1
|
Li W, Xu X, Song Y, Fan L, Huang J, Yang L, Liu Y, Xu H. POD-like nanozyme constructed from perspective of charge transfer engineering for biosensing of magnetic separation treated Listeria monocytogenes. Food Chem 2025; 463:141495. [PMID: 39362102 DOI: 10.1016/j.foodchem.2024.141495] [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: 07/25/2024] [Revised: 09/07/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
For foodborne pathogens pose a serious threat to public health, a magnetic separation strategy and a nanozyme-based biosensor are proposed for biosensing of Listeria monocytogenes (L. monocytogenes). In this work, doripenem is selected as a recognized molecule for the modification of magnetic beads to capture L.monocytogenes in food and environmental samples. Furthermore, the POD-like MXene-Hemin-Au is constructed from perspective of charge transfer engineering which provides a vivid example to rational design of nanozymes. Finally, the captured L.monocytogenes is labeled with MXene-Hemin-Au@mAb, forming the sandwich complexes for quantitative determination. The current signals that generated by the complexes exhibit a good linear relationship with a limit of detection of 2.3 × 101 CFU/mL. The biosensor shows a satisfactory applicability in real samples with recoveries of 91.19% to 102.98%. Overall, the biosensor with integrated magnetic separation strategy presents a potential approach for high sensitivity biosensing of foodborne pathogens.
Collapse
Affiliation(s)
- Weiqiang Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; International Institute of Food Innovation Co., Ltd., Nanchang University, Nanchang 330200, PR China
| | - Xiaoyun Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China
| | - Yang Song
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China
| | - Linping Fan
- Departments of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, the 1(st) affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang 330209, PR China
| | - Jin Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China
| | - Luyu Yang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China
| | - Yang Liu
- Departments of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, the 1(st) affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang 330209, PR China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; International Institute of Food Innovation Co., Ltd., Nanchang University, Nanchang 330200, PR China.
| |
Collapse
|
2
|
Robinson TE, Clark C, Moakes RJA, Schofield Z, Moiemen N, Geoghegan JA, Grover LM. Simultaneous viscoelasticity and sprayability in antimicrobial acetic acid-alginate fluid gels. BIOMATERIALS ADVANCES 2025; 166:214051. [PMID: 39357110 DOI: 10.1016/j.bioadv.2024.214051] [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: 05/22/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
Acetic acid is a promising alternative to antibiotics for topical applications, particularly burn wounds, however its site specificity and retention are impaired by poor material properties. In this study, acetic acid was investigated as both the gelling agent and antimicrobial active in alginate fluid gels. The formed microstructure was found to be directly dependent on acetic acid concentration, leading to highly tuneable material properties. At clinically relevant concentrations of 2.5-5 % acetic acid, the fluid gels were elastically dominated at rest, with viscosities up to 7 orders of magnitude greater than acetic acid alone. These material properties imparted long term surface retention and microparticle barrier function, not seen with either acetic acid or alginate solutions. Most notably, sprayability was enhanced simultaneously with the increased viscosity and elasticity due to the introduction of a discretised microstructure, leading to a remarkable tenfold increase in spray coverage. Formulation was found not to inhibit antimicrobial activity, despite the less acidic pH, with common burn wound pathogens Staphylococcus aureus and Pseudomonas aeruginosa being equally susceptible to the fluid gels as to acetic acid solutions.
Collapse
Affiliation(s)
- Thomas E Robinson
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, UK.
| | - Callum Clark
- Department of Microbes, Infection & Microbiomes, College of Medicine & Health, University of Birmingham, UK; Institute of Microbiology & Infection, University of Birmingham, UK
| | - Richard J A Moakes
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, UK
| | - Zoe Schofield
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, UK
| | - Naiem Moiemen
- Department of Burns and Plastic Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Joan A Geoghegan
- Department of Microbes, Infection & Microbiomes, College of Medicine & Health, University of Birmingham, UK; Institute of Microbiology & Infection, University of Birmingham, UK
| | - Liam M Grover
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, UK
| |
Collapse
|
3
|
Zhang XT, Lin S, Wang XY, Guo HL, Cong YY, He X, Zhang CF, Yuan CS. Construction of a one-stop N-doped negatively charged carbon dot nanoplatform with antibacterial and anti-inflammatory dual activities for wound infection based on biocompatibility. J Colloid Interface Sci 2025; 677:1061-1074. [PMID: 39180841 DOI: 10.1016/j.jcis.2024.08.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
The development of bacterial resistance significantly contributes to the persistence of infections. Although previous studies have highlighted the benefits of metal-doped positive carbon nanodots in managing bacterial wound infections, their mechanism of action is relatively simple and they may pose potential hazards to human cells. Therefore, it is essential to develop a one-stop carbon dot nanoplatform that offers high biocompatibility, antibacterial properties, and anti-inflammatory activities for wound infection management. This study explores the antibacterial efficacy, without detectable resistance, and wound-healing potential of nitrogen-doped (N-doped) negatively charged carbon dots (TPP-CDs). These carbon dots are synthesized using tannic acid (TA), polyethylene polyamine, and polyethylene glycol (PEG) as precursors, with a focus on their biocompatibility. Numerous systematic studies have shown that TPP-CDs can effectively destroy bacterial biofilms and deoxyribonucleic acid (DNA), while also inducing oxidative stress, leading to a potent antimicrobial effect. TPP-CDs also demonstrate the ability to scavenge excess free radicals, promote cellular proliferation, and inhibit inflammatory factors, all of which contribute to improved wound healing. TPP-CDs also demonstrate favorable cell imaging capabilities. These findings suggest that N-doped negatively charged TPP-CDs hold significant potential for treating bacterial infections and offer practical insights for their application in the medical field.
Collapse
Affiliation(s)
- Xiao-Tong Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shuai Lin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Yan Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hui-Ling Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan-Yuan Cong
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xin He
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Chun-Feng Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Chun-Su Yuan
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
4
|
Uchiyama H, Kudo T, Yamaguchi T, Obana N, Watanabe K, Abe K, Miyazaki H, Toyofuku M, Nomura N, Akeda Y, Nakao R. Mucosal adjuvanticity and mucosal booster effect of colibactin-depleted probiotic Escherichia coli membrane vesicles. Hum Vaccin Immunother 2024; 20:2337987. [PMID: 38658133 PMCID: PMC11057659 DOI: 10.1080/21645515.2024.2337987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024] Open
Abstract
There is a growing interest in development of novel vaccines against respiratory tract infections, due to COVID-19 pandemic. Here, we examined mucosal adjuvanticity and the mucosal booster effect of membrane vesicles (MVs) of a novel probiotic E. coli derivative lacking both flagella and potentially carcinogenic colibactin (ΔflhDΔclbP). ΔflhDΔclbP-derived MVs showed rather strong mucosal adjuvanticity as compared to those of a single flagellar mutant strain (ΔflhD-MVs). In addition, glycoengineered ΔflhDΔclbP-MVs displaying serotype-14 pneumococcal capsular polysaccharide (CPS14+MVs) were well-characterized based on biological and physicochemical parameters. Subcutaneous (SC) and intranasal (IN) booster effects of CPS14+MVs on systemic and mucosal immunity were evaluated in mice that have already been subcutaneously prime-immunized with the same MVs. With a two-dose regimen, an IN boost (SC-IN) elicited stronger IgA responses than homologous prime-boost immunization (SC-SC). With a three-dose regimen, serum IgG levels were comparable among all tested regimens. Homologous immunization (SC-SC-SC) elicited the highest IgM responses among all regimens tested, whereas SC-SC-SC failed to elicit IgA responses in blood and saliva. Furthermore, serum IgA and salivary SIgA levels were increased with an increased number of IN doses administrated. Notably, SC-IN-IN induced not only robust IgG response, but also the highest IgA response in both serum and saliva among the groups. The present findings suggest the potential of a heterologous three-dose administration for building both systemic and mucosal immunity, e.g. an SC-IN-IN vaccine regimen could be beneficial. Another important observation was abundant packaging of colibactin in MVs, suggesting increased applicability of ΔflhDΔclbP-MVs in the context of vaccine safety.
Collapse
Affiliation(s)
- Hiroki Uchiyama
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
- Department of Vascular Surgery, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Toshifumi Kudo
- Department of Vascular Surgery, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Takehiro Yamaguchi
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Nozomu Obana
- Tsukuba Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Microbiology Research Center for Sustainability, University of Tsukuba, Tsukuba, Japan
| | - Kenji Watanabe
- Department of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kimihiro Abe
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Hidetaka Miyazaki
- Department of Oculoplastic, Orbital and Lacrimal Surgery, Aichi Medical University, Nagakute, Japan
- Department of Oral and Maxillofacial Surgery, Division of Oral Health Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Masanori Toyofuku
- Microbiology Research Center for Sustainability, University of Tsukuba, Tsukuba, Japan
| | - Nobuhiko Nomura
- Microbiology Research Center for Sustainability, University of Tsukuba, Tsukuba, Japan
| | - Yukihiro Akeda
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Ryoma Nakao
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
5
|
Ge M, Jiang F, Lin H. Nanocatalytic medicine enabled next-generation therapeutics for bacterial infections. Mater Today Bio 2024; 29:101255. [PMID: 39381264 PMCID: PMC11459013 DOI: 10.1016/j.mtbio.2024.101255] [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: 07/01/2024] [Revised: 09/08/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024] Open
Abstract
The rapid rise of antibiotic-resistant strains and the persistence of biofilm-associated infections have significantly challenged global public health. Unfortunately, current clinical high-dose antibiotic regimens and combination therapies often fail to completely eradicate these infections, which can lead to adverse side effects and further drug resistance. Amidst this challenge, however, the burgeoning development in nanotechnology and nanomaterials brings hopes. This review provides a comprehensive summary of recent advancements in nanomaterials for treating bacterial infections. Firstly, the research progress of catalytic therapies in the field of antimicrobials is comprehensively discussed. Thereafter, we systematically discuss the strategies of nanomaterials for anti-bacterial infection therapies, including endogenous response catalytic therapy, exogenous stimulation catalytic therapy, and catalytic immunotherapy, in order to elucidate the mechanism of nanocatalytic anti-infections. Based on the current state of the art, we conclude with insights on the remaining challenges and future prospects in this rapidly emerging field.
Collapse
Affiliation(s)
- Min Ge
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Feng Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Han Lin
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, China
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200331, China
| |
Collapse
|
6
|
Panhwar S, Çelikkan H, Evran E, Ekiz E, Ozkan Hukum K, Çetin D, Suludere Z, Hakki Boyaci I, Tamer U. Phage probe on RAFT polymer surface for rapid enumeration of E. coli K12. Bioelectrochemistry 2024; 160:108785. [PMID: 39094446 DOI: 10.1016/j.bioelechem.2024.108785] [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: 03/09/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
This study presents a simple, fast, and sensitive label-free sensing assay for the precise enumeration of modeled pathogenic Escherichia coli K12 (E. coli K12) bacteria for the first time. The method employs the covalent binding bacteriophage technique on the surface of a reversible addition-fragmentation chain transfer (RAFT) polymer film. The Nyquist plots obtained from electrochemical impedance spectroscopy (EIS) identified the charge transfer resistance Rct was calculated from a suitable electrochemical circuit model through an evaluation of the relevant parameter after the immobilization of the bacteriophage and the binding of specific E. coli K12. The impedimetric biosensor reveals specific and reproducible detection with sensitivity in the linear working range of 104.2-107.0 CFU/mL, a limit of detection (LOD) of 101.3 CFU/mL, and a short response time of 15 min. The SERS response validates the surface roughness and interaction of the SERS-tag with E. coli K12-modified electrodes. Furthermore, the covalently immobilized active phage selectivity was proved against various non-targeting bacterial strains in the presence of targeted E.coli K12 with a result of 94 % specificity and 98 % sensitivity. Therefore, the developed phage-based electrode surface can be used as a disposable, label-free impedimetric biosensor for rapid and real-time monitoring of serum samples.
Collapse
Affiliation(s)
- Sallahuddin Panhwar
- Department of Analytical Chemistry, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye; Department of Civil Engineering, National University of Sciences and Technology, Quetta 24090, Balochistan, Pakistan
| | - Hüseyin Çelikkan
- Gazi University, Faculty of Science, Department of Chemistry, Ankara, Türkiye
| | - Eylul Evran
- Department of Food Engineering, Faculty of Engineering, Hacettepe University, Beytepe 06800 Ankara, Türkiye
| | - Esra Ekiz
- Department of Food Engineering, Faculty of Engineering, Hacettepe University, Beytepe 06800 Ankara, Türkiye
| | - Kubra Ozkan Hukum
- Gazi University, Faculty of Science, Department of Chemistry, Ankara, Türkiye
| | - Demet Çetin
- Department of Mathematics and Science Education, Gazi Faculty of Education, Gazi University, Besevler, Ankara 06500, Türkiye
| | - Zekiye Suludere
- Faculty of Science, Department of Biology, Gazi University, 06500 Ankara, Türkiye
| | - Ismail Hakki Boyaci
- Department of Food Engineering, Faculty of Engineering, Hacettepe University, Beytepe 06800 Ankara, Türkiye
| | - Uğur Tamer
- Department of Analytical Chemistry, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye; METU MEMS Research and Application Center, Ankara, Türkiye.
| |
Collapse
|
7
|
Vargas-Cuebas GG, Sanchez CA, Bezold EL, Walker GM, Siddiqui S, Minbiole KPC, Wuest WM. Adaptations to cationic biocide exposure differentially influence virulence factors and pathogenicity in Pseudomonas aeruginosa. Virulence 2024; 15:2397503. [PMID: 39282885 PMCID: PMC11407422 DOI: 10.1080/21505594.2024.2397503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Cationic biocides (CBs), which include quaternary ammonium compounds (QACs), are employed to mitigate the spread of infectious bacteria, but resistance to such surface disinfectants is rising. CB exposure can have profound phenotypic implications that extend beyond allowing microorganisms to persist on surfaces. Pseudomonas aeruginosa is a deadly bacterial pathogen that is intrinsically tolerant to a wide variety of antimicrobials and is commonly spread in healthcare settings. In this study, we pursued resistance selection assays to the QAC benzalkonium chloride and quaternary phosphonium compound P6P-10,10 to assess the phenotypic effects of CB exposure in P. aeruginosa PAO1 and four genetically diverse, drug-resistant clinical isolates. In particular, we sought to examine how CB exposure affects defensive strategies and the virulence-associated "offensive" strategies in P. aeruginosa. We demonstrated that development of resistance to BAC is associated with increased production of virulence-associated pigments and alginate as well as pellicle formation. In an in vivo infection model, CB-resistant PAO1 exhibited a decreased level of virulence compared to wild type, potentially due to an observed fitness cost in these strains. Taken together, these results illustrate the significant consequence CB resistance exerts on the virulence-associated phenotypes of P. aeruginosa.
Collapse
Affiliation(s)
- Germán G Vargas-Cuebas
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Elise L Bezold
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
8
|
Wassil J, Sisti M, Fairman J, Rankin B, Clark J, Bennett S, Johnson D, Migone TS, Nguyen K, Paschenko A, Sauer P, Iki S, Hanson ME, Simon JK. A phase 2, randomized, blinded, dose-finding, controlled clinical trial to evaluate the safety, tolerability, and immunogenicity of a 24-valent pneumococcal conjugate vaccine (VAX-24) in healthy adults 65 years and older. Vaccine 2024; 42:126124. [PMID: 39025698 DOI: 10.1016/j.vaccine.2024.07.025] [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: 02/14/2024] [Revised: 06/21/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024]
Abstract
Despite current polysaccharide and conjugate vaccine use, pneumococcal diseases remain prevalent in older adults. VAX-24 is a 24-valent pneumococcal conjugate vaccine (PCV) containing eCRM, a proprietary carrier protein with non-native amino acids (para-azidomethyl-L-phenylalanine) that undergo site-specific conjugation to pneumococcal polysaccharides that have been activated with a small-molecule linker (dibenzocyclooctyne). Site-specific conjugation utilizing click chemistry enables consistent exposure of T-cell epitopes, reduction in carrier protein to pneumococcal polysaccharide ratio, and enhances manufacturing process consistency to improve PCVs by increasing serotype coverage while minimizing carrier suppression. Healthy adults aged 65 or older were randomized in a 1:1:1:1 ratio to receive a single injection of VAX-24 at 1 of 3 dose levels (1.1, 2.2, or a mixed dose of 2.2 or 4.4 mcg) or Prevnar 20® (PCV20) in a phase 2, blinded study. Primary outcome measures were solicited local and systemic events within 7 days post-vaccination, unsolicited adverse events (AEs) within 1 month, and serious AEs, medically attended AEs, or new onset of chronic disease within 6 months of vaccination. Serotype-specific opsonophagocytic activity (OPA) and immunoglobulin G (IgG) were measured pre-vaccination and at 1 month post-vaccination. Of 207 participants enrolled, 200 completed the trial. Safety profiles were comparable across the three VAX-24 doses and PCV20. Robust OPA and IgG immune responses were seen for all 24 serotypes. On average, immune responses to VAX-24 2.2 mcg dose were similar or higher compared to PCV20. In adults ≥ 65 years, VAX-24 had a safety profile similar to PCV20 through six months post-vaccination and induced robust OPA and IgG responses to all 24 serotypes, supporting prior data showing that site-specific conjugation allows for increased serotype coverage with similar or higher immune response vs other PCVs. The outcome of this phase 2 study further supports use of VAX-24 2.2 mcg dose in phase 3 trials. Clinicaltrials.gov: NCT05297578.
Collapse
Affiliation(s)
- J Wassil
- Vaxcyte, Inc., San Carlos, CA, USA.
| | - M Sisti
- Vaxcyte, Inc., San Carlos, CA, USA
| | | | | | - J Clark
- Charlottesville Medical Research, Charlottesville, VA, USA
| | | | | | | | - K Nguyen
- Vaxcyte, Inc., San Carlos, CA, USA
| | | | - P Sauer
- Vaxcyte, Inc., San Carlos, CA, USA
| | - S Iki
- Vaxcyte, Inc., San Carlos, CA, USA
| | | | | |
Collapse
|
9
|
Mohamadou M, Kagning Tsinda E, Hina Q, Javed K, Dongang Nana RR, Ngonde Essome MC, Essama SR, Sattar S, Kamga HG, Javed S. Genome sequence of multi-drug-resistant Staphyloccocus aureus isolated from a clinical sample collected in Maroua, Cameroon. Microbiol Resour Announc 2024; 13:e0065624. [PMID: 39345203 PMCID: PMC11556012 DOI: 10.1128/mra.00656-24] [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: 06/22/2024] [Accepted: 08/24/2024] [Indexed: 10/01/2024] Open
Abstract
Staphylococcus aureus is a major pathogen of public health concern due to its implications in pathologies and its increasing antimicrobial resistance. Here, we present the draft genome of a 2.7-Mbp S. aureus isolate obtained from a pus swab sampled in Cameroon. The GC content of the draft genome is 32%.
Collapse
Affiliation(s)
- Mansour Mohamadou
- Centre for Research on Health and Priority Pathologies, Institute of Medical Research and Medicinal Plants Studies, Yaoundé, Cameroon
- Biosciences Department, COMSATS University Islamabad, Islamabad, Pakistan
- Department of Microbiology, Faculty of Science, University of Yaounde, Yaoundé, Cameroon
| | - Emmanuel Kagning Tsinda
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Qudsia Hina
- Biosciences Department, COMSATS University Islamabad, Islamabad, Pakistan
| | - Kashaf Javed
- Biosciences Department, COMSATS University Islamabad, Islamabad, Pakistan
| | - Rodrigue Roman Dongang Nana
- Centre for Research on Health and Priority Pathologies, Institute of Medical Research and Medicinal Plants Studies, Yaoundé, Cameroon
| | - Marie Chantal Ngonde Essome
- Centre for Research on Health and Priority Pathologies, Institute of Medical Research and Medicinal Plants Studies, Yaoundé, Cameroon
| | - Sara Riwom Essama
- Department of Microbiology, Faculty of Science, University of Yaounde, Yaoundé, Cameroon
| | - Sadia Sattar
- Biosciences Department, COMSATS University Islamabad, Islamabad, Pakistan
| | - Hortense Gonsu Kamga
- Department of Microbiology, Hematology and Infectious Diseases, Faculty of Medicine and Biomedical Sciences, University of Yaounde, Yaoundé, Cameroon
| | - Sundus Javed
- Biosciences Department, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
10
|
Sun R, Zhao D, Yu X, Zhang F, You R, Luo X, Li L. Discovery of a family of menaquinone-targeting cyclic lipodepsipeptides for multidrug-resistant Gram-positive pathogens. Commun Biol 2024; 7:1453. [PMID: 39506023 PMCID: PMC11541763 DOI: 10.1038/s42003-024-07159-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Menaquinone (MK) in bacterial membrane is an attractive target for the development of novel therapeutic agents. Mining the untapped chemical diversity encoded by Gram-negative bacteria presents an opportunity to identify additional MK-binding antibiotics (MBAs). By MK-binding motif searching of bioinformatically predicted linear non-ribosomal peptides from 14,298 sequenced genomes of 45 underexplored Gram-negative bacterial genera, here we identify a novel MBA structural family, including silvmeb and pseudomeb, using structure prediction-guided chemical synthesis. Both MBAs show rapid bacteriolysis by MK-dependent membrane depolarization to achieve their potent activities against a panel of Gram-positive pathogens. Furthermore, both MBAs are proven to be effective against methicillin-resistant Staphylococcus aureus in a murine peritonitis-sepsis model. Our findings suggest that MBAs are a kind of structurally diverse and still underexplored antibacterial lipodepsipeptide class. The interrogation of underexplored bacterial taxa using synthetic bioinformatic natural product methods is an appealing strategy for discovering novel biomedically relevant agents to confront the crisis of antimicrobial resistance.
Collapse
Affiliation(s)
- Runze Sun
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
| | - Di Zhao
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
| | - Xuchang Yu
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
- State Key Laboratory of Bioreactor Engineering and School of Biotechnology, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Fei Zhang
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
- College of Life Science, Tarim University, E1487 Tarim Avenue, Alar, 43300, China
| | - Ruixiang You
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China
| | - Xiaoxia Luo
- College of Life Science, Tarim University, E1487 Tarim Avenue, Alar, 43300, China
| | - Lei Li
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai, 200240, China.
| |
Collapse
|
11
|
Borhade P, LeBoa C, Jayaprasad N, Date K, Haldar P, Harvey P, Shimpi R, An Q, Zhang C, Horng L, Fagerli K, Yewale VN, Daruwalla S, Dharmapalan D, Gavhane J, Joshi S, Rai R, Rathod V, Shetty K, Warrier DS, Yadav S, Chakraborty D, Bahl S, Katkar A, Kunwar A, Andrews JR, Bhatnagar P, Dutta S, Luby SP, Hoffman SA. Factors Influencing Vaccine Receipt During a 2018 Pediatric Typhoid Conjugate Vaccine Campaign in Navi Mumbai, India. Am J Trop Med Hyg 2024; 111:1060-1065. [PMID: 39348826 PMCID: PMC11542534 DOI: 10.4269/ajtmh.24-0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/31/2024] [Indexed: 10/02/2024] Open
Abstract
In 2018, the Navi Mumbai Municipal Corporation implemented phase 1 of a public sector typhoid conjugate vaccine campaign in Navi Mumbai, India, targeting all children aged 9 months to 14 years within its administrative boundaries. To assess associations with receipt of vaccine in phase 1, we used generalized estimating equations to calculate estimates of vaccination by child-, household-, and community-level demographics (child education and age; household head education, income, and occupation; community informal settlement percent). Campaign vaccine receipt was most associated with children enrolled in school (odds ratio [OR] = 3.84, 95% CI: 2.18-6.77), the lowest household income tertile when divided into three equal parts (OR = 1.64, 95% CI: 1.43-1.84), and lower community-level socioeconomic status (OR = 1.06, 95% CI: 1.04-1.08 per 10% informal settlement proportion). The campaign was successful in reaching the most underserved populations of its target communities.
Collapse
Affiliation(s)
- Priyanka Borhade
- World Health Organization–Country Office for India, National Public Health Surveillance Project, New Delhi, India
| | - Christopher LeBoa
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Niniya Jayaprasad
- World Health Organization–Country Office for India, National Public Health Surveillance Project, New Delhi, India
| | - Kashmira Date
- Global Immunization Division, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Pradeep Haldar
- Ministry of Health & Family Welfare, Government of India, New Delhi, India
| | - Pauline Harvey
- World Health Organization–Country Office for India, National Public Health Surveillance Project, New Delhi, India
| | - Rahul Shimpi
- World Health Organization–Country Office for India, National Public Health Surveillance Project, New Delhi, India
| | - Qian An
- Global Immunization Division, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Chenhua Zhang
- Global Immunization Division, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Lily Horng
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kirsten Fagerli
- Global Immunization Division, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Vijay N. Yewale
- Dr. Yewale Multispecialty Hospital for Children, Navi Mumbai, India
| | - Savita Daruwalla
- Department of Pediatrics, NMMC General Hospital, Navi Mumbai, India
| | | | - Jeetendra Gavhane
- Department of Pediatrics, MGM New Bombay Hospital, MGM Medical College, Navi Mumbai, India
| | - Shrikrishna Joshi
- Dr. Joshi’s Central Clinical Microbiology Laboratory, Navi Mumbai, India
| | - Rajesh Rai
- Department of Pediatrics & Neonatology, Dr. D. Y. Patil Medical College and Hospital, Navi Mumbai, India
| | - Varsha Rathod
- Rajmata Jijau Hospital, Airoli (NMMC), Navi Mumbai, India
| | - Keertana Shetty
- Department of Microbiology, Dr. D. Y. Patil Medical College and Hospital, Navi Mumbai, India
| | | | - Shalini Yadav
- Department of Microbiology, MGM New Bombay Hospital, Navi Mumbai, India
| | - Debjit Chakraborty
- National Institute of Cholera and Enteric Diseases, Indian Council of Medical Research, Kolkata, India
| | - Sunil Bahl
- World Health Organization South-East Asia Regional Office, New Delhi, India
| | - Arun Katkar
- Ministry of Health & Family Welfare, Government of India, New Delhi, India
| | - Abhishek Kunwar
- Ministry of Health & Family Welfare, Government of India, New Delhi, India
| | - Jason R. Andrews
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Pankaj Bhatnagar
- Ministry of Health & Family Welfare, Government of India, New Delhi, India
| | - Shanta Dutta
- National Institute of Cholera and Enteric Diseases, Indian Council of Medical Research, Kolkata, India
| | - Stephen P. Luby
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Seth A. Hoffman
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
12
|
Narciso AR, Dookie R, Nannapaneni P, Normark S, Henriques-Normark B. Streptococcus pneumoniae epidemiology, pathogenesis and control. Nat Rev Microbiol 2024:10.1038/s41579-024-01116-z. [PMID: 39506137 DOI: 10.1038/s41579-024-01116-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Infections caused by Streptococcus pneumoniae (also known as pneumococci) pose a threat to human health. Pneumococcal infections are the most common cause of milder respiratory tract infections, such as otitis and sinusitis, and of more severe diseases, including pneumonia (with or without septicaemia) and meningitis. The introduction of pneumococcal conjugate vaccines in the childhood vaccination programme in many countries has led to a notable decrease of severe invasive pneumococcal disease in vaccinated children. However, infections caused by non-vaccine types have concurrently increased, causing invasive pneumococcal disease in unvaccinated populations (such as older adults), which has hampered the effect of these vaccines. Moreover, emerging antibiotic resistance is threatening effective therapy. Thus, new approaches are needed for the treatment and prevention of pneumococcal infections, and recent advances in the field may pave the way for new strategies. Recently, several important findings have been gained regarding pneumococcal epidemiology, genomics and the effect of the introduction of pneumococcal conjugate vaccines and of the COVID-19 pandemic. Moreover, elucidative pathogenesis studies have shown that the interactions between pneumococcal virulence factors and host receptors may be exploited for new therapies, and new vaccine candidates have been suggested. In this Review, we summarize some recent findings from clinical disease to basic pathogenesis studies that may be of importance for future control strategies.
Collapse
Affiliation(s)
- Ana Rita Narciso
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rebecca Dookie
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Priyanka Nannapaneni
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Staffan Normark
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
13
|
Shi N, Wang ZJ, Shi YZ, Jiang L, Zhu YY, He XC, Zhou ZS, Wei MZ, Zhao YL, Luo XD. New resorcylic acid derivatives of Lysimachia tengyuehensis against MRSA and VRE by interfering with bacterial metabolic imbalance. Eur J Med Chem 2024; 277:116714. [PMID: 39096819 DOI: 10.1016/j.ejmech.2024.116714] [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: 05/19/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/05/2024]
Abstract
The abuse of antibiotics leads to the rapid spread of bacterial resistance, which seriously threatens human life and health. Now, 8 resorcylic acid derivatives, including 4 new compounds (1-4) were isolated from Lysimachia tengyuehensis by bio-guided isolation, and they inhibited both methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE) (MIC = 4-8 μg/mL). Notably, 1 and 2 rapidly killed MRSA and VRE within 40 min without drug resistance in 20 days. Mechanically, they potently disrupted biofilm and cell membrane by interfering with bacterial metabolic imbalance. The structure-activity relationship (SAR) revealed that the lipophilic long carbon chains (C-5/C-6) and hydrophilic hydroxyl/carboxyl groups were essential for the anti-MRSA and VRE bioactivity. Additionally, they effectively recovered MRSA-infected skin wounds and VRE-infected peritoneal in vivo. Resorcylic acid derivatives showed significant anti-MRSA and VRE bioactivity in vitro and in vivo with potential application for the first time.
Collapse
Affiliation(s)
- Nian Shi
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yang-Zhu Shi
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Ling Jiang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Xing-Chao He
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Zhong-Shun Zhou
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Mei-Zhen Wei
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China.
| |
Collapse
|
14
|
Le Brun G, Nuytten M, Leprince A, Glinel K, Gillis A, Mahillon J, Raskin JP. Rapid and Specific Detection of Bacillus cereus Using Phage Protein-Based Lateral Flow Assays. ACS APPLIED BIO MATERIALS 2024. [PMID: 39498971 DOI: 10.1021/acsabm.4c00965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Rapid and precise diagnostic techniques are essential for identifying foodborne pathogens, including Bacillus cereus (B. cereus), which poses significant challenges to food safety. Traditional detection methods are limited by long incubation times and high costs. In this context, gold nanoparticle (AuNP)-based lateral flow assays (LFAs) are emerging as valuable tools for rapid screening. However, the use of antibodies in LFAs faces challenges, including complex production processes, ethical concerns, or variability. Here, we address these challenges by proposing an innovative approach using bacteriophage-derived proteins for pathogen detection on LFAs. We used the engineered endolysin cell-wall-binding domain (CBD) and distal tail proteins (Dit) from bacteriophages that specifically target B. cereus. The protein-binding properties, essential for the formation of efficient capture and detection biointerfaces in LFAs, were extensively characterized from the microstructural to the LFA device level. Machine-learning models leverage knowledge of the protein sequence to predict advantageous protein orientations on the nitrocellulose membrane and AuNPs. The study of the biointerface binding quantified the degree of attachment of AuNPs to bacteria, providing, for the first time, a microscopic model of the number of AuNPs binding to bacteria. It highlighted the binding of up to one hundred 40 nm AuNPs per bacterium in conditions mimicking LFAs. Eventually, phage proteins were demonstrated as efficient bioreceptors in a straightforward LFA prototype combining the two proteins, providing a rapid colorimetric response within 15 min upon the detection of 105 B. cereus cells. Recombinantly produced phage binding proteins present an opportunity to generate a customizable library of proteins with precise binding capabilities, offering a cost-effective and ethical alternative to antibodies. This study enhances our understanding of phage protein biointerfaces, laying the groundwork for their utilization as efficient bioreceptors in LFAs and rapid point-of-care diagnostic assays, thus potentially strengthening public health measures.
Collapse
Affiliation(s)
- Grégoire Le Brun
- Institute of Information and Communication Technologies, Electronics and Applied Mathematics, UCLouvain, Place du Levant 1, 1348 Louvain-la-Neuve, Belgium
| | - Manon Nuytten
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, UCLouvain, Croix du Sud 2/12, 1348 Louvain-la-Neuve, Belgium
| | - Audrey Leprince
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, UCLouvain, Croix du Sud 2/12, 1348 Louvain-la-Neuve, Belgium
| | - Karine Glinel
- Institute of Condensed Matter and Nanosciences, UCLouvain, Croix du Sud 1, 1348 Louvain-la-Neuve, Belgium
| | - Annika Gillis
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, UCLouvain, Croix du Sud 2/12, 1348 Louvain-la-Neuve, Belgium
| | - Jacques Mahillon
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, UCLouvain, Croix du Sud 2/12, 1348 Louvain-la-Neuve, Belgium
| | - Jean-Pierre Raskin
- Institute of Information and Communication Technologies, Electronics and Applied Mathematics, UCLouvain, Place du Levant 1, 1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
15
|
Sun M, Ma C, Emran MY, Kotb A, Bai J, Zhou M. A fully integrated wireless microfluidic immunosensing system for portable monitoring of Staphylococcus aureus. Talanta 2024; 283:127158. [PMID: 39515059 DOI: 10.1016/j.talanta.2024.127158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/25/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
The advanced devices that function fully without the need for external accessories are regarded as a pinnacle goal in the design and construction of modern ones. Staphylococcus aureus (S. aureus), a prominent human pathogen, is responsible for causing a wide variety of infections and chronic diseases. Herein, we present the first instance of a fully integrated wireless microfluidic immunosensing system (FIWMIS) capable of conducting point-of-care S. aureus monitoring in real samples of S. aureus-spiked commercial purified drinking water and S. aureus-spiked watermelon juice. The development of the proposed FIWMIS became a reality by conquering significant engineering hurdles in seamlessly integrating a microfluidic unit for liquid sample transport without the need of an external pump, an immunosensing unit for S. aureus monitoring, and an electronic control unit for signal conversion and wireless transmission. Such full integration culminated in a FIWMIS that upholds its pump-free, wireless, and low-cost characteristics for portable monitoring of S. aureus.
Collapse
Affiliation(s)
- Mimi Sun
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Analysis and Testing Center, Department of Chemistry, Northeast Normal University, Changchun, Jilin Province, 130024, China
| | - Chongbo Ma
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Analysis and Testing Center, Department of Chemistry, Northeast Normal University, Changchun, Jilin Province, 130024, China
| | - Mohammed Y Emran
- Chemistry Department, Faculty of Science, Al-Azhar University, Assiut, 71524, Egypt
| | - Ahmed Kotb
- Chemistry Department, Faculty of Science, Al-Azhar University, Assiut, 71524, Egypt
| | - Jing Bai
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Analysis and Testing Center, Department of Chemistry, Northeast Normal University, Changchun, Jilin Province, 130024, China.
| | - Ming Zhou
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Analysis and Testing Center, Department of Chemistry, Northeast Normal University, Changchun, Jilin Province, 130024, China.
| |
Collapse
|
16
|
Cao Z, Wong F, Choudhury AK, Kamath PS, Topazian M, Torre A, Hayes PC, George J, Idilman R, Seto WK, Desalegn H, Alvares-da-Silva MR, Bush BJ, Thacker LR, Xie Q, Bajaj JS. Global prevalence and characteristics of infections and clinical outcomes in hospitalised patients with cirrhosis: a prospective cohort study for the CLEARED Consortium. Lancet Gastroenterol Hepatol 2024; 9:997-1009. [PMID: 39243795 DOI: 10.1016/s2468-1253(24)00224-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Infections have a poor prognosis in inpatients with cirrhosis. We aimed to determine regional variations in infections and their association with clinical outcomes in a global cohort of inpatients with cirrhosis. METHODS In this prospective cohort study initiated by the CLEARED Consortium, we enrolled adults (aged >18 years) with cirrhosis who were non-electively admitted to 98 hospitals from 26 countries or regions across six continents between Nov 5, 2021, and Dec 10, 2022. Data at admission, during hospitalisation, and for 30 days after discharge were collected through patient reports and chart reviews. Collected data included demographics; country and country income level per World Bank classifications (high-income countries [HICs], upper-middle-income countries [UMICs], and low-income or lower-middle-income countries [L-LMICs]); comorbidities; characteristics related to cirrhosis and the infections, including types, culture results, and drug resistance profile; antibiotic use; and disease course while hospitalised and for 30 days post-discharge. The primary outcome was in-hospital death or hospice referral in those with versus those without an admission infection (defined by the presence of infection on or within 48 h of admission). Multivariable log-binomial regression for in-hospital death or hospice referral was performed to identify risk factors. FINDINGS Of 4550 patients screened, 4238 patients (mean age 56·1 years [SD 13·3]; 2711 [64·0%] male and 1527 [36·0%] female) with complete data were enrolled. 1351 (31·9%) had admission infections. A higher proportion of patients in L-LMICs had infections (318 [41·7%] of 762 vs 444 [58·3%] without infection) than in UMICs (588 [30·6%] of 1922 vs 1334 [69·4%]) or HICs (445 [28·6%] of 1554 vs 1109 [71·4%]). Patients with admission infections had worse severity of cirrhosis and were more likely to have had an infection or been hospitalised in the preceding 6 months. The most common specific infection types were spontaneous bacterial peritonitis (391 [28·9%] of 1351), pneumonia (233 [17·2%]), and urinary tract infections (193 [14·3%]). 549 (40·6%) patients were culture-positive for bacterial or fungal infections, with the lowest culture-positive rates in Africa and mainland China. Most of the isolated organisms were Gram-negative (345 [63%] of 549), then Gram-positive (157 [29%]), and then fungi or mixed (47 [9%]), with Escherichia coli, Klebsiella pneumoniae, and Enterococcus spp being the top three isolated pathogens. The overall rate of drug resistance was 40% (220 of 549 with positive cultures), being highest in UMICs. The most used empirical antimicrobials were third-generation cephalosporins (453 [37%] of 1241), followed by the broad-spectrum β-lactams and β-lactamase inhibitors (289 [23%]). De-escalation was observed in 62 (20%) of 304 patients who had their antibiotics changed. Patients with versus without admission infections had a higher rate of in-hospital death or hospice transfer (299 [22·1%] of 1351 vs 232 [8·0%] of 2887; p<0·0001), a result replicated in multivariable analysis (adjusted risk ratio 1·75 [95% CI 1·42-2·06]; p<0·0001). Older age, self-reported female gender, not being in a HIC, lactulose use, and higher MELD-Na score were also associated with in-hospital death or hospice transfer on multivariable analysis. INTERPRETATION In the CLEARED Consortium cohort of inpatients with cirrhosis, the rates and types of infections, causative organisms, and culture-positivity varied substantially across regions, and infections were associated with a higher mortality risk. Culture positivity, which guides appropriate antibiotic use, was low. Taking a global perspective, considering regional variations in infections, drug resistance, and resources, could help to alleviate disparities in burden and outcomes. FUNDING US Department of Veterans Affairs, the Richmond Institute for Veterans Research, the National Natural Science Foundation of China, Shanghai Rising-Star Program, the National Council for Scientific and Technological Development of Brazil, and Shanghai Municipal Key Clinical Specialty.
Collapse
Affiliation(s)
- Zhujun Cao
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Florence Wong
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ashok K Choudhury
- Department of Hepatology, Institute for Liver and Biliary Sciences, New Delhi, India
| | - Patrick S Kamath
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mark Topazian
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Gastroenterology and Hepatology Unit, St Paul's Hospital, Millennium Medical College, Addis Ababa, Ethiopia
| | - Aldo Torre
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Peter C Hayes
- Hepatology, Division of Health Sciences, Deanery of Clinical Sciences, University of Edinburgh, Edinburgh, UK
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, NSW, Australia
| | - Ramazan Idilman
- Department of Gastroenterology, Ankara University School of Medicine, Ankara, Turkey
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Hailemichael Desalegn
- Gastroenterology and Hepatology Unit, St Paul's Hospital, Millennium Medical College, Addis Ababa, Ethiopia
| | - Mario Reis Alvares-da-Silva
- Department of Hepatology, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Brian J Bush
- Department of Population Health, Virginia Commonwealth University, Richmond, VA, USA
| | - Leroy R Thacker
- Department of Population Health, Virginia Commonwealth University, Richmond, VA, USA
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology, and Nutrition and Richmond VA Medical Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
17
|
Cai S, Wang L, Cui X, Zou X, Zheng S, Liu F, Shi C, Li Y, Zhang Z. Rhein-loaded chitosan nanoparticles for treatment of MRSA-infected wound. Int J Biol Macromol 2024; 279:135360. [PMID: 39242008 DOI: 10.1016/j.ijbiomac.2024.135360] [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: 07/21/2024] [Revised: 08/25/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The multi-drug resistance of methicillin-resistant Staphylococcus aureus (MRSA) and complex wound microenvironment challenge the repair of MRSA infected wound. Herein, in this study, α-tocopherol modified glycol chitosan (TG) nanoparticles encapsulated with phytochemical rhein (Rhein@TG NPs) were prepared for comprehensive anti-infection and promotion of MRSA infected wound healing. Rhein@TG NPs could not only specifically release rhein in the infection site in response to low pH and lipase of infectious microenvironment, but also up-regulated M1 macrophage polarization in the infection stage, thus achieving synergistically bacterial elimination with low possibility of developing resistance. Additionally, the NPs reduced the levels of pro-inflammatory factors in the post-infection stage, scavenged the ROS, promoted cell migration and angiogenesis, which significantly improved the microenvironment of infected wound healing. Therefore, this antibiotic-free NPs enabling anti-infection and promotion of wound healing provides a new and long-term strategy for the treatment of MRSA infected wound.
Collapse
Affiliation(s)
- Shuang Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Linlin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Xilong Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Xinshu Zou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Sidi Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Fanhui Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Chun Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China.
| | - Zhiyun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China.
| |
Collapse
|
18
|
Lipworth S, Crook D, Walker AS, Peto T, Stoesser N. Exploring uncatalogued genetic variation in antimicrobial resistance gene families in Escherichia coli: an observational analysis. THE LANCET. MICROBE 2024; 5:100913. [PMID: 39378891 DOI: 10.1016/s2666-5247(24)00152-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Antimicrobial resistance (AMR) in Escherichia coli is a global problem associated with substantial morbidity and mortality. AMR-associated genes are typically annotated based on similarity to variants in a curated reference database, with the implicit assumption that uncatalogued genetic variation within these is phenotypically unimportant. In this study, we evaluated the performance of the AMRFinder tool and, subsequently, the potential for discovering new AMR-associated gene families and characterising variation within existing ones to improve genotype-to-susceptibility phenotype predictions in E coli. METHODS In this cross-sectional study of international genome sequence data, we assembled a global dataset of 9001 E coli sequences from five publicly available data collections predominantly deriving from human bloodstream infections from: Norway, Oxfordshire (UK), Thailand, the UK, and Sweden. 8555 of these sequences had linked antibiotic susceptibility data. Raw reads were assembled using Shovill and AMR genes (relevant to amoxicillin-clavulanic acid, ampicillin, ceftriaxone, ciprofloxacin, gentamicin, piperacillin-tazobactam, and trimethoprim) extracted using the National Center for Biotechnology Information AMRFinder tool (using both default and strict [100%] coverage and identity filters). We assessed the predictive value of the presence of these genes for predicting resistance or susceptibility against US Food and Drug Administration thresholds for major and very major errors. Mash was used to calculate the similarity between extracted genes using Jaccard distances. We empirically reclustered extracted gene sequences into AMR-associated gene families (≥70% match) and antibiotic-resistance genes (ARGs; 100% match) and categorised these according to their frequency in the dataset. Accumulation curves were simulated and correlations between gene frequency in the Oxfordshire and other datasets calculated using the Spearman coefficient. Firth regression was used to model the association between the presence of blaTEM-1 variants and amoxicillin-clavulanic acid or piperacillin-tazobactam resistance, adjusted for the presence of other relevant ARGs. FINDINGS The performance of the AMRFinder database for genotype-to-phenotype predictions using strict 100% identity and coverage thresholds did not meet US Food and Drug Administration thresholds for any of the seven antibiotics evaluated. Relaxing filters to default settings improved sensitivity with a specificity cost. For all antibiotics, most explainable resistance was associated with the presence of a small number of genes. There was a proportion of resistance that could not be explained by known ARGs; this ranged from 75·1% for amoxicillin-clavulanic acid to 3·4% for ciprofloxacin. Only 18 199 (51·5%) of the 35 343 ARGs detected had a 100% identity and coverage match in the AMRFinder database. After empirically reclassifying genes at 100% nucleotide sequence identity, we identified 1042 unique ARGs, of which 126 (12·1%) were present ten times or more, 313 (30·0%) were present between two and nine times, and 603 (57·9%) were present only once. Simulated accumulation curves revealed that discovery of new (100% match) ARGs present more than once in the dataset plateaued relatively quickly, whereas new singleton ARGs were discovered even after many thousands of isolates had been included. We identified a strong correlation (Spearman coefficient 0·76 [95% CI 0·73-0·80], p<0·0001) between the number of times an ARG was observed in Oxfordshire and the number of times it was seen internationally, with ARGs that were observed six times in Oxfordshire always being found elsewhere. Finally, using the example of blaTEM-1, we showed that uncatalogued variation, including synonymous variation, is associated with potentially important phenotypic differences; for example, two common, uncatalogued blaTEM-1 alleles with only synonymous mutations compared with the known reference were associated with reduced resistance to amoxicillin-clavulanic acid (adjusted odds ratio 0·58 [95% CI 0·35-0·95], p=0·031) and piperacillin-tazobactam (0·50 [95% CI 0·29-0·82], p=0·005). INTERPRETATION We highlight substantial uncatalogued genetic variation with respect to known ARGs, although a relatively small proportion of these alleles are repeatedly observed in a large international dataset suggesting strong selection pressures. The current approach of using fuzzy matching for ARG detection, ignoring the unknown effects of uncatalogued variation, is unlikely to be acceptable for future clinical deployment. The association of synonymous mutations with potentially important phenotypic differences suggests that relying solely on amino acid-based gene detection to predict resistance is unlikely to be sufficient. Finally, the inability to explain all resistance using existing knowledge highlights the importance of new target gene discovery. FUNDING National Institute for Health and Care Research, Wellcome, and UK Medical Research Council.
Collapse
Affiliation(s)
- Samuel Lipworth
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Derrick Crook
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK; NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at University of Oxford in partnership with UKHSA, Oxford, UK
| | - A Sarah Walker
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK; NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at University of Oxford in partnership with UKHSA, Oxford, UK
| | - Tim Peto
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Nicole Stoesser
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK; NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at University of Oxford in partnership with UKHSA, Oxford, UK
| |
Collapse
|
19
|
Jayaraman A, Walachowski S, Bosmann M. The complement system: A key player in the host response to infections. Eur J Immunol 2024; 54:e2350814. [PMID: 39188171 DOI: 10.1002/eji.202350814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024]
Abstract
Infections are one of the most significant healthcare and economic burdens across the world as underscored by the recent coronavirus pandemic. Moreover, with the increasing incidence of antimicrobial resistance, there is an urgent need to better understand host-pathogen interactions to design effective treatment strategies. The complement system is a key arsenal of the host defense response to pathogens and bridges both innate and adaptive immunity. However, in the contest between pathogens and host defense mechanisms, the host is not always victorious. Pathogens have evolved several approaches, including co-opting the host complement regulators to evade complement-mediated killing. Furthermore, deficiencies in the complement proteins, both genetic and therapeutic, can lead to an inefficient complement-mediated pathogen eradication, rendering the host more susceptible to certain infections. On the other hand, overwhelming infection can provoke fulminant complement activation with uncontrolled inflammation and potentially fatal tissue and organ damage. This review presents an overview of critical aspects of the complement-pathogen interactions during infection and discusses perspectives on designing therapies to mitigate complement dysfunction and limit tissue injury.
Collapse
Affiliation(s)
- Archana Jayaraman
- Department of Medicine, Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Sarah Walachowski
- Department of Medicine, Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Markus Bosmann
- Department of Medicine, Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
20
|
Passaris I, Depickère S, Braeye T, Mukovnikova M, Vodolazkaia A, Abels C, Cuypers L, Desmet S, Ceyssens PJ. Non-invasive Streptococcus pneumoniae infections are associated with different serotypes than invasive infections, Belgium, 2020 to 2023. Euro Surveill 2024; 29:2400108. [PMID: 39512163 PMCID: PMC11544722 DOI: 10.2807/1560-7917.es.2024.29.45.2400108] [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: 02/15/2024] [Accepted: 05/02/2024] [Indexed: 11/15/2024] Open
Abstract
BackgroundDespite widely implemented pneumococcal vaccination programmes, Streptococcus pneumoniae remains a global risk for human health. Streptococcus pneumoniae can cause invasive (IPD) or non-invasive pneumococcal disease (NIPD). Surveillance is mainly focusing on IPD, assessing the full impact of pneumococcal vaccination programmes on pneumococcal disease is challenging.AimWe aimed to prospectively investigate serotype distribution and antimicrobial resistance (AMR) of S. pneumoniae isolates from patients with NIPD and compare with data on IPD isolates and with a 2007-2008 dataset on NIPD.MethodsBetween September 2020 and April 2023, we collected isolates and patient data from patients with NIPD from 23 clinical laboratories in Belgium. Capsular typing was performed by a validated Fourier-Transform Infrared spectroscopic method, and AMR was assessed with broth microdilution, using the European Committee on Antimicrobial Susceptibility Testing (EUCAST) clinical breakpoints.ResultsWe received S. pneumoniae isolates from 1,008 patients with lower respiratory tract infections (n = 760), otitis media (n = 190) and sinusitis (n = 58). Serotype 3 was the most prevalent serotype among the NIPD isolates. Serotypes not included in the 20-valent pneumococcal conjugate vaccine (PCV20) were significantly more common among the NIPD than among the IPD isolates. Antimicrobial resistance levels were significantly higher among the NIPD isolates (n = 539; 2020-2022) compared with the IPD isolates (n = 2,344; 2021-2022). Resistance to several β-lactam antimicrobials had increased significantly compared with 15 years before.ConclusionsThe NIPD isolates were strongly associated with non-vaccine serotypes and with increased AMR levels. This underlines the importance of continued NIPD surveillance for informed policy making on vaccination programmes.
Collapse
Affiliation(s)
| | | | - Toon Braeye
- Epidemiology of Infectious Diseases, Sciensano, Brussels, Belgium
| | | | | | | | - Lize Cuypers
- Laboratory of Clinical Microbiology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- National Reference Centre for invasive Streptococcus pneumoniae, UZ Leuven, Leuven, Belgium
| | - Stefanie Desmet
- Laboratory of Clinical Microbiology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- National Reference Centre for invasive Streptococcus pneumoniae, UZ Leuven, Leuven, Belgium
| | | |
Collapse
|
21
|
McEwan TBD, De Oliveira DMP, Stares EK, Hartley-Tassell LE, Day CJ, Proctor EJ, Nizet V, Walker MJ, Jennings MP, Sluyter R, Sanderson-Smith ML. M proteins of group A Streptococcus bind hyaluronic acid via arginine-arginine/serine-arginine motifs. FASEB J 2024; 38:e70123. [PMID: 39436142 DOI: 10.1096/fj.202401301r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/31/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Tissue injury, including extracellular matrix (ECM) degradation, is a hallmark of group A Streptococcus (GAS) skin infection and is partially mediated by M proteins which possess lectin-like properties. Hyaluronic acid is a glycosaminoglycan enriched in the cutaneous ECM, yet an interaction with M proteins has yet to be explored. This study revealed that hyaluronic acid binding was conserved across phylogenetically diverse M proteins, mediated by RR/SR motifs predominantly localized in the C repeat region. Keratinocyte wound healing was decreased through the recruitment of hyaluronic acid by M proteins in an M type-specific manner. GAS strains 5448 (M1 serotype) and ALAB49 (M53 serotype) also bound hyaluronic acid via M proteins, but hyaluronic acid could increase bacterial adherence independently of M proteins. The identification of host-pathogen mechanisms that affect ECM composition and cell repair responses may facilitate the development of nonantibiotic therapeutics that arrest GAS disease progression in the skin.
Collapse
Affiliation(s)
- Tahnee B-D McEwan
- Molecular Horizons Research Institute and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - David M P De Oliveira
- Molecular Horizons Research Institute and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
- Institute for Molecular Biosciences, The Centre for Superbug Solutions, The University of Queensland, St Lucia, Queensland, Australia
| | - Emily K Stares
- Molecular Horizons Research Institute and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | | | - Christopher J Day
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Emma-Jayne Proctor
- Molecular Horizons Research Institute and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, California, USA
| | - Mark J Walker
- Institute for Molecular Biosciences, The Centre for Superbug Solutions, The University of Queensland, St Lucia, Queensland, Australia
| | - Michael P Jennings
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Ronald Sluyter
- Molecular Horizons Research Institute and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - Martina L Sanderson-Smith
- Molecular Horizons Research Institute and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| |
Collapse
|
22
|
Jones SU, Kee BP, Chew CH, Yeo CC, Chua KH, Puah SM. Differential expression of small RNAs in biofilm-producing clinical methicillin-susceptible Staphylococcus aureus recovered from human urine. Heliyon 2024; 10:e39634. [PMID: 39506957 PMCID: PMC11538773 DOI: 10.1016/j.heliyon.2024.e39634] [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: 07/17/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
Bacterial small RNAs (sRNAs) play crucial roles in coordinating gene regulatory networks in various physiological processes, including biofilm formation. In this study, RNA sequencing was performed on biofilm (n = 4) and planktonic (n = 4) cells harvested at 10 h (pre-stationary phase of biofilm development) to identify biofilm-associated sRNAs in human methicillin-susceptible Staphylococcus aureus (MSSA) recovered from urine isolate. A total of 56 highly expressed sRNAs were identified with 15 overlapping sRNA genes (srn_9348, sprD, sRNA205, sRNA288, srn_2467, Sau-25, srn_2468, sRNA260, sRNA200, RsaE, sRNA397, Teg55, Teg60, RsaX05 and Teg140). Further validation through RT-qPCR analysis of nine sRNAs revealed that srn_9348 and sRNA260 were significantly expressed in the biofilm cells of urine sample. Both sRNAs were predicted to interact with mRNA genes including intracellular adhesin A (icaA) and host factor protein (hfq) involved in biofilm formation via cis-acting and trans-acting using CopraRNA analysis. Therefore, both sRNAs merit further investigations via reverse genetic approaches to elucidate their mechanism of translational regulation. In summary, the transcriptomic analysis conducted in this study offers new insights into the potential regulatory roles of sRNAs in MSSA biofilm development within the urinary environment.
Collapse
Affiliation(s)
- Sherry Usun Jones
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Boon Pin Kee
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ching Hoong Chew
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, 21300, Kuala Nerus, Terengganu, Malaysia
| | - Chew Chieng Yeo
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, 20400, Kuala Terengganu, Terengganu, Malaysia
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Suat Moi Puah
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Fayet-Moore F, Robinson SR. A breath of fresh air: Perspectives on inhaled nutrients and bacteria to improve human health. Adv Nutr 2024:100333. [PMID: 39486624 DOI: 10.1016/j.advnut.2024.100333] [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: 04/17/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
We propose that the human respiratory system and olfactory pathways sequester airborne nutrients (vitamins, fatty acids and trace minerals) that are beneficial for health, which we term 'aeronutrients'. In addition, airborne bacteria, termed 'aeromicrobes', have the potential for positive health effects by improving species diversity in the microbiotas of the respiratory and gastrointestinal tracts. These concepts have implications for people living in urban areas or those who have limited access to nature, such as astronauts exposed for long periods to highly filtered air which may be depleted of aeronutrients and aeromicrobes. The possibility that fresh air contributes to human nutrition and health may stimulate a re-evaluation of guidelines pertaining to nutrition and access to natural environments, and will open new avenues of scientific enquiry.
Collapse
Affiliation(s)
- Flávia Fayet-Moore
- FOODiQ Global, Sydney, New South Wales, 2000, Australia; School of Environmental and Life Sciences, The University of Newcastle, Ourimbah 2258, Australia
| | - Stephen R Robinson
- School of Health & Biomedical Sciences, Royal Melbourne Institute of Technology, Bundoora, Victoria, 3083, Australia; Institute for Breathing and Sleep (IBAS), Austin Health, Heidelberg, Victoria, 3084, Australia.
| |
Collapse
|
24
|
Li A, Chen C, Li Y, Wang Y, Li X, Zhu Q, Zhang Y, Tian S, Xia Q. Characterisation of a new virulent phage isolated from Hainan Island with potential against multidrug-resistant Pseudomonas aeruginosa infections. Res Microbiol 2024:104250. [PMID: 39477080 DOI: 10.1016/j.resmic.2024.104250] [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: 06/22/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Multidrug-resistant (MDR) Pseudomonas aeruginosa is a serious life-threatening pathogen. The rise in P. aeruginosa resistance rates has renewed interest in phages as an alternative therapeutic approach for treating bacterial infections. In this study, we investigated the characteristics of the first Pseudomonas phage, vB_PaP_HN01, isolated from Hainan, the only tropical island in China. The lytic rate of this phage against P. aeruginosa reached 64.3 % (27/42). Under the optimal multiplicity of infection (MOI) of 0.1, more than 90 % of phage particles absorb onto the host cell within 10 min, with an eclipse period of around 15 min, and a high titer phage production (1011 PFU/ml) within 90 min was demonstrated. vB_PaP_HN01 maintains a robust titer after 1 h exposure to pH values and temperatures (up to 50 °C). Genome annotation revealed that vB_PaP_HN01 did not contain drug-resistance or lysogeny-associated genes. It can effectively inhibit the formation of biofilms of MDR P. aeruginosa and eliminated aggressive biofilms (removal rate about 70 %). In the in vivo infection models, it was demonstrated that the survival rate and lifespan of Galleria mellonella larvae were increased alongside the injection of vB_PaP_HN01. These data revealed the potential of vB_PaP_HN01 against P. aeruginosa in clinic.
Collapse
Affiliation(s)
- Anyang Li
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine and the Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Chen Chen
- Medical Laboratory Department, Traditional Chinese Medicine Hospital of Yaan, Sichuan, China
| | - Yanmei Li
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine and the Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Yanshuang Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Xuemiao Li
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine and the Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Qiao Zhu
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine and the Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Yue Zhang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine and the Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Shen Tian
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine and the Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China.
| | - Qianfeng Xia
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine and the Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
25
|
Jurado-Martín I, Tomás-Cortázar J, Hou Y, Sainz-Mejías M, Mysior MM, Sadonès O, Huebner J, Romero-Saavedra F, Simpson JC, Baugh JA, McClean S. Proteomic approach to identify host cell attachment proteins provides protective Pseudomonas aeruginosa vaccine antigen FtsZ. NPJ Vaccines 2024; 9:204. [PMID: 39468053 PMCID: PMC11519640 DOI: 10.1038/s41541-024-00994-x] [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: 04/24/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic Gram-negative pathogen that causes severe nosocomial infections in susceptible individuals due to the emergence of multidrug-resistant strains. There are no approved vaccines against P. aeruginosa infections nor candidates in active clinical development, highlighting the need for novel candidates and strategies. Using a cell-blot proteomic approach, we reproducibly identified 49 proteins involved in interactions with human lung epithelial cells across four P. aeruginosa strains. Among these were cell division protein FtsZ and outer membrane protein OpmH. Escherichia coli BL21 cells overexpressing recombinant FtsZ or rOpmH showed a 66- and 15-fold increased ability to attach to 16HBE14o- cells, further supporting their involvement in host cell attachment. Both antigens led to proliferation of NK and CD8+ cytotoxic T cells, significant increases in the production of IFN-γ, IL-17A, TNF and IL-4 in immunised mice and elicited strong antigen-specific serological IgG1 and IgG2c responses. Immunisation with FtsZ significantly reduced bacterial burden in the lungs by 1.9-log CFU and dissemination to spleen by 1.8-log CFU. The protective antigen candidate, FtsZ, would not have been identified by traditional approaches relying on either virulence mechanisms or sequence-based predictions, opening new avenues in the development of an anti-P. aeruginosa vaccine.
Collapse
Affiliation(s)
- Irene Jurado-Martín
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Julen Tomás-Cortázar
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Yueran Hou
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Maite Sainz-Mejías
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Margaritha M Mysior
- Cell Screening Laboratory, School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Océane Sadonès
- Division of Pediatric Infectious Disease, Hauner Children's Hospital, LMU, Munich, Germany
| | - Johannes Huebner
- Division of Pediatric Infectious Disease, Hauner Children's Hospital, LMU, Munich, Germany
| | - Felipe Romero-Saavedra
- Division of Pediatric Infectious Disease, Hauner Children's Hospital, LMU, Munich, Germany
| | - Jeremy C Simpson
- Cell Screening Laboratory, School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - John A Baugh
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Siobhán McClean
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
26
|
Jakubczak M, Bury D, Montes-García V, Ciesielski A, Naguib M, Jastrzębska AM. Bacterial Responses and Material-Cell Interplays With Novel MoAlB@MBene. Adv Healthc Mater 2024:e2402323. [PMID: 39460417 DOI: 10.1002/adhm.202402323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/08/2024] [Indexed: 10/28/2024]
Abstract
Developing efficient antibacterial nanomaterials has potential across diverse fields, but it requires a deeper understanding of material-bacteria interactions. In this study, a novel 2D core-shell MoAlB@MBene structure is synthesized using a mild wet-chemical etching approach. The growth of E. coli, S. aureus, and B. subtilis bacteria in the presence of MoAlB@MBene decreased in a concentration-dependent manner, with a prolonged lag phase in the initial 6 h of incubation. Even under dark conditions, MoAlB@MBene triggered the formation of intercellular reactive oxygen species (ROS) and singlet oxygen (1O2) in bacteria, while the bacteria protected themselves by forming biofilm and altering cell morphology. The MoAlB@MBene shows consistent light absorption across the visible range, along with a distinctive UV absorption edge. Two types of band gaps are identified: direct (1.67 eV) and indirect (0.74 eV), which facilitate complex light interactions with MoAlB@MBene. Exposure to simulated white light led to decreased viability rates of E. coli (20.6%), S. aureus (22.9%), and B. subtilis (21.4%). Altogether, the presented study enhances the understanding of bacteria responses in the presence of light-activated 2D nanomaterials.
Collapse
Affiliation(s)
- Michał Jakubczak
- Faculty of Mechatronics, Warsaw University of Technology, św. Andrzeja Boboli 8, Warsaw, 02-525, Poland
| | - Dominika Bury
- Faculty of Mechatronics, Warsaw University of Technology, św. Andrzeja Boboli 8, Warsaw, 02-525, Poland
| | - Verónica Montes-García
- Institut de Science et d'Ingénierie Supramoléculaires (I.S.I.S.), Université de Strasbourg & CNRS, 8 allée Gaspard Monge, Strasbourg, 67000, France
| | - Artur Ciesielski
- Institut de Science et d'Ingénierie Supramoléculaires (I.S.I.S.), Université de Strasbourg & CNRS, 8 allée Gaspard Monge, Strasbourg, 67000, France
- Center for Advanced Technologies, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, Poznań, 61-614, Poland
| | - Michael Naguib
- Department of Physics and Engineering Physics, Tulane University, New Orleans, LA, 70118, USA
- Department of Chemistry, Tulane University, New Orleans, LA, 70118, USA
| | - Agnieszka M Jastrzębska
- Faculty of Mechatronics, Warsaw University of Technology, św. Andrzeja Boboli 8, Warsaw, 02-525, Poland
| |
Collapse
|
27
|
Jara-Collao A, Poli MC, Bain W, Peñaloza HF. Editorial: Immune response to gram-negative bacteria in the lungs. Front Cell Infect Microbiol 2024; 14:1503892. [PMID: 39512593 PMCID: PMC11540822 DOI: 10.3389/fcimb.2024.1503892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] Open
Affiliation(s)
- Agnes Jara-Collao
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M Cecilia Poli
- Facultad de Medicina Clínica Alemana de Santiago, Universidad del Desarrollo, Santiago, Chile
- Programa de Inmunogenética e Inmunología Traslacional, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Santiago, Chile
- Unidad de Inmunología y Reumatología, Hospital Roberto del Río, Santiago, Chile
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hernán F. Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Laboratorios Clínicos, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
28
|
Hasan A, Ibrahim M, Alonazi WB, Shen J. Application of immunoinformatics to develop a novel and effective multiepitope chimeric vaccine against Variovorax durovernensis. Comput Biol Chem 2024; 113:108266. [PMID: 39504600 DOI: 10.1016/j.compbiolchem.2024.108266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/31/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024]
Abstract
Bloodstream infections pose a significant public health challenge caused by resistant bacteria such as Variovorax durovernensis, a recently reported Gram-negative bacterium, worsening the burden on healthcare systems. The design of a vaccine using chimeric peptides derived from a representative V. durovernensis strain holds significant promise for preventing disease onset. The current study aimed to employ reverse vaccinology (RV) approaches such as the retrieval of V. durovernensis proteomics data, removal of redundant proteins by CD-HIT, filtering of non-homologous proteins to humans and essential proteins, identification of outer membrane (OM) proteins by CELLO and PSORTb. Following these steps immunoinformatic approaches were applied, such as epitope prediction by IEDB, vaccine design using linkers and adjuvant and analysis of antigenicity, allergenicity, safety and stability. Among the 4208 nonredundant proteins, an OmpA family protein (A0A940EKP4) was designated a potential candidate for the development of a multiepitope vaccine construct. Upon analysis of OM protein, six immunodominant (B cell) epitopes were found on the basis of the chimeric construct following the prediction of CTL stands cytotoxic T lymphocyte and HTL stands helper T lymphocyte epitopes. To ensure comprehensive population coverage globally, the CTL and HTL coverage rates were 58.18 % and 46.56 %, respectively, and 77.23 % overall. By utilizing EAAAK, GPGPG, and AAY linkers, Cholera toxin B subunit adjuvants, and appropriate epitopes were smoothly incorporated into a chimeric vaccine effectively triggering both adaptive and innate immune responses. For example, the administered antigen showed a peak in counts on the fifthday post injection and then gradually declined until the fifteenth day. Elevated levels of several antibodies (IgG + IgM > 700,000; IgM > 600,000; IgG1 + IgG2; IgG1 > 500,000) were observed as decreased in the antigen concentration. Molecular dynamics simulations carried out via iMODS revealed strong correlations between residue pairs, highlighting the stability of the docked complex. The designed vaccine has promising potential in eliciting specific immunogenic responses, thereby facilitating future research for vaccine development against V. durovernensis.
Collapse
Affiliation(s)
- Ahmad Hasan
- Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, PR China
| | - Muhammad Ibrahim
- Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, PR China
| | - Wadi B Alonazi
- Health Administration Department, College of Business Administration, King Saud University, Riyadh, Saudi Arabia
| | - Jian Shen
- Department of Medical Administration, Zhejiang Province People Hospital, Affiliated People Hospital, Hangzhou Medical College Hangzhou, Zhejiang, PR China.
| |
Collapse
|
29
|
Mayattu K, Rajwade J, Ghormade V. Development of erythromycin loaded PLGA nanoparticles for improved drug efficacy and sustained release against bacterial infections and biofilm formation. Microb Pathog 2024; 197:107083. [PMID: 39454804 DOI: 10.1016/j.micpath.2024.107083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 10/28/2024]
Abstract
Bacterial infections are a common cause of sepsis, often leading to high patient mortality. Such infections are challenging to treat due to bacterial resistance to many existing drugs. Erythromycin (Ery) is a macrolide antibiotic used against bacterial infections with reported resistance. Recently, synthetic poly-lactide co-glycolic acid (PLGA) polymer nanoparticles (NPs) have displayed improved drug delivery characteristics and biocompatibility. In this study, PLGA-Ery NPs were synthesized by the o/w emulsion diffusion method, having a particle size of 159 ± 23 nm and displayed 71.89 % of encapsulation efficiency. The PLGA-Ery NPs showed 1.5, 2.1 and 1.5-fold improved MIC and antibacterial efficacy against E. coli, S. aureus, and P. aeruginosa, respectively than the pure drug. As illustrated by scanning electron microscopy, PLGA-Ery NPs caused damage to the bacterial cell walls. Furthermore, a surface coating with PLGA-Ery NPs on a glass surface showed efficient inhibition (>90 %) of the biofilm formation by P. aeruginosa, as determined by fluorescence microscopy and MTT assay. This study demonstrates that PLGA-Ery NPs can increase the efficiency of erythromycin and can suppress the growth and biofilm formation of P. aeruginosa. Such polymeric nanoparticles drug nanoformulations have potential as an antimicrobial and as a surface coating for medical devices.
Collapse
Affiliation(s)
- Kamal Mayattu
- Nanobioscience Group, Agharkar Research Institute, GG Agarkar Road, Pune, 411004, India
| | - Jyutika Rajwade
- Nanobioscience Group, Agharkar Research Institute, GG Agarkar Road, Pune, 411004, India
| | - Vandana Ghormade
- Nanobioscience Group, Agharkar Research Institute, GG Agarkar Road, Pune, 411004, India.
| |
Collapse
|
30
|
Myers MJ, Xu Z, Ryan BJ, DeMars ZR, Ridder MJ, Johnson DK, Krute CN, Flynn TS, Kashipathy MM, Battaile KP, Schnicker N, Lovell S, Freudenthal BD, Bose JL. Molecular insights into the structure and function of the Staphylococcus aureus fatty acid kinase. J Biol Chem 2024:107920. [PMID: 39454961 DOI: 10.1016/j.jbc.2024.107920] [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: 07/24/2024] [Revised: 09/30/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Gram-positive bacteria utilize a Fatty Acid Kinase (FAK) complex to harvest fatty acids from the environment. This complex consists of the fatty acid kinase, FakA, and an acyl carrier protein, FakB, and is known to impact virulence and disease outcomes. Despite some recent studies, there remains many outstanding questions as to the enzymatic mechanism and structure of FAK . To better address this gap in knowledge, we used a combination of modeling, biochemical, and cell-based approaches to build on prior proposed models and identify critical details of FAK activity. Using bio-layer interferometry, we demonstrated nanomolar affinity between FakA and FakB that also indicates that FakA is dimer when binding FakB. Additionally, targeted mutagenesis of the FakA Middle domain demonstrates it possesses a metal binding pocket that is critical for FakA dimer stability and FAK function in vitro and in vivo. Lastly, we solved structures of the apo and ligand-bound FakA kinase domain to capture the molecular changes in the protein following ATP binding and hydrolysis. Together, these data provide critical insight into the structure and function of the FAK complex which is essential for understanding its mechanism.
Collapse
Affiliation(s)
- Megan J Myers
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Zhen Xu
- Protein and Crystallography Facility, University of Iowa, Iowa City, Iowa, USA
| | - Benjamin J Ryan
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Zachary R DeMars
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Miranda J Ridder
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - David K Johnson
- Computational Chemical Biology Core, University of Kansas, Lawrence, Kansas, USA
| | - Christina N Krute
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Tony S Flynn
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Maithri M Kashipathy
- Protein Structure & X-Ray Crystallography Laboratory, University of Kansas, Lawrence, Kansas, USA
| | | | - Nicholas Schnicker
- Protein and Crystallography Facility, University of Iowa, Iowa City, Iowa, USA
| | - Scott Lovell
- Protein Structure & X-Ray Crystallography Laboratory, University of Kansas, Lawrence, Kansas, USA
| | - Bret D Freudenthal
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jeffrey L Bose
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
31
|
Tieu MV, Abafogi AT, Hoang TX, Pham DT, Park J, Park S, Park S, Cho S. Impedimetric Gram-Positive Bacteria Biosensor Using Vancomycin-Coated Silica Nanoparticles with a Gold Nanocluster-Deposited Electrode. Anal Chem 2024; 96:16658-16667. [PMID: 39279360 DOI: 10.1021/acs.analchem.4c02852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
We introduce a swift, label-free electrochemical biosensor designed for the precise on-site detection of Gram-positive bacteria via electrochemical impedance spectroscopy. The biosensor was prepared by electroplating the electrode surface with gold nanoclusters (AuNCs) on the gold-interdigitated wave-shaped electrode with a printed circuit board (Au-PCB) electrode, which plays a role in cost-effective and promising lab-on-a-chip microsystems and integrated biosensing systems. This was followed by the application of silica nanoparticle-modified vancomycin (SiNPs-VAN) that binds to Gram-positive bacteria and facilitates their detection on the AuNC-coated surface. The biosensor demonstrated remarkable sensitivity and specificity. It could detect as few as 102 colony-forming units (CFU)/mL of Staphylococcus aureus, 101 CFU/mL of Bacillus cereus, and 102 CFU/mL of Micrococcus luteus within 20 min. Additionally, SiNPs-VAN is also known for its high stability, low cost, and ease of preparation. It is effective in identifying Gram-positive bacteria in water samples across a concentration range of 102-105 CFU/mL and shows selective identification of Gram-positive bacteria with minimal interference from Gram-negative bacteria like Escherichia coli. The ability of the biosensor to quantify Gram-positive bacteria aligns well with the results obtained from the quantitative real-time polymerase chain reaction (qRT-PCR). These findings highlight the potential of electrochemical biosensors for the detection of pathogens and other biological entities, marking a significant advancement in this field.
Collapse
Affiliation(s)
- My-Van Tieu
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Korea
| | - Abdurhaman Teyib Abafogi
- School of Mechanical Engineering, Sungkyunkwan University, Seobu-ro 2066, Jangan-gu, Suwon 16419, Korea
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| | - Thi Xoan Hoang
- Department of Life Science, Gachon University, Seongnam-si 13120, Korea
| | - Duc-Trung Pham
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Korea
| | - Jaehwan Park
- Department of Semiconductor Engineering, Gachon University, Seongnam-si 13120, Korea
| | - Sungho Park
- Department of Semiconductor Engineering, Gachon University, Seongnam-si 13120, Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University, Seobu-ro 2066, Jangan-gu, Suwon 16419, Korea
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| | - Sungbo Cho
- Department of Semiconductor Engineering, Gachon University, Seongnam-si 13120, Korea
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| |
Collapse
|
32
|
Kolenda C, Bonhomme M, Medina M, Pouilly M, Rousseau C, Troesch E, Martins-Simoes P, Stegger M, Verhoeven PO, Laumay F, Laurent F. Potential of training of anti- Staphylococcus aureus therapeutic phages against Staphylococcus epidermidis multidrug-resistant isolates is restricted by inter- and intra-sequence type specificity. mSystems 2024; 9:e0085024. [PMID: 39248470 PMCID: PMC11494967 DOI: 10.1128/msystems.00850-24] [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: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 09/10/2024] Open
Abstract
Phage therapy appears to be a promising approach to tackle multidrug-resistant bacteria, including staphylococci. However, most anti-staphylococcal phages have been characterized in Staphylococcus aureus, while a limited number of studies investigated phage activity against S. epidermidis. We studied the potential of phage training to extend the host range of two types of anti-S. aureus phages against S. epidermidis isolates. The Appelmans protocol was applied to a mixture of Kayvirus and a mixture of Silviavirus phages repeatedly exposed to seven S. epidermidis strains representative of nosocomial-associated sequence types (ST), including the world-wide disseminated ST2. We observed increased activity only for the Kayvirus mixture against two of these strains (ST2 or ST35). Phage subpopulations isolated from the training mixture using these two strains (five/strain) exhibited different evolved phenotypes, active only against their isolation strain or strains of the same ST. Of note, 16/47 ST2 strains were susceptible to one of the groups of trained phages. A comparative genomic analysis of ancestral and trained phage genomes, conducted to identify potential bacterial determinants of such specific activity, found numerous recombination events between two of the three ancestors. However, a small number of trained phage genes had nucleotide sequence modifications impacting the corresponding protein compared to ancestral phages, two to four of them per phage genome being specific of each group of phage subpopulations exhibiting different host range. The results suggest that anti-S. aureus phages can be adapted to S. epidermidis isolates but with inter- and intra-ST specificity.ImportanceS. epidermidis is increasingly recognized as a threat for public health. Its clinical importance is notably related to multidrug resistance. Phage therapy is one of the most promising alternative therapeutic strategies to antibiotics. Nonetheless, only very few phages active against this bacterial species have been described. In the present study, we showed that phage training can be used to extend the host range of polyvalent Kayvirus phages within the Staphylococcus genera to include S. epidermidis species. In the context of rapid development of phage therapy, in vitro forced adaptation of previously characterized phages could be an appealing alternative to fastidious repeated isolation of new phages to improve the therapeutic potential of a phage collection.
Collapse
Affiliation(s)
- Camille Kolenda
- Service de bactériologie, Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Equipe StaPath, CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Mélanie Bonhomme
- Service de bactériologie, Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Equipe StaPath, CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Mathieu Medina
- Service de bactériologie, Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Equipe StaPath, CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Mateo Pouilly
- Service de bactériologie, Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Clara Rousseau
- Service de bactériologie, Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Emma Troesch
- Service de bactériologie, Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Patricia Martins-Simoes
- Service de bactériologie, Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Equipe StaPath, CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Marc Stegger
- Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
- Antimicrobial Resistance and Infectious Diseases Laboratory, Harry Butler Institute, Murdoch University, Perth, Australia
| | - Paul O. Verhoeven
- GIMAP Team, CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Agents and Hygiene, University Hospital of St-Etienne, St-Etienne, France
| | - Floriane Laumay
- Service de bactériologie, Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Equipe StaPath, CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
- Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France
| | - Frédéric Laurent
- Service de bactériologie, Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Equipe StaPath, CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
- Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
33
|
You J, Xu A, Wang Y, Tu G, Huang R, Wu S. The STING signaling pathways and bacterial infection. Apoptosis 2024:10.1007/s10495-024-02031-7. [PMID: 39428409 DOI: 10.1007/s10495-024-02031-7] [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] [Accepted: 10/06/2024] [Indexed: 10/22/2024]
Abstract
As antibiotic-resistant bacteria continue to emerge frequently, bacterial infections have become a significant and pressing challenge to global public health. Innate immunity triggers the activation of host responses by sensing "non-self" components through various pattern recognition receptors (PRRs), serving as the first line of antibacterial defense. Stimulator of interferon genes (STING) is a PRR that binds with cyclic dinucleotides (CDN) to exert effects against bacteria, viruses, and cancer by inducing the production of type I interferon and inflammatory cytokines, and facilitating regulated cell death. Currently, drugs targeting the STING signaling pathway are predominantly applied in the fields of modulating host immune defense against cancer and viral infections, with relatively limited application in treating bacterial infections. Given the significant immunomodulatory functions of STING in the interaction between bacteria and hosts, this review summarizes the research progress on STING signaling pathways and their roles in bacterial infection, as well as the novel functions of STING modulators, aiming to offer insights for the development of antibacterial drugs.
Collapse
Affiliation(s)
- Jiayi You
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Ailing Xu
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Ye Wang
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Guangmin Tu
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Rui Huang
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Shuyan Wu
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China.
| |
Collapse
|
34
|
De Plano LM, Caratozzolo M, Conoci S, Guglielmino SPP, Franco D. Impact of Nutrient Starvation on Biofilm Formation in Pseudomonas aeruginosa: An Analysis of Growth, Adhesion, and Spatial Distribution. Antibiotics (Basel) 2024; 13:987. [PMID: 39452253 PMCID: PMC11504098 DOI: 10.3390/antibiotics13100987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Objectives: This study investigates the impact of nutrient availability on the growth, adhesion, and biofilm formation of Pseudomonas aeruginosa ATCC 27853 under static conditions. Methods: Bacterial behaviour was evaluated in nutrient-rich Luria-Bertani (LB) broth and nutrient-limited M9 media, specifically lacking carbon (M9-C), nitrogen (M9-N), or phosphorus (M9-P). Bacterial adhesion was analysed microscopically during the transition from reversible to irreversible attachment (up to 120 min) and during biofilm production/maturation stages (up to 72 h). Results: Results demonstrated that LB and M9 media supported bacterial growth, whereas nutrient-starved conditions halted growth, with M9-C and M9-N inducing stationary phases and M9-P leading to cell death. Fractal analysis was employed to characterise the spatial distribution and complexity of bacterial adhesion patterns, revealing that nutrient-limited conditions affected both adhesion density and biofilm architecture, particularly in M9-C. In addition, live/dead staining confirmed a higher proportion of dead cells in M9-P over time (at 48 and 72 h). Conclusions: This study highlights how nutrient starvation influences biofilm formation and bacterial dispersion, offering insights into the survival strategies of P. aeruginosa in resource-limited environments. These findings should contribute to a better understanding of biofilm dynamics, with implications for managing biofilm-related infections and industrial biofouling.
Collapse
Affiliation(s)
- Laura Maria De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Manuela Caratozzolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Sabrina Conoci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy
- LAB Sense Beyond Nano—URT Department of Sciences Physics and Technologies of Matter (DSFTM) CNR, 98166 Messina, Italy
| | - Salvatore P. P. Guglielmino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Domenico Franco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
35
|
de Kretser D, Mora J, Bloomfield M, Campbell A, Cheng MP, Guy S, Hensgens M, Kalimuddin S, Lee TC, Legg A, Mahar RK, Marks M, Marsh J, McGlothin A, Morpeth SC, Sud A, Ten Oever J, Yahav D, Bonten M, Bowen AC, Daneman N, van Hal SJ, Heriot GS, Lewis RJ, Lye DC, McQuilten Z, Paterson DL, Owen Robinson J, Roberts JA, Scarborough M, Webb SA, Whiteway L, Tong SYC, Davis JS, Walls G, Goodman AL. Early Oral Antibiotic Switch in Staphylococcus aureus Bacteraemia: The Staphylococcus aureus Network Adaptive Platform (SNAP) Trial Early Oral Switch Protocol. Clin Infect Dis 2024; 79:871-887. [PMID: 37921609 PMCID: PMC11478773 DOI: 10.1093/cid/ciad666] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/07/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Staphylococcus aureus bloodstream infection (bacteremia) is traditionally treated with at least 2 weeks of intravenous (IV) antibiotics in adults, 3-7 days in children, and often longer for those with complicated disease. The current practice of treating S. aureus bacteremia (SAB) with prolonged IV antibiotics (rather than oral antibiotics) is based on historical observational research and expert opinion. Prolonged IV antibiotic therapy has significant disadvantages for patients and healthcare systems, and there is growing interest in whether a switch to oral antibiotics following an initial period of IV therapy is a safe alternative for clinically stable patients. PROTOCOL The early oral switch (EOS) domain of the S. aureus Network Adaptive Platform (SNAP) trial will assess early switch to oral antibiotics compared with continued IV treatment in clinically stable patients with SAB. The primary endpoint is 90-day all-cause mortality. Hospitalised SAB patients are assessed at platform day 7 ±2 (uncomplicated SAB) and day 14 ±2 (complicated SAB) to determine their eligibility for randomization to EOS (intervention) or continued IV treatment (current standard of care). DISCUSSION Recruitment is occurring in the EOS domain of the SNAP trial. As of August 2023, 21% of all SNAP participants had been randomized to the EOS domain, a total of 264 participants across 77 centers, with an aim to recruit at least 1000 participants. We describe challenges and facilitators to enrolment in this domain to aid those planning similar trials.
Collapse
Affiliation(s)
- Dana de Kretser
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Jocelyn Mora
- Department of Infectious Diseases University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Max Bloomfield
- Department of Infection Services, Wellington Regional Hospital, New Zealand
| | - Anita Campbell
- Telethon Kids Institute, Wesfarmers Center of Infectious Diseases and Vaccines, The University of Western Australia, Perth, Australia
| | - Matthew P Cheng
- Divisions of Infectious Diseases and Medical Microbiology, McGill University Health Center, Montreal, Canada
| | - Stephen Guy
- Department of Infectious Diseases, Eastern Health, Box Hill, Australia
- Monash University (including Australian and New Zealand Intensive Care Research Centre), Clayton, Australia
| | - Marjolein Hensgens
- UMC Utrecht, Utrecht University, Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Todd C Lee
- Clinical Practice Assessment Unit and Division of Infectious Diseases, McGill University, Montreal, Canada
| | - Amy Legg
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Herston Infectious Diseases Institute, Herston, Brisbane, Australia
| | - Robert K Mahar
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Australia
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, Australia
| | - Michael Marks
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Hospital for Tropical Diseases, University College London Hospital, London
- Division of Infection and Immunity, University College London, London
| | - Julie Marsh
- Telethon Kids Institute &/Department of Infectious Diseases &/Wesfarmers Centre for Vaccines and Infectious Diseases, Perth Children's Hospital, Perth, Australia
| | | | - Susan C Morpeth
- Department of Infectious Diseases, Middlemore Hospital, Auckland, New Zealand
| | - Archana Sud
- Department of Infectious Diseases, University of Sydney, Nepean Hospital, Kingswood, New South Wales, Australia
| | - Jaap Ten Oever
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Medical CenterNijmegen, The Netherlands
| | - Dafna Yahav
- Infectious Diseases Unit, Sheba Medical Center, Ramat-Gan, Israel
| | - Marc Bonten
- UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Asha C Bowen
- Telethon Kids Institute &/Department of Infectious Diseases &/Wesfarmers Centre for Vaccines and Infectious Diseases, Perth Children's Hospital, Perth, Australia
| | - Nick Daneman
- Division of Infectious Diseases, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Sebastiaan J van Hal
- Department of Microbiology and Infectious Diseases, Royal Prince Alfred Hospital, Sydney, Australia
- School of Medicine, University of Sydney, Sydney, Australia
| | - George S Heriot
- Department of Infectious Diseases University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | | - David C Lye
- National Center for Infectious Diseases, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
- Yong Loo Lin School of Medicine, Singapore
- Lee Kong Chian School of Medicine, Singapore
| | - Zoe McQuilten
- Monash University (including Australian and New Zealand Intensive Care Research Centre), Clayton, Australia
- Department of Haematology, Monash Health, Melbourne, Australia
| | - David L Paterson
- University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia
| | - J Owen Robinson
- Department of Infectious Diseases, Royal Perth Hospital, Perth, Australia
- Department of Infectious Diseases, Fiona Stanley Hospital, Murdoch, Australia
- PathWest Laboratory Medicine, Perth, Australia
- College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Australia
| | - Jason A Roberts
- Herston Infectious Diseases Institute, Herston, Brisbane, Australia
- University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia
- Metro North Health, Brisbane, Australia
- Department of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
- Division of Anesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | - Matthew Scarborough
- Department of Infectious Diseases, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | - Steve A Webb
- Department of Infectious Diseases, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | | | - Steven Y C Tong
- Department of Infectious Diseases University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Joshua S Davis
- School of Medicine and Public Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Genevieve Walls
- Department of Infectious Diseases, Middlemore Hospital, Auckland, New Zealand
| | - Anna L Goodman
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
- Department of Infectious Diseases, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
- Centre for Clinical Infection and Diagnostics Research, Guy's and St Thomas' Foundation NHS Trust, King's College, London, United Kingdom
| |
Collapse
|
36
|
Yu X, Wang H, Ma S, Chen W, Sun L, Zou Z. Estimating the global and regional burden of lower respiratory infections attributable to leading pathogens and the protective effectiveness of immunization programs. Int J Infect Dis 2024; 149:107268. [PMID: 39413960 DOI: 10.1016/j.ijid.2024.107268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024] Open
Abstract
OBJECTIVES Reducing mortality from infectious diseases is an urgent global public health priority. Streptococcus pneumoniae, H. influenzae, and influenza virus are the three leading causes of lower respiratory infections (LRIs) death worldwide. Our objective was to assess the global burden of LRIs attributable to S. pneumoniae, H. influenzae, and influenza virus and explore the protective effectiveness of immunization programs. METHODS Data were retrieved from the Global Burden of Disease Study 2021 and World Health Organization United Nations Children's Fund Estimates of National Immunization Coverage. Locally weighted linear regression and Spearman correlation analysis were used to examine the associations between LRI mortality and vaccination coverage. Mixed-effects regression models were used to estimate the reduction in deaths that would be reduced by pneumococcal conjugate vaccine (PCV) and the H. influenzae type b (Hib) vaccine if all countries realized the Immunization Agenda 2030 (IA2030). RESULTS In 2021, about 30.2% of the 2.18 million LRI-related deaths were attributed to three studied pathogens. From 1990 to 2021, the age-standardized mortality rate of LRIs was attributable to three pathogens decreased by more than half. In 2022, the global vaccination rates for PCV and Hib vaccines were 60.0% and 76.0%, respectively. The LRIs attributable to S. pneumoniae (rs = -0.45, P <0.001) and H. influenzae (rs = -0.47, P <0.001) decreased with the increasing vaccination coverage of PCV and Hib. By 2030, approximately 59.9% and 70.8% of countries worldwide will reach the IA2030 targets of 90% coverage for PCV and Hib, respectively. By that time, the number of deaths from LRIs in children attributable to S. pneumoniae and H. influenzae will decrease by 54.8% and 24.4%, respectively. CONCLUSIONS Despite the declines in LRI mortality attributed to respiratory pathogens, substantial deaths still occurred in 2021. To advance toward achieving the IA2030 targets and further mitigate mortality associated with LRIs, intensified efforts by the international community and national health systems are imperative.
Collapse
Affiliation(s)
- Xiaoran Yu
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Huan Wang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Sheng Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Wanning Chen
- Beijing Children's Hospital, Capital Medical University, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, National Center for Children's Health, Beijing, China
| | - Lin Sun
- Beijing Children's Hospital, Capital Medical University, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, National Center for Children's Health, Beijing, China
| | - Zhiyong Zou
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China.
| |
Collapse
|
37
|
McMahon F, Ware RS, Grimwood K, Atack JM. Haemophilus influenzae and pneumococci: Co-colonization, interactions, cooperation and competition. Pediatr Pulmonol 2024. [PMID: 39392258 DOI: 10.1002/ppul.27318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
Nontypeable Haemophilus influenzae (NTHi) and Streptococcus pneumoniae (pneumococcus) are pathobionts that share common environmental niches within the upper respiratory tract. They can form part of the resident upper airway microbiota, but under certain environmental circumstances become pathogenic and induce disease. In children, both organisms have a considerable impact on the healthcare system, commonly causing acute otitis media and pneumonia. They are also associated with chronic biofilm-mediated respiratory infections, such as persistent middle ear effusions and chronic suppurative otitis media, and in the lower airways with protracted bacterial bronchitis and bronchiectasis. Consequently, both organisms are responsible for large numbers of antibiotic prescriptions and substantial healthcare costs. The complex relationship between NTHi and pneumococcal co-interaction during colonization, infection and biofilm formation is poorly understood and a greater understanding is needed to facilitate development of future therapies, and novel interventions and prevention strategies. Co-infections with both bacteria can result in more severe disease, with disease severity likely mediated by their ability to cooperate in some in vivo niches. However, this relationship is not always straightforward, as under certain conditions, these two bacteria compete rather than cooperate. Current opinion supports developing a vaccine targeting NTHi strains, as well as a combined vaccine targeting both NTHi and pneumococci to decrease the respiratory disease burden in young children. This review summarizes our current knowledge of the interactions between NTHi and pneumococci and speculates on the future directions of research to understand how these bacteria co-exist and how to better prevent and treat NTHi and pneumococcal infection.
Collapse
Affiliation(s)
- Finn McMahon
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Robert S Ware
- Health Group, Griffith University, Gold Coast, Queensland, Australia
| | - Keith Grimwood
- School of Medicine and Dentistry, Griffith University, Gold Coast, Queensland, Australia
- NHMRC Centre for Research Excellence in Paediatric Bronchiectasis (AusBREATHE), Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - John M Atack
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
- School of Environment and Science, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
38
|
Mielniczuk S, Hoff K, Baselious F, Li Y, Haupenthal J, Kany AM, Riedner M, Rohde H, Rox K, Hirsch AKH, Krimm I, Sippl W, Holl R. Development of Fragment-Based Inhibitors of the Bacterial Deacetylase LpxC with Low Nanomolar Activity. J Med Chem 2024; 67:17363-17391. [PMID: 39303295 PMCID: PMC11472313 DOI: 10.1021/acs.jmedchem.4c01262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
In a fragment-based approach using NMR spectroscopy, benzyloxyacetohydroxamic acid-derived inhibitors of the bacterial deacetylase LpxC bearing a substituent to target the uridine diphosphate-binding site of the enzyme were developed. By appending privileged fragments via a suitable linker, potent LpxC inhibitors with promising antibacterial activities could be obtained, like the one-digit nanomolar LpxC inhibitor (S)-13j [Ki (EcLpxC C63A) = 9.5 nM; Ki (PaLpxC): 5.6 nM]. To rationalize the observed structure-activity relationships, molecular docking and molecular dynamics studies were performed. Initial in vitro absorption-distribution-metabolism-excretion-toxicity (ADMET) studies of the most potent compounds have paved the way for multiparameter optimization of our newly developed isoserine-based amides.
Collapse
Affiliation(s)
- Sebastian Mielniczuk
- Institute
of Organic Chemistry, Universität
Hamburg, Martin-Luther-King-Platz
6, 20146 Hamburg, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
| | - Katharina Hoff
- Institute
of Organic Chemistry, Universität
Hamburg, Martin-Luther-King-Platz
6, 20146 Hamburg, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
| | - Fady Baselious
- Institute
of Pharmacy, Martin-Luther-University of
Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120 Halle (Saale), Germany
| | - Yunqi Li
- Team
“Small Molecules for Biological Targets”, Institut Convergence
Plascan, Centre de Recherche en Cancérologie de Lyon, INSERM
U1052-CNRS UMR5286, Centre Léon Bérard, Université
de Lyon, Université Claude Bernard
Lyon1, 69008 Lyon, France
- Shanghai
Key Laboratory of Regulatory Biology, The Institute of Biomedical
Sciences & School of Life Sciences, East China Normal University, 200241 Shanghai, China
| | - Jörg Haupenthal
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Andreas M. Kany
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Maria Riedner
- Technology
Platform Mass Spectrometry, Universität
Hamburg, Mittelweg 177, 20148 Hamburg, Germany
| | - Holger Rohde
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
- Institute
of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Katharina Rox
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Anna K. H. Hirsch
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Helmholtz International Lab for Anti-infectives, Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Isabelle Krimm
- Team
“Small Molecules for Biological Targets”, Institut Convergence
Plascan, Centre de Recherche en Cancérologie de Lyon, INSERM
U1052-CNRS UMR5286, Centre Léon Bérard, Université
de Lyon, Université Claude Bernard
Lyon1, 69008 Lyon, France
| | - Wolfgang Sippl
- Institute
of Pharmacy, Martin-Luther-University of
Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120 Halle (Saale), Germany
| | - Ralph Holl
- Institute
of Organic Chemistry, Universität
Hamburg, Martin-Luther-King-Platz
6, 20146 Hamburg, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
| |
Collapse
|
39
|
Zhang Y, Zhao C, Guo Z, Yang T, Zhang X, Huang X, Shi J, Gao S, Zou X. Ultrasensitive Analysis of Escherichia coli O157:H7 Based on Immunomagnetic Separation and Labeled Surface-Enhanced Raman Scattering with Minimized False Positive Identifications. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:22349-22359. [PMID: 39327911 DOI: 10.1021/acs.jafc.4c06311] [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: 09/28/2024]
Abstract
It is a big challenge to monitor pathogens in food with high selectivity. In this study, we reported an ultrasensitive method for Escherichia coli O157:H7 detection based on immunomagnetic separation and labeled surface-enhanced Raman scattering (SERS). The bacterium was identified by heterogeneous recognition elements, monoclonal antibody (mAb), and aptamer. E. coli O157:H7 was separated and enriched by magnetic nanoparticles modified by mAb, and then a plasmonic nanostructure functionalized by aptamers with embedded Raman tags and interior gaps was utilized for further discrimination and detection. The selectivity was enhanced by two binding sites. The higher Raman enhancement was obtained by strong local electromagnetic field oscillation in the gap and the firm embedment of 4-mercaptopyridine (4-Mpy). Optimum experiments created that SERS signals of 4-Mpy at 1010 cm-1 had a good linearity with E. coli O157:H7 at a large range of 10 to 107 CFU/mL with a limit of detection of 2 CFU/mL. This method has great potential for on-site food pathogenic bacterial detection.
Collapse
Affiliation(s)
- Yang Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Chuping Zhao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zhiming Guo
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Tianxi Yang
- Nutrition and Health, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Xinai Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xiaowei Huang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jiyong Shi
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Shipeng Gao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xiaobo Zou
- International Joint Research Laboratory of Intelligent Agriculture and Agriproducts Processing, China Light Industry Key Laboratory of Food Intelligent Detection & Processing, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| |
Collapse
|
40
|
Jiang Y, Wang Z, Yang W, Yang P, Feng X, Qin P, Huang F. Lead-Free Cs 2AgBiBr 6/TiO 2 S-Scheme Heterojunction for Efficient Photocatalytic Antibiotic Rifampicin Degradation. NANO LETTERS 2024; 24:12597-12604. [PMID: 39329391 DOI: 10.1021/acs.nanolett.4c03648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Exploring efficient and stable halide perovskite-based photocatalysts is a great challenge due to the balance between the photocatalytic performance, toxicity, and intrinsic chemical instability of the materials. Here, the environmentally friendly lead-free perovskite Cs2AgBiBr6 confined in the mesoporous TiO2 crystal matrix has been designed to enhance the charge carrier extraction and utilization for efficient photocatalytic rifampicin degradation. The as-prepared Cs2AgBiBr6/TiO2 catalyst was stable in air for over 500 days. An S-scheme heterojunction was formed between the (004) plane of Cs2AgBiBr6 and the (101) plane of TiO2 through the Bi-O-Br bonds. The built-in electric field at the interface efficiently promoted the photoinduced charge separation and carrier extraction. The Cs2AgBiBr6/TiO2-200 showed a 92.83% degradation efficiency of rifampicin within 80 min under simulated sunlight illumination (AM 1.5G 100 mW cm-2). This work offers an effective way for the construction of halide perovskite-based photocatalysts with high photocatalytic performance, good stability, and low toxicity simultaneously.
Collapse
Affiliation(s)
- Yin Jiang
- Director, Key Laboratory of Advanced Technique & Preparation for Renewable Energy Materials, Ministry of Education, Yunnan Normal University, Kunming 650500, China
| | - Zhaoyang Wang
- Director, Key Laboratory of Advanced Technique & Preparation for Renewable Energy Materials, Ministry of Education, Yunnan Normal University, Kunming 650500, China
| | - Wen Yang
- Director, Key Laboratory of Advanced Technique & Preparation for Renewable Energy Materials, Ministry of Education, Yunnan Normal University, Kunming 650500, China
| | - Peizhi Yang
- Director, Key Laboratory of Advanced Technique & Preparation for Renewable Energy Materials, Ministry of Education, Yunnan Normal University, Kunming 650500, China
| | - Xiaobo Feng
- Director, Key Laboratory of Advanced Technique & Preparation for Renewable Energy Materials, Ministry of Education, Yunnan Normal University, Kunming 650500, China
| | - Peng Qin
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Science, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Science, Beijing 100049, China
| | - Fuqiang Huang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Dongchuan Road 800, 200240 Shanghai, China
| |
Collapse
|
41
|
Selva Sharma A, Ryou SM, Lee JH, Lee NY. New insights into the photophysical properties and interaction mechanisms of Janus green blue dye with polyanions and its applications in colorimetric visualization of loop-mediated isothermal amplification and polymerase chain reaction. J Mater Chem B 2024; 12:10082-10092. [PMID: 39268583 DOI: 10.1039/d4tb01623h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
In this investigation, the photophysical properties and interaction mechanisms of Janus green blue (JGB) dye with polyanions were systematically studied using spectroscopic techniques. The absorption spectral analysis revealed that JGB binds cooperatively to sodium alginate, leading to dye stacking along the polymer chain. The interaction of JGB dye with DNA was characterized by the emergence of a metachromatic peak at 564 nm, indicating the formation of dye aggregates. The analysis of absorption data reveals that JGB dye interacts with DNA at multiple binding sites, including at least one high-affinity site. The AutoDock Vina based blind docking approach was used to analyze the most probable binding location of JGB dye in DNA. By making use of the DNA-induced metachromasia, a colorimetric approach was developed for the visualization of loop-mediated isothermal amplification (LAMP) and polymerase chain reaction (PCR). The LAMP-colorimetric assay, targeting the Streptococcus pneumoniae gene, demonstrated a noticeable colour change with a detection limit of 1 pg μL-1. The practical applicability was validated by detecting S. pneumoniae in artificial urine. In addition to LAMP, we tested the JGB dye based colorimetric assay for applicability in PCR reactions. The colorimetric PCR assay using the metal-responsive transcription factor (MTF-1) gene achieved a detection limit as low as 0.1 pg μL-1. The study highlights the potential of DNA binding metachromic dye to significantly enhance colorimetric assays, offering a robust and sensitive tool for molecular diagnostics.
Collapse
Affiliation(s)
- Arumugam Selva Sharma
- Department of Nanoscience and Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do 13120, South Korea
| | - Sung Min Ryou
- Department of BioNano Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do 13120, South Korea.
| | - Ji Hyeok Lee
- Department of BioNano Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do 13120, South Korea.
| | - Nae Yoon Lee
- Department of BioNano Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do 13120, South Korea.
| |
Collapse
|
42
|
Nasr J, Abdessamad H, Mina J, Haykal T, Jamil Y, Abboud E, Mahdi A, Asmar R, Abi Assaad R, Alameddine D, Bourji A, Mahdi M, Abdulaal R, Tomassian S, El Ahmadieh H, Azzam W, Mokhbat JE, Moghnieh R, Rodriguez-Morales AJ, Husni R. The epidemiology of gram-negative bacteremia in Lebanon: a study in four hospitals. Ann Clin Microbiol Antimicrob 2024; 23:90. [PMID: 39385237 PMCID: PMC11465513 DOI: 10.1186/s12941-024-00740-0] [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: 03/05/2024] [Accepted: 08/16/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Gram-negative bacteremia is a life-threatening infection with high morbidity and mortality. Its incidence is rising worldwide, and treatment has become more challenging due to emerging bacterial resistance. Little data is available on the burden and outcome of such infections in Lebanon. METHODS We conducted this retrospective study in four Lebanese hospitals. Data on medical conditions and demographics of 2400 patients diagnosed with a bloodstream infection based on a positive blood culture were collected between January 2014 and December 2020. RESULTS Most bacteremias were caused by Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter baumannii, with the more resistant organisms being hospital-acquired. Third-generation cephalosporin and quinolone resistance was steady throughout the study, but carbapenem resistance increased. Mortality with such infections is high, but carbapenem resistance or infection with Pseudomonas or Acinetobacter species were significant risk factors for poor outcomes. CONCLUSION This is the first multi-center study from Lebanon on gram-negative bacteremia, resistance patterns, and factors associated with a poor outcome. More surveillance is needed to provide data to guide empirical treatment for bacteremia in Lebanon.
Collapse
Affiliation(s)
- Janane Nasr
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Hilal Abdessamad
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Johnathan Mina
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Tony Haykal
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Yasser Jamil
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Emma Abboud
- Laboratory Director, Mount Lebanon Hospital University Medical Center, Beirut, 1102, Lebanon
| | - Ahmad Mahdi
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rana Asmar
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rawad Abi Assaad
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Dana Alameddine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Alaa Bourji
- Department of Surgery, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Mahmoud Mahdi
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Razan Abdulaal
- Department of Internal Medicine, University of Balamand, Balamand, Lebanon
| | - Serge Tomassian
- Department of Internal Medicine, University of Balamand, Balamand, Lebanon
| | - Hanane El Ahmadieh
- Infection Control Coordination, Mount Lebanon Hospital University Medical Center, Beirut, 1102, Lebanon
| | - Wael Azzam
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Jacques E Mokhbat
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rima Moghnieh
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Alfonso J Rodriguez-Morales
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
- Master of Clinical Epidemiology and Biostatistics, Universidad Cientifica del Sur, Lima, 15067, Peru
| | - Rola Husni
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon.
| |
Collapse
|
43
|
Ashmi M, He C, Drobniewski F. Can immuno-PCR (IPCR) transform bacterial disease diagnostics? Expert Rev Mol Diagn 2024:1-10. [PMID: 39381910 DOI: 10.1080/14737159.2024.2413556] [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: 07/18/2024] [Accepted: 10/03/2024] [Indexed: 10/10/2024]
Abstract
INTRODUCTION Approximately 15 million deaths occur globally each year due to infectious diseases. Timely diagnosis is crucial in promoting cure and preventing disease transmission. Currently, molecular diagnostics have replaced many conventional diagnostic tools due to their inherent limitations. However, the full potential of Immuno Polymerase Chain Reaction (IPCR) remains largely untapped. AREAS COVERED This review focuses on the use of IPCR in the diagnosis of different bacterial diseases, highlighting its advantages over traditional methods. EXPERT OPINION Early and accurate diagnosis of infectious diseases is crucial because it enhances treatment effectiveness, reduces morbidity and mortality, helps identify potential causes of sepsis earlier, and reduces the risk of unknowingly spreading the disease to others. IPCR in turn has shown promise for the early diagnosis of bacterial diseases as an alternative to conventional culture-based or serological diagnostic assays leading to delayed diagnosis and treatment. IPCR has the potential to revolutionize the diagnostic field due to its increased sensitivity and specificity. Although efforts are needed to reduce the time of the assay and to reduce background noise, IPCR can be combined with other platforms like lateral flow assay/biosensors/automation to improve its use as a point-of-care assay, especially in resource-limited settings.
Collapse
Affiliation(s)
- Marcia Ashmi
- Adult Infectious Disease, Department of Medicine, Imperial College London, London, UK
| | - Changchunzi He
- Adult Infectious Disease, Department of Medicine, Imperial College London, London, UK
| | - Francis Drobniewski
- Adult Infectious Disease, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
44
|
Tomaszewski KL, Blanchard M, Olaniyi R, Brenton HR, Hayes S, Fatma F, Amarasinghe GK, Cho BK, Goo YA, DeDent AC, Fritz SA, Wardenburg JB. Enhanced Staphylococcus aureus protection by uncoupling of the α-toxin-ADAM10 interaction during murine neonatal vaccination. Nat Commun 2024; 15:8702. [PMID: 39379345 PMCID: PMC11461939 DOI: 10.1038/s41467-024-52714-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Staphylococcus aureus remains a leading global cause of bacterial infection-associated mortality and has eluded prior vaccine development efforts. S. aureus α-toxin (Hla) is an essential virulence factor in disease, impairing the T cell response to infection. The anti-Hla antibody response is a correlate of human protective immunity. Here we observe that this response is limited early in human life and design a vaccine strategy to elicit immune protection against Hla in a neonatal mice. By targeted disruption of the interaction of Hla with its receptor ADAM10, we identify a vaccine antigen (HlaH35L/R66C/E70C, HlaHRE) that elicits an ~100-fold increase in the neutralizing anti-Hla response. Immunization with HlaHRE enhances the T follicular helper (TFH) cell response to S. aureus infection, correlating with the magnitude of the neutralizing anti-toxin response and disease protection. Furthermore, maternal HlaHRE immunization confers protection to offspring. Together, these findings illuminate a path for S. aureus vaccine development at the maternal-infant interface.
Collapse
Affiliation(s)
- Kelly L Tomaszewski
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Forward Defense, LLC, St. Louis, MO, USA
| | - Meagan Blanchard
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Reuben Olaniyi
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Merck & Co, West Point, PA, USA
| | - Hannah R Brenton
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Samantha Hayes
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Farheen Fatma
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Byoung-Kyu Cho
- Mass Spectrometry Technology Access Center at the McDonnell Genome Institute - Washington University School of Medicine, St. Louis, MO, USA
| | - Young Ah Goo
- Mass Spectrometry Technology Access Center at the McDonnell Genome Institute - Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea C DeDent
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
- Cleveland Clinic Innovations, Cleveland Clinic, Cleveland, OH, USA
| | - Stephanie A Fritz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Juliane Bubeck Wardenburg
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA.
- Forward Defense, LLC, St. Louis, MO, USA.
| |
Collapse
|
45
|
Tejeda MI, Fernández J, Valledor P, Almirall C, Barberán J, Romero-Brufau S. Retrospective validation study of a machine learning-based software for empirical and organism-targeted antibiotic therapy selection. Antimicrob Agents Chemother 2024; 68:e0077724. [PMID: 39194206 PMCID: PMC11460031 DOI: 10.1128/aac.00777-24] [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: 05/27/2024] [Accepted: 07/16/2024] [Indexed: 08/29/2024] Open
Abstract
Errors in antibiotic prescriptions are frequent, often resulting from the inadequate coverage of the infection-causative microorganism. The efficacy of iAST, a machine-learning-based software offering empirical and organism-targeted antibiotic recommendations, was assessed. The study was conducted in a 12-hospital Spanish institution. After model fine-tuning with 27,531 historical antibiograms, 325 consecutive patients with acute infections were selected for retrospective validation. The primary endpoint was comparing each of the top three of iAST's antibiotic recommendations' success rates (confirmed by antibiogram results) with the antibiotic prescribed by the physicians. Secondary endpoints included examining the same hypothesis within specific study population subgroups and assessing antibiotic stewardship by comparing the percentage of antibiotics recommended that belonged to different World Health Organization AWaRe groups within each arm of the study. All of iAST first three recommendations were non-inferior to doctor prescription in the primary endpoint analysis population as well as the secondary endpoint. The overall success rate of doctors' empirical treatment was 68.93%, while that of the first three iAST options was 91.06% (P < 0.001), 90.63% (P < 0.001), and 91.06% (P < 001), respectively. For organism-targeted therapy, the doctor's overall success rate was 84.16%, and that of the first three ranked iAST options was 97.83% (P < 0.001), 94.09% (P < 0.001), and 91.30% (P < 0.001), respectively. In empirical therapy, compared to physician prescriptions, iAST demonstrated a greater propensity to recommend access antibiotics, fewer watch antibiotics, and higher reserve antibiotics. In organism-targeted therapy, iAST advised a higher utilization of access antibiotics. The present study demonstrates iAST accuracy in predicting antibiotic susceptibility, showcasing its potential to promote effective antibiotic stewardship. CLINICAL TRIALS This study is registered with ClinicalTrials.gov as NCT06174519.
Collapse
Affiliation(s)
- Maria Isabel Tejeda
- Infectious Diseases Unit, Hospital Universitario HM Montepríncipe, Madrid, Spain
| | - Javier Fernández
- Research and Innovation Department, Pragmatech AI Solutions, Oviedo, Spain
- Microbiology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
- Microbiology and Infectious Pathology, ISPA, Oviedo, Spain
- Functional Biology Department, Universidad de Oviedo, Oviedo, Spain
| | - Pablo Valledor
- Research and Innovation Department, Pragmatech AI Solutions, Oviedo, Spain
| | | | - José Barberán
- Infectious Diseases Unit, Hospital Universitario HM Montepríncipe, Madrid, Spain
- HM Faculty of Health Sciences, University Camilo Jose Cela, Madrid, Spain
| | - Santiago Romero-Brufau
- Research and Innovation Department, Pragmatech AI Solutions, Oviedo, Spain
- Department of Otorhinolaryngology–Head & Neck Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Antoniuk O, Maranha A, Salvador JAR, Empadinhas N, Moreira VM. Bi- and tricyclic diterpenoids: landmarks from a decade (2013-2023) in search of leads against infectious diseases. Nat Prod Rep 2024. [PMID: 39371026 DOI: 10.1039/d4np00021h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Covering: 2013 to 2023In an era where antimicrobial resistance severely threatens our ability to treat infections, the discovery of new drugs that belong to different chemical classes and/or bear original modes of action is urgently needed. In this case, diterpenoids comprise a productive field with a proven track record in providing new anti-infectives to tackle bacterial infections and malaria. This review highlights the potential of both naturally occurring and semi-synthetic bi- and tricyclic diterpenoids to become leads in search of new drugs to treat infections caused by bacteria, fungi, viruses and protozoan parasites. The literature from the last decade (2013-2023) is covered, focusing on naturally occurring and semi-synthetic bicyclic (labdanes and labdane-type) and tricyclic (all classes) diterpenoids, detailing their relevant biological activities in the context of infection, which are explained through structure-activity relationships.
Collapse
Affiliation(s)
- Olha Antoniuk
- Faculty of Pharmacy, University of Coimbra, Portugal.
- Centre for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Ana Maranha
- Centre for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Jorge A R Salvador
- Faculty of Pharmacy, University of Coimbra, Portugal.
- Centre for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Nuno Empadinhas
- Centre for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Vânia M Moreira
- Faculty of Pharmacy, University of Coimbra, Portugal.
- Centre for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
47
|
Fritz SA, Bubeck Wardenburg J. A path forward for Staphylococcus aureus vaccine development. J Exp Med 2024; 221:e20240002. [PMID: 39150449 PMCID: PMC11329773 DOI: 10.1084/jem.20240002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024] Open
Abstract
The pursuit of a vaccine to quell Staphylococcus aureus disease has been unfruitful. In this Viewpoint, we explore the biological linkage between microbial niche acquisition and host immunity as a basis to guide future vaccine efforts.
Collapse
Affiliation(s)
- Stephanie A Fritz
- Departments of Pediatrics and Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Juliane Bubeck Wardenburg
- Departments of Pediatrics and Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
48
|
Li Y, Wang Y, Wu Q, Qi R, Li L, Xu L, Yuan H. High-throughput fluorescence sensing array based on tetraphenylethylene derivatives for detecting and distinguishing pathogenic microbes. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 318:124435. [PMID: 38796890 DOI: 10.1016/j.saa.2024.124435] [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: 03/21/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
Infections induced by pathogenic microorganisms will bring negative effects such as diseases that damage health and result in heavy economic burden. Therefore, it is very important to detect and identify the pathogens in time. Moreover, traditional clinical diagnosis or food testing often faces the problem of dealing with a large number of samples. Here, we designed a high-throughput fluorescent sensor array based on the different binding ability of five tetraphenylethylene derivatives (TPEs) with various side chains to different kinds of pathogenic microbes, which is used to detect and distinguish various species, so as to realize rapid mass diagnosis, and hopefully provide guidance for further determination of microbial infections and clinical treatment.
Collapse
Affiliation(s)
- Yutong Li
- Department of Chemistry, Beijing Technology and Business University, Beijing 100048, China
| | - Yi Wang
- Department of Chemistry, Beijing Technology and Business University, Beijing 100048, China
| | - Qiaoyue Wu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ruilian Qi
- Department of Chemistry, Beijing Technology and Business University, Beijing 100048, China.
| | - Li Li
- Department of Chemistry, Beijing Technology and Business University, Beijing 100048, China
| | - Li Xu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China.
| | - Huanxiang Yuan
- Department of Chemistry, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
49
|
Yu L, Han X, Zhang W, Fu Y, Yang S, Wu S, Jin J, Li S, Chen Y, Jiang Y, Yu Y. The two-component sensor factor envZ influences antibiotic resistance and virulence in the evolutionary dynamics of multidrug-resistant Salmonella enteritidis causing multisite invasive infections. J Antimicrob Chemother 2024:dkae355. [PMID: 39365636 DOI: 10.1093/jac/dkae355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024] Open
Abstract
OBJECTIVES To assess the impact of mutations in the two-component sensor envZ on antibiotic resistance and virulence in the evolutionary dynamics of MDR Salmonella enteritidis (S. enteritidis). METHODS Five S. enteritidis isolates obtained from a patient with multisite invasive infections were analysed. Analysis of antibiotic resistance genes, virulence genes and SNP was performed through WGS. RNA sequencing, quantitative RT-PCR, virulence testing in a Galleria mellonella (G. mellonella) infection model and in vitro cell experiments were used to examine the effects of envZ mutations. RESULTS WGS revealed identical resistance and virulence genes on an IncFIB(S)/IncFII(S)/IncX1 fusion plasmid in all strains. The faecal strains harboured envZ mutations, reducing outer membrane protein ompD and ompF transcriptional level. Virulence testing demonstrated elevated virulence in envZ mutant strains. In vitro experiments revealed increased adhesion, invasion and phagocytosis resistance in envZ mutants, along with reduced biofilm formation and growth rates. CONCLUSIONS These findings highlight novel genetic locations on envZ influencing antibiotic resistance and virulence in clinical S. enteritidis strains. envZ mutations impact antibiotic resistance by down-regulating ompD and ompF expression and enhance virulence, contributing to multisite infections with increased fitness costs.
Collapse
Affiliation(s)
- Lifei Yu
- Department of Infectious Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xinhong Han
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Wang Zhang
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Ying Fu
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Shaoxue Yang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Shenghai Wu
- Department of Clinical Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Jie Jin
- Department of Infectious Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Siying Li
- Department of Infectious Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Yan Chen
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310016, China
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yan Jiang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310016, China
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yunsong Yu
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310016, China
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| |
Collapse
|
50
|
Deventer AT, Stevens CE, Stewart A, Hobbs JK. Antibiotic tolerance among clinical isolates: mechanisms, detection, prevalence, and significance. Clin Microbiol Rev 2024:e0010624. [PMID: 39364999 DOI: 10.1128/cmr.00106-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
SUMMARYAntibiotic treatment failures in the absence of resistance are not uncommon. Recently, attention has grown around the phenomenon of antibiotic tolerance, an underappreciated contributor to recalcitrant infections first detected in the 1970s. Tolerance describes the ability of a bacterial population to survive transient exposure to an otherwise lethal concentration of antibiotic without exhibiting resistance. With advances in genomics, we are gaining a better understanding of the molecular mechanisms behind tolerance, and several studies have sought to examine the clinical prevalence of tolerance. Attempts have also been made to assess the clinical significance of tolerance through in vivo infection models and prospective/retrospective clinical studies. Here, we review the data available on the molecular mechanisms, detection, prevalence, and clinical significance of genotypic tolerance that span ~50 years. We discuss the need for standardized methodology and interpretation criteria for tolerance detection and the impact that methodological inconsistencies have on our ability to accurately assess the scale of the problem. In terms of the clinical significance of tolerance, studies suggest that tolerance contributes to worse outcomes for patients (e.g., higher mortality, prolonged hospitalization), but historical data from animal models are varied. Furthermore, we lack the necessary information to effectively treat tolerant infections. Overall, while the tolerance field is gaining much-needed traction, the underlying clinical significance of tolerance that underpins all tolerance research is still far from clear and requires attention.
Collapse
Affiliation(s)
- Ashley T Deventer
- School of Biology, Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Claire E Stevens
- School of Biology, Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Amy Stewart
- School of Biology, Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Joanne K Hobbs
- School of Biology, Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|