molecular scaffold and developed a Ru-based metalloantibiotic, termed Ru1. Notably, Ru1 exerted remarkable bactericidal activity (MICS = 460 nM) and attenuated bacterial virulence as well. Mechanistic studies demonstrated that Ru1 had two independent targets: CcpA and bacterial membrane integrity. Based on its dual mechanism of action, Ru1 effectively overcame S. aureus resistance and showed high efficacy in a mouse infection model against S. aureus. This study provides a promising approach to confronting bacterial infections.
Collapse
Affiliation(s)
- Jing Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Yun Song
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Ziying Huang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Wenjing Lin
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Guangying Yu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Yanshi Xiong
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Guijuan Jiang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Yanhui Tan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jintao Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Xiangwen Liao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| |
Collapse
|
17
|
Liu W, Ou P, Tian F, Liao J, Ma Y, Wang J, Jin X. Anti- Vibrio parahaemolyticus compounds from Streptomyces parvus based on Pan-genome and subtractive proteomics. Front Microbiol 2023; 14:1218176. [PMID: 37485508 PMCID: PMC10361664 DOI: 10.3389/fmicb.2023.1218176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Vibrio parahaemolyticus is a foodborne pathogen commonly found in seafood, and drug resistance poses significant challenges to its control. This study aimed to identify novel drug targets for antibacterial drug discovery. Methods To identify drug targets, we performed a pan-genome analysis on 58 strains of V. parahaemolyticus genomes to obtain core genes. Subsequently, subtractive proteomics and physiochemical checks were conducted on the core proteins to identify potential therapeutic targets. Molecular docking was then employed to screen for anti-V. parahaemolyticus compounds using a in-house compound library of Streptomyces parvus, chosen based on binding energy. The anti-V. parahaemolyticus efficacy of the identified compounds was further validated through a series of experimental tests. Results and Discussion Pangenome analysis of 58 V. parahaemolyticus genomes revealed that there were 1,392 core genes. After Subtractive proteomics and physiochemical checks, Flagellar motor switch protein FliN was selected as a therapeutic target against V. parahaemolyticus. FliN was modeled and docked with Streptomyces parvus source compounds, and Actinomycin D was identified as a potential anti-V. parahaemolyticus agent with a strong binding energy. Experimental verification confirmed its effectiveness in killing V. parahaemolyticus and significantly inhibiting biofilm formation and motility. This study is the first to use pan-genome and subtractive proteomics to identify new antimicrobial targets for V. parahaemolyticus and to identify the anti-V. parahaemolyticus effect of Actinomycin D. These findings suggest potential avenues for the development of new antibacterial drugs to control V. parahaemolyticus infections.
Collapse
Affiliation(s)
- Wenbin Liu
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Peiyu Ou
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Fangyuan Tian
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jingyang Liao
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan Ma
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie Wang
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaobao Jin
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
18
|
Gao Y, Wang H, Li X, Niu X. Molecular mechanism of green tea polyphenol epicatechin gallate attenuating Staphylococcus aureus pathogenicity by targeting Ser/Thr phosphatase Stp1. Food Funct 2023; 14:4792-4806. [PMID: 37128867 DOI: 10.1039/d3fo00170a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In this study, through virtual screening and in vitro bioactivity assays, we discovered that (-)-epicatechin gallate (ECG), a polyphenol compound extracted from green tea, demonstrated marked anti-Ser/Thr phosphatase (Stp1) activity towards Staphylococcus aureus (S. aureus) with an IC50 value of 8.35 μM. By targeting S. aureus Stp1, ECG prevented the up-regulation of virulence gene and the formation of antibody membrane and protected the mice from S. aureus infection. Through MD simulation, the allosteric inhibitory mechanism of ECG on Stp1 was determined. The Stp1-ECG complex model underwent a significant change in conformation; its flap subdomain changed from opening to closing, whereas Stp1 activity was lost when bound to ECG. In addition, the MD simulation results of Stp1 and several tea polyphenol compounds showed that gallate groups and fewer adjacent phenolic hydroxyl groups contributed to the binding of Stp1 and inhibitors. As an inhibitor targeting S. aureus Stp1, ECG reduced the pathogenicity of S. aureus without inhibiting S. aureus, which largely reduced the possibility of drug resistance. Our findings demonstrated a novel molecular mechanism of green tea as the usual drink against S. aureus infection and elucidated the future design of allosteric inhibitors targeting Stp1.
Collapse
Affiliation(s)
- Yawen Gao
- College of Food Science and Engineering, Jilin University, Changchun 130062, P.R. China.
| | - Hongsu Wang
- College of Food Science and Engineering, Jilin University, Changchun 130062, P.R. China.
| | - Xuening Li
- College of Food Science and Engineering, Jilin University, Changchun 130062, P.R. China.
| | - Xiaodi Niu
- College of Food Science and Engineering, Jilin University, Changchun 130062, P.R. China.
| |
Collapse
|
19
|
Lettl C, Schindele F, Mehdipour AR, Steiner T, Ring D, Brack-Werner R, Stecher B, Eisenreich W, Bilitewski U, Hummer G, Witschel M, Fischer W, Haas R. Selective killing of the human gastric pathogen Helicobacter pylori by mitochondrial respiratory complex I inhibitors. Cell Chem Biol 2023; 30:499-512.e5. [PMID: 37100053 DOI: 10.1016/j.chembiol.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/16/2023] [Accepted: 04/05/2023] [Indexed: 04/28/2023]
Abstract
Respiratory complex I is a multicomponent enzyme conserved between eukaryotic cells and many bacteria, which couples oxidation of electron donors and quinone reduction with proton pumping. Here, we report that protein transport via the Cag type IV secretion system, a major virulence factor of the Gram-negative bacterial pathogen Helicobacter pylori, is efficiently impeded by respiratory inhibition. Mitochondrial complex I inhibitors, including well-established insecticidal compounds, selectively kill H. pylori, while other Gram-negative or Gram-positive bacteria, such as the close relative Campylobacter jejuni or representative gut microbiota species, are not affected. Using a combination of different phenotypic assays, selection of resistance-inducing mutations, and molecular modeling approaches, we demonstrate that the unique composition of the H. pylori complex I quinone-binding pocket is the basis for this hypersensitivity. Comprehensive targeted mutagenesis and compound optimization studies highlight the potential to develop complex I inhibitors as narrow-spectrum antimicrobial agents against this pathogen.
Collapse
Affiliation(s)
- Clara Lettl
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Pettenkoferstrasse 9a, 80336 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Franziska Schindele
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Pettenkoferstrasse 9a, 80336 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Ahmad Reza Mehdipour
- Center for Molecular Modeling, Ghent University, 9052 Zwijnaarde, Belgium; Department of Theoretical Biophysics, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Thomas Steiner
- Bavarian NMR Center-Structural Membrane Biochemistry, Department of Chemistry, Technical University Munich, 85748 Garching, Germany
| | - Diana Ring
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Pettenkoferstrasse 9a, 80336 Munich, Germany
| | - Ruth Brack-Werner
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany; German Research Center for Environmental Health, Institute of Virology, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Pettenkoferstrasse 9a, 80336 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Wolfgang Eisenreich
- Bavarian NMR Center-Structural Membrane Biochemistry, Department of Chemistry, Technical University Munich, 85748 Garching, Germany
| | - Ursula Bilitewski
- Helmholtz Center for Infection Research, 38124 Braunschweig, Germany; German Center for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Braunschweig, Germany
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany; Institute for Biophysics, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | | | - Wolfgang Fischer
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Pettenkoferstrasse 9a, 80336 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| | - Rainer Haas
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Pettenkoferstrasse 9a, 80336 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
20
|
Shen Z, Pan Y, Yan D, Wang D, Tang BZ. AIEgen-Based Nanomaterials for Bacterial Imaging and Antimicrobial Applications: Recent Advances and Perspectives. Molecules 2023; 28:2863. [PMID: 36985835 PMCID: PMC10057855 DOI: 10.3390/molecules28062863] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Microbial infections have always been a thorny problem. Multi-drug resistant (MDR) bacterial infections rendered the antibiotics commonly used in clinical treatment helpless. Nanomaterials based on aggregation-induced emission luminogens (AIEgens) recently made great progress in the fight against microbial infections. As a family of photosensitive antimicrobial materials, AIEgens enable the fluorescent tracing of microorganisms and the production of reactive oxygen (ROS) and/or heat upon light irradiation for photodynamic and photothermal treatments targeting microorganisms. The novel nanomaterials constructed by combining polymers, antibiotics, metal complexes, peptides, and other materials retain the excellent antimicrobial properties of AIEgens while giving other materials excellent properties, further enhancing the antimicrobial effect of the material. This paper reviews the research progress of AIEgen-based nanomaterials in the field of antimicrobial activity, focusing on the materials' preparation and their related antimicrobial strategies. Finally, it concludes with an outlook on some of the problems and challenges still facing the field.
Collapse
Affiliation(s)
- Zipeng Shen
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yinzhen Pan
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Dingyuan Yan
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Ben Zhong Tang
- Shenzhen Institute of Molecular Aggregate Science and Engineering, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
21
|
Liu M, Lv Q, Xu J, Liu B, Zhou Y, Zhang S, Shen X, Wang L. Isoflavone glucoside genistin, an inhibitor targeting Sortase A and Listeriolysin O, attenuates the virulence of Listeria monocytogenes in vivo and in vitro. Biochem Pharmacol 2023; 209:115447. [PMID: 36746262 DOI: 10.1016/j.bcp.2023.115447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
As a common intracellular facultative anaerobic Gram-positive bacterium, Listeria monocytogenes (L. monocytogenes) exhibits strong resistance to extreme environments, such as low temperature and a wide range of pH values, causing contamination in food production and processing. Sortase A (SrtA) and listeriolysin O (LLO), two crucial virulence factors of L. monocytogenes, are widely recognized as potential targets for the development of anti-L. monocytogenes infection drugs. In this study, we found that genistin simultaneously inhibits the peptidase activity of SrtA and the hemolytic activity of LLO without affecting the growth of L. monocytogenes, alleviating concerns about developing resistance. Furthermore, we demonstrated that genistin reduces L. monocytogenes biofilm formation and invasion of human colorectal cancer (Caco-2) cells. Subsequent mechanistic studies revealed that genistin inhibited LLO-mediated Caco-2 cell damage by blocking LLO oligomerization. Fluorescence quenching assay revealed the potential binding mode of SrtA and LLO to genistin. Genistin might bind to the active pocket of SrtA through residues Leu33, Asn29, and Met40, interacting with D1 domain of LLO involved in oligomerization and pore formation through residues Asn259. Studies in infection models revealed that genistin reduces mortality and pathological damage in mice infected with L. monocytogenes. These results indicate that genistin is a promising anti-virulence agent that could be considered an alternative candidate for the treatment of L. monocytogenes infection.
Collapse
Affiliation(s)
- Minda Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China
| | - Qianghua Lv
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, P.R.China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, P.R.China
| | - Jingwen Xu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Baichen Liu
- The Second Bethune Clinical Medical College of Jilin University, Changchun 130012, Jilin, People's Republic of China
| | - Yonglin Zhou
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China
| | - Siqi Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China
| | - Xue Shen
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Food Science, College of Food Science and Engineering, Jilin University, Changchun, China.
| | - Lin Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
22
|
Niu Z, Xie M, Wei Z, Guo Y, Han M, Ding Y, Huang J, Zheng K, Zhang Y, Song Y, Niu D, Li Y, Wen G, Li X, Shi J. In Situ Structure Transformation of a Sprayed Gel for pH-Ultrasensitive Nano-Catalytic Antibacterial Therapy. Adv Healthc Mater 2022; 12:e2202441. [PMID: 36577136 DOI: 10.1002/adhm.202202441] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/24/2022] [Indexed: 12/29/2022]
Abstract
Nano-catalytic bacterial killing provides new opportunities to address ever-increasing antibiotic resistance. However, the intrinsic catalytic activity usually depends on a much lower pH conditions (pH = 2-5) than that in the weakly acidic bacterial microenvironments (pH = 6-7) for reactive oxygen species production by Fenton reactions. Herein, a MnSiO3 -based pH-ultrasensitive "in situ structure transformation" is first reported to significantly promote the adhesion between material and bacteria, and shorten the diffusion distance (<20 nm) to compensate ultra-short life (<200 ns) of ·OH generated by Mn2+ -mediated Fenton-like reaction, finally enhancing its nano-catalytic antibacterial performance in weakly acidic conditions. A separated spray bottle is further designed to achieve in situ gelation at the wound site, which demonstrates excellent shape adaptability to complicated and rough surfaces of wounds, allowing for long-term nano-catalyst release. As a result, bacterial-infected wound healing is efficiently promoted. Herein, the in situ sprayed nano-catalytic antibacterial gel presents a promising paradigm for bacterial infection treatment.
Collapse
Affiliation(s)
- Zhihui Niu
- School of Physics and Optoelectronic Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Mingxiao Xie
- Institute of Engineering Ceramics, School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Zicheng Wei
- Institute of Engineering Ceramics, School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Yang Guo
- Institute of Engineering Ceramics, School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Mengxuan Han
- Institute of Engineering Ceramics, School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Yingying Ding
- Institute of Engineering Ceramics, School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Jianyu Huang
- Institute of Engineering Ceramics, School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Kang Zheng
- Institute of Engineering Ceramics, School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Yao Zhang
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Yuanda Song
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Dechao Niu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yongsheng Li
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Guangwu Wen
- Institute of Engineering Ceramics, School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Xiaowei Li
- Institute of Engineering Ceramics, School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
23
|
Jia XM, Cheng C, Liu T, Zhao YL, Guo B, Tang L, Yang YY. Synthesis and antibiofilm evaluation of N-acyl-2-aminopyrimidine derivatives against Acinetobacter baumannii. Bioorg Med Chem 2022; 76:117095. [PMID: 36442439 DOI: 10.1016/j.bmc.2022.117095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/15/2022]
Abstract
The overuse of antibiotics will led to the increase of drug resistance. Especially, the multidrug-resistant A. baumannii became the leading cause of nosocomial infections with high rates of morbimortality. The drug resistance of A. baumannii is greatly attributed to its biofilm. To alleviate the burden of drug resistance, the anti-virulence signaling strategies was developed. By specifically interfering with the ability of the bacteria to recognize host signals that are needed to establish infection, the bacteria are less able to colonize the host. In this paper, 39 N-acyl-2-aminopyrimidine derivatives were synthesized and tested for their biofilm inhibition efficacy. The screening results reveal that some of the analogues (3ac, 8d) efficiently inhibited the biofilm formation of A. baumannii (IC50 as low as 3.8 μM), and the biofilm inhibition ability was further demonstrated with laser confocal results and extracellular polysaccharides inhibition test. Further motility test reveals our compounds are quorum sensing inhibitors. Besides, the synergistic effect of compounds 3ac and 8d with different antibiotics suggest its potential clinical significance, which was further enhanced by the long time biofilm inhibition test after coating with PLGA. Finally, we also look into the safety of the compounds with cytotoxicity assay.
Collapse
Affiliation(s)
- Xue-Min Jia
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang 550004, China
| | - Cheng Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang 550004, China
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang 550004, China
| | - Yong-Long Zhao
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang 550004, China
| | - Bing Guo
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Lei Tang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang 550004, China
| | - Yuan-Yong Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
24
|
Thorave RG, Shinde AP, Kadam NS, Doshi PJ, Gawhale ST, Malkhede DD. Moxifloxacin embedded p-Sulfonatocalix[6]arene: Multispectroscopic studies to evaluate its cytotoxicity, antibacterial efficacy, and molecular docking. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.121035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Wei R, Wang X, Wang Q, Qiang G, Zhang L, Hu HY. Hyperglycemia in Diabetic Skin Infections Promotes Staphylococcus aureus Virulence Factor Aureolysin: Visualization by Molecular Imaging. ACS Sens 2022; 7:3416-3421. [PMID: 36351204 DOI: 10.1021/acssensors.2c01565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Bacterial skin infections are common in diabetic patients, with Staphylococcus aureus (S. aureus) being the most commonly isolated, causing comorbidities such as increased mortality and long-term hospitalization. While precise mechanisms remain to be determined, hyperglycemia represents an important pathogenetic factor responsible for the increased risk of S. aureus infection. Herein, we constructed a series of ratiometric fluorescent molecular probes for aureolysin (Aur), a major virulence factor in S. aureus. Using probe 1, we were able to determine specific Aur activity in both cells and tissues. We also observed that elevated glucose levels led to 2-fold higher Aur expression in S. aureus cultures. In a diabetic mouse model, we used molecular imaging to demonstrate that hyperglycemia tripled S. aureus Aur virulence compared to nondiabetic mice, resulting in more severe infections.
Collapse
Affiliation(s)
- Rao Wei
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substance Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substance Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qinghua Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substance Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Guifen Qiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Leilei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substance Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substance Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
26
|
He J, Hong M, Xie W, Chen Z, Chen D, Xie S. Progress and prospects of nanomaterials against resistant bacteria. J Control Release 2022; 351:301-323. [PMID: 36165865 DOI: 10.1016/j.jconrel.2022.09.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 12/18/2022]
Abstract
Drug-resistant bacterial infections are increasingly heightening, which lead to more severe illness, higher cost of treatment and increased risk of death. Nanomaterials-based therapy, an "outrider", serving as a kind of innovative antimicrobial therapeutics, showing promise in replacing antimicrobial agents and enhancing the activity of antibiotics, generally bases on the various inorganic and/or organic materials. When the size of those materials is below to a certain nano-level and the content of nanomaterials is above a certain amount, they are lethal to the resistant bacteria, which bypass the traditional bacterial resistance mechanisms. This review highlights the effect of nanomaterials in combating extracellular/intracellular bacteria and eradicating biofilms. Based on the studies searched on the Web of Science through relevant keywords, this review article starts with analyzing the current situation, resistance mechanisms, and treatment difficulties of bacteria resistance. Then, the efficacy of nanomaterials against resistant bacteria and their mechanisms (e.g., physical impairment, biofilm lysis, regulating bacterial metabolism, protein and DNA replication as well as enhancing the antibiotics concentration in infected cells) are collected. Lastly, the factors affecting the antibacterial efficacy are argued from the side of nanomatrials and bacterium, which followed by the emerging challenges and recent perspectives of achieving higher targeting released nanomaterials as antibacterial therapeutics.
Collapse
Affiliation(s)
- Jian He
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Mian Hong
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, China
| | - Wenqing Xie
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhen Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, China
| | - Dongmei Chen
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, China.
| |
Collapse
|
27
|
Patel EN, Lin L, Sneller MM, Eubanks LM, Tepp WH, Pellett S, Janda KD. Investigation of Salicylanilides as Botulinum Toxin Antagonists. ACS Infect Dis 2022; 8:1637-1645. [PMID: 35877209 PMCID: PMC9592073 DOI: 10.1021/acsinfecdis.2c00230] [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] [Indexed: 11/28/2022]
Abstract
Botulinum neurotoxin serotype A (BoNT/A) is recognized by the Centers for Disease Control and Prevention (CDC) as the most potent toxin and as a Tier 1 biowarfare agent. The severity and longevity of botulism stemming from BoNT/A is of significant therapeutic concern, and early administration of antitoxin-antibody therapy is the only approved pharmaceutical treatment for botulism. Small molecule therapeutic strategies have targeted both the heavy chain (HC) and the light chain (LC) catalytic active site and α-/β-exosites. The LC translocation mechanism has also been studied, but an effective, nontoxic inhibitor remains underexplored. In this work, we screened a library of salicylanilides as potential translocation inhibitors. Potential leads following a primary screen were further scrutinized to identify sal30, which has a cellular minimal concentration of a drug that is required for 50% inhibition (IC50) value of 141 nM. The inquiry of salicylanilide sal30's mechanism of action was explored through a self-quenched fluorogenic substrate conjugated to bovine serum albumin (DQ-BSA) fluorescence, confocal microscopy, and vacuolar H+-ATPase (V-ATPase) inhibition assays. The summation of these findings imply that endolysosomal proton translocation through the protonophore mechanism of sal30 causes endosome pH to increase, which in turn prevents LC translocation into cytosol, a process that requires an acidic pH. Thus, the inhibition of BoNT/A activity by salicylanilides likely occurs through disruption of pH-dependent endosomal LC translocation. We further probed BoNT inhibition by sal30 using additivity analysis studies with bafilomycin A1, a known BoNT/A LC translocation inhibitor, which indicated the absence of synergy between the two ionophores.
Collapse
Affiliation(s)
- Ealin N. Patel
- Department of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California, 92037, United States
| | - Lucy Lin
- Department of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California, 92037, United States
| | - Molly M. Sneller
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, Wisconsin, 53706, United States
| | - Lisa M. Eubanks
- Department of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California, 92037, United States
| | - William H. Tepp
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, Wisconsin, 53706, United States
| | - Sabine Pellett
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, Wisconsin, 53706, United States
| | - Kim D. Janda
- Department of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California, 92037, United States
| |
Collapse
|
28
|
Lin X, Zhang H, Li S, Huang L, Zhang R, Zhang L, Yu A, Duan B. Polyphenol-driving assembly for constructing chitin-polyphenol-metal hydrogel as wound dressing. Carbohydr Polym 2022; 290:119444. [DOI: 10.1016/j.carbpol.2022.119444] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/18/2022] [Accepted: 03/30/2022] [Indexed: 12/20/2022]
|
29
|
Chen Y, Liu P, Wu J, Yan W, Xie S, Sun X, Ye BC, Chu X. N-acylhomoserine lactonase-based hybrid nanoflowers: a novel and practical strategy to control plant bacterial diseases. J Nanobiotechnology 2022; 20:347. [PMID: 35883097 PMCID: PMC9327166 DOI: 10.1186/s12951-022-01557-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The disease caused by plant pathogenic bacteria in the production, transportation, and storage of many crops has brought huge losses to agricultural production. N-acylhomoserine lactonases (AHLases) can quench quorum-sensing (QS) by hydrolyzing acylhomoserine lactones (AHLs), which makes them the promising candidates for controlling infections of QS-dependent pathogenic bacteria. Although many AHLases have been isolated and considered as a potentially effective preventive and therapeutic agents for bacterial diseases, the intrinsically poor ambient stability has seriously restricted its application. RESULTS Herein, we showed that a spheroid enzyme-based hybrid nanoflower (EHNF), AhlX@Ni3(PO4)2, can be easily synthesized, and it exhibited 10 times AHL (3OC8-HSL) degradation activity than that with free AhlX (a thermostable AHL lactonase). In addition, it showed intriguing stability even at the working concentration, and retained ~ 100% activity after incubation at room temperature (25 °C) for 40 days and approximately 80% activity after incubation at 60 °C for 48 h. Furthermore, it exhibited better organic solvent tolerance and long-term stability in a complicated ecological environment than that of AhlX. To reduce the cost and streamline production processes, CSA@Ni3(PO4)2, which was assembled from the crude supernatants of AhlX and Ni3(PO4)2, was synthesized. Both AhlX@Ni3(PO4)2 and CSA@Ni3(PO4)2 efficiently attenuated pathogenic bacterial infection. CONCLUSIONS In this study, we have developed N-acylhomoserine lactonase-based hybrid nanoflowers as a novel and efficient biocontrol reagent with significant control effect, outstanding environmental adaptability and tolerance. It was expected to overcome the bottlenecks of poor stability and limited environmental tolerance that have existed for over two decades and pioneered the practical application of EHNFs in the field of biological control.
Collapse
Affiliation(s)
- Yan Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
| | - Pengfu Liu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
| | - Jiequn Wu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
| | - Wanqing Yan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
| | - Saixue Xie
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
| | - Xuanrong Sun
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
| | - Bang-Ce Ye
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China.
| | - Xiaohe Chu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
30
|
Abstract
AB toxins are protein virulence factors secreted by many bacterial pathogens, contributing to the pathogenicity of the cognate bacteria. AB toxins consist of two functionally distinct components: the enzymatic "A" component for pathogenicity and the receptor-binding "B" component for toxin delivery. Consistently, unlike other virulence factors such as effectors, AB toxins do not require additional systems to deliver them to the target host cells. Target host cells are located in the infection site and/or located distantly from infected host cells. The first part of this review discusses the structural and functional features of single-peptide and multiprotein AB toxins in the context of host-microbe interactions, using several well-characterized examples. The second part of this review discusses toxin neutralization strategies, as well as applications of AB toxins relevant to developing intervention strategies against diseases.
Collapse
Affiliation(s)
- Jeongmin Song
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
31
|
Guo S, He Y, Zhu Y, Tang Y, Yu B. Combatting Antibiotic Resistance Using Supramolecular Assemblies. Pharmaceuticals (Basel) 2022; 15:ph15070804. [PMID: 35890105 PMCID: PMC9322166 DOI: 10.3390/ph15070804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Antibiotic resistance has posed a great threat to human health. The emergence of antibiotic resistance has always outpaced the development of new antibiotics, and the investment in the development of new antibiotics is diminishing. Supramolecular self-assembly of the conventional antibacterial agents has been proved to be a promising and versatile strategy to tackle the serious problem of antibiotic resistance. In this review, the recent development of antibacterial agents based on supramolecular self-assembly strategies will be introduced.
Collapse
Affiliation(s)
- Shuwen Guo
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an 710100, China;
- Correspondence: (S.G.); (Y.T.); (B.Y.)
| | - Yuling He
- Institute of Basic and Translational Medicine, Xi’an Medical University, No. 1 Xinwang Road, Xi’an 710021, China;
| | - Yuanyuan Zhu
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an 710100, China;
| | - Yanli Tang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an 710100, China;
- Correspondence: (S.G.); (Y.T.); (B.Y.)
| | - Bingran Yu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing 100029, China
- Correspondence: (S.G.); (Y.T.); (B.Y.)
| |
Collapse
|
32
|
Xue Y, Qiu Z, Zhao Z, Wang C, Cui R, Shen S, Zhao Y, Zhou S, Fang L, Chen Z, Zhu H, Zhu B. Secondary Ammonium-Based Hyperbranched Poly(amidoamine) with Excellent Membrane-Active Property for Multidrug-Resistant Bacterial Infection. ACS APPLIED BIO MATERIALS 2022; 5:3384-3395. [PMID: 35765122 DOI: 10.1021/acsabm.2c00356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
With the rapid emergence of microbial infections induced by "superbugs", public health and the global economy are threatened by the lack of effective and biocompatible antibacterial agents. Herein, we systematically design a series of secondary ammonium-based hyperbranched poly(amidoamine) (SAHBP) with different alkyl chain lengths for probing high-efficacy antibacterial agents. SAHBP modified with alkyl tails at the hyperbranched core could efficiently kill Escherichia coli and Staphylococcus aureus, two types of clinically important bacteria worldwide. The best SAHBP with 12-carbon-long alkyl tails (SAHBP-12) also showed high activity against problematic multidrug-resistant bacteria, including Pseudomonas aeruginosa and methicillin-resistant S. aureus (MRSA). Based on ζ potential, isothermal titration microcalorimetry (ITC), and membrane integrity assays, it is found that SAHBP-12 could attach to the cell membrane via electrostatic adsorption and hydrophobic interactions, following which the integrity of the bacterial cell wall and the cell membrane is disrupted, resulting in severe cell membrane damage and the leakage of cytoplasmic contents, finally causing bacterial cell death. Impressively, benefiting from excellent membrane-active property, SAHBP-12 exhibited robust therapeutic efficacy in MRSA-infected mice wounds. Moreover, SAHBP-12 also showed excellent biosafety in vitro and in vivo, which undoubtedly distinguished it as a potent weapon in combating the growing threat of problematic multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Yunyun Xue
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zelin Qiu
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zihao Zhao
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Chuyao Wang
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ronglu Cui
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Shuyang Shen
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yu Zhao
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Shien Zhou
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Lifeng Fang
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Haihong Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Baoku Zhu
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
33
|
Wang S, Cong Z, Xu Z, Ban S, Song H. Fluorescent dyes with multiple quaternary ammonium centers for specific image discrimination and Gram-positive antibacterial activity. Org Biomol Chem 2022; 20:3980-3987. [PMID: 35502882 DOI: 10.1039/d2ob00399f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Three quaternary ammonium compounds (QACs), TPQA, T2PQA, and T3PQA, were synthesized and employed in antimicrobial tests against E. coli and S. aureus. It was confirmed that they exhibit selective bacteriostasis against S. aureus. The antibacterial activities of the compounds were evaluated via determining their minimum inhibitory concentrations (MICs) and minimal bactericidal concentrations (MBCs) against S. aureus using the 2,3,5-triphenyltetrazolium chloride (TTC) coloration method. Notably, T2PQA exhibited far better properties than TPQA and T3PQA, with the activity found to be dependent on the structure of the QA and the exposed hydrophobic groups. All three compounds showed promising potential for killing Gram-positive bacteria, efficiently guided by fluorescence imaging.
Collapse
Affiliation(s)
- Siqi Wang
- College of Chemistry and Molecular Sciences, Wuhan University, 299 Bayi Road, Wuhan, Hubei Province, China.
| | - Zisong Cong
- College of Chemistry and Molecular Sciences, Wuhan University, 299 Bayi Road, Wuhan, Hubei Province, China.
| | - Zhiqin Xu
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Shurong Ban
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Heng Song
- College of Chemistry and Molecular Sciences, Wuhan University, 299 Bayi Road, Wuhan, Hubei Province, China.
| |
Collapse
|
34
|
Li Z, Zhao L, Bian Y, Li Y, Qu J, Song F. The antibacterial activity of quinazoline and quinazolinone hybrids. Curr Top Med Chem 2022; 22:1035-1044. [PMID: 35255796 DOI: 10.2174/1568026622666220307144015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 11/22/2022]
Abstract
Bacterial infections cause substantial morbidity and mortality across the world and pose serious threats to humankind. Drug resistance, especially multidrug resistance resulting from different defensive mechanisms in bacteria, is the leading cause of failure the chemotherapy, making it an urgent need to develop more effective antibacterials. Quinazoline and quinazolinone frameworks have received considerable attention due to their diversified therapeutic potential. In particular, quinazoline/quinazolinone hybrids could exert antibacterial activity through various mechanisms and are useful scaffolds for the discovery of novel antibacterials. This review principally emphases on the antibacterial potential, structure-activity relationships (SARs), and mechanism of action of quinazoline and quinazolinone hybrids, covering articles published between 2017 and 2021.
Collapse
Affiliation(s)
- Zhenghua Li
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Li Zhao
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Yunqiang Bian
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Yu Li
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| | - Jie Qu
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| | - Feng Song
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| |
Collapse
|
35
|
You K, Gao B, Wang M, Wang X, Okoro KC, Rakhimbekzoda A, Feng Y. Versatile polymer-based strategies for antibacterial drug delivery systems and antibacterial coatings. J Mater Chem B 2022; 10:1005-1018. [DOI: 10.1039/d1tb02417e] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Human health damage and economic losses due to bacterial infections are very serious worldwide. Excessive use of antibiotics has caused an increase in bacterial resistance. Fortunately, various non-antibiotic antibacterial materials...
Collapse
|
36
|
Sun H, Huang SY, Jeyakkumar P, Cai GX, Fang B, Zhou CH. Natural Berberine-derived Azolyl Ethanols as New Structural Antibacterial Agents against Drug-Resistant Escherichia coli. J Med Chem 2021; 65:436-459. [PMID: 34964345 DOI: 10.1021/acs.jmedchem.1c01592] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Natural berberine-derived azolyl ethanols as new structural antibacterial agents were designed and synthesized for fighting with dreadful bacterial resistance. Partial target molecules exhibited potent activity against the tested strains, particularly, nitroimidazole derivative 4d and benzothiazole-2-thoil compound 18b, with low cytotoxicity both exerted strong antibacterial activities against multidrug-resistant Escherichia coli at low concentrations as 0.007 and 0.006 mM, respectively. Meanwhile, the active compounds 4d and 18b possessed the ability to rapidly kill bacteria and observably eradicate the E. coli biofilm by reducing exopolysaccharide content to prevent bacterial adhesion, which was conducive to alleviating the development of E. coli resistance. Preliminary mechanistic explorations suggested that the excellent antibacterial potential of molecules 4d and 18b might be attributed to their ability to disintegrate membrane, accelerate ROS accumulation, reduce bacterial metabolism, and intercalate into DNA groove. These results provided powerful information for the further exploitation of natural berberine derivatives against bacterial pathogens.
Collapse
Affiliation(s)
- Hang Sun
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Shi-Yu Huang
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ponmani Jeyakkumar
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Gui-Xin Cai
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Bo Fang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China
| | - Cheng-He Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| |
Collapse
|
37
|
Dai X, Zhang B, Yu Q, Liu Y. Multicharged Supramolecular Assembly Mediated by Polycationic Cyclodextrin for Efficiently Photodynamic Antibacteria. ACS APPLIED BIO MATERIALS 2021; 4:8536-8542. [PMID: 35005946 DOI: 10.1021/acsabm.1c01018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Supramolecular antimicrobial materials based on synthetic macrocycles have recently aroused enormous interests due to their controllable and effective antibacterial treatment. Herein, a multicharged supramolecular assembly was fabricated employing the moderate host-guest interaction between hexa-adamantane-appended ruthenium polypyridyl (Ru2) and polycationic cyclodextrin (CD-QAS) in water. The positively multicharged feature of supramolecular assembly could remarkably enhance the specific intercalation and accumulation in negatively charged bacteria membrane leading to the physical membrane damage. Subsequently, the assembly could efficiently initiate the significant generation of singlet oxygen (1O2) in situ when irradiated with white light thus exhibiting a highly efficient antibacterial capability. Significantly, antibacterial experiments indicated that Ru2/CD-QAS displayed less effect on suppressing the growth of E. coli only about 25% in the absence of light while they exhibited excellent killing efficiency more than 99% toward E. coli under light irradiation. This work provides a simple approach for constructing supramolecular antimicrobial materials for synergistic photodynamic antibacteria.
Collapse
Affiliation(s)
- Xianyin Dai
- Department College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| | - Bing Zhang
- Department College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yu Liu
- Department College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
38
|
Abd El-Aleam RH, George RF, Georgey HH, Abdel-Rahman HM. Bacterial virulence factors: a target for heterocyclic compounds to combat bacterial resistance. RSC Adv 2021; 11:36459-36482. [PMID: 35494393 PMCID: PMC9043591 DOI: 10.1039/d1ra06238g] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/01/2021] [Indexed: 12/17/2022] Open
Abstract
Antibiotic resistance is one of the most important challenges of the 21st century. However, the growing understanding of bacterial pathogenesis and cell-to-cell communication has revealed many potential strategies for the discovery of drugs that can be used for the treatment of bacterial infections. Interfering with bacterial virulence and/or quorum sensing could be a particularly interesting approach, because it is believed to exert less selective pressure on the bacterial resistance than with traditional strategies, geared toward killing bacteria or preventing their growth. Here, we discuss the mechanism of bacterial virulence, presenting promising strategies and recently synthesized heterocyclic compounds to combat future bacterial infections.
Collapse
Affiliation(s)
- Rehab H Abd El-Aleam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information MTI Cairo 11571 Egypt
| | - Riham F George
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University Cairo 11562 Egypt
| | - Hanan H Georgey
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University Cairo 11562 Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University Cairo 11786 Egypt
| | - Hamdy M Abdel-Rahman
- Medicinal Chemistry Department, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University Beni Suef Egypt
| |
Collapse
|
39
|
|
40
|
Wang C, Lu H, Liu M, Wang G, Li X, Lu W, Dong W, Wang X, Chen H, Tan C. Effective Antibacterial and Antihemolysin Activities of Ellipticine Hydrochloride against Streptococcus suis in a Mouse Model. Appl Environ Microbiol 2021; 87:e03165-20. [PMID: 33674433 PMCID: PMC8117749 DOI: 10.1128/aem.03165-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/31/2021] [Indexed: 02/03/2023] Open
Abstract
Streptococcal toxic shock-like syndrome (STSLS) caused by the epidemic strain of Streptococcus suis leads to severe inflammation and high mortality. The life and health of humans and animals are also threatened by the increasingly severe antimicrobial resistance in Streptococcus suis There is an urgent need to discover novel strategies for the treatment of S. suis infection. Suilysin (SLY) is considered to be an important virulence factor in the pathogenesis of S. suis In this study, ellipticine hydrochloride (EH) was reported as a compound that antagonizes the hemolytic activity of SLY. In vitro, EH was found to effectively inhibit SLY-mediated hemolytic activity. Furthermore, EH had a strong affinity for SLY, thereby directly binding to SLY to interfere with the hemolytic activity. Meanwhile, it was worth noting that EH was also found to have a significant antibacterial activity. In vivo, compared with traditional ampicillin, EH not only significantly improved the survival rate of mice infected with S. suis 2 strain Sc19 but also relieved lung pathological damage. Furthermore, EH effectively decreased the levels of inflammatory cytokines (interleukin-6 [IL-6], tumor necrosis factor alpha [TNF-α]) and blood biochemistry enzymes (alanine transaminase [ALT], aspartate transaminase [AST], creatine kinase [CK]) in Sc19-infected mice. Additionally, EH markedly reduced the bacterial load of tissues in Sc19-infected mice. In conclusion, our findings suggest that EH can be a potential compound for treating S. suis infection in view of its antibacterial and antihemolysin activity.IMPORTANCE In recent years, the inappropriate use of antibiotics has unnecessarily caused the continuous emergence of resistant bacteria. The antimicrobial resistance of Streptococcus suis has also become an increasingly serious problem. Targeting virulence can reduce the selective pressure of bacteria on antibiotics, thereby alleviating the development of bacterial resistance to a certain extent. Meanwhile, the excessive inflammatory response caused by S. suis infection is considered the primary cause of acute death. Here, we found that ellipticine hydrochloride (EH) exhibited effective antibacterial and antihemolysin activities against S. suisin vitroIn vivo, compared with ampicillin, EH had a significant protective effect on S. suis serotype 2 strain Sc19-infected mice. Our results indicated that EH, with dual antibacterial and antivirulence effects, will contribute to treating S. suis infections and alleviating the antimicrobial resistance of S. suis to a certain extent. More importantly, EH may develop into a promising drug for the prevention of acute death caused by excessive inflammation.
Collapse
Affiliation(s)
- Chenchen Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Hao Lu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Manli Liu
- Hubei Biopesticide Engineering Research Centre, Wuhan, Hubei, China
| | - Gaoyan Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xiaodan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Wenjia Lu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Wenqi Dong
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xiangru Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, Hubei, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, Hubei, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, Hubei, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, Hubei, China
| | - Chen Tan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, Hubei, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, Hubei, China
| |
Collapse
|
41
|
Maisetta G, Piras AM, Motta V, Braccini S, Mazzantini D, Chiellini F, Zambito Y, Esin S, Batoni G. Antivirulence Properties of a Low-Molecular-Weight Quaternized Chitosan Derivative against Pseudomonas aeruginosa. Microorganisms 2021; 9:912. [PMID: 33923269 PMCID: PMC8145479 DOI: 10.3390/microorganisms9050912] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 01/21/2023] Open
Abstract
The co-occurrence of increasing rates of resistance to current antibiotics and the paucity of novel antibiotics pose major challenges for the treatment of bacterial infections. In this scenario, treatments targeting bacterial virulence have gained considerable interest as they are expected to exert a weaker selection for resistance than conventional antibiotics. In a previous study, we demonstrated that a low-molecular-weight quaternized chitosan derivative, named QAL, displays antibiofilm activity against the major pathogen Pseudomonas aeruginosa at subinhibitory concentrations. The aim of this study was to investigate whether QAL was able to inhibit the production of relevant virulence factors of P. aeruginosa. When tested in vitro at subinhibiting concentrations (0.31-0.62 mg/mL), QAL markedly reduced the production of pyocyanin, pyoverdin, proteases, and LasA, as well as inhibited the swarming motility of three out of four P. aeruginosa strains tested. Furthermore, quantitative reverse transcription PCR (qRT-PCR) analyses demonstrated that expression of lasI and rhlI, two QS-related genes, was highly downregulated in a representative P. aeruginosa strain. Confocal scanning laser microscopy analysis suggested that FITC-labelled QAL accumulates intracellularly following incubation with P. aeruginosa. In contrast, the reduced production of virulence factors was not evidenced when QAL was used as the main polymeric component of polyelectrolyte-based nanoparticles. Additionally, combination of sub-MIC concentrations of QAL and tobramycin significantly reduced biofilm formation of P. aeruginosa, likely due to a synergistic activity towards planktonic bacteria. Overall, the results obtained demonstrated an antivirulence activity of QAL, possibly due to polymer intracellular localization and QS-inhibition, and its ability to inhibit P. aeruginosa growth synergizing with tobramycin.
Collapse
Affiliation(s)
- Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (V.M.); (D.M.); (S.E.); (G.B.)
| | - Anna Maria Piras
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (A.M.P.); (Y.Z.)
| | - Vincenzo Motta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (V.M.); (D.M.); (S.E.); (G.B.)
| | - Simona Braccini
- Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM PISA, 56124 Pisa, Italy; (S.B.); (F.C.)
| | - Diletta Mazzantini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (V.M.); (D.M.); (S.E.); (G.B.)
| | - Federica Chiellini
- Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM PISA, 56124 Pisa, Italy; (S.B.); (F.C.)
| | - Ylenia Zambito
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (A.M.P.); (Y.Z.)
- Interdepartmental Research Centre “Nutraceuticals and Food for Health”, University of Pisa, 56100 Pisa, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (V.M.); (D.M.); (S.E.); (G.B.)
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (V.M.); (D.M.); (S.E.); (G.B.)
| |
Collapse
|
42
|
Spirescu VA, Chircov C, Grumezescu AM, Andronescu E. Polymeric Nanoparticles for Antimicrobial Therapies: An Up-To-Date Overview. Polymers (Basel) 2021; 13:724. [PMID: 33673451 PMCID: PMC7956825 DOI: 10.3390/polym13050724] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Despite the many advancements in the pharmaceutical and medical fields and the development of numerous antimicrobial drugs aimed to suppress and destroy pathogenic microorganisms, infectious diseases still represent a major health threat affecting millions of lives daily. In addition to the limitations of antimicrobial drugs associated with low transportation rate, water solubility, oral bioavailability and stability, inefficient drug targeting, considerable toxicity, and limited patient compliance, the major cause for their inefficiency is the antimicrobial resistance of microorganisms. In this context, the risk of a pre-antibiotic era is a real possibility. For this reason, the research focus has shifted toward the discovery and development of novel and alternative antimicrobial agents that could overcome the challenges associated with conventional drugs. Nanotechnology is a possible alternative, as there is significant evidence of the broad-spectrum antimicrobial activity of nanomaterials and nanoparticles in particular. Moreover, owing to their considerable advantages regarding their efficient cargo dissolving, entrapment, encapsulation, or surface attachment, the possibility of forming antimicrobial groups for specific targeting and destruction, biocompatibility and biodegradability, low toxicity, and synergistic therapy, polymeric nanoparticles have received considerable attention as potential antimicrobial drug delivery agents. In this context, the aim of this paper is to provide an up-to-date overview of the most recent studies investigating polymeric nanoparticles designed for antimicrobial therapies, describing both their targeting strategies and their effects.
Collapse
Affiliation(s)
- Vera Alexandra Spirescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| | - Cristina Chircov
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| |
Collapse
|
43
|
Annunziato G, Spadini C, Franko N, Storici P, Demitri N, Pieroni M, Flisi S, Rosati L, Iannarelli M, Marchetti M, Magalhaes J, Bettati S, Mozzarelli A, Cabassi CS, Campanini B, Costantino G. Investigational Studies on a Hit Compound Cyclopropane-Carboxylic Acid Derivative Targeting O-Acetylserine Sulfhydrylase as a Colistin Adjuvant. ACS Infect Dis 2021; 7:281-292. [PMID: 33513010 DOI: 10.1021/acsinfecdis.0c00378] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Antibacterial adjuvants are of great significance, since they allow the therapeutic dose of conventional antibiotics to be lowered and reduce the insurgence of antibiotic resistance. Herein, we report that an O-acetylserine sulfhydrylase (OASS) inhibitor can be used as a colistin adjuvant to treat infections caused by Gram-positive and Gram-negative pathogens. A compound that binds OASS with a nM dissociation constant was tested as an adjuvant of colistin against six critical pathogens responsible for infections spreading worldwide, Escherichia coli, Salmonella enterica serovar Typhimurium, Klebisiella pneumoniae, Staphylococcus aureus, methicillin-resistant Staphylococcus aureus, and Staphylococcus pseudintermedius. The compound showed promising synergistic or additive activities against all of them. Knockout experiments confirmed the intracellular target engagement supporting the proposed mechanism of action. Moreover, compound toxicity was evaluated by means of its hemolytic activity against sheep defibrinated blood cells, showing a good safety profile. The 3D structure of the compound in complex with OASS was determined at 1.2 Å resolution by macromolecular crystallography, providing for the first time structural insights about the nature of the interaction between the enzyme and this class of competitive inhibitors. Our results provide a robust proof of principle supporting OASS as a potential nonessential antibacterial target to develop a new class of adjuvants and the structural basis for further structure-activity relationship studies.
Collapse
Affiliation(s)
- Giannamaria Annunziato
- P4T Group, Department of Food and Drugs, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Costanza Spadini
- Operative Unit of Animals Infectious Diseases, Department of Veterinary Science, University of Parma, via del Taglio, 8, 43126 Parma, Italy
| | - Nina Franko
- Laboratory of Biochemistry and Molecular Biology, Department of Food and Drugs, University of Parma, via Parco Area delle Scienze 23/A, 43124 Parma, Italy
| | - Paola Storici
- Elettra - Sincrotrone Trieste S.C.p.A., SS 14
- km 163,5 in AREA Science Park, 34149 Trieste, Italy
| | - Nicola Demitri
- Elettra - Sincrotrone Trieste S.C.p.A., SS 14
- km 163,5 in AREA Science Park, 34149 Trieste, Italy
| | - Marco Pieroni
- P4T Group, Department of Food and Drugs, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Sara Flisi
- Operative Unit of Animals Infectious Diseases, Department of Veterinary Science, University of Parma, via del Taglio, 8, 43126 Parma, Italy
| | - Lucrezia Rosati
- Operative Unit of Animals Infectious Diseases, Department of Veterinary Science, University of Parma, via del Taglio, 8, 43126 Parma, Italy
| | - Mattia Iannarelli
- Operative Unit of Animals Infectious Diseases, Department of Veterinary Science, University of Parma, via del Taglio, 8, 43126 Parma, Italy
| | - Marialaura Marchetti
- Biopharmanet-TEC Interdepartmental Center, University of Parma, 43124 Parma, Italy
| | - Joana Magalhaes
- P4T Group, Department of Food and Drugs, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Stefano Bettati
- Department of Medicine and Surgery, University of Parma, via Volturno, 39, 43125 Parma, Italy
- Biopharmanet-TEC Interdepartmental Center, University of Parma, 43124 Parma, Italy
- Institute of Biophysics, CNR, 56124 Pisa, Italy
| | - Andrea Mozzarelli
- Laboratory of Biochemistry and Molecular Biology, Department of Food and Drugs, University of Parma, via Parco Area delle Scienze 23/A, 43124 Parma, Italy
- Biopharmanet-TEC Interdepartmental Center, University of Parma, 43124 Parma, Italy
- Institute of Biophysics, CNR, 56124 Pisa, Italy
| | - Clotilde Silvia Cabassi
- Operative Unit of Animals Infectious Diseases, Department of Veterinary Science, University of Parma, via del Taglio, 8, 43126 Parma, Italy
| | - Barbara Campanini
- Laboratory of Biochemistry and Molecular Biology, Department of Food and Drugs, University of Parma, via Parco Area delle Scienze 23/A, 43124 Parma, Italy
- Biopharmanet-TEC Interdepartmental Center, University of Parma, 43124 Parma, Italy
| | - Gabriele Costantino
- P4T Group, Department of Food and Drugs, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| |
Collapse
|
44
|
Streicher LM. Exploring the future of infectious disease treatment in a post-antibiotic era: A comparative review of alternative therapeutics. J Glob Antimicrob Resist 2021; 24:285-295. [PMID: 33484895 DOI: 10.1016/j.jgar.2020.12.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/19/2020] [Accepted: 12/26/2020] [Indexed: 12/23/2022] Open
Abstract
Antibiotic resistance is projected to be one of the greatest healthcare challenges of the 21st century. As the efficacy of these critical drugs wanes and the discovery of new antibiotics stagnates, exploration of alternative therapies could offer a much needed solution. Although numerous alternative therapies are currently under investigation, three in particular appear poised for long-term success, namely antimicrobial oligonucleotides, monoclonal antibodies and phage therapy. Antimicrobial oligonucleotides could conceivably offer the greatest spectrum of activity while having the lowest chance of unrecoverable resistance. Bacteriophages, while most susceptible to resistance, are inexhaustible, inexpensive and exceptionally adept at eliminating biofilm-associated infections. And although monoclonal antibodies may have limited access to such recalcitrant bacteria, these agents are uniquely able to neutralise exotoxins and other diffusible virulence factors. This comparative review seeks to illuminate these promising therapies and to encourage the scientific and financial support necessary to usher in the next generation of infectious disease treatment.
Collapse
|
45
|
Thioether-linked dihydropyrrol-2-one analogues as PqsR antagonists against antibiotic resistant Pseudomonas aeruginosa. Bioorg Med Chem 2021; 31:115967. [PMID: 33434766 DOI: 10.1016/j.bmc.2020.115967] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023]
Abstract
The Pseudomonas quinolone system (pqs) is one of the key quorum sensing systems in antibiotic-resistant P. aeruginosa and is responsible for the production of virulence factors and biofilm formation. Thus, synthetic small molecules that can target the PqsR (MvfR) receptor can be utilized as quorum sensing inhibitors to treat P. aeruginosa infections. In this study, we report the synthesis of novel thioether-linked dihydropyrrol-2-one (DHP) analogues as PqsR antagonists. Compound 7g containing a 2-mercaptopyridyl linkage effectively inhibited the pqs system with an IC50 of 32 µM in P. aeruginosa PAO1. Additionally, these inhibitors significantly reduced bacterial aggregation and biofilm formation without affecting planktonic growth. The molecular docking study suggest that these inhibitors bind with the ligand binding domain of the MvfR as a competitive antagonist.
Collapse
|
46
|
Does targeting Arg98 of FimH lead to high affinity antagonists? Eur J Med Chem 2020; 211:113093. [PMID: 33340913 DOI: 10.1016/j.ejmech.2020.113093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/20/2020] [Accepted: 12/06/2020] [Indexed: 11/23/2022]
Abstract
Bacterial resistance has become an important challenge in the treatment of urinary tract infections. The underlying resistance mechanisms can most likely be circumvented with an antiadhesive approach, antagonizing the lectin FimH located at the tip of fimbriae of uropathogenic E. coli. Here we report on a novel series of FimH antagonists based on the 1-(α-d-mannopyranosyl)-4-phenyl-1,2,3-triazole scaffold, designed to incorporate carboxylic acid or ester functions to interact with FimH Arg98. The most potent representative of the series, ester 11e, displayed a Kd value of 7.6 nM for the lectin domain of FimH with a general conclusion that all esters outperform carboxylates in terms of affinity. Surprisingly, all compounds from this new series exhibited improved binding affinities also for the R98A mutant, indicating another possible interaction contributing to binding. Our study on 1-(α-d-mannopyranosyl)-4-phenyl-1,2,3-triazole-based FimH antagonists offers proof that targeting Arg98 side chain by a "chemical common sense", i.e. by introduction of the acidic moiety to form ionic bond with Arg98 is most likely unsuitable approach to boost FimH antagonists' potency.
Collapse
|
47
|
Corrêa RCG, Heleno SA, Alves MJ, Ferreira ICFR. Bacterial Resistance: Antibiotics of Last Generation used in Clinical Practice and the Arise of Natural Products as New Therapeutic Alternatives. Curr Pharm Des 2020; 26:815-837. [PMID: 32091328 DOI: 10.2174/1381612826666200224105153] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/29/2020] [Indexed: 01/13/2023]
Abstract
Bacterial resistance to therapeutical drugs has been a serious issue over the last decades. In fact, the quick development of resistance mechanisms by the microorganisms has been fatal for millions of people around the world, turning into a public health issue. The major cause of the resistance mechanisms is the overuse of antimicrobials. European countries try to implement mechanisms to overcome antimicrobial resistance in the community through the rational use of antimicrobials. The scientific community has been exhaustively dedicated to the discovering of new, safer and efficient drugs, being the exploitation of natural resources, mainly plants and fungi, considered as a hot topic in the field of antimicrobial agents. Innumerous reports have already shown the promising capacity of natural products or molecules extracted from these natural resources, to act as bacteriostatic and bactericidal agents. More importantly, these natural agents present significantly lower harmful effects. Bearing that in mind, this review aims at giving a contribution to the knowledge about the synthetic antibiotics of the last generation. Moreover, it is intended to provide information about the last advances regarding the discovery of new antimicrobial agents. Thus, a compilation of the chemical characteristics, efficiency, harmful outcomes and resistance mechanisms developed by the microorganisms can be consulted in the following sections together with a critical discussion, in line with the recent approaches. Furthermore, modern strategies for the prospection of novel anti-infective compounds for tackling resistant bacteria have been considered as also a current synopsis of plants and mushrooms with relevant antimicrobial potentials.
Collapse
Affiliation(s)
- Rúbia C G Corrêa
- Centro de Investigacao de Montanha (CIMO), Instituto Politecnico de Braganca, Campus de Santa Apolonia, Braganca, Portugal.,Program of Master in Science, Technology and Food Safety, Cesumar Institute of Science Technology and Innovation (ICETI), University Center of Maringa (UNICESUMAR), Maringa, Parana, Brazil
| | - Sandrina A Heleno
- Centro de Investigacao de Montanha (CIMO), Instituto Politecnico de Braganca, Campus de Santa Apolonia, Braganca, Portugal
| | - Maria J Alves
- Centro de Investigacao de Montanha (CIMO), Instituto Politecnico de Braganca, Campus de Santa Apolonia, Braganca, Portugal
| | - Isabel C F R Ferreira
- Centro de Investigacao de Montanha (CIMO), Instituto Politecnico de Braganca, Campus de Santa Apolonia, Braganca, Portugal
| |
Collapse
|
48
|
Qian Y, Deng S, Lu Z, She Y, Xie J, Cong Z, Zhang W, Liu R. Using In Vivo Assessment on Host Defense Peptide Mimicking Polymer-Modified Surfaces for Combating Implant Infections. ACS APPLIED BIO MATERIALS 2020; 4:3811-3829. [DOI: 10.1021/acsabm.0c01066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Yuxin Qian
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Shuai Deng
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, Key Laboratory of Specially Functional Polymeric Materials and Related Technology (ECUST) Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ziyi Lu
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, Key Laboratory of Specially Functional Polymeric Materials and Related Technology (ECUST) Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yunrui She
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, Key Laboratory of Specially Functional Polymeric Materials and Related Technology (ECUST) Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jiayang Xie
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, Key Laboratory of Specially Functional Polymeric Materials and Related Technology (ECUST) Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Zihao Cong
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, Key Laboratory of Specially Functional Polymeric Materials and Related Technology (ECUST) Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wenjing Zhang
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, Key Laboratory of Specially Functional Polymeric Materials and Related Technology (ECUST) Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, Key Laboratory of Specially Functional Polymeric Materials and Related Technology (ECUST) Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
49
|
Song F, Li Z, Bian Y, Huo X, Fang J, Shao L, Zhou M. Indole/isatin-containing hybrids as potential antibacterial agents. Arch Pharm (Weinheim) 2020; 353:e2000143. [PMID: 32667714 DOI: 10.1002/ardp.202000143] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
The emergence and worldwide spread of drug-resistant bacteria have already posed a serious threat to human life, creating the urgent need to develop potent and novel antibacterial drug candidates with high efficacy. Indole and isatin (indole-2,3-dione) present a wide structural and mechanistic diversity, so their derivatives possess various pharmacological properties and occupy a salient place in the development of new drugs. Indole/isatin-containing hybrids, which demonstrate a promising activity against a panel of clinically important Gram-positive and Gram-negative bacteria, are privileged scaffolds for the discovery of novel antibacterial candidates. This review, covering articles published between January 2015 and May 2020, focuses on the development and structure-activity relationship (SAR) of indole/isatin-containing hybrids with potential application for fighting bacterial infections, to facilitate further rational design of novel drug candidates.
Collapse
Affiliation(s)
- Feng Song
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, Shandong, China.,School of Life Sciences, Dezhou University, Dezhou, Shandong, China
| | - Zhenghua Li
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, Shandong, China
| | - Yunqiang Bian
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, Shandong, China
| | - Xiankai Huo
- Department of Medical Imaging, Dezhou People's Hospital, Dezhou, Shandong, China
| | - Junman Fang
- School of Life Sciences, Dezhou University, Dezhou, Shandong, China
| | - Linlin Shao
- School of Life Sciences, Dezhou University, Dezhou, Shandong, China
| | - Meng Zhou
- School of Life Sciences, Dezhou University, Dezhou, Shandong, China
| |
Collapse
|
50
|
Yan H, Ni H, Yang Y, Shan C, Yang X, Li X, Cao J, Wu W, Liu W, Tang Y. Smart nanoprobe based on two-photon sensitized terbium-carbon dots for dual-mode fluorescence thermometer and antibacterial. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2019.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|