1
|
Deng J, Yuan Y, Wu Y, Wen F, Yang X, Gou S, Chu Y, Zhao K. Isovanillin decreases the virulence regulated by the quorum sensing system of Pseudomonas aeruginosa. Microb Pathog 2024; 196:107010. [PMID: 39396686 DOI: 10.1016/j.micpath.2024.107010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
The quorum-sensing (QS) system of Pseudomonas aeruginosa dominates the pathogenicity of the acute or chronic infection process. Hence, curbing the pathogenicity of P. aeruginosa by targeting QS system is an ideal strategy. This study aims to identify potential anti-virulence compounds that can effectively disrupt the QS system of P. aeruginosa using a combination of virtual screening and experimental validation techniques. We explored inhibitory effect of isovanillin obtained by virtual screening on P. aeruginosa QS regulated virulence factors extracellular protease, biofilm, and pyocyanin. Results displayed that isovanillin could inhibit the virulence phenotypes regulated by the las- and pqs-QS systems of P. aeruginosa. The synthesis of extracellular proteases, pyocyanin, and biofilm formation by P. aeruginosa were dramatically inhibited by sub-MICs doses of isovanillin. The results of RNA sequencing and quantitative PCR revealed that the QS-activated genes down-regulated by subinhibitory isovanillin in the transcriptional evels. Furthermore, the presence of isovanillin increased the susceptibility of drug-resistant P. aeruginosa to kanamycin, meropenem, and polymyxin B. Treatment of isovanillin as a monotherapy significantly decreased the mortality of C. elegans in P. aeruginosa PAO1 or UCBPP-PA14 (PA14) infection. Our study reported the anti-virulence activity of isovanillin against P. aeruginosa, and provided a structural foundation for developing anti-virulence drugs targeting the QS system of P. aeruginosa.
Collapse
Affiliation(s)
- Junfeng Deng
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, 610106, China
| | - Yang Yuan
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, 610106, China; Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Yi Wu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, 610106, China
| | - Fulong Wen
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, 610106, China
| | - Xiting Yang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, 610106, China
| | - Shiyi Gou
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, 610106, China
| | - Yiwen Chu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, 610106, China.
| | - Kelei Zhao
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, 610106, China.
| |
Collapse
|
2
|
Elghali F, Ibrahim I, Guesmi M, Frikha F, Mnif S. Unveiling the impact of selected essential oils on MRSA strain ATCC 33591: antibacterial efficiency, biofilm disruption, and staphyloxanthin inhibition. Braz J Microbiol 2024; 55:2057-2069. [PMID: 38775905 PMCID: PMC11405564 DOI: 10.1007/s42770-024-01374-2] [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: 12/16/2023] [Accepted: 05/06/2024] [Indexed: 09/18/2024] Open
Abstract
This work aimed to evaluate the effects of 4 selected essential oils on planktonic cells and microbial biofilms of the Staphylococcus aureus strain (MRSA ATCC 33591). The antibacterial activities of the four essential oils Geranium (Pelargonium graveolens), PgEO, Tea Tree (Melaleuca alternifolia) MaEO, Lemon peel (Citrus limon) ClEO and Peppermint (Mentha piperita) MpEO had MICs ranging from 1.56 to 12.5 µl/ml. The evaluation of the antibiofilm activities of the 4 EOs revealed that they had antiadhesive activities against S. aureus MRSA biofilms; the activity reached 60% (the EO of MpEO peppermint at a concentration of 3.12 µl/ml), and the eradication activity was 80% (the EO of PgEO and MpEO at 3.12 µl/ml). The antibiofilm activity of S. aureus has been explained by the binding of several essential oil bioactive molecules to the SarA protein, the main target protein involved in biofilm formation. The synthesis of the virulence factor staphyloxanthin by S. aureus MRSA ATCC 33591 was significantly inhibited in the presence of PgEO at a concentration of MIC/2. This inhibition was explained by the binding of the main PgEO molecules (β-citronellol and geraniol) to the CrTM protein involved in the staphyloxanthin synthesis pathway. There is evidence that these essential oils could be used as potential anti-virulents to control Staphylococcus biofilm formation.
Collapse
Affiliation(s)
- Fares Elghali
- Laboratory of Molecular and Cellular Screening Processes at Center of Biotechnology of Sfax, Sfax, Tunisia
| | - Ibtissem Ibrahim
- Laboratory of Molecular and Cellular Screening Processes at Center of Biotechnology of Sfax, Sfax, Tunisia
| | - Maha Guesmi
- Laboratory of Molecular and Cellular Screening Processes at Center of Biotechnology of Sfax, Sfax, Tunisia
| | - Fakher Frikha
- Laboratory of Molecular and Cellular Screening Processes at Center of Biotechnology of Sfax, Sfax, Tunisia
| | - Sami Mnif
- Laboratory of Molecular and Cellular Screening Processes at Center of Biotechnology of Sfax, Sfax, Tunisia.
| |
Collapse
|
3
|
Pylkkö T, Schneider YKH, Rämä T, Andersen JH, Tammela P. Bioprospecting of inhibitors of EPEC virulence from metabolites of marine actinobacteria from the Arctic Sea. Front Microbiol 2024; 15:1432475. [PMID: 39282555 PMCID: PMC11392781 DOI: 10.3389/fmicb.2024.1432475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/28/2024] [Indexed: 09/19/2024] Open
Abstract
A considerable number of antibacterial agents are derived from bacterial metabolites. Similarly, numerous known compounds that impede bacterial virulence stem from bacterial metabolites. Enteropathogenic Escherichia coli (EPEC) is a notable human pathogen causing intestinal infections, particularly affecting infant mortality in developing regions. These infections are characterized by microvilli effacement and intestinal epithelial lesions linked with aberrant actin polymerization. This study aimed to identify potential antivirulence compounds for EPEC infections among bacterial metabolites harvested from marine actinobacteria (Kocuria sp. and Rhodococcus spp.) from the Arctic Sea by the application of virulence-based screening assays. Moreover, we demonstrate the suitability of these antivirulence assays to screen actinobacteria extract fractions for the bioassay-guided identification of metabolites. We discovered a compound in the fifth fraction of a Kocuria strain that interferes with EPEC-induced actin polymerization without affecting growth. Furthermore, a growth-inhibiting compound was identified in the fifth fraction of a Rhodococcus strain. Our findings include the bioassay-guided identification, HPLC-MS-based dereplication, and isolation of a large phospholipid and a likely antimicrobial peptide, demonstrating the usefulness of this approach in screening for compounds capable of inhibiting EPEC virulence.
Collapse
Affiliation(s)
- Tuomas Pylkkö
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | - Teppo Rämä
- Marbio, Faculty for Fisheries, Biosciences and Economy, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Jeanette Hammer Andersen
- Marbio, Faculty for Fisheries, Biosciences and Economy, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Päivi Tammela
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Hintzen JCJ, Abujubara H, Tietze D, Tietze AA. The Complete Assessment of Small Molecule and Peptidomimetic Inhibitors of Sortase A Towards Antivirulence Treatment. Chemistry 2024; 30:e202401103. [PMID: 38716707 DOI: 10.1002/chem.202401103] [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: 03/19/2024] [Indexed: 06/20/2024]
Abstract
This review covers the most recent advances in the development of inhibitors for the bacterial enzyme sortase A (SrtA). Sortase A (SrtA) is a critical virulence factor, present ubiquitously in Gram-positive bacteria of which many are pathogenic. Sortases are key enzymes regulating bacterial adherence to host cells, by anchoring extracellular matrix-binding proteins to the bacterial outer cell wall. By targeting virulence factors, effective treatment can be achieved, without inducing antibiotic resistance to the treatment. This is a potentially more sustainable, long-term approach to treating bacterial infections, including ones that display multiple resistance to current therapeutics. There are many promising approaches available for SrtA inhibition, some of which have the potential to advance into further clinical development, with peptidomimetic and in vivo active small molecules being among the most promising. There are currently no approved drugs on the market targeting SrtA, despite its promise, adding to the relevance of this review article, as it extends to the pharmaceutical industry additionally to academic researchers.
Collapse
Affiliation(s)
- Jordi C J Hintzen
- University of Gothenburg, Department of Chemistry and Molecular Biology, Wallenberg Centre for Molecular and Translational Medicine, Kemigården 4, 412 96, Göteborg, Sweden
| | - Helal Abujubara
- University of Gothenburg, Department of Chemistry and Molecular Biology, Wallenberg Centre for Molecular and Translational Medicine, Kemigården 4, 412 96, Göteborg, Sweden
| | - Daniel Tietze
- University of Gothenburg, Department of Chemistry and Molecular Biology, Wallenberg Centre for Molecular and Translational Medicine, Kemigården 4, 412 96, Göteborg, Sweden
| | - Alesia A Tietze
- University of Gothenburg, Department of Chemistry and Molecular Biology, Wallenberg Centre for Molecular and Translational Medicine, Kemigården 4, 412 96, Göteborg, Sweden
| |
Collapse
|
5
|
Savitskii MV, Moskaleva NE, Brito A, Zigangirova NA, Soloveva AV, Sheremet AB, Bondareva NE, Lubenec NL, Kuznetsov RM, Samoylov VM, Tagliaro F, Appolonova SA. Pharmacokinetics, quorum-sensing signal molecules and tryptophan-related metabolomics of the novel anti-virulence drug Fluorothiazinon in a Pseudomonas aeruginosa-induced pneumonia murine model. J Pharm Biomed Anal 2023; 236:115739. [PMID: 37778200 DOI: 10.1016/j.jpba.2023.115739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
Pseudomonas aeruginosa (PA) infection is commonly associated with hospital-acquired infections in patients with immune deficiency and/or severe lung diseases. Managing this bacterium is complex due to drug resistance and high adaptability. Fluorothiazinon (FT) is an anti-virulence drug developed to suppress the virulence of bacteria as opposed to bacterial death increasing host's immune response to infection and improving treatment to inhibit drug resistant bacteria. We aimed to evaluate FT pharmacokinetics, quorum sensing signal molecules profiling and tryptophan-related metabolomics in blood, liver, kidneys, and lungs of mice. Study comprised three groups: a group infected with PA that was treated with 400 mg/kg FT ("infected treated group"); a non-infected group, but also treated with the same single drug dose ("non-infected treated group"); and an infected group that received a vehicle ("infected non-treated group"). PA-mediated infection blood pharmacokinetics profiling was indicative of increased drug concentrations as shown by increased Cmax and AUCs. Tissue distribution in liver, kidneys, and lungs, showed that liver presented the most consistently higher concentrations of FT in the infected versus non-infected mice. FT showed that HHQ levels were decreased at 1 h after dosing in lungs while PQS levels were lower across time in lungs of infected treated mice in comparison to infected non-treated mice. Metabolomics profiling performed in lungs and blood of infected treated versus infected non-treated mice revealed drug-associated metabolite alterations, especially in the kynurenic and indole pathways.
Collapse
Affiliation(s)
- Mark V Savitskii
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
| | - Natalia E Moskaleva
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alex Brito
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Nailya A Zigangirova
- National Research Center for Epidemiology and Microbiology Named after N. F. Gamaleya, Russian Health Ministry, Moscow, Russia
| | - Anna V Soloveva
- National Research Center for Epidemiology and Microbiology Named after N. F. Gamaleya, Russian Health Ministry, Moscow, Russia
| | - Anna B Sheremet
- National Research Center for Epidemiology and Microbiology Named after N. F. Gamaleya, Russian Health Ministry, Moscow, Russia
| | - Natalia E Bondareva
- National Research Center for Epidemiology and Microbiology Named after N. F. Gamaleya, Russian Health Ministry, Moscow, Russia
| | - Nadezhda L Lubenec
- National Research Center for Epidemiology and Microbiology Named after N. F. Gamaleya, Russian Health Ministry, Moscow, Russia
| | - Roman M Kuznetsov
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Viktor M Samoylov
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Franco Tagliaro
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, 37129 Verona, Italy
| | - Svetlana A Appolonova
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
6
|
Murase LS, Perez de Souza JV, Meneguello JE, Palomo CT, Fernandes Herculano Ramos Milaré ÁC, Negri M, Dias Siqueira VL, Demarchi IG, Vieira Teixeira JJ, Cardoso RF. Antibacterial and immunological properties of piperine evidenced by preclinical studies: a systematic review. Future Microbiol 2023; 18:1279-1299. [PMID: 37882762 DOI: 10.2217/fmb-2023-0101] [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: 05/02/2023] [Accepted: 08/23/2023] [Indexed: 10/27/2023] Open
Abstract
Aim: To review in vitro, in vivo, and in silico studies examining the antibacterial and immunomodulatory properties of piperine (PPN). Methods: This systematic review followed PRISMA guidelines, and five databases were searched. Results: A total of 40 articles were included in this study. Six aspects of PPN activity were identified, including antibacterial spectrum, association with antibiotics, efflux pump inhibition, biofilm effects, protein target binding, and modulation of immune functions/virulence factors. Most studies focused on Mycobacterium spp. and Staphylococcus aureus. Cell lineages and in vivo models were employed to study PPN antibacterial effects. Conclusion: We highlight PPN as a potential adjuvant in the treatment of bacterial infections. PPN possesses several antibacterial properties that need further exploration to determine the mechanisms behind its pharmacological activity.
Collapse
Affiliation(s)
- Letícia Sayuri Murase
- Postgraduate Program in Health Sciences, State University of Maringa, Maringá, Paraná, 87020-900, Brazil
| | - João Vítor Perez de Souza
- Postgraduate Program in Biosciences and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Jean Eduardo Meneguello
- Postgraduate Program in Biosciences and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Carolina Trevisolli Palomo
- Postgraduate Program in Health Sciences, State University of Maringa, Maringá, Paraná, 87020-900, Brazil
| | | | - Melyssa Negri
- Postgraduate Program in Health Sciences, State University of Maringa, Maringá, Paraná, 87020-900, Brazil
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Vera Lúcia Dias Siqueira
- Postgraduate Program in Biosciences and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Izabel Galhardo Demarchi
- Department of Clinical Analysis, Federal University of Santa Catarina, Florianopólis, Santa Catarina, 88040-900, Brazil
| | - Jorge Juarez Vieira Teixeira
- Postgraduate Program in Health Sciences, State University of Maringa, Maringá, Paraná, 87020-900, Brazil
- Postgraduate Program in Biosciences and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Rosilene Fressatti Cardoso
- Postgraduate Program in Health Sciences, State University of Maringa, Maringá, Paraná, 87020-900, Brazil
- Postgraduate Program in Biosciences and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| |
Collapse
|
7
|
Stelitano G, Cocorullo M, Mori M, Villa S, Meneghetti F, Chiarelli LR. Iron Acquisition and Metabolism as a Promising Target for Antimicrobials (Bottlenecks and Opportunities): Where Do We Stand? Int J Mol Sci 2023; 24:ijms24076181. [PMID: 37047161 PMCID: PMC10094389 DOI: 10.3390/ijms24076181] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
The emergence of multidrug-resistant (MDR) and extensively drug-resistant (XDR) infections is one of the most crucial challenges currently faced by the scientific community. Developments in the fundamental understanding of their underlying mechanisms may open new perspectives in drug discovery. In this review, we conducted a systematic literature search in PubMed, Web of Science, and Scopus, to collect information on innovative strategies to hinder iron acquisition in bacteria. In detail, we discussed the most interesting targets from iron uptake and metabolism pathways, and examined the main chemical entities that exhibit anti-infective activities by interfering with their function. The mechanism of action of each drug candidate was also reviewed, together with its pharmacodynamic, pharmacokinetic, and toxicological properties. The comprehensive knowledge of such an impactful area of research will hopefully reflect in the discovery of newer antibiotics able to effectively tackle the antimicrobial resistance issue.
Collapse
|
8
|
Recchia D, Stelitano G, Stamilla A, Gutierrez DL, Degiacomi G, Chiarelli LR, Pasca MR. Mycobacterium abscessus Infections in Cystic Fibrosis Individuals: A Review on Therapeutic Options. Int J Mol Sci 2023; 24:ijms24054635. [PMID: 36902066 PMCID: PMC10002592 DOI: 10.3390/ijms24054635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/02/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Mycobacterium abscessus is an opportunistic pathogen that mainly colonizes and infects cystic fibrosis patients' lungs. M. abscessus is naturally resistant to many antibiotics such as rifamycin, tetracyclines and β-lactams. The current therapeutic regimens are not very effective and are mostly based on repurposed drugs used against Mycobacterium tuberculosis infections. Thus, new approaches and novel strategies are urgently needed. This review aims to provide an overview of the latest ongoing findings to fight M. abscessus infections by analyzing emerging and alternative treatments, novel drug delivery strategies, and innovative molecules.
Collapse
|
9
|
Bai YB, Yang XR, Li B, Zhou XZ, Wang WW, Cheng FS, Zhang JY. Virtual Screening and In Vitro Experimental Verification of LuxS Inhibitors for Escherichia coli O157:H7. Microbiol Spectr 2023; 11:e0350222. [PMID: 36809060 PMCID: PMC10100900 DOI: 10.1128/spectrum.03502-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/02/2023] [Indexed: 02/23/2023] Open
Abstract
Enterohemorrhagic Escherichia coli O157:H7 is an important foodborne pathogen that forms biofilms. In this study, three quorum-sensing (QS) inhibitors (M414-3326, 3254-3286, and L413-0180) were obtained through virtual screening, and their in vitro antibiofilm activities were validated. Briefly, the three-dimensional structure model of LuxS was constructed and characterized using the SWISS-MODEL. High-affinity inhibitors were screened from the ChemDiv database (1,535,478 compounds) using LuxS as a ligand. Five compounds (L449-1159, L368-0079, M414-3326, 3254-3286, and L413-0180) with a good inhibitory effect (50% inhibitory concentration <10 μM) on type II QS signal molecule autoinducer-2 (AI-2) were obtained using a AI-2 bioluminescence assay. The absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties predicated that the five compounds had high intestinal absorption levels (high) and plasma protein binding (absorbent strong) and did not inhibit the metabolism of CYP2D6 metabolic enzymes. In addition, molecular dynamics simulation showed that compounds L449-1159 and L368-0079 could not stably bind with LuxS. Thus, these compounds were excluded. Furthermore, surface plasmon resonance results showed that the three compounds could specifically bind to LuxS. IN addition, the three compounds could effectively inhibit the biofilm formation without affecting the growth and metabolism of the bacteria. Finally, the reverse transcription-quantitative PCR results showed that the three compounds downregulated the expression of the LuxS gene. Overall, these results revealed that the three compounds obtained through virtual screening could inhibit biofilm formation of E. coli O157:H7 and are potential LuxS inhibitors that can be used to treat E. coli O157:H7 infections. IMPORTANCE E. coli O157:H7 is a foodborne pathogen of public health importance. Quorum sensing (QS) is a form of bacterial communication that can regulate various group behaviors, including biofilm formation. Here, we identified three QS AI-2 inhibitors (M414-3326, 3254-3286, and L413-0180) that can stably and specifically bind to LuxS protein. The three QS AI-2 inhibitors inhibited biofilm formation without affecting the growth and metabolic activity of E. coli O157:H7. The three QS AI-2 inhibitors are promising agents for treating E. coli O157:H7 infections. Further studies to identify the mechanism of the three QS AI-2 inhibitors are needed to develop new drugs to overcome antibiotic resistance.
Collapse
Affiliation(s)
- Yu-Bin Bai
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, Gansu, People’s Republic of China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, Gansu, People’s Republic of China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Xiao-Rong Yang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, Gansu, People’s Republic of China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, Gansu, People’s Republic of China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Bing Li
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, Gansu, People’s Republic of China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, Gansu, People’s Republic of China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Xu-Zheng Zhou
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, Gansu, People’s Republic of China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, Gansu, People’s Republic of China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Wei-Wei Wang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, Gansu, People’s Republic of China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, Gansu, People’s Republic of China
| | - Fu-Sheng Cheng
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, Gansu, People’s Republic of China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, Gansu, People’s Republic of China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Ji-Yu Zhang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, Gansu, People’s Republic of China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, Gansu, People’s Republic of China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| |
Collapse
|
10
|
Targeting Siderophore-Mediated Iron Uptake in M. abscessus: A New Strategy to Limit the Virulence of Non-Tuberculous Mycobacteria. Pharmaceutics 2023; 15:pharmaceutics15020502. [PMID: 36839823 PMCID: PMC9966845 DOI: 10.3390/pharmaceutics15020502] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Targeting pathogenic mechanisms, rather than essential processes, represents a very attractive approach for the development of new antimycobacterial drugs. In this context, iron acquisition routes have recently emerged as potentially druggable pathways. However, the importance of siderophore biosynthesis in the virulence and pathogenicity of M. abscessus (Mab) is still poorly understood. In this study, we investigated the Salicylate Synthase (SaS) of Mab as an innovative molecular target for the development of inhibitors of siderophore production. Notably, Mab-SaS does not have any counterpart in human cells, making it an interesting candidate for drug discovery. Starting from the analysis of the binding of a series of furan-based derivatives, previously identified by our group as inhibitors of MbtI from M. tuberculosis (Mtb), we successfully selected the lead compound 1, exhibiting a strong activity against Mab-SaS (IC50 ≈ 5 µM). Computational studies characterized the key interactions between 1 and the enzyme, highlighting the important roles of Y387, G421, and K207, the latter being one of the residues involved in the first step of the catalytic reaction. These results support the hypothesis that 5-phenylfuran-2-carboxylic acids are also a promising class of Mab-SaS inhibitors, paving the way for the optimization and rational design of more potent derivatives.
Collapse
|
11
|
Poyraz S, Döndaş HA, Sansano JM, Belveren S, Yamali C, Ülger M, Döndaş NY, Sağlık BN, Pask CM. N-Benzoylthiourea-pyrrolidine carboxylic acid derivatives bearing an imidazole moiety: Synthesis, characterization, crystal structure, in vitro ChEs inhibition, and antituberculosis, antibacterial, antifungal studies. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
12
|
Zhang W, Gong Q, Tang Z, Ma X, Wang Z, Guan J, Wang L, Zhao Y, Yan M. The natural product, echinatin, protects mice from methicillin-resistant Staphylococcus aureus pneumonia by inhibition of alpha-hemolysin expression. Front Microbiol 2023; 14:1128144. [PMID: 37125192 PMCID: PMC10140358 DOI: 10.3389/fmicb.2023.1128144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/06/2023] [Indexed: 05/02/2023] Open
Abstract
Antimicrobial resistance (AMR) is a global, multifaceted crisis that poses significant challenges to the successful eradication of devastating pathogens, particularly methicillin-resistant Staphylococcus aureus (MRSA), a persistent superbug that causes devastating infections. The scarcity of new antibacterial drugs is obvious, and antivirulence strategies that reduce the pathogenicity of bacteria by weakening their virulence have become the subject of intense investigation. Alpha-hemolysin (Hla), a cytolytic pore-forming toxin, has a pivotal role in S. aureus pathogenesis. Here, we demonstrated that echinatin, a natural compound isolated from licorice, effectively inhibited the hemolytic activity of MRSA at 32 μg/mL. In addition, echinatin did not interfere with bacterial growth and had no significant cytotoxicity at the inhibitory concentration of S. aureus hemolysis. Heptamer formation tightly correlated with Hla-mediated cell invasion, whereas echinatin did not affect deoxycholic acid-induced oligomerization of Hla. Echinatin affected hemolytic activity through indirect binding to Hla as confirmed by the neutralization assay and cellular thermal shift assay (CETSA). Furthermore, qRT-PCR and western blot analyses revealed that echinatin suppressed Hla expression at both the mRNA and protein levels as well as the transcript levels of Agr quorum-sensing system-related genes. Additionally, when echinatin was added to a coculture system of A549 cells and S. aureus, it significantly reduced cell damage. Importantly, echinatin exhibited a significant therapeutic effect in an MRSA-induced mouse pneumonia model. In conclusion, the present findings demonstrated that echinatin significantly inhibits the hemolysin effect and may be a potential candidate compound for combating drug-resistant MRSA infections.
Collapse
Affiliation(s)
- Wei Zhang
- The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Qing Gong
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Zhitong Tang
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xin Ma
- Jilin Provincial Cancer Hospital, Changchun, China
| | - Zhuoer Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Jiyu Guan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Li Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Yicheng Zhao
- Changchun University of Chinese Medicine, Changchun, China
| | - Ming Yan
- The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Ming Yan,
| |
Collapse
|
13
|
5-(4-Nitrophenyl)furan-2-carboxylic Acid. MOLBANK 2022. [DOI: 10.3390/m1515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The ever-evolving research in the field of antitubercular agents has led to the identification of several new potential drug classes. Among them, 5-phenyl-furan-2-carboxylic acids have emerged as innovative potential therapeutics, targeting iron acquisition in mycobacterial species. In our efforts to characterize the molecular interactions between these compounds and their protein target (MbtI from M. tuberculosis) by means of co-crystallization experiments, we unexpectedly obtained the structure of 5-(4-nitrophenyl)furan-2-carboxylic acid (1). Herein, we describe the preparation of the compound and its analysis by 1H NMR, 13C NMR, HRMS, and SC-XRD.
Collapse
|
14
|
Methyl 5-(2-Fluoro-4-nitrophenyl)furan-2-carboxylate. MOLBANK 2022. [DOI: 10.3390/m1492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
5-Phenyl-furan-2-carboxylic acids have emerged as a new, promising class of antimycobacterial agents that have the ability to interfere with iron homeostasis. Considering the lack of structural data on these compounds, we analyzed the crystal of a fluorinated ester derivative of 5-(4-nitrophenyl)furan-2-carboxylic acid, one of the most potent candidates in the series. Here, we describe the preparation of methyl 5-(2-fluoro-4-nitrophenyl)furan-2-carboxylate (1) and its analysis by 1H-NMR, 13C-NMR, HRMS, and SC-XRD.
Collapse
|
15
|
Bai YB, Shi MY, Wang WW, Wu LY, Bai YT, Li B, Zhou XZ, Zhang JY. Novel quorum sensing inhibitor Echinatin as an antibacterial synergist against Escherichia coli. Front Microbiol 2022; 13:1003692. [PMID: 36386683 PMCID: PMC9663819 DOI: 10.3389/fmicb.2022.1003692] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/14/2022] [Indexed: 12/05/2022] Open
Abstract
A new antibacterial strategy based on inhibiting bacterial quorum sensing (QS) has emerged as a promising method of attenuating bacterial pathogenicity and preventing bacterial resistance to antibiotics. In this study, we screened Echinatin (Ech) with high-efficiency anti-QS from 13 flavonoids through the AI-2 bioluminescence assay. Additionally, crystal violet (CV) staining combined with confocal laser scanning microscopy (CLSM) was used to evaluate the effect of anti-biofilm against Escherichia coli (E. coli). Further, the antibacterial synergistic effect of Ech and marketed antibiotics were measured by broth dilution and Alamar Blue Assay. It was found that Ech interfered with the phenotype of QS, including biofilm formation, exopolysaccharide (EPS) production, and motility, without affecting bacterial growth and metabolic activity. Moreover, qRT-PCR exhibited that Ech significantly reduced the expression of QS-regulated genes (luxS, pfs, lsrB, lsrK, lsrR, flhC, flhD, fliC, csgD, and stx2). More important, Ech with currently marketed colistin antibiotics (including colistin B and colistin E) showed significantly synergistically increased antibacterial activity in overcoming antibiotic resistance of E. coli. In summary, these results suggested the potent anti-QS and novel antibacterial synergist candidate of Ech for treating E. coli infections.
Collapse
Affiliation(s)
- Yu-Bin Bai
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, China,Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Meng-Yan Shi
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, China,Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wei-Wei Wang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, China,Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Ling-Yu Wu
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, China,Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yu-Ting Bai
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, China,Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Bing Li
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, China,Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xu-Zheng Zhou
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, China,Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Ji-Yu Zhang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou, China,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, China,Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China,*Correspondence: Ji-Yu Zhang,
| |
Collapse
|
16
|
Novel Antibiofilm Inhibitor Ginkgetin as an Antibacterial Synergist against Escherichia coli. Int J Mol Sci 2022; 23:ijms23158809. [PMID: 35955943 PMCID: PMC9369100 DOI: 10.3390/ijms23158809] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 11/27/2022] Open
Abstract
As an opportunistic pathogen, Escherichia coli (E. coli) forms biofilm that increases the virulence of bacteria and antibiotic resistance, posing a serious threat to human and animal health. Recently, ginkgetin (Gin) has been discovered to have antiinflammatory, antioxidant, and antitumor properties. In the present study, we evaluated the antibiofilm and antibacterial synergist of Gin against E. coli. Additionally, Alamar Blue assay combined with confocal laser scanning microscope (CLSM) and crystal violet (CV) staining was used to evaluate the effect of antibiofilm and antibacterial synergist against E. coli. Results showed that Gin reduces biofilm formation, exopolysaccharide (EPS) production, and motility against E. coli without limiting its growth and metabolic activity. Furthermore, we identified the inhibitory effect of Gin on AI-2 signaling molecule production, which showed apparent anti-quorum sensing (QS) properties. The qRT-PCR also indicated that Gin reduced the transcription of curli-related genes (csgA, csgD), flagella-formation genes (flhC, flhD, fliC, fliM), and QS-related genes (luxS, lsrB, lsrK, lsrR). Moreover, Gin showed obvious antibacterial synergism to overcome antibiotic resistance in E. coli with marketed antibiotics, including gentamicin, colistin B, and colistin E. These results suggested the potent antibiofilm and novel antibacterial synergist effect of Gin for treating E. coli infections.
Collapse
|
17
|
Gao P, Wei Y, Tai SSC, Halebeedu Prakash P, Iu HTV, Li Y, Yam HCB, Chen JHK, Ho PL, Davies J, Kao RYT. Antivirulence Agent as an Adjuvant of β-Lactam Antibiotics in Treating Staphylococcal Infections. Antibiotics (Basel) 2022; 11:antibiotics11060819. [PMID: 35740225 PMCID: PMC9219823 DOI: 10.3390/antibiotics11060819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 11/27/2022] Open
Abstract
Staphylococcus aureus can cause a plethora of life-threatening infections. Antibiotics have been extensively used to treat S. aureus infections. However, when antibiotics are used at sub-inhibitory concentrations, especially for β-lactam antibiotics, they may enhance staphylococcal pathogenicity and exacerbate the infection. The combination of antivirulence agents and antibiotics may be a novel approach to controlling antibiotic-induced S. aureus pathogenicity. We have illustrated that under in vitro conditions, antivirulence agent M21, when administered concurrently with ampicillin, suppressed the expression and production of virulence factors induced by ampicillin. In a mouse peritonitis model, M21 reduced bacterial load irrespective of administration of ampicillin. In a bacteremia model, combinatorial treatment consisting of ampicillin or ceftazidime and M21 increased the survival rate of mice and reduced cytokine abundance, suggesting the suppression of antibiotic-induced virulence by M21. Different from traditional antibiotic adjuvants, an antivirulence agent may not synergistically inhibit bacterial growth in vitro, but effectively benefit the host in vivo. Collectively, our findings from this study demonstrated the benefits of antivirulence–antibiotic combinatorial treatment against S. aureus infections and provide a new perspective on the development of antibiotic adjuvants.
Collapse
Affiliation(s)
- Peng Gao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
- Correspondence: (P.G.); (R.Y.T.K.)
| | - Yuanxin Wei
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
| | - Sherlock Shing Chiu Tai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
| | - Pradeep Halebeedu Prakash
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
| | - Ho Ting Venice Iu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
| | - Yongli Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
| | - Hin Cheung Bill Yam
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
| | - Jonathan Hon Kwan Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
- Department of Microbiology, Queen Mary Hospital, Hong Kong, China
| | - Pak Leung Ho
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
- Department of Microbiology, Queen Mary Hospital, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases and the Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, China
| | - Julian Davies
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| | - Richard Yi Tsun Kao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Y.W.); (S.S.C.T.); (P.H.P.); (H.T.V.I.); (Y.L.); (H.C.B.Y.); (J.H.K.C.); (P.L.H.)
- State Key Laboratory of Emerging Infectious Diseases and the Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Correspondence: (P.G.); (R.Y.T.K.)
| |
Collapse
|
18
|
Bové M, Coenye T. The anti-virulence activity of the non-mevalonate pathway inhibitor FR900098 towards Burkholderia cenocepacia is maintained during experimental evolution. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 35358034 DOI: 10.1099/mic.0.001170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Burkholderia cenocepacia infections are difficult to treat and there is an urgent need for alternative (combination) treatments. The use of anti-virulence therapies in combination with antibiotics is a possible strategy to increase the antimicrobial susceptibility of the pathogen and to slow down the development of resistance. In the present study we evaluated the β-lactam and colistin-potentiating activity, and anti-virulence effect of the non-mevalonate pathway inhibitor FR900098 against B. cenocepacia in various in vitro and in vivo models. In addition, we evaluated whether repeated exposure to FR900098 alone or when combined with ceftazidime leads to increased resistance. FR900098 potentiated the activity of colistin and several β-lactam antibiotics (aztreonam, cefepime, cefotaxime, ceftazidime, mecillinam and piperacillin) but not of imipenem and meropenem. When used alone or in combination with ceftazidime, FR900098 increased the survival of infected Galleria mellonella and Caenorhabditis elegans. Furthermore, combining ceftazidime with FR900098 resulted in a significant inhibition of the biofilm formation of B. cenocepacia. Repeated exposure to FR900098 in the C. elegans infection model did not lead to decreased activity, and the susceptibility of the evolved B. cenocepacia HI2424 lineages to ceftazidime, FR900098 and the combination of both remained unchanged. In conclusion, FR900098 reduces B. cenocepacia virulence and potentiates ceftazidime in an in vivo C. elegans model, and this activity is not lost during the experimental evolution experiment carried out in the present study.
Collapse
Affiliation(s)
- Mona Bové
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| |
Collapse
|
19
|
Wolfmeier H, Wardell SJT, Liu LT, Falsafi R, Draeger A, Babiychuk EB, Pletzer D, Hancock REW. Targeting the Pseudomonas aeruginosa Virulence Factor Phospholipase C With Engineered Liposomes. Front Microbiol 2022; 13:867449. [PMID: 35369481 PMCID: PMC8971843 DOI: 10.3389/fmicb.2022.867449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Engineered liposomes composed of the naturally occurring lipids sphingomyelin (Sm) and cholesterol (Ch) have been demonstrated to efficiently neutralize toxins secreted by Gram-positive bacteria such as Streptococcus pneumoniae and Staphylococcus aureus. Here, we hypothesized that liposomes are capable of neutralizing cytolytic virulence factors secreted by the Gram-negative pathogen Pseudomonas aeruginosa. We used the highly virulent cystic fibrosis P. aeruginosa Liverpool Epidemic Strain LESB58 and showed that sphingomyelin (Sm) and a combination of sphingomyelin with cholesterol (Ch:Sm; 66 mol/% Ch and 34 mol/% Sm) liposomes reduced lysis of human bronchial and red blood cells upon challenge with the Pseudomonas secretome. Mass spectrometry of liposome-sequestered Pseudomonas proteins identified the virulence-promoting hemolytic phospholipase C (PlcH) as having been neutralized. Pseudomonas aeruginosa supernatants incubated with liposomes demonstrated reduced PlcH activity as assessed by the p-nitrophenylphosphorylcholine (NPPC) assay. Testing the in vivo efficacy of the liposomes in a murine cutaneous abscess model revealed that Sm and Ch:Sm, as single dose treatments, attenuated abscesses by >30%, demonstrating a similar effect to that of a mutant lacking plcH in this infection model. Thus, sphingomyelin-containing liposome therapy offers an interesting approach to treat and reduce virulence of complex infections caused by P. aeruginosa and potentially other Gram-negative pathogens expressing PlcH.
Collapse
Affiliation(s)
- Heidi Wolfmeier
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Salzburg, Austria
| | - Samuel J. T. Wardell
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Leo T. Liu
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Reza Falsafi
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | | | | | - Daniel Pletzer
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- *Correspondence: Daniel Pletzer,
| | - Robert E. W. Hancock
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
- Robert E. W. Hancock,
| |
Collapse
|
20
|
Scutellarin potentiates vancomycin against lethal pneumonia caused by methicillin-resistant Staphylococcus aureus through dual inhibition of sortase A and caseinolytic peptidase P. Biochem Pharmacol 2022; 199:114982. [PMID: 35247333 DOI: 10.1016/j.bcp.2022.114982] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/12/2022] [Accepted: 02/22/2022] [Indexed: 01/09/2023]
Abstract
The strategy of targeting virulence factor has received great attention as it barely develops bacterial resistance. Sortase A (SrtA) and caseinolytic peptidase P (ClpP), as important virulence factors, are considered to be ideal pharmacological targets for methicillin-resistant Staphylococcus aureus (MRSA) infection. Through screening hundreds of compounds, we found scutellarin, a natural flavonoid, markedly inhibited SrtA and ClpP activities of MRSA strain USA300 with an IC50 of 53.64 μg/mL and 107.00 μg/mL, respectively. Subsequently, we observed that scutellarin could inhibit the SrtA-related virulence of MRSA. To demonstrate whether scutellarin directly binding to SrtA, fluorescence quenching assay and molecular docking were performed and the results indicated that scutellarin directly bonded to SrtA molecule with a KA value of 7.58 × 104 L/mol. In addition to direct SrtA inhibition, scutellarin could also inhibit hemolytic activity of S. aureus by inhibiting the expression of Hla in a SrtA-independent manner. Further assays confirmed that scutellarin inhibited hemolysis by inhibiting ClpP. The combination of scutellarin and vancomycin showed enhancing inhibition of USA300 in vitro and in vivo, evidenced by decreased MIC from 3 μg/mL to 0.5 μg/mL and increased survival and improvement of lung pathology in pneumonia mice. Taken together, these results suggest that scutellarin exhibited di-inhibitory effects on SrtA and ClpP of USA300. The di-inhibition of virulence factors by scutellarin combined with vancomycin to prevent MRSA invasion of A549 cells and pneumonia in mice, indicating that scutellarin is expected to be a potential adjuvant against MRSA in the future.
Collapse
|
21
|
Murugaiyan J, Kumar PA, Rao GS, Iskandar K, Hawser S, Hays JP, Mohsen Y, Adukkadukkam S, Awuah WA, Jose RAM, Sylvia N, Nansubuga EP, Tilocca B, Roncada P, Roson-Calero N, Moreno-Morales J, Amin R, Kumar BK, Kumar A, Toufik AR, Zaw TN, Akinwotu OO, Satyaseela MP, van Dongen MBM. Progress in Alternative Strategies to Combat Antimicrobial Resistance: Focus on Antibiotics. Antibiotics (Basel) 2022; 11:200. [PMID: 35203804 PMCID: PMC8868457 DOI: 10.3390/antibiotics11020200] [Citation(s) in RCA: 181] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/24/2022] Open
Abstract
Antibiotic resistance, and, in a broader perspective, antimicrobial resistance (AMR), continues to evolve and spread beyond all boundaries. As a result, infectious diseases have become more challenging or even impossible to treat, leading to an increase in morbidity and mortality. Despite the failure of conventional, traditional antimicrobial therapy, in the past two decades, no novel class of antibiotics has been introduced. Consequently, several novel alternative strategies to combat these (multi-) drug-resistant infectious microorganisms have been identified. The purpose of this review is to gather and consider the strategies that are being applied or proposed as potential alternatives to traditional antibiotics. These strategies include combination therapy, techniques that target the enzymes or proteins responsible for antimicrobial resistance, resistant bacteria, drug delivery systems, physicochemical methods, and unconventional techniques, including the CRISPR-Cas system. These alternative strategies may have the potential to change the treatment of multi-drug-resistant pathogens in human clinical settings.
Collapse
Affiliation(s)
- Jayaseelan Murugaiyan
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - P. Anand Kumar
- Department of Veterinary Microbiology, NTR College of Veterinary Science, Sri Venkateswara Veterinary University, Gannavaram 521102, India;
| | - G. Srinivasa Rao
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati 517502, India;
| | - Katia Iskandar
- Department of Mathématiques Informatique et Télécommunications, Université Toulouse III, Paul Sabatier, INSERM, UMR 1295, 31000 Toulouse, France;
- INSPECT-LB: Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573, Lebanon
- Faculty of Pharmacy, Lebanese University, Beirut 6573, Lebanon
| | | | - John P. Hays
- Department of Medical Microbiology, Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), 3015 GD Rotterdam, The Netherlands;
| | - Yara Mohsen
- Department of Epidemiology, High Institute of Public Health, Alexandria University, Alexandria 21544, Egypt;
- Infectious Disease Clinical Pharmacist, Antimicrobial Stewardship Department, International Medical Center Hospital, Cairo 11511, Egypt
| | - Saranya Adukkadukkam
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - Wireko Andrew Awuah
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Ruiz Alvarez Maria Jose
- Research Coordination and Support Service, National Institute of Health (ISS) Viale Regina -Elena, 299, 00161 Rome, Italy;
| | - Nanono Sylvia
- Infectious Diseases Institute (IDI), College of Health Sciences, Makerere University, Kampala 7072, Uganda;
| | | | - Bruno Tilocca
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Paola Roncada
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Natalia Roson-Calero
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Javier Moreno-Morales
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Rohul Amin
- James P Grant School of Public Health, BRAC University, Dhaka 1212, Bangladesh;
| | - Ballamoole Krishna Kumar
- Nitte (Deemed to be University), Division of Infectious Diseases, Nitte University Centre for Science Education and Research, Deralakatte, Mangalore 575018, India;
| | - Abishek Kumar
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Abdul-Rahman Toufik
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Thaint Nadi Zaw
- Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK;
| | - Oluwatosin O. Akinwotu
- Department of Microbiology and Biotechnology Centre, Maharaja Sayajirao University of Baroda, Vadodara 390002, India;
- Environmental and Biotechnology Unit, Department of Microbiology, University of Ibadan, 200132 Ibadan, Nigeria
| | | | | |
Collapse
|
22
|
Preventative treatment with Fluorothiazinon suppressed Acinetobacter baumannii-associated septicemia in mice. J Antibiot (Tokyo) 2022; 75:155-163. [PMID: 35064243 PMCID: PMC8777177 DOI: 10.1038/s41429-022-00504-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/07/2021] [Accepted: 11/10/2021] [Indexed: 02/01/2023]
Abstract
The high prevalence of multidrug-resistant Acinetobacter baumannii has emerged as a serious problem in the treatment of nosocomial infections in the past three decades. Recently, we developed a new small-molecule inhibitor belonging to a class of 2,4-disubstituted-4H-[1,3,4]-thiadiazine-5-ones, Fluorothiazinon (FT, previously called CL-55). FT effectively suppressed the T3SS of Chlamydia spp., Pseudomonas aeruginosa, and Salmonella sp. without affecting bacterial growth in vitro. In this study, we describe that prophylactic use of FT for 4 days prior to challenge with resistant clinical isolates of A. baumannii (ABT-897-17 and 52TS19) suppressed septic infection in mice, resulting in improved survival, limited bacteraemia and decreased bacterial load in the organs of the mice. We show that FT had an inhibitory effect on A. baumannii biofilm formation in vitro and, to a greater extent, on biofilm maturation. In addition, FT inhibited Acinetobacter isolate-induced death of HeLa cells, which morphologically manifested as apoptosis. The mechanism of FT action on A. baumannii is currently being studied. FT may be a promising candidate for the development of a broad-spectrum anti-virulence drug to use in the prevention of nosocomial infections.
Collapse
|
23
|
Cazzaniga G, Mori M, Chiarelli LR, Gelain A, Meneghetti F, Villa S. Natural products against key Mycobacterium tuberculosis enzymatic targets: Emerging opportunities for drug discovery. Eur J Med Chem 2021; 224:113732. [PMID: 34399099 DOI: 10.1016/j.ejmech.2021.113732] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/15/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022]
Abstract
For centuries, natural products (NPs) have served as powerful therapeutics against a variety of human ailments. Nowadays, they still represent invaluable resources for the treatment of many diseases, including bacterial infections. After nearly three decades since the World Health Organization's (WHO) declaration of tuberculosis (TB) as a global health emergency, Mycobacterium tuberculosis (Mtb) continues to claim millions of lives, remaining among the leading causes of death worldwide. In the last years, several efforts have been devoted to shortening and improving treatment outcomes, and to overcoming the increasing resistance phenomenon. Nature has always provided a virtually unlimited source of bioactive molecules, which have inspired the development of new drugs. NPs are characterized by an exceptional chemical and structural diversity, the result of millennia of evolutionary responses to various stimuli. Thanks to their favorable structural features and their enzymatic origin, they are naturally prone to bind proteins and exhibit bioactivities. Furthermore, their worldwide distribution and ease of accessibility has contributed to promote investigations on their activity. Overall, these characteristics make NPs excellent models for the design of novel therapeutics. This review offers a critical and comprehensive overview of the most promising NPs, isolated from plants, fungi, marine species, and bacteria, endowed with inhibitory properties against traditional and emerging mycobacterial enzymatic targets. A selection of 86 compounds is here discussed, with a special emphasis on their biological activity, structure-activity relationships, and mechanism of action. Our study corroborates the antimycobacterial potential of NPs, substantiating their relevance in future drug discovery and development efforts.
Collapse
Affiliation(s)
- Giulia Cazzaniga
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy
| | - Matteo Mori
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy
| | - Laurent Roberto Chiarelli
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, via A. Ferrata 9, 27100, Pavia, Italy
| | - Arianna Gelain
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy
| | - Fiorella Meneghetti
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy.
| | - Stefania Villa
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy
| |
Collapse
|
24
|
Wang L, Wang G, Qu H, Wang K, Jing S, Guan S, Su L, Li Q, Wang D. Taxifolin, an Inhibitor of Sortase A, Interferes With the Adhesion of Methicillin-Resistant Staphylococcal aureus. Front Microbiol 2021; 12:686864. [PMID: 34295320 PMCID: PMC8290497 DOI: 10.3389/fmicb.2021.686864] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/14/2021] [Indexed: 11/30/2022] Open
Abstract
The evolution and spread of methicillin-resistant Staphylococcus aureus (MRSA) poses a significant hidden risk to human public health. The majority of antibiotics used clinically have become mostly ineffective, and so the development of novel anti-infection strategies is urgently required. Since Staphylococcus aureus (S. aureus) cysteine transpeptidase sortase A (SrtA) mediates the surface-anchoring of proteins to its surface, compounds that inhibit SrtA are considered potential antivirulence treatments. Herein, we report on the efficacy of the potent SrtA inhibitor taxifolin (Tax), a flavonoid compound isolated from Chinese herbs. It was able to reversibly block the activity of SrtA with an IC50 of 24.53 ± 0.42 μM. Tax did not display toxicity toward mammalian cells or S. aureus at a concentration of 200 μM. In addition, Tax attenuated the virulence-related phenotype of SrtA in vitro by decreasing the adherence of S. aureus, reducing the formation of a biofilm, and anchoring of S. aureus protein A on its cell wall. The mechanism of the SrtA-Tax interaction was determined using a localized surface plasmon resonance assay. Subsequent mechanistic studies confirmed that Asp-170 and Gln-172 were the principal sites on SrtA with which it binds to Tax. Importantly, in vivo experiments demonstrated that Tax protects mice against pneumonia induced by lethal doses of MRSA, significantly improving their survival rate and reducing the number of viable S. aureus in the lung tissue. The present study indicates that Tax is a useful pioneer compound for the development of novel agents against S. aureus infections.
Collapse
Affiliation(s)
- Li Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Guangming Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Han Qu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Kai Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Shisong Jing
- College of Animal Science, Jilin University, Changchun, China
| | - Shuhan Guan
- College of Animal Science, Jilin University, Changchun, China
| | - Liyan Su
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qianxue Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Dacheng Wang
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
25
|
Pylkkö T, Ilina P, Tammela P. Development and validation of a high-content screening assay for inhibitors of enteropathogenic E. coli adhesion. J Microbiol Methods 2021; 184:106201. [PMID: 33713725 DOI: 10.1016/j.mimet.2021.106201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 11/25/2022]
Abstract
Enteropathogenic E. coli (EPEC) causes intestinal infections leading to severe diarrhea. EPEC attaches to the host cell causing lesions to the intestinal epithelium coupled with the effacement of microvilli. In the process, actin accumulates into a pedestal-like structure under bacterial microcolonies. We designed an automated fluorescence microscopy-based screening method for discovering compounds capable of inhibiting EPEC adhesion and virulence using aurodox, a type three secretion system (T3SS) inhibitor, as a positive control. The screening assay employs an EPEC strain (2348/69) expressing a fluorescent protein and actin staining for monitoring the bacteria and their pedestals respectively, analyzing these with a custom image analysis pipeline. The assay allows for the discovery of compounds capable of preventing the formation of pathogenic actin rearrangements. These compounds may be interfering with virulence-related molecular pathways relevant for developing antivirulence leads.
Collapse
Affiliation(s)
- Tuomas Pylkkö
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, P.O. Box 56, FI-00014, University of Helsinki, Finland
| | - Polina Ilina
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, P.O. Box 56, FI-00014, University of Helsinki, Finland
| | - Päivi Tammela
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, P.O. Box 56, FI-00014, University of Helsinki, Finland.
| |
Collapse
|
26
|
Krzyżek P, Migdał P, Paluch E, Karwańska M, Wieliczko A, Gościniak G. Myricetin as an Antivirulence Compound Interfering with a Morphological Transformation into Coccoid Forms and Potentiating Activity of Antibiotics against Helicobacter pylori. Int J Mol Sci 2021; 22:ijms22052695. [PMID: 33800082 PMCID: PMC7962197 DOI: 10.3390/ijms22052695] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 02/28/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori, a gastric pathogen associated with a broad range of stomach diseases, has a high tendency to become resistant to antibiotics. One of the most important factors related to therapeutic failures is its ability to change from a spiral to a coccoid form. Therefore, the main aim of our original article was to determine the influence of myricetin, a natural compound with an antivirulence action, on the morphological transformation of H. pylori and check the potential of myricetin to increase the activity of antibiotics against this pathogen. We observed that sub-minimal inhibitory concentrations (sub-MICs) of this compound have the ability to slow down the process of transformation into coccoid forms and reduce biofilm formation of this bacterium. Using checkerboard assays, we noticed that the exposure of H. pylori to sub-MICs of myricetin enabled a 4–16-fold reduction in MICs of all classically used antibiotics (amoxicillin, clarithromycin, tetracycline, metronidazole, and levofloxacin). Additionally, RT-qPCR studies of genes related to the H. pylori morphogenesis showed a decrease in their expression during exposure to myricetin. This inhibitory effect was more strongly seen for genes involved in the muropeptide monomers shortening (csd3, csd6, csd4, and amiA), suggesting their significant participation in the spiral-to-coccoid transition. To our knowledge, this is the first research showing the ability of any compound to synergistically interact with all five antibiotics against H. pylori and the first one showing the capacity of a natural substance to interfere with the morphological transition of H. pylori from spiral to coccoid forms.
Collapse
Affiliation(s)
- Paweł Krzyżek
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (E.P.); (G.G.)
- Correspondence:
| | - Paweł Migdał
- Department of Environment, Hygiene and Animal Welfare, Wroclaw University of Environmental and Life Sciences, 51-630 Wroclaw, Poland;
| | - Emil Paluch
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (E.P.); (G.G.)
| | - Magdalena Karwańska
- Department of Epizootiology and Veterinary Administration with Clinic of Infectious Diseases, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, 50-366 Wroclaw, Poland; (M.K.); (A.W.)
| | - Alina Wieliczko
- Department of Epizootiology and Veterinary Administration with Clinic of Infectious Diseases, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, 50-366 Wroclaw, Poland; (M.K.); (A.W.)
| | - Grażyna Gościniak
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (E.P.); (G.G.)
| |
Collapse
|
27
|
Yang Q, Zou P, Cao Z, Wang Q, Fu S, Xie G, Huang J. QseC Inhibition as a Novel Antivirulence Strategy for the Prevention of Acute Hepatopancreatic Necrosis Disease (AHPND)-Causing Vibrio parahaemolyticus. Front Cell Infect Microbiol 2021; 10:594652. [PMID: 33553003 PMCID: PMC7859628 DOI: 10.3389/fcimb.2020.594652] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/04/2020] [Indexed: 11/13/2022] Open
Abstract
Acute hepatopancreatic necrosis disease (AHPND) caused by Vibrio parahaemolyticus resulted in great economic losses in global shrimp aquaculture. There is an urgent need for development of novel strategies to combat AHPND-causing V. parahaemolyticus (Vp AHPND), given that one of the greatest challenges currently is the widespread use of antibiotics and subsequent emergence of multidrug-resistant bacteria. Here, we proposed a broad-spectrum antivirulence approach targeting a conserved histidine kinase, QseC, which has been demonstrated to activate virulence expression in several Gram-negative pathogens. Our results showed that QseC mediated the catecholamine stimulated effects on growth and flagellar motility of Vp AHPND. Transcriptome analysis revealed that QseC was involved in the global regulation of the virulence of Vp AHPND as the ΔqseC mutant exhibited a decreased expression of genes related to type IV pilin, flagellar motility, and biofilm formation, while an overexpression of type VI secretion system and cell wall biosynthesis. Subsequently, the bacterial catecholamine receptor antagonist LED209 not only neutralized the stimulatory effects of host catecholamines on the growth and motility of Vp AHPND in vitro, but also attenuated the virulence of Vp AHPND towards brine shrimp larvae and white shrimp in vivo. Additionally, LED209 presented no interference with pathogen growth, nor the toxicity to the experimental animals. These results suggest that QseC can be an attractive antivirulence therapy target, and LED209 is a promising candidate for development of broad-spectrum antivirulence agents. This is the first study that demonstrated the role of QseC in the global regulation of Vp AHPND infection and demonstrated the antivirulence potential of LED209, which provides insight into the use of an antivirulence approach for targeting not only Vp AHPND, but also a much larger collection of pathogenic bacteria.
Collapse
Affiliation(s)
- Qian Yang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao, China.,Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China.,Center for Microbial Ecology and Technology (CMET), Ghent University, Gent, Belgium
| | - Peizhuo Zou
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao, China.,Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Zhi Cao
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao, China.,Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Qingyao Wang
- College of Marine Science and Environment, Dalian Ocean University, Dalian, China.,Key Laboratory of Environment Controlled Aquaculture (KLECA), Ministry of Education, Dalian, China
| | - Songzhe Fu
- College of Marine Science and Environment, Dalian Ocean University, Dalian, China.,Key Laboratory of Environment Controlled Aquaculture (KLECA), Ministry of Education, Dalian, China
| | - Guosi Xie
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao, China.,Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Jie Huang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao, China.,Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China.,Network of Aquaculture Centers in Asia-Pacific, Bangkok, Thailand
| |
Collapse
|
28
|
Investigation of Virulence Genes Detected in Antimicrobial-Resistance Pathogens Isolates for Five Countries across the World. Processes (Basel) 2020. [DOI: 10.3390/pr8121589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A large portion of annual deaths worldwide are due to infections caused by disease-causing pathogens. These pathogens contain virulence genes, which encode mechanisms that facilitate infection and microbial survival in hosts. More recently, antimicrobial resistance (AMR) genes, also found in these pathogens, have become an increasingly large issue. While the National Center for Biotechnology Information (NCBI) Pathogen Detection Isolates Browser (NPDIB) database has been compiling genes involved in microbial virulence and antimicrobial resistance through isolate samples, few studies have identified the genes primarily responsible for virulence and compared them to those responsible for AMR. This study performed the first multivariate statistical analysis of the multidimensional NPDIB data to identify the major virulence genes from historical pathogen isolates for Australia, China, South Africa, UK, and US—the largely populated countries from five of the six major continents. The important virulence genes were then compared with the AMR genes to study whether there is correlation between their occurrences. Among the significant genes and pathogens associated with virulence, it was found that the genes fdeC, iha, iss, iutA, lpfA, sslE, ybtP, and ybtQ are shared amongst all five countries. The pathogens E. coli and Shigella, Salmonella enterica, and Klebsiella pneumoniae mostly contained these genes and were common among four of the five studied countries. Additionally, the trend of virulence was investigated by plotting historical occurrences of gene and pathogen frequency in the annual samples. These plots showed that the trends of E. coli and Shigella and Salmonella enterica were similar to the trends of certain virulence genes, confirming the two pathogens do indeed carry important virulence genes. While the virulence genes in the five countries are not significantly different, the US and the UK share the largest amount of important virulence genes. The plots from principal component analysis and hierarchical clustering show that the important virulence and AMR genes were not significantly correlated, with only few genes from both types of genes clustered into the same groups.
Collapse
|