1
|
Zhu L, He Z, Li M, Xu J, Ding W, Zeng W, Jiang X. Antimicrobial and antivirulence function of cinnamaldehyde against Streptococcus suis type 2. Microbiol Spectr 2025; 13:e0256124. [PMID: 39945529 PMCID: PMC11960091 DOI: 10.1128/spectrum.02561-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/20/2025] [Indexed: 04/03/2025] Open
Abstract
Streptococcus suis type 2 (SS2) is an important zoonotic pathogen for swine and humans. The increasing prevalence of antimicrobial resistance in S. suis isolates poses a threat to public health. This study investigated the antimicrobial activity and therapeutic potential of cinnamaldehyde (CA), a natural compound from cinnamon, against SS2. CA showed significant antimicrobial activity with a minimal inhibition concentration of 0.25 µg/mL and prolonged post-antibiotic effect of over 7 h in SS2. Increased bacterial cell membrane permeability and blocked protein synthesis of SS2 were observed after being treated with CA. CA could effectively prevent biofilm formation. CA treatment reduced the crucial virulence factor of suilysin expression and secretion in SS2 cells through a probable interaction with the suilysin protein. CA treatment could prominently alleviate both epithelial HEp-2 and phagocytic RAW264.7 cell cytotoxicity induced by SS2. The pathogenic SS2 strain was attenuated by CA, as demonstrated by the diminished adherence in HEp-2 cells, increased clearance by RAW264.7 and mice whole blood, and improved survival rate in a mouse infection model. CA possessed therapeutic efficacy since the CA treatment exhibited a 50% improvement in mouse survival rate, which surpassed the traditional ampicillin therapy control group. Alleviated clinical symptoms and histopathological phenotypes, with reduced bacterial burden in mouse organs after CA treatment, were examined. Overall, this study identified cinnamaldehyde as a novel antibacterial compound against SS2 with potential therapeutic protective effects, offering an alternative drug for controlling SS2 prevalence and infection. IMPORTANCE Widespread infections caused by Streptococcus suis type 2 (SS2) have garnered significant attention in the realm of public health due to their zoonotic nature. In recent years, antimicrobial resistance phenotypes in SS2 have emerged and intensified within the context of animal husbandry. Herbal compounds and medicinal plants are increasingly recognized as promising therapeutic alternatives for mitigating or addressing the challenges posed by antimicrobial resistance. The aim of this present study was to explore a novel compound of cinnamaldehyde, which obtained significant antimicrobial activity and potential therapeutic protective effect against SS2 infection. The research has made an innovative discovery that the bactericidal effect of cinnamaldehyde is associated with its antivirulence strategies, such as targeting the key virulence factors of SS2 and countering the bacterial infection process.
Collapse
Affiliation(s)
- Lexin Zhu
- College of Medicine, Yichun University, Yichun, Jiangxi, China
| | - Zhishu He
- College of Medicine, Yichun University, Yichun, Jiangxi, China
| | - Mengqing Li
- College of Medicine, Yichun University, Yichun, Jiangxi, China
| | - Jixin Xu
- College of Medicine, Yichun University, Yichun, Jiangxi, China
| | - Wei Ding
- College of Medicine, Yichun University, Yichun, Jiangxi, China
| | - Wenzhen Zeng
- College of Medicine, Yichun University, Yichun, Jiangxi, China
| | - Xiaowu Jiang
- College of Medicine, Yichun University, Yichun, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Active Component of Natural Drugs, Poster-Doctoral Research Center, Yichun, Jiangxi, China
| |
Collapse
|
2
|
Wong BC, Ling FY, Ayub Q, Tan HS. Transposon mutagenesis identifies acid resistance and biofilm genes as Shigella sonnei virulence factors in Caenorhabditis elegans infection. Biochem Biophys Res Commun 2025; 754:151546. [PMID: 40023989 DOI: 10.1016/j.bbrc.2025.151546] [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: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Identifying essential genes in bacterial pathogens during infection can enhance knowledge and provide novel targets for antimicrobial agents' development. Currently, only Shigella flexneri essential genes during in vitro growth have been experimentally identified. This study used transposon insertion sequencing (TIS) to identify Shigella sonnei essential genes during Caenorhabditis elegans infection. 498 genes were predicted to be essential in S. sonnei during growth on nutrient-rich media. Some genes previously predicted to be essential in Shigella were found non-essential in S. sonnei, such as acetyl metabolism genes (aceEF, lpdA) and sulphate transport genes (cysA, cyst, cysW). Finally, 217 genes were predicted as S. sonnei virulence genes during infection, including acid resistance and biofilm formation genes which was not linked to S. sonnei virulence previously.
Collapse
Affiliation(s)
- Bao Chi Wong
- School of Science, Monash University Malaysia, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
| | - Fong Yoke Ling
- Monash University Malaysia Genomics Platform, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
| | - Qasim Ayub
- School of Science, Monash University Malaysia, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia; Monash University Malaysia Genomics Platform, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
| | - Hock Siew Tan
- School of Science, Monash University Malaysia, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
3
|
Cazzaniga G, Mori M, Griego A, Scarpa E, Moschetti G, Muzzioli S, Stelitano G, Chiarelli LR, Cocorullo M, Casali E, Porta A, Zanoni G, Tresoldi A, Pini E, Batalha ÍL, Battaglia G, Tuccinardi T, Rizzello L, Villa S, Meneghetti F. Nanoenabling MbtI Inhibitors for Next-Generation Tuberculosis Therapy. J Med Chem 2025; 68:5312-5332. [PMID: 40029993 PMCID: PMC11912484 DOI: 10.1021/acs.jmedchem.4c02386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025]
Abstract
The urgent need for safer and innovative antitubercular agents remains a priority for the scientific community. In pursuit of this goal, we designed and evaluated novel 5-phenylfuran-2-carboxylic acid derivatives targeting Mycobacterium tuberculosis (Mtb) salicylate synthase (MbtI), a key enzyme, absent in humans, that plays a crucial role in Mtb virulence. Several potent MbtI inhibitors demonstrating significant antitubercular activity and a favorable safety profile were identified. Structure-guided optimization yielded 5-(3-cyano-5-isobutoxyphenyl)furan-2-carboxylic acid (1e), which exhibited strong MbtI inhibition (IC50 = 11.2 μM) and a promising in vitro antitubercular activity (MIC99 = 32 μM against M. bovis BCG). Esters of 1e were effectively loaded into poly(2-methacryloyloxyethyl phosphorylcholine)-poly(2-(diisopropylamino)ethyl methacrylate) (PMPC-PDPA) polymersomes (POs) and delivered to intracellular mycobacteria, resulting in reduced Mtb viability. This study provides a foundation for the use of POs in the development of future MbtI-targeted therapies for tuberculosis.
Collapse
Affiliation(s)
- Giulia Cazzaniga
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- Department of Science and High Technology, University of Insubria, via Valleggio 9, 22100 Como, Italy
| | - Matteo Mori
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Anna Griego
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Edoardo Scarpa
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Giorgia Moschetti
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Stefano Muzzioli
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Giovanni Stelitano
- Department of Biology and Biotechnology "Lazzaro Spallanzani″, University of Pavia, via A. Ferrata 9, 27100 Pavia, Italy
| | - Laurent R Chiarelli
- Department of Biology and Biotechnology "Lazzaro Spallanzani″, University of Pavia, via A. Ferrata 9, 27100 Pavia, Italy
| | - Mario Cocorullo
- Department of Biology and Biotechnology "Lazzaro Spallanzani″, University of Pavia, via A. Ferrata 9, 27100 Pavia, Italy
| | - Emanuele Casali
- Department of Chemistry, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy
| | - Alessio Porta
- Department of Chemistry, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy
| | - Giuseppe Zanoni
- Department of Chemistry, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy
| | - Andrea Tresoldi
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Elena Pini
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Íris L Batalha
- Department of Life Sciences, University of Bath, Claverton Down, BA2 7AY Bath, U.K
| | - Giuseppe Battaglia
- Molecular Bionics Group, Institute for Bioengineering of Catalonia (IBEC), C. Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Catalan Institution of Research and Advanced Studies, (ICREA), Passeig de Lluís Companys, 23, 08010 Barcelona, Spain
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy
| | - Loris Rizzello
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Stefania Villa
- 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
| |
Collapse
|
4
|
Schneider G, Schweitzer B, Steinbach AS, Hodován ÁS, Horváth M, Bakó E, Mayer A, Pál S. The Therapeutic Potential of West Indian Lemongrass ( Cymbopogon citratus) Essential Oil-Based Ointment in the Treatment of Pitted Keratolysis. Antibiotics (Basel) 2025; 14:241. [PMID: 40149054 PMCID: PMC11939757 DOI: 10.3390/antibiotics14030241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Due to their antibacterial activities, essential oils can be potential alternatives to antibiotics in certain cases. West Indian lemongrass (Cymbopogon citratus) essential oil (LEO) is effective against a broad range of bacteria by inhibiting spore formation, and is considered safe. In this study, we demonstrated its therapeutical potential in the treatment of pitted keratolysis (PK), a superficial skin infection affecting the pressure-bearing areas of plantar surfaces. Methods: For in vitro antibacterial efficacy testing, LEO was mixed into different ointment bases, including Hydrogelum methylcellulose FoNo VIII., Ungentum oleosum FoNo VIII. (Ung. oleoso), Unguentum stearini FoNo VIII. (Ung. stearin), and Vaselinum cholesterinatum FoNo VIII. (Vasel. cholest.), at different concentrations of 1, 3, and 5%. These formulations were tested on representatives of three bacterial species associated with PK: Kytococcus sedentarius, Dermatophilus congolensis, and Bacillus thuringiensis. Results: In the in vitro tests, Hydrogelum methylcellulose (HM) gel best supported the antibacterial effects of LEO, reducing the number of living bacteria on agar plates by 4-5 orders of magnitude in a concentration-dependent manner during the 30 min exposure times. This was also confirmed by the Franz diffusion cell drug release test; after 30 min, several active compounds could be detected in the HM samples, in contrast to the other bases. Shelf-life experiments showed that the HM base supported the antibacterial features of 3% LEO for at least 2 years without significant loss of efficacy. Conclusions: Our study highlights that ointments containing essential oils potentially have a place in the treatment of PK. Therefore, antibiotics may potentially be replaced for the treatment of PK, thereby reducing environmental antibiotic pressure, which is one of the driving forces behind the spread of antibiotic resistance.
Collapse
Affiliation(s)
- György Schneider
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Szigeti st. 12., H-7624 Pécs, Hungary; (B.S.); (A.S.S.); (Á.S.H.); (M.H.)
| | - Bettina Schweitzer
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Szigeti st. 12., H-7624 Pécs, Hungary; (B.S.); (A.S.S.); (Á.S.H.); (M.H.)
| | - Anita S. Steinbach
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Szigeti st. 12., H-7624 Pécs, Hungary; (B.S.); (A.S.S.); (Á.S.H.); (M.H.)
| | - Ágnes S. Hodován
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Szigeti st. 12., H-7624 Pécs, Hungary; (B.S.); (A.S.S.); (Á.S.H.); (M.H.)
| | - Marianna Horváth
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Szigeti st. 12., H-7624 Pécs, Hungary; (B.S.); (A.S.S.); (Á.S.H.); (M.H.)
| | - Eszter Bakó
- Department of Pharmacognosy, Semmelweis University, Üllői st. 26., H-1085 Budapest, Hungary;
- Centre for Translational Medicine, Semmelweis University, Üllői st. 26., H-1085 Budapest, Hungary
| | - Anna Mayer
- Institute of Clinical Pharmacy, Medical School, University of Pécs, Szigeti st. 12., H-7624 Pécs, Hungary;
| | - Szilárd Pál
- Institute of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, University of Pécs, Rókus str. 2., H-7624 Pécs, Hungary;
| |
Collapse
|
5
|
Gajic I, Tomic N, Lukovic B, Jovicevic M, Kekic D, Petrovic M, Jankovic M, Trudic A, Mitic Culafic D, Milenkovic M, Opavski N. A Comprehensive Overview of Antibacterial Agents for Combating Multidrug-Resistant Bacteria: The Current Landscape, Development, Future Opportunities, and Challenges. Antibiotics (Basel) 2025; 14:221. [PMID: 40149033 PMCID: PMC11939824 DOI: 10.3390/antibiotics14030221] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Antimicrobial resistance poses a major public health challenge. The World Health Organization has identified 15 priority pathogens that require prompt development of new antibiotics. This review systematically evaluates the antibacterial resistance of the most significant bacterial pathogens, currently available treatment options, as well as complementary approaches for the management of infections caused by the most challenging multidrug-resistant (MDR) bacteria. For carbapenem-resistant Gram-negative bacteria, treatment options include combinations of beta-lactam antibiotics and beta-lactamase inhibitors, a novel siderophore cephalosporin, known as cefiderocol, as well as older antibiotics like polymixins and tigecycline. Treatment options for Gram-positive bacteria are vancomycin, daptomycin, linezolid, etc. Although the development of new antibiotics has stagnated, various agents with antibacterial properties are currently in clinical and preclinical trials. Non-antibiotic strategies encompass antibiotic potentiators, bacteriophage therapy, antivirulence therapeutics, antimicrobial peptides, antibacterial nanomaterials, host-directed therapy, vaccines, antibodies, plant-based products, repurposed drugs, as well as their combinations, including those used alongside antibiotics. Significant challenges exist in developing new antimicrobials, particularly related to scientific and technical issues, along with policy and economic factors. Currently, most of the alternative options are not part of routine treatment protocols. Conclusions and Future Directions: There is an urgent need to expedite the development of new strategies for treating infections caused by MDR bacteria. This requires a multidisciplinary approach that involves collaboration across research, healthcare, and regulatory bodies. Suggested approaches are crucial for addressing this challenge and should be backed by rational antibiotic use, enhanced infection control practices, and improved surveillance systems for emerging pathogens.
Collapse
Affiliation(s)
- Ina Gajic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.); (D.K.); (M.J.)
| | - Nina Tomic
- Group for Biomedical Engineering and Nanobiotechnology, Institute of Technical Sciences of SASA, Kneza Mihaila 35/IV, 11000 Belgrade, Serbia;
| | - Bojana Lukovic
- Academy of Applied Studies Belgrade, College of Health Sciences, 11000 Belgrade, Serbia;
| | - Milos Jovicevic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.); (D.K.); (M.J.)
| | - Dusan Kekic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.); (D.K.); (M.J.)
| | - Milos Petrovic
- University Clinical Hospital Center “Dr. Dragisa Misovic-Dedinje”, 11040 Belgrade, Serbia;
| | - Marko Jankovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.); (D.K.); (M.J.)
| | - Anika Trudic
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia;
- Institute for Pulmonary Diseases of Vojvodina, Sremska Kamenica, 21204 Novi Sad, Serbia
| | | | - Marina Milenkovic
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia;
| | - Natasa Opavski
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.J.); (D.K.); (M.J.)
| |
Collapse
|
6
|
Ju Y, Li L, Zhang J, Yusuf B, Zeng S, Fang C, Tian X, Han X, Ding J, Zhang H, Ma W, Wang S, Chen X, Zhang T. The gene MAB_2362 is responsible for intrinsic resistance to various drugs and virulence in Mycobacterium abscessus by regulating cell division. Antimicrob Agents Chemother 2025; 69:e0043324. [PMID: 39699214 PMCID: PMC11823648 DOI: 10.1128/aac.00433-24] [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: 03/21/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Mycobacterium abscessus exhibits intrinsic resistance to most antibiotics, hence leading to infections that are difficult to treat. To address this issue, the identification of new molecular targets is essential for the development or repositioning of therapeutic agents. This study demonstrated that the MAB_2362-knockout strain, MabΔ2362, became significantly susceptible to a range of antibiotics, not only in vitro but also exhibited susceptibility to rifabutin, bedaquiline, and linezolid in vivo. While the bacterial burden of the wild-type M. abscessus (MabWt) increased by over 1 log10 CFU/lung in a murine infection model 16 days post-infection, that of MabΔ2362 strain decreased by more than 1 log10 CFU/lung, which suggests that the disruption leads to attenuation. Bioinformatics analysis revealed that MAB_2362 shares the highest similarity (41.35%) with SteA, a protein known to influence cell division in Corynebacterium glutamicum, suggesting that MAB_2362 might be involved in cell division. MabΔ2362 cells exhibited a median length of 2.62 µm, which was substantially longer than the 1.44 µm recorded for MabWt cells. Additionally, multiple cell division septa were observed in 42% of MabΔ2362 cells, whereas none were seen in MabWt cells. An ethidium bromide uptake assay further suggested a higher cell envelope permeability in MabΔ2362 compared to MabWt. Collectively, these findings underscore the role of MAB_2362 in intrinsic resistance and virulence of M. abscessus possibly through the regulation of cell division. Thus, MAB_2362 emerges as a promising candidate for targeted interventions in the pursuit of novel antimicrobials against M. abscessus.
Collapse
Affiliation(s)
- Yanan Ju
- School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Lijie Li
- School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jingran Zhang
- School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Buhari Yusuf
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Sanshan Zeng
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Cuiting Fang
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xirong Tian
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xingli Han
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jie Ding
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Han Zhang
- School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wanli Ma
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shuai Wang
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, China
| | - Tianyu Zhang
- School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Institute of Physical Science and Information Technology, Anhui University, Hefei, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
7
|
Fihn CA, Lembke HK, Gaulin J, Bouchard P, Villarreal AR, Penningroth MR, Crone KK, Vogt GA, Gilbertsen AJ, Ayotte Y, Coutinho de Oliveira L, Serrano-Wu MH, Drouin N, Hung DT, Hunter RC, Carlson EE. Evaluation of expanded 2-aminobenzothiazole library as inhibitors of a model histidine kinase and virulence suppressors in Pseudomonas aeruginosa. Bioorg Chem 2024; 153:107840. [PMID: 39362083 PMCID: PMC11614690 DOI: 10.1016/j.bioorg.2024.107840] [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: 01/25/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024]
Abstract
Bacterial resistance to antibiotics is a rapidly increasing threat to human health. New strategies to combat resistant organisms are desperately needed. One potential avenue is targeting two-component systems, which are the main bacterial signal transduction pathways used to regulate development, metabolism, virulence, and antibiotic resistance. These systems consist of a homodimeric membrane-bound sensor histidine kinase, and a cognate effector, the response regulator. Histidine kinases play an essential role in the regulation of multiple virulence mechanisms including toxin production, immune evasion, and antibiotic resistance. Targeting virulence, as opposed to development of bactericidal compounds, could reduce evolutionary pressure for acquired resistance. Additionally, compounds targeting the highly conserved catalytic and adenosine triphosphate-binding (CA) domain have the potential to impair multiple two-component systems that regulate virulence in one or more pathogens. We conducted in vitro structure-activity relationship studies of 2-aminobenzothiazole-based inhibitors designed to target the CA domain. We found that these compounds, which inhibit the model histidine kinase, HK853 from Thermotoga maritima, have anti-virulence activities inPseudomonas aeruginosa, reducing motility phenotypes and toxin production associated with the pathogenic functions of this bacterium.
Collapse
Affiliation(s)
- Conrad A Fihn
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, United States
| | - Hannah K Lembke
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, MN 55454, United States
| | - Jeffrey Gaulin
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, United States
| | - Patricia Bouchard
- NMX Research and Solution Inc., 500 Cartier Boulevard W., Suite 6000, Laval, Quebec H1Y 2R1, Canada
| | - Alex R Villarreal
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave SE Minneapolis, MN 55455, United States
| | - Mitchell R Penningroth
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave SE Minneapolis, MN 55455, United States
| | - Kathryn K Crone
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, MN 55455, United States
| | - Grace A Vogt
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave SE Minneapolis, MN 55455, United States
| | - Adam J Gilbertsen
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave SE Minneapolis, MN 55455, United States
| | - Yann Ayotte
- NMX Research and Solution Inc., 500 Cartier Boulevard W., Suite 6000, Laval, Quebec H1Y 2R1, Canada
| | | | | | - Nathalie Drouin
- NMX Research and Solution Inc., 500 Cartier Boulevard W., Suite 6000, Laval, Quebec H1Y 2R1, Canada
| | - Deborah T Hung
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, United States
| | - Ryan C Hunter
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave SE Minneapolis, MN 55455, United States
| | - Erin E Carlson
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, United States; Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, MN 55454, United States; Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, MN 55455, United States.
| |
Collapse
|
8
|
Herzberg C, van Meegen EN, van Hasselt JGC. Interplay of virulence factors shapes ecology and treatment outcomes in polymicrobial infections. Math Biosci 2024; 377:109293. [PMID: 39245301 DOI: 10.1016/j.mbs.2024.109293] [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: 05/07/2024] [Revised: 08/11/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Polymicrobial infections, caused by a community of multiple micro-organisms, are often associated with increased infection severity and poorer patient outcomes. The design of improved antimicrobial treatment strategies for PMIs can be supported by an understanding of their ecological and evolutionary dynamics. Bacterial species present in polymicrobial infections can produce virulence factors to inhibit host immune responses, such as neutrophil recruitment and phagocytosis. The presence of virulence factors can indirectly affect other bacterial species acting as a type of host-mediated interspecies interaction. The aim of this study was to assess how bacterial virulence factors targeting neutrophil function influence ecology and treatment outcomes of PMIs. An agent-based model was constructed which describes a dual-species bacterial population in the presence of neutrophils and a bacteriostatic drug. Our analysis has revealed unforeseen dynamics of the interplay of multiple virulence factors acting as interspecies interaction. We found that the distribution of two phagocytosis-inhibiting virulence factors amongst species can impact whether they have a mutually protective effect for both species. The addition of a virulence factor inhibiting neutrophil recruitment was found to reduce the protective effect of phagocytosis-inhibiting virulence factors. Furthermore we demonstrate the importance of virulence strength of a species relative to other virulent species to determine the fate of a species. We conclude that virulence factors are an important driver of population dynamics in polymicrobial infections, and may be a relevant therapeutic target for treatment of polymicrobial infections.
Collapse
Affiliation(s)
- C Herzberg
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - E N van Meegen
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - J G C van Hasselt
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands.
| |
Collapse
|
9
|
Peng Y, Yan Y, Ma X, Jiang B, Chen R, Feng H, Xia Y. Efficient electrochemical oxidation of antibiotic wastewater using a graphene-loaded PbO 2 membrane anode: Mechanisms and applications. ENVIRONMENTAL RESEARCH 2024; 259:119517. [PMID: 38964585 DOI: 10.1016/j.envres.2024.119517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
This paper aims to develop a flow-through electrochemical system with a series of graphene nanoparticles loaded PbO2 reactive electrochemical membrane electrodes (GNPs-PbO2 REMs) on porous Ti substrates with pore sizes of 100, 150, 300 and 600 μm, and apply them to treat antibiotic wastewater. Among them, the GNPs-PbO2 with Ti substrate of 150 μm (Ti-150/GNPs-PbO2) had superior electrochemical degradation performance over the REMs with other pore sizes due to its smaller crystal size, larger electrochemical active specific area, lower charge-transfer impedance and larger oxygen evolution potential. Under the relatively optimized conditions of initial pH of 5, current density of 15 mA cm-2, and membrane flux of 4.20 m3 (m2·h)-1, the Ti-150/GNPs-PbO2 REM realized 99.34% of benzylpenicillin sodium (PNG) removal with an EE/O of 6.52 kWh m-3. Its excellent performance could be explained as the increased mass transfer. Then three plausible PNG degradation pathways in the flow-through electrochemical system were proposed, and great stability and safety of Ti-150/GNPs-PbO2 REM were demonstrated. Moreover, a single-pass Ti-150/GNPs-PbO2 REM system with five-modules in series was designed, which could consistently treat real antibiotic wastewater in compliance with disposal requirements of China. Thus, this study evidenced that the flow-through electrochemical system with the Ti-150/GNPs-PbO2 REM is an efficient alternative for treating antibiotic wastewater.
Collapse
Affiliation(s)
- Yifei Peng
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Yan Yan
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Xiangjuan Ma
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Bowen Jiang
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Ruya Chen
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Huajun Feng
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310018, China; College of Environment and Resources, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yijing Xia
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310018, China.
| |
Collapse
|
10
|
Campos DL, Canales CSC, Demarqui FM, Fernandes GFS, dos Santos CG, Prates JLB, da Silva IGM, Barros-Cordeiro KB, Báo SN, de Andrade LN, Abichabki N, Zacharias LV, de Campos MMA, dos Santos JL, Pavan FR. Screening of novel narrow-spectrum benzofuroxan derivatives for the treatment of multidrug-resistant tuberculosis through in silico, in vitro, and in vivo approaches. Front Microbiol 2024; 15:1487829. [PMID: 39464394 PMCID: PMC11502347 DOI: 10.3389/fmicb.2024.1487829] [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: 08/28/2024] [Accepted: 10/01/2024] [Indexed: 10/29/2024] Open
Abstract
Tuberculosis remains a serious global health threat, exacerbated by the rise of resistant strains. This study investigates the potential of two benzofuroxan (Bfx) derivatives, 5n and 5b, as targeted treatments for MDR-TB using in silico, in vitro, and in vivo methodologies. In vitro analyses showed that Bfx compounds have significant activity against Mtb H37Rv, with Bfx 5n standing out with a MIC90 of 0.09 ± 0.04 μM. Additionally, their efficacy against MDR and pre-XDR strains was superior compared to commercial drugs. These Bfx compounds have a narrow spectrum for mycobacteria, which helps avoid dysbiosis of the gut microbiota, and they also exhibit high selectivity and low toxicity. Synergism studies indicate that Bfx derivatives could be combined with rifampicin to enhance treatment efficacy and reduce its duration. Scanning electron microscopy revealed severe damage to the morphology of Mtb following treatment with Bfx 5n, showing significant distortions in the bacillary structures. Whole-genome sequencing of the 5n-resistant isolate suggests resistance mechanisms mediated by the Rv1855c gene, supported by in silico studies. In vivo studies showed that the 5n compound reduced the pulmonary load by 3.0 log10 CFU/mL, demonstrating superiority over rifampicin, which achieved a reduction of 1.23 log10 CFU/mL. In conclusion, Bfx derivatives, especially 5n, effectively address resistant infections caused by Mtb, suggesting they could be a solid foundation for future therapeutic developments against MDR-TB.
Collapse
Affiliation(s)
- Débora Leite Campos
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - Christian Shleider Carnero Canales
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
- School of Pharmacy, Biochemistry and Biotechnology, Santa Maria Catholic University, Arequipa, Peru
| | - Fernanda Manaia Demarqui
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - Guilherme F. S. Fernandes
- Medicinal Chemistry Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
- School of Pharmacy, University College London, London, United Kingdom
| | - Camila Gonçalves dos Santos
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - João Lucas B. Prates
- Medicinal Chemistry Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - Ingrid Gracielle Martins da Silva
- Microscopy and Microanalysis Laboratory, Cell Biology Department, Institute of Biological Sciences, University of Brasilia, Brasília, Brazil
| | - Karine Brenda Barros-Cordeiro
- Microscopy and Microanalysis Laboratory, Cell Biology Department, Institute of Biological Sciences, University of Brasilia, Brasília, Brazil
| | - Sônia Nair Báo
- Microscopy and Microanalysis Laboratory, Cell Biology Department, Institute of Biological Sciences, University of Brasilia, Brasília, Brazil
| | - Leonardo Neves de Andrade
- University of São Paulo – USPSchool of Pharmaceutical Sciences of Ribeirão Preto, , São Paulo, Brazil
| | - Nathália Abichabki
- University of São Paulo – USPSchool of Pharmaceutical Sciences of Ribeirão Preto, , São Paulo, Brazil
| | - Luísa Vieira Zacharias
- University of São Paulo – USPSchool of Pharmaceutical Sciences of Ribeirão Preto, , São Paulo, Brazil
| | - Marli Matiko Anraku de Campos
- Mycobacteriology Laboratory, Department of Clinical and Toxicological Analysis, Federal University of Santa Maria, Santa Maria, Brazil
| | - Jean Leandro dos Santos
- Medicinal Chemistry Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - Fernando Rogério Pavan
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| |
Collapse
|
11
|
Scott A, Khoruts A, Freeman ML, Beilman G, Ramanathan K, Bellin MD, Trikudanathan G. Successful Use of Fecal Microbiota Transplantation in Management of Nonobstructive Recurrent Cholangitis Following Total Pancreatectomy and Islet Autotransplant. ACG Case Rep J 2024; 11:e01527. [PMID: 39399248 PMCID: PMC11469898 DOI: 10.14309/crj.0000000000001527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024] Open
Abstract
Alterations in the gut microbiome have been implicated in various pathologies. Fecal microbiota transplantation (FMT) has been offered as a novel treatment for conditions implicated in the disruption of the gut-microbiota axis. This case report details the successful treatment of recurrent nonobstructive cholangitis following a single FMT application in a patient who had previously undergone a hepatobiliary tract surgical diversion. Cholangitis was suspected secondary to reflux of an altered microbiome into the surgically reanastomosed biliary tract, and FMT was justified based on the history of recurrent Clostridioides difficile infections. This case supports the further evaluation of the utility of FMT as one potential treatment of post hepatobiliary surgical diversion cholangitis.
Collapse
Affiliation(s)
- Adam Scott
- University of Minnesota Medical School, Minneapolis, MN
| | | | | | - Greg Beilman
- University of Minnesota Medical Center, Minneapolis, MN
| | | | | | | |
Collapse
|
12
|
Elalouf A, Yaniv-Rosenfeld A, Maoz H. Immune response against bacterial infection in organ transplant recipients. Transpl Immunol 2024; 86:102102. [PMID: 39094907 DOI: 10.1016/j.trim.2024.102102] [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: 05/17/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
This comprehensive review delves into the intricate dynamics between the immune system and bacterial infections in organ transplant recipients. Its primary objective is to fill existing knowledge gaps while critically assessing the strengths and weaknesses of current research. The paper accentuates the delicate balance that must be struck between preventing graft rejection through immunosuppression and maintaining robust immunity against bacterial threats. In this context, personalized medicine emerges as a transformative concept, offering the potential to revolutionize clinical outcomes by tailoring immunosuppressive regimens and vaccination strategies to the unique profiles of transplant recipients. By emphasizing the pivotal role of continuous monitoring, the review underscores the necessity for vigilant surveillance of transplant recipients to detect bacterial infections and associated immune responses early, thereby reducing the risk of severe infections and ultimately improving patient outcomes. Furthermore, the study highlights the significance of the host microbiome in shaping immune responses, suggesting that interventions targeting the microbiome hold promise for enhancing bacterial immunity in transplant recipients, both in research and clinical practice. In terms of future research directions, the review advocates for large-scale, longitudinal studies encompassing diverse patient cohorts to provide more comprehensive insights into post-transplant immune responses. It also advocates integrating multi-omics approaches, including genomics, transcriptomics, proteomics, and microbiome data, to understand immune responses and their underlying mechanisms. In conclusion, this review significantly enriches our understanding of immune responses in transplant recipients. It paves the way for more effective and personalized approaches to managing infections in this complex setting.
Collapse
Affiliation(s)
- Amir Elalouf
- Bar-Ilan University, Department of Management, Ramat Gan 5290002, Israel.
| | | | - Hanan Maoz
- Bar-Ilan University, Department of Management, Ramat Gan 5290002, Israel
| |
Collapse
|
13
|
Fieldman T. Evolutionary principles for modifying pathogen virulence. Crit Rev Microbiol 2024; 50:385-396. [PMID: 37146153 DOI: 10.1080/1040841x.2023.2203766] [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: 12/21/2022] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 05/07/2023]
Abstract
Current methods for combatting infectious diseases are largely limited to the prevention of infection, enhancing host immunity (via vaccination), and administration of small molecules to slow the growth of or kill pathogens (e.g. antimicrobials). Beyond efforts to deter the rise of antimicrobial resistance, little consideration is given to pathogen evolution. Natural selection will favor different levels of virulence under different circumstances. Experimental studies and a wealth of theoretical work have identified many likely evolutionary determinants of virulence. Some of these, such as transmission dynamics, are amenable to modification by clinicians and public health practitioners. In this article, we provide a conceptual overview of virulence, followed by an analysis of modifiable evolutionary determinants of virulence including vaccinations, antibiotics, and transmission dynamics. Finally, we discuss both the importance and limitations of taking an evolutionary approach to reducing pathogen virulence.
Collapse
Affiliation(s)
- Tom Fieldman
- Clinical Microbiology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
14
|
Eisenbraun EL, Vulpis TD, Prosser BN, Horswill AR, Blackwell HE. Synthetic Peptides Capable of Potent Multigroup Staphylococcal Quorum Sensing Activation and Inhibition in Both Cultures and Biofilm Communities. J Am Chem Soc 2024; 146:15941-15954. [PMID: 38832917 PMCID: PMC11321086 DOI: 10.1021/jacs.4c02694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The pathogen Staphylococcus epidermidis uses a chemical signaling process, i.e., quorum sensing (QS), to form robust biofilms and cause human infection. Many questions remain about QS in S. epidermidis, as it uses this intercellular communication pathway to both negatively and positively regulate virulence traits. Herein, we report synthetic multigroup agonists and antagonists of the S. epidermidis accessory gene regulator (agr) QS system capable of potent superactivation and complete inhibition, respectively. These macrocyclic peptides maintain full efficacy across the three major agr specificity groups, and their activity can be "mode-switched" from agonist to antagonist via subtle residue-specific structural changes. We describe the design and synthesis of these non-native peptides and demonstrate that they can appreciably decrease biofilm formation on abiotic surfaces, underscoring the potential for agr agonism as a route to block S. epidermidis virulence. Additionally, we show that both the S. epidermidis agonists and antagonists are active in S. aureus, another common pathogen with a related agr system, yet only as antagonists. This result not only revealed one of the most potent agr inhibitors known in S. aureus but also highlighted differences in the mechanisms of agr agonism and antagonism between these related bacteria. Finally, our investigations reveal unexpected inhibitory behavior for certain S. epidermidis agr agonists at sub-activating concentrations, an observation that can be leveraged for the design of future probes with enhanced potencies. Together, these peptides provide a powerful tool set to interrogate the role of QS in S. epidermidis infections and in Staphylococcal pathogenicity in general.
Collapse
Affiliation(s)
- Emma L. Eisenbraun
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| | - Troy D. Vulpis
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| | - Brendan N. Prosser
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Helen E. Blackwell
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| |
Collapse
|
15
|
MacNair CR, Rutherford ST, Tan MW. Alternative therapeutic strategies to treat antibiotic-resistant pathogens. Nat Rev Microbiol 2024; 22:262-275. [PMID: 38082064 DOI: 10.1038/s41579-023-00993-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 04/19/2024]
Abstract
Resistance threatens to render antibiotics - which are essential for modern medicine - ineffective, thus posing a threat to human health. The discovery of novel classes of antibiotics able to overcome resistance has been stalled for decades, with the developmental pipeline relying almost entirely on variations of existing chemical scaffolds. Unfortunately, this approach has been unable to keep pace with resistance evolution, necessitating new therapeutic strategies. In this Review, we highlight recent efforts to discover non-traditional antimicrobials, specifically describing the advantages and limitations of antimicrobial peptides and macrocycles, antibodies, bacteriophages and antisense oligonucleotides. These approaches have the potential to stem the tide of resistance by expanding the physicochemical property space and target spectrum occupied by currently approved antibiotics.
Collapse
Affiliation(s)
- Craig R MacNair
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA, USA
| | - Steven T Rutherford
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA, USA
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
16
|
Curcic J, Dinic M, Novovic K, Vasiljevic Z, Kojic M, Jovcic B, Malesevic M. A novel thermostable YtnP lactonase from Stenotrophomonas maltophilia inhibits Pseudomonas aeruginosa virulence in vitro and in vivo. Int J Biol Macromol 2024; 264:130421. [PMID: 38423425 DOI: 10.1016/j.ijbiomac.2024.130421] [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: 08/25/2023] [Revised: 01/18/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024]
Abstract
Infections caused by multidrug-resistant pathogens are one of the biggest challenges facing the healthcare system today. Quorum quenching (QQ) enzymes have the potential to be used as innovative enzyme-based antivirulence therapeutics to combat infections caused by multidrug-resistant pathogens. The main objective of this research was to describe the novel YtnP lactonase derived from the clinical isolate Stenotrophomonas maltophilia and to investigate its antivirulence potential against multidrug-resistant Pseudomonas aeruginosa MMA83. YtnP lactonase, the QQ enzyme, belongs to the family of metallo-β-lactamases. The recombinant enzyme has several advantageous biotechnological properties, such as high thermostability, activity in a wide pH range, and no cytotoxic effect. High-performance liquid chromatography analysis revealed the activity of recombinant YtnP lactonase toward a wide range of N-acyl-homoserine lactones (AHLs), quorum sensing signaling molecules, with a higher preference for long-chain AHLs. Recombinant YtnP lactonase was shown to inhibit P. aeruginosa MMA83 biofilm formation, induce biofilm decomposition, and reduce extracellular virulence factors production. Moreover, the lifespan of MMA83-infected Caenorhabditis elegans was prolonged with YtnP lactonase treatment. YtnP lactonase showed synergistic inhibitory activity in combination with gentamicin and acted additively with meropenem against MMA83. The described properties make YtnP lactonase a promising therapeutic candidate for the development of next-generation antivirulence agents.
Collapse
Affiliation(s)
- Jovana Curcic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | - Miroslav Dinic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | - Katarina Novovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | - Zorica Vasiljevic
- Institute for Mother and Child Health Care of Serbia "Dr Vukan Čupić", Radoja Dakića 8, 11070 Belgrade, Serbia
| | - Milan Kojic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; Institute of Virology, Vaccines and Sera "Torlak", Vojvode Stepe 448, 11042 Belgrade, Serbia
| | - Branko Jovcic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
| | - Milka Malesevic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia.
| |
Collapse
|
17
|
Wang L, Li H, Chen J, Wang Y, Gu Y, Jiu M. Antibacterial Mechanisms and Antivirulence Activities of Oridonin against Pathogenic Aeromonas hydrophila AS 1.1801. Microorganisms 2024; 12:415. [PMID: 38399819 PMCID: PMC10891661 DOI: 10.3390/microorganisms12020415] [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: 01/18/2024] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Aeromonas hydrophila, a Gram-negative bacterium widely found in freshwater environments, acts as a common conditional pathogen affecting humans, livestock, and aquatic animals. In this study, the impact of oridonin, an ent-kaurane diterpenoid compound derived from Rabdosia rubescens, on the virulence factors of A. hydrophila AS 1.1801 and its antibacterial mechanism was elucidated. The minimum inhibitory concentration (MIC) of oridonin against A. hydrophila AS 1.1801 was 100 μg/mL. Oridonin at inhibitory concentrations could significantly increase the electrical conductivity in the supernatant and escalate nucleic acid leakage (p < 0.01). This effect was concomitant with observed distortions in bacterial cells, the formation of cytoplasmic cavities, cellular damage, and pronounced inhibition of protein and nucleic acid synthesis. Additionally, oridonin at inhibitory levels exhibited a noteworthy suppressive impact on A. hydrophila AS 1.1801 across biofilm formation, motility, hemolytic activity, lipase activity, and protease activity (p < 0.05), demonstrating a dose-dependent enhancement. qRT-PCR analysis showed that the gene expression of luxR, qseB and omp were significantly downregulated after oridonin treatment in A. hydrophila AS 1.1801 (p < 0.05). Our results indicated that oridonin possessed significant antibacterial and anti-virulence effects on A. hydrophila AS 1.1801.
Collapse
Affiliation(s)
- Lunji Wang
- Key Laboratory of Microbial Resources Development and Utilization, Henan University of Science and Technology, Luoyang 471023, China; (L.W.); (J.C.)
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (H.L.); (Y.W.); (Y.G.)
| | - Huijuan Li
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (H.L.); (Y.W.); (Y.G.)
| | - Jinhao Chen
- Key Laboratory of Microbial Resources Development and Utilization, Henan University of Science and Technology, Luoyang 471023, China; (L.W.); (J.C.)
| | - Yi Wang
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (H.L.); (Y.W.); (Y.G.)
| | - Yuqing Gu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; (H.L.); (Y.W.); (Y.G.)
| | - Min Jiu
- Key Laboratory of Microbial Resources Development and Utilization, Henan University of Science and Technology, Luoyang 471023, China; (L.W.); (J.C.)
| |
Collapse
|
18
|
Fihn CA, Lembke HK, Gaulin J, Bouchard P, Villarreal AR, Penningroth MR, Crone KK, Vogt GA, Gilbertsen AJ, Ayotte Y, de Oliveira LC, Serrano-Wu MH, Drouin N, Hung DT, Hunter RC, Carlson EE. Evaluation of Expanded 2-Aminobenzothiazole Library for Inhibition of Pseudomonas aeruginosa Virulence Phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.02.539119. [PMID: 37205454 PMCID: PMC10187220 DOI: 10.1101/2023.05.02.539119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Bacterial resistance to antibiotics is a rapidly increasing threat to human health. New strategies to combat resistant organisms are desperately needed. One potential avenue is targeting two-component systems, which are the main bacterial signal transduction pathways used to regulate development, metabolism, virulence, and antibiotic resistance. These systems consist of a homodimeric membrane-bound sensor histidine kinase, and a cognate effector, the response regulator. The high sequence conservation in the catalytic and adenosine triphosphate-binding (CA) domain of histidine kinases and their essential role in bacterial signal transduction could enable broad-spectrum antibacterial activity. Through this signal transduction, histidine kinases regulate multiple virulence mechanisms including toxin production, immune evasion, and antibiotic resistance. Targeting virulence, as opposed to development of bactericidal compounds, could reduce evolutionary pressure for acquired resistance. Additionally, compounds targeting the CA domain have the potential to impair multiple two-component systems that regulate virulence in one or more pathogens. We conducted structure-activity relationship studies of 2-aminobenzothiazole-based inhibitors designed to target the CA domain of histidine kinases. We found these compounds have anti-virulence activities in Pseudomonas aeruginosa, reducing motility phenotypes and toxin production associated with the pathogenic functions of this bacterium.
Collapse
Affiliation(s)
- Conrad A. Fihn
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, Minnesota 55455, United States
| | - Hannah K. Lembke
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55454, United States
| | - Jeffrey Gaulin
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Patricia Bouchard
- NMX Research and Solution Inc., 500 Cartier Boulevard W., Suite 6000, Laval, Quebec, Canada, H1Y 2R1
| | - Alex R. Villarreal
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Mitchell R. Penningroth
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Kathryn K. Crone
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55455, United States
| | - Grace A. Vogt
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Adam J. Gilbertsen
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Yann Ayotte
- NMX Research and Solution Inc., 500 Cartier Boulevard W., Suite 6000, Laval, Quebec, Canada, H1Y 2R1
| | | | | | - Nathalie Drouin
- NMX Research and Solution Inc., 500 Cartier Boulevard W., Suite 6000, Laval, Quebec, Canada, H1Y 2R1
| | - Deborah T. Hung
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Ryan C. Hunter
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Erin E. Carlson
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55454, United States
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
19
|
Vergoz D, Le H, Bernay B, Schaumann A, Barreau M, Nilly F, Desriac F, Tahrioui A, Giard JC, Lesouhaitier O, Chevalier S, Brunel JM, Muller C, Dé E. Antibiofilm and Antivirulence Properties of 6-Polyaminosteroid Derivatives against Antibiotic-Resistant Bacteria. Antibiotics (Basel) 2023; 13:8. [PMID: 38275318 PMCID: PMC10812528 DOI: 10.3390/antibiotics13010008] [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: 11/29/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The emergence of multi-drug resistant pathogens is a major public health problem, leading us to rethink and innovate our bacterial control strategies. Here, we explore the antibiofilm and antivirulence activities of nineteen 6-polyaminosterol derivatives (squalamine-based), presenting a modulation of their polyamine side chain on four major pathogens, i.e., carbapenem-resistant A. baumannii (CRAB) and P. aeruginosa (CRPA), methicillin-resistant S. aureus (MRSA), and vancomycin-resistant E. faecium (VRE) strains. We screened the effect of these derivatives on biofilm formation and eradication. Derivatives 4e (for CRAB, VRE, and MRSA) and 4f (for all the strains) were the most potent ones and displayed activities as good as those of conventional antibiotics. We also identified 11 compounds able to decrease by more than 40% the production of pyocyanin, a major virulence factor of P. aeruginosa. We demonstrated that 4f treatment acts against bacterial infections in Galleria mellonella and significantly prolonged larvae survival (from 50% to 80%) after 24 h of CRAB, VRE, and MRSA infections. As shown by proteomic studies, 4f triggered distinct cellular responses depending on the bacterial species but essentially linked to cell envelope. Its interesting antibiofilm and antivirulence properties make it a promising a candidate for use in therapeutics.
Collapse
Affiliation(s)
- Delphine Vergoz
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000 Rouen, France; (D.V.); (H.L.); (A.S.)
| | - Hung Le
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000 Rouen, France; (D.V.); (H.L.); (A.S.)
| | - Benoit Bernay
- Univ Caen Normandie, Proteogen Platform, US EMERODE, F-14000 Caen, France;
| | - Annick Schaumann
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000 Rouen, France; (D.V.); (H.L.); (A.S.)
| | - Magalie Barreau
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Flore Nilly
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Florie Desriac
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Ali Tahrioui
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | | | - Olivier Lesouhaitier
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Sylvie Chevalier
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | | | - Cécile Muller
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Emmanuelle Dé
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000 Rouen, France; (D.V.); (H.L.); (A.S.)
| |
Collapse
|
20
|
Francis D, Veeramanickathadathil Hari G, Koonthanmala Subash A, Bhairaddy A, Joy A. The biofilm proteome of Staphylococcus aureus and its implications for therapeutic interventions to biofilm-associated infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:327-400. [PMID: 38220430 DOI: 10.1016/bs.apcsb.2023.08.002] [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: 01/16/2024]
Abstract
Staphylococcus aureus is a major healthcare concern due to its ability to inflict life-threatening infections and evolve antibiotic resistance at an alarming pace. It is frequently associated with hospital-acquired infections, especially device-associated infections. Systemic infections due to S. aureus are difficult to treat and are associated with significant mortality and morbidity. The situation is worsened by the ability of S. aureus to form social associations called biofilms. Biofilms embed a community of cells with the ability to communicate with each other and share resources within a polysaccharide or protein matrix. S. aureus establish biofilms on tissues and conditioned abiotic surfaces. Biofilms are hyper-tolerant to antibiotics and help evade host immune responses. Biofilms exacerbate the severity and recalcitrance of device-associated infections. The development of a biofilm involves various biomolecules, such as polysaccharides, proteins and nucleic acids, contributing to different structural and functional roles. Interconnected signaling pathways and regulatory molecules modulate the expression of these molecules. A comprehensive understanding of the molecular biology of biofilm development would help to devise effective anti-biofilm therapeutics. Although bactericidal agents, antimicrobial peptides, bacteriophages and nano-conjugated anti-biofilm agents have been employed with varying levels of success, there is still a requirement for effective and clinically viable anti-biofilm therapeutics. Proteins that are expressed and utilized during biofilm formation, constituting the biofilm proteome, are a particularly attractive target for anti-biofilm strategies. The proteome can be explored to identify potential anti-biofilm drug targets and utilized for rational drug discovery. With the aim of uncovering the biofilm proteome, this chapter explores the mechanism of biofilm formation and its regulation. Furthermore, it explores the antibiofilm therapeutics targeted against the biofilm proteome.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India.
| | | | | | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| |
Collapse
|
21
|
Alanko I, Sandberg R, Brockmann E, de Haas CJC, van Strijp JAG, Lamminmäki U, Salo‐Ahen OMH. Isolation and functional analysis of phage-displayed antibody fragments targeting the staphylococcal superantigen-like proteins. Microbiologyopen 2023; 12:e1371. [PMID: 37642487 PMCID: PMC10350561 DOI: 10.1002/mbo3.1371] [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: 02/08/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 08/31/2023] Open
Abstract
Staphylococcus aureus produces numerous virulence factors that manipulate the immune system, helping the bacteria avoid phagocytosis. In this study, we are investigating three immune evasion molecules called the staphylococcal superantigen-like proteins 1, 5, and 10 (SSL1, SSL5, and SSL10). All three SSLs inhibit vital host immune processes and contribute to S. aureus immune evasion. This study aimed to identify single-chain variable fragment (scFvs) antibodies from synthetic antibody phage libraries, which can recognize either of the three SSLs and could block the interaction between the SSLs and their respective human targets. The antibodies were isolated after three rounds of panning against SSL1, SSL5, and SSL10, and their ability to bind to the SSLs was studied using a time-resolved fluorescence-based immunoassay. We successfully obtained altogether 44 unique clones displaying binding activity to either SSL1, SSL5, or SSL10. The capability of the SSL-recognizing scFvs to inhibit the SSLs' function was tested in an MMP9 enzymatic activity assay, a P-selectin glycoprotein ligand 1 competitive binding assay, and an IgG1-mediated phagocytosis assay. We could show that one scFv was able to inhibit SSL1 and maintain MMP9 activity in a concentration-dependent manner. Finally, the structure of this inhibiting scFv was modeled and used to create putative scFv-SSL1-complex models by protein-protein docking. The complex models were subjected to a 100-ns molecular dynamics simulation to assess the possible binding mode of the antibody.
Collapse
Affiliation(s)
- Ida Alanko
- Faculty of Sciences and Engineering, Pharmaceutical Sciences Laboratory (Pharmacy) & Structural Bioinformatics Laboratory (Biochemistry) TurkuÅbo Akademi UniversityTurkuFinland
| | - Rebecca Sandberg
- Faculty of Sciences and Engineering, Pharmaceutical Sciences Laboratory (Pharmacy) & Structural Bioinformatics Laboratory (Biochemistry) TurkuÅbo Akademi UniversityTurkuFinland
| | | | - Carla J. C. de Haas
- Department of Medical Microbiology, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Jos A. G. van Strijp
- Department of Medical Microbiology, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Urpo Lamminmäki
- Department of Life TechnologiesUniversity of TurkuTurkuFinland
| | - Outi M. H. Salo‐Ahen
- Faculty of Sciences and Engineering, Pharmaceutical Sciences Laboratory (Pharmacy) & Structural Bioinformatics Laboratory (Biochemistry) TurkuÅbo Akademi UniversityTurkuFinland
| |
Collapse
|
22
|
Harnagel AP, Sheshova M, Zheng M, Zheng M, Skorupinska-Tudek K, Swiezewska E, Lupoli TJ. Preference of Bacterial Rhamnosyltransferases for 6-Deoxysugars Reveals a Strategy To Deplete O-Antigens. J Am Chem Soc 2023. [PMID: 37437030 PMCID: PMC10375533 DOI: 10.1021/jacs.3c03005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Bacteria synthesize hundreds of bacteria-specific or "rare" sugars that are absent in mammalian cells and enriched in 6-deoxy monosaccharides such as l-rhamnose (l-Rha). Across bacteria, l-Rha is incorporated into glycans by rhamnosyltransferases (RTs) that couple nucleotide sugar substrates (donors) to target biomolecules (acceptors). Since l-Rha is required for the biosynthesis of bacterial glycans involved in survival or host infection, RTs represent potential antibiotic or antivirulence targets. However, purified RTs and their unique bacterial sugar substrates have been difficult to obtain. Here, we use synthetic nucleotide rare sugar and glycolipid analogs to examine substrate recognition by three RTs that produce cell envelope components in diverse species, including a known pathogen. We find that bacterial RTs prefer pyrimidine nucleotide-linked 6-deoxysugars, not those containing a C6-hydroxyl, as donors. While glycolipid acceptors must contain a lipid, isoprenoid chain length, and stereochemistry can vary. Based on these observations, we demonstrate that a 6-deoxysugar transition state analog inhibits an RT in vitro and reduces levels of RT-dependent O-antigen polysaccharides in Gram-negative cells. As O-antigens are virulence factors, bacteria-specific sugar transferase inhibition represents a novel strategy to prevent bacterial infections.
Collapse
Affiliation(s)
- Alexa P Harnagel
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Mia Sheshova
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Meng Zheng
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Maggie Zheng
- Department of Chemistry, New York University, New York, New York 10003, United States
| | | | - Ewa Swiezewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Tania J Lupoli
- Department of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
23
|
Escobar-Salom M, Barceló IM, Jordana-Lluch E, Torrens G, Oliver A, Juan C. Bacterial virulence regulation through soluble peptidoglycan fragments sensing and response: knowledge gaps and therapeutic potential. FEMS Microbiol Rev 2023; 47:fuad010. [PMID: 36893807 PMCID: PMC10039701 DOI: 10.1093/femsre/fuad010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 02/10/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Given the growing clinical-epidemiological threat posed by the phenomenon of antibiotic resistance, new therapeutic options are urgently needed, especially against top nosocomial pathogens such as those within the ESKAPE group. In this scenario, research is pushed to explore therapeutic alternatives and, among these, those oriented toward reducing bacterial pathogenic power could pose encouraging options. However, the first step in developing these antivirulence weapons is to find weak points in the bacterial biology to be attacked with the goal of dampening pathogenesis. In this regard, during the last decades some studies have directly/indirectly suggested that certain soluble peptidoglycan-derived fragments display virulence-regulatory capacities, likely through similar mechanisms to those followed to regulate the production of several β-lactamases: binding to specific transcriptional regulators and/or sensing/activation of two-component systems. These data suggest the existence of intra- and also intercellular peptidoglycan-derived signaling capable of impacting bacterial behavior, and hence likely exploitable from the therapeutic perspective. Using the well-known phenomenon of peptidoglycan metabolism-linked β-lactamase regulation as a starting point, we gather and integrate the studies connecting soluble peptidoglycan sensing with fitness/virulence regulation in Gram-negatives, dissecting the gaps in current knowledge that need filling to enable potential therapeutic strategy development, a topic which is also finally discussed.
Collapse
Affiliation(s)
- María Escobar-Salom
- Research Unit and Microbiology Department, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Crtra. Valldemossa 79, 07010 Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC). Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Isabel María Barceló
- Research Unit and Microbiology Department, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Crtra. Valldemossa 79, 07010 Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC). Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Elena Jordana-Lluch
- Research Unit and Microbiology Department, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Crtra. Valldemossa 79, 07010 Palma, Spain
| | - Gabriel Torrens
- Research Unit and Microbiology Department, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Crtra. Valldemossa 79, 07010 Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC). Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University. Försörjningsvägen 2A, SE-901 87 Umeå, Sweden
| | - Antonio Oliver
- Research Unit and Microbiology Department, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Crtra. Valldemossa 79, 07010 Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC). Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Carlos Juan
- Research Unit and Microbiology Department, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Crtra. Valldemossa 79, 07010 Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC). Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
| |
Collapse
|
24
|
Rodríguez Á, Maneiro M, Lence E, Otero JM, van Raaij MJ, Thompson P, Hawkins AR, González-Bello C. Quinate-based ligands for irreversible inactivation of the bacterial virulence factor DHQ1 enzyme-A molecular insight. Front Mol Biosci 2023; 10:1111598. [PMID: 36762206 PMCID: PMC9902378 DOI: 10.3389/fmolb.2023.1111598] [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: 11/29/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
Irreversible inhibition of the enzyme type I dehydroquinase (DHQ1), a promising target for anti-virulence drug development, has been explored by enhancing the electrophilicity of specific positions of the ligand towards covalent lysine modification. For ligand design, we made use of the advantages offered by the intrinsic acid-base properties of the amino substituents introduced in the quinate scaffold, namely compounds 6-7 (R configuration at C3), to generate a potential leaving group, as well as the recognition pattern of the enzyme. The reactivity of the C2-C3 bond (Re face) in the scaffold was also explored using compound 8. The results of the present study show that replacement of the C3 hydroxy group of (-)-quinic acid by a hydroxyamino substituent (compound 6) provides a time-dependent irreversible inhibitor, while compound 7, in which the latter functionality was substituted by an amino group, and the introduction of an oxirane ring at C2-C3 bond, compound 8, do not allow covalent modification of the enzyme. These outcomes were supported by resolution of the crystal structures of DHQ1 from Staphylococcus aureus (Sa-DHQ1) and Salmonella typhi (St-DHQ1) chemically modified by 6 at a resolution of 1.65 and 1.90 Å, respectively, and of St-DHQ1 in the complex with 8 (1.55 Å). The combination of these structural studies with extensive molecular dynamics simulation studies allowed us to understand the molecular basis of the type of inhibition observed. This study is a good example of the importance of achieving the correct geometry between the reactive center of the ligand (electrophile) and the enzyme nucleophile (lysine residue) to allow selective covalent modification. The outcomes obtained with the hydroxyamino derivative 6 also open up new possibilities in the design of irreversible inhibitors based on the use of amino substituents.
Collapse
Affiliation(s)
- Ángela Rodríguez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - María Maneiro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Emilio Lence
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - José M. Otero
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mark J. van Raaij
- Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CSIC), Madrid, Spain
| | - Paul Thompson
- Newcastle University Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alastair R. Hawkins
- Newcastle University Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Concepción González-Bello
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain,*Correspondence: Concepción González-Bello,
| |
Collapse
|
25
|
Stoitsova S, Paunova-Krasteva T, Dimitrova PD, Damyanova T. The concept for the antivirulence therapeutics approach as alternative to antibiotics: hope or still a fiction? BIOTECHNOL BIOTEC EQ 2022. [DOI: 10.1080/13102818.2022.2106887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Affiliation(s)
- Stoyanka Stoitsova
- Department of General Microbiology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tsvetelina Paunova-Krasteva
- Department of General Microbiology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Petya D. Dimitrova
- Department of General Microbiology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tsvetozara Damyanova
- Department of General Microbiology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
26
|
Research Progress on Small Molecular Inhibitors of the Type 3 Secretion System. Molecules 2022; 27:molecules27238348. [PMID: 36500441 PMCID: PMC9740592 DOI: 10.3390/molecules27238348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
The overuse of antibiotics has led to severe bacterial drug resistance. Blocking pathogen virulence devices is a highly effective approach to combating bacterial resistance worldwide. Type three secretion systems (T3SSs) are significant virulence factors in Gram-negative pathogens. Inhibition of these systems can effectively weaken infection whilst having no significant effect on bacterial growth. Therefore, T3SS inhibitors may be a powerful weapon against resistance in Gram-negative bacteria, and there has been increasing interest in the research and development of T3SS inhibitors. This review outlines several reported small-molecule inhibitors of the T3SS, covering those of synthetic and natural origin, including their sources, structures, and mechanisms of action.
Collapse
|
27
|
Jabbari S. Unravelling microbial efflux through mathematical modelling. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 36409600 DOI: 10.1099/mic.0.001264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
AbstractMathematical modelling is a useful tool that is increasingly used in the life sciences to understand and predict the behaviour of biological systems. This review looks at how this interdisciplinary approach has advanced our understanding of microbial efflux, the process by which microbes expel harmful substances. The discussion is largely in the context of antimicrobial resistance, but applications in synthetic biology are also touched upon. The goal of this paper is to spark further fruitful collaborations between modellers and experimentalists in the efflux community and beyond.
Collapse
Affiliation(s)
- Sara Jabbari
- School of Mathematics and Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| |
Collapse
|
28
|
Seixas AMM, Sousa SA, Leitão JH. Antibody-Based Immunotherapies as a Tool for Tackling Multidrug-Resistant Bacterial Infections. Vaccines (Basel) 2022; 10:1789. [PMID: 36366297 PMCID: PMC9695245 DOI: 10.3390/vaccines10111789] [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: 09/15/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 07/27/2023] Open
Abstract
The discovery of antimicrobials is an outstanding achievement of mankind that led to the development of modern medicine. However, increasing antimicrobial resistance observed worldwide is rendering commercially available antimicrobials ineffective. This problem results from the bacterial ability to adapt to selective pressure, leading to the development or acquisition of multiple types of resistance mechanisms that can severely affect the efficacy of antimicrobials. The misuse, over-prescription, and poor treatment adherence by patients are factors strongly aggravating this issue, with an epidemic of infections untreatable by first-line therapies occurring over decades. Alternatives are required to tackle this problem, and immunotherapies are emerging as pathogen-specific and nonresistance-generating alternatives to antimicrobials. In this work, four types of antibody formats and their potential for the development of antibody-based immunotherapies against bacteria are discussed. These antibody isotypes include conventional mammalian polyclonal antibodies that are used for the neutralization of toxins; conventional mammalian monoclonal antibodies that currently have 100 IgG mAbs approved for therapeutic use; immunoglobulin Y found in birds and an excellent source of high-quality polyclonal antibodies able to be purified noninvasively from egg yolks; and single domain antibodies (also known as nanobodies), a recently discovered antibody format (found in camelids and nurse sharks) that allows for a low-cost synthesis in microbial systems, access to hidden or hard-to-reach epitopes, and exhibits a high modularity for the development of complex structures.
Collapse
Affiliation(s)
- António M. M. Seixas
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Sílvia A. Sousa
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Jorge H. Leitão
- Department of Bioengineering, IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
29
|
Berdida DJE, Grande RAN, Lopez V, Ramirez SH, Manting MME, Berdida MME, Bañas CB. A national online survey of Filipinos' knowledge, attitude, and awareness of antibiotic use and resistance: A cross-sectional study. Nurs Forum 2022; 57:1299-1313. [PMID: 36161465 DOI: 10.1111/nuf.12803] [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: 07/17/2022] [Revised: 08/23/2022] [Accepted: 09/15/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Antibiotic resistance is one of the 21st century's most challenging clinical and public health issues. However, this health issue remains underreported in the Philippines. AIM This study examined Filipinos' knowledge, attitude, and awareness regarding antibiotic use and resistance and the associated predictive variables of antibiotic resistance. METHODS A cross-sectional design and Strengthening the Reporting of Observational Studies in Epidemiology guidelines were used in this study. Convenience sampling of 3767 participants completed the Eurobarometer survey on antibiotic resistance from October 2021 to February 2022. χ2 and regression analysis were used to analyze the data. RESULTS Most participants were familiar with the popular types of available antibiotics. Males and healthcare workers had a higher percentage of correct responses on antibiotic resistance knowledge. Participants had moderate to high knowledge levels of antibiotic resistance. Age, educational attainment, profession, antibiotic use in the previous year, and household members taking antibiotics were significant predictors of the level of knowledge of antibiotic resistance. There was a significant difference in participants' sex, age, and educational attainment in their attitudes toward acquisition, hygienic practices, and the role of health professionals in antibiotic resistance. CONCLUSION Government agencies and policymakers should consider the identified predictors when establishing policies on antibiotic resistance. This will ensure that antibiotic use is safe and effective.
Collapse
Affiliation(s)
| | - Rizal Angelo N Grande
- Mental Health Nursing Department, College of Nursing, University of Ha'il, Ha'il City, Saudi Arabia
| | - Violeta Lopez
- School of Nursing, Midwifery and Social Sciences, Central Queensland University, Rockhampton, Queensland, Australia
| | - Sheryl H Ramirez
- University Research Innovation and Extension, Universidad de Manila, Manila, Philippines
| | - Muhmin Michael E Manting
- Department of Biological Sciences, Mindanao State University-Iligan Institute of Technology, Iligan City, Lanao del Norte, Philippines
| | | | | |
Collapse
|
30
|
Liu PX, Zhang XY, Wang Q, Li YY, Sun WD, Qi Y, Zhou K, Han XG, Chen ZG, Fang WH, Jiang W. Biological and transcriptional studies reveal VmeL is involved in motility, biofilm formation and virulence in Vibrio parahaemolyticus. Front Microbiol 2022; 13:976334. [PMID: 36016795 PMCID: PMC9397117 DOI: 10.3389/fmicb.2022.976334] [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: 06/23/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
Vibrio parahaemolyticus is a marine pathogen thought to be the leading cause of seafood-borne gastroenteritis globally, urgently requiring efficient management methods. V. parahaemolyticus encodes 12 resistance/nodulation/division (RND) efflux systems. However, research on these systems is still in its infancy. In this study, we discovered that the inactivation of VmeL, a membrane fusion protein within the RND efflux systems, led to reduction of the ability of biofilm formation. Further results displayed that the decreased capacity of Congo red binding and the colony of ΔvmeL is more translucent compared with wild type strains, suggested reduced biofilm formation due to decreased production of biofilm exopolysaccharide upon vmeL deletion. In addition, the deletion of vmeL abolished surface swarming and swimming motility of V. parahaemolyticus. Additionally, deletion of vmeL weakened the cytotoxicity of V. parahaemolyticus towards HeLa cells, and impaired its virulence in a murine intraperitoneal infection assay. Finally, through RNA-sequencing, we ascertained that there were 716 upregulated genes and 247 downregulated genes in ΔvmeL strain. KEGG enrichment analysis revealed that quorum sensing, bacterial secretion systems, ATP-binding cassette transporters, and various amino acid metabolism pathways were altered due to the inactivation of vmeL. qRT-PCR further confirmed that genes accountable to the type III secretion system (T3SS1) and lateral flagella were negatively affected by vmeL deletion. Taken together, our results suggest that VmeL plays an important role in pathogenicity, making it a good target for managing infection with V. parahaemolyticus.
Collapse
Affiliation(s)
- Peng-xuan Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiao-yun Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai, China
| | - Quan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai, China
| | - Yang-yang Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wei-dong Sun
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yu Qi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, The First Affiliated Hospital (Shenzhen People’s Hospital), Shenzhen, China
| | - Xian-gan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai, China
| | - Zhao-guo Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai, China
| | - Wei-huan Fang
- Institute of Preventive Veterinary Medicine and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Wei Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai, China
- *Correspondence: Wei Jiang,
| |
Collapse
|
31
|
Martínez OF, Duque HM, Franco OL. Peptidomimetics as Potential Anti-Virulence Drugs Against Resistant Bacterial Pathogens. Front Microbiol 2022; 13:831037. [PMID: 35516442 PMCID: PMC9062693 DOI: 10.3389/fmicb.2022.831037] [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: 12/07/2021] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
The uncontrollable spread of superbugs calls for new approaches in dealing with microbial-antibiotic resistance. Accordingly, the anti-virulence approach has arisen as an attractive unconventional strategy to face multidrug-resistant pathogens. As an emergent strategy, there is an imperative demand for discovery, design, and development of anti-virulence drugs. In this regard, peptidomimetic compounds could be a valuable source of anti-virulence drugs, since these molecules circumvent several shortcomings of natural peptide-based drugs like proteolytic instability, immunogenicity, toxicity, and low bioavailability. Some emerging evidence points to the feasibility of peptidomimetics to impair pathogen virulence. Consequently, in this review, we shed some light on the potential of peptidomimetics as anti-virulence drugs to overcome antibiotic resistance. Specifically, we address the anti-virulence activity of peptidomimetics against pathogens' secretion systems, biofilms, and quorum-sensing systems.
Collapse
Affiliation(s)
- Osmel Fleitas Martínez
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil.,Programa de Pós-Graduação em Biotecnologia, S-Inova Biotech, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Harry Morales Duque
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
| | - Octávio Luiz Franco
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil.,Programa de Pós-Graduação em Biotecnologia, S-Inova Biotech, Universidade Católica Dom Bosco, Campo Grande, Brazil
| |
Collapse
|
32
|
Law SKK, Tan HS. The Role of Quorum Sensing, Biofilm Formation, and Iron Acquisition as Key Virulence Mechanisms in Acinetobacter baumannii and the Corresponding Anti-virulence Strategies. Microbiol Res 2022; 260:127032. [DOI: 10.1016/j.micres.2022.127032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/15/2022]
|
33
|
McCarthy M, Goncalves M, Powell H, Morey B, Turner M, Merrill AR. A Structural Approach to Anti-Virulence: A Discovery Pipeline. Microorganisms 2021; 9:microorganisms9122514. [PMID: 34946116 PMCID: PMC8704661 DOI: 10.3390/microorganisms9122514] [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: 10/31/2021] [Revised: 11/27/2021] [Accepted: 11/30/2021] [Indexed: 11/25/2022] Open
Abstract
The anti-virulence strategy is designed to prevent bacterial virulence factors produced by pathogenic bacteria from initiating and sustaining an infection. One family of bacterial virulence factors is the mono-ADP-ribosyltransferase toxins, which are produced by pathogens as tools to compromise the target host cell. These toxins are bacterial enzymes that exploit host cellular NAD+ as the donor substrate to modify an essential macromolecule acceptor target in the host cell. This biochemical reaction modifies the target macromolecule (often protein or DNA) and functions in a binary fashion to turn the target activity on or off by blocking or impairing a critical process or pathway in the host. A structural biology approach to the anti-virulence method to neutralize the cytotoxic effect of these factors requires the search and design of small molecules that bind tightly to the enzyme active site and prevent catalytic function essentially disarming the pathogen. This method requires a high-resolution structure to serve as the model for small molecule inhibitor development, which illuminates the path to drug development. This alternative strategy to antibiotic therapy represents a paradigm shift that may circumvent multi-drug resistance in the offending microbe through anti-virulence therapy. In this report, the rationale for the anti-virulence structural approach will be discussed along with recent efforts to apply this method to treat honey bee diseases using natural products.
Collapse
|