1
|
Wang C, Sheng W, Zhou Y, Hang X, Zhao J, Gu Y, Meng X, Bai Y, Li W, Zhang Y, Zhang L, Yu J, Zhou Z, Li X, Sun H, Xue Y, Xu T, Zen K, Ling H, Zhang CY, Bi H, Wang H. siRNA-AGO2 complex inhibits bacterial gene translation: A promising therapeutic strategy for superbug infection. Cell Rep Med 2025; 6:101997. [PMID: 40054457 PMCID: PMC11970400 DOI: 10.1016/j.xcrm.2025.101997] [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: 05/05/2023] [Revised: 10/15/2024] [Accepted: 02/11/2025] [Indexed: 03/21/2025]
Abstract
Silencing resistance genes of pathogenic bacteria by RNA interference (RNAi) is a potential strategy to fight antibiotic-resistant bacterial infections. Currently, RNAi cannot be achieved in bacteria due to the lack of RNA-induced silencing complex machinery and the difficulty of small interfering RNA (siRNA) delivery. Here, we show that exosomal siRNAs can be efficiently delivered into bacterial cells and can silence target genes primarily through translational repression without mRNA degradation. The exosomal Argonaute 2 (AGO2) protein forms a complex with siRNAs, which is essential for bacterial gene silencing. Both in vitro and in vivo-generated exosome-packaged siRNAs resensitize methicillin-resistant Staphylococcus aureus (MRSA) to methicillin treatment by silencing the mecA gene, which is the primary beta-lactam resistance determinant of MRSA. This approach significantly enhances the therapeutic effect in a mouse model of MRSA infection. In summary, our study provides a method for siRNA delivery to bacteria that may facilitate the treatment of antibiotic-resistant bacterial infection.
Collapse
Affiliation(s)
- Chen Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China; School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Wangjian Sheng
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yu Zhou
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Xudong Hang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan 571199, China
| | - Jiayi Zhao
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yuanyuan Gu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Xiangfeng Meng
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yuefan Bai
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Weili Li
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yujing Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Linlin Zhang
- Department of Microbiology, Wu Lien-Teh Institute, Heilongjiang Provincial Key Laboratory of Infection and Immunity, Key Laboratory of Pathogen Biology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Jing Yu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Zhen Zhou
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Xiaona Li
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Haorui Sun
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yanhong Xue
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ke Zen
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Hong Ling
- Department of Microbiology, Wu Lien-Teh Institute, Heilongjiang Provincial Key Laboratory of Infection and Immunity, Key Laboratory of Pathogen Biology, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Chen-Yu Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of Artificial Intelligence Biomedicine and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China.
| | - Hongkai Bi
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan 571199, China; Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| | - Huan Wang
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
2
|
Müller E, Monecke S, Armengol Porta M, Narvaez Encalada MV, Reissig A, Rüttiger L, Schröttner P, Schwede I, Söffing HH, Thürmer A, Ehricht R. Rapid Detection of Panton-Valentine Leukocidin Production in Clinical Isolates of Staphylococcus aureus from Saxony and Brandenburg and Their Molecular Characterisation. Pathogens 2025; 14:238. [PMID: 40137723 PMCID: PMC11945114 DOI: 10.3390/pathogens14030238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Panton-Valentine leukocidin (PVL) is a staphylococcal toxin associated with chronic/recurrent skin and soft tissue infections (SSTIs) and necrotizing pneumonia. Its detection in clinical isolates of Staphylococcus aureus warrants aggressive therapy and infection control measures. However, PVL detection relies on molecular methods of limited use, especially in outpatient or resource-poor settings. In order to aid the development of a lateral flow (LF) test for PVL, clinical isolates from SSTIs were collected in 2020/21 at three laboratories in two cities in the Eastern part of Germany. After the exclusion of duplicate and serial isolates, 83 isolates were eligible. These were tested using an experimental LF test for PVL production. They were also characterized using DNA microarrays, facilitating the detection of virulence and resistance markers as well as the assignment to clonal complexes and epidemic/pandemic strains. Thirty-nine isolates (47%) were PVL-positive, and the LF results were in 81 cases (97.6%) concordant with genotyping. One false-positive and one false-negative case were observed. This translated into a diagnostic sensitivity of 0.974 and a diagnostic specificity of 0.977. The most common PVL-positive MSSA lineages were CC152 (n = 6), CC121 (n = 4), and CC5 and CC30 (each n = 2). Thirty isolates (36%) were mecA-positive. The MRSA rate among PVL-negatives was 20% (nine isolates), but among the PVL-positives, it was as high as 54% (n = 21). The most common PVL-MRSA strains were CC398-MRSA-VT (n = 5), CC5-MRSA-IV "Sri Lanka Clone" (n = 4), CC8-MRSA-[mec IV+Hg] "Latin American USA300" (n = 4), and CC22-MRSA-IV (PVL+/tst+) (n = 2). While the PVL rate was similar just like the German isolates from a previous study a decade before, the MRSA rate among PVL-positives was clearly higher. All PVL-MRSA strains detected, as well as the most common methicillin-susceptible lineage (CC152), are known to be common locally in other parts of the world, and might, thus, be regarded as travel-associated. Therefore, patients with suspected PVL-associated disease should be asked for their history of travel or migration, and, in case of hospitalization, they should be treated as MRSA cases until proven otherwise.
Collapse
Affiliation(s)
- Elke Müller
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), Leibniz Center for Photonics in Infection Research (LPI), Germany and InfectoGnostics Research Campus, 07745 Jena, Germany
- InfectoGnostics Research Campus, Centre for Applied Research, 07745 Jena, Germany
| | - Stefan Monecke
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), Leibniz Center for Photonics in Infection Research (LPI), Germany and InfectoGnostics Research Campus, 07745 Jena, Germany
- InfectoGnostics Research Campus, Centre for Applied Research, 07745 Jena, Germany
| | | | | | - Annett Reissig
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), Leibniz Center for Photonics in Infection Research (LPI), Germany and InfectoGnostics Research Campus, 07745 Jena, Germany
- InfectoGnostics Research Campus, Centre for Applied Research, 07745 Jena, Germany
| | - Lukas Rüttiger
- Senova Gesellschaft für Biowissenschaft und Technik mbH, 99427 Weimar, Germany
| | - Percy Schröttner
- Institute for Medical Microbiology and Virology, Faculty of Medicine and University Hospital “Carl Gustav Carus”, Technische Universität Dresden, 01307 Dresden, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital “Carl Gustav Carus”, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ilona Schwede
- IMD Labor Oderland GmbH, 15230 Frankfurt (Oder), Germany
| | - Hans-Herman Söffing
- Senova Gesellschaft für Biowissenschaft und Technik mbH, 99427 Weimar, Germany
| | | | - Ralf Ehricht
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), Leibniz Center for Photonics in Infection Research (LPI), Germany and InfectoGnostics Research Campus, 07745 Jena, Germany
- InfectoGnostics Research Campus, Centre for Applied Research, 07745 Jena, Germany
- Institute of Physical Chemistry, Friedrich Schiller University Jena, 07745 Jena, Germany
| |
Collapse
|
3
|
Turner AB, Zermeño-Pérez D, Mysior MM, Giraldo-Osorno PM, García B, O'Gorman E, Oubihi S, Simpson JC, Lasa I, Ó Cróinín T, Trobos M. Biofilm morphology and antibiotic susceptibility of methicillin-resistant Staphylococcus aureus (MRSA) on poly-D,L-lactide- co-poly(ethylene glycol) (PDLLA-PEG) coated titanium. Biofilm 2024; 8:100228. [PMID: 39830519 PMCID: PMC11740804 DOI: 10.1016/j.bioflm.2024.100228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 01/22/2025] Open
Abstract
Biodegradable polymeric coatings are being explored as a preventive strategy for orthopaedic device-related infection. In this study, titanium surfaces (Ti) were coated with poly-D,L-lactide (PDLLA, (P)), polyethylene-glycol poly-D,L-lactide (PEGylated-PDLLA, (PP20)), or multi-layered PEGylated-PDLLA (M), with or without 1 % silver sulfadiazine. The aim was to evaluate their cytocompatibility, resistance to Staphylococcus aureus biofilm formation, and their potential to enhance the susceptibility of any biofilm formed to antibiotics. Using automated high-content screening confocal microscopy, biofilm formation of a clinical methicillin-resistant Staphylococcus aureus (MRSA) isolate expressing GFP was quantified, along with isogenic mutants that were unable to form polysaccharidic or proteinaceous biofilm matrices. The results showed that PEGylated-PDLLA coatings exhibited significant antibiofilm properties, with M showing the highest effect. This inhibitory effect was stronger in S. aureus biofilms with a matrix composed of proteins compared to those with an exopolysaccharide (PIA) biofilm matrix. Our data suggest that the antibiofilm effect may have been due to (i) inhibition of the initial attachment through microbial surface components recognising adhesive matrix molecules (MSCRAMMs), since PEG reduces protein surface adsorption via surface hydration layer and steric repulsion; and (ii) mechanical disaggregation and dispersal of microcolonies due to the bioresorbable/degradable nature of the polymers, which undergo hydration and hydrolysis over time. The disruption of biofilm morphology by the PDLLA-PEG co-polymers increased S. aureus susceptibility to antibiotics like rifampicin and fusidic acid. Adding 1 % AgSD provided additional early bactericidal effects on both biofilm and planktonic S. aureus. Additionally, the coatings were cytocompatible with immune cells, indicating their potential to enhance bacterial clearance and reduce bacterial colonisation of titanium-based orthopaedic biomaterials.
Collapse
Affiliation(s)
- Adam Benedict Turner
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| | - David Zermeño-Pérez
- Ashland Specialties Ireland Ltd., Mullingar, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Margaritha M. Mysior
- Cell Screening Laboratory, UCD School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | - Paula Milena Giraldo-Osorno
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| | - Begoña García
- Microbial Pathogenesis Laboratory. Navarrabiomed-Complejo Hospitalario de Navarra (CHN)-Universidad Pública de Navarra (UPNA), IDISNA, Pamplona, Navarra, Spain
| | - Elizabeth O'Gorman
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Shafik Oubihi
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Jeremy C. Simpson
- Cell Screening Laboratory, UCD School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | - Iñigo Lasa
- Microbial Pathogenesis Laboratory. Navarrabiomed-Complejo Hospitalario de Navarra (CHN)-Universidad Pública de Navarra (UPNA), IDISNA, Pamplona, Navarra, Spain
| | - Tadhg Ó Cróinín
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Margarita Trobos
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| |
Collapse
|
4
|
Shin S, Yu J, Tae H, Zhao Y, Jiang D, Qiao Y, Kim W, Cho NJ. Exploring the Membrane-Active Interactions of Antimicrobial Long-Chain Fatty Acids Using a Supported Lipid Bilayer Model for Gram-Positive Bacterial Membranes. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56705-56717. [PMID: 39388376 DOI: 10.1021/acsami.4c11158] [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: 10/12/2024]
Abstract
The dynamic nature of bacterial lipid membranes significantly impacts the efficacy of antimicrobial therapies. However, traditional assay methods often fall short in replicating the complexity of these membranes, necessitating innovative approaches. Herein, we successfully fabricated model bacterially supported lipid bilayers (SLBs) that closely mimic the characteristics of Gram-positive bacteria using the solvent-assisted lipid bilayer (SALB) technique. By employing a quartz crystal microbalance with dissipation and fluorescence microscopy, we investigated the interactions between these bacterial mimetic membranes and long-chain unsaturated fatty acids. Specifically, linolenic acid (LNA) and linoleic acid (LLA) demonstrated interaction behaviors correlated with the critical micelle concentration (CMC) on Gram-positive membranes, resulting in membrane remodeling and removal at concentrations above their respective CMC values. In contrast, oleic acid (OA), while showing similar membrane remodeling patterns to LNA and LLA, exhibited membrane insertion and CMC-independent activity on the Gram-positive membranes. Particularly, LNA and LLA demonstrated bactericidal effects and promoted membrane permeability and ATP leakage in the bacterial membranes. OA, characterized by a CMC-independent activity profile, exhibited potent bactericidal effects due to its robust penetration into the SLBs, also enhancing membrane permeability and ATP leakage. These findings shed light on the intricate molecular mechanisms governing the interactions between long-chain unsaturated fatty acids and bacterial membranes. Importantly, this study underscores the potential of using biologically relevant model bacterial membrane systems to develop innovative strategies for combating bacterial infections and designing effective therapeutic agents.
Collapse
Affiliation(s)
- Sungmin Shin
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- Singapore-HUJ Alliance for Research and Enterprise, Singapore HUJ Alliance Research Enterprise (SHARE) 1 CREATE Way, #03-09 Innovation Wing, Singapore 138602, Singapore
| | - Jingyeong Yu
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hyunhyuk Tae
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Yilin Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 62 Nanyang Avenue, Singapore 637459, Singapore
| | - Dongping Jiang
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Yuan Qiao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 62 Nanyang Avenue, Singapore 637459, Singapore
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- Singapore-HUJ Alliance for Research and Enterprise, Singapore HUJ Alliance Research Enterprise (SHARE) 1 CREATE Way, #03-09 Innovation Wing, Singapore 138602, Singapore
| |
Collapse
|
5
|
Ighem Chi S, Flint A, Weedmark K, Pagotto F, Ramirez-Arcos S. Comparative genome analyses of Staphylococcus aureus from platelet concentrates reveal rearrangements involving loss of type VII secretion genes. Access Microbiol 2024; 6:000820.v4. [PMID: 39697362 PMCID: PMC11652724 DOI: 10.1099/acmi.0.000820.v4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 08/02/2024] [Indexed: 12/20/2024] Open
Abstract
Staphylococcus aureus has been involved in transfusion-transmitted fatalities associated with platelet concentrates (PCs) due to its heightened pathogenicity enhanced by genome-encoded virulence and antibiotic resistance genes. This may be facilitated by mobile genetic elements (MGEs) that can cause rearrangements. Several factors contribute to S. aureus virulence, including the type VII secretion system (T7SS), composed of six core genes conserved across S. aureus strains. In this study, we conducted comparative genome analyses of five S. aureus isolates from PCs (CI/BAC/25/13 /W, PS/BAC/169/17 /W and PS/BAC/317/16 /W were detected during PCs screening with the BACT/ALERT automated culture system, and ATR-20003 and CBS2016-05 were missed during screening and caused septic transfusion reactions). Multiple alignments of the genomes revealed evidence of rearrangements involving phage Sa3int in PS/BAC/169/17 /W and PS/BAC/317/16 /W. While the former had undergone translocation of its immune evasion cluster (IEC), the latter had lost part of the phage, leaving behind the IEC. This observation highlights S. aureus genome plasticity. Unexpectedly, strain CBS2016-05 was found to encode a pseudo-type VII secretion system (T7SS) that had lost five of the conserved core genes (esxA, esaA, essA, esaB and essB) and contained a 5' truncated essC. Since these genes are essential for the function of the T7SS protein transport machinery, which plays a key role in S. aureus virulence, CBS2016-05 probably compensates by recruiting other export mechanisms and/or alternative virulence factors, such as neu-tralizing immunity proteins. This study unravels genome rearrangements in S. aureus isolated from PCs and reports the first S. aureus isolate lacking conserved T7SS core genes.
Collapse
Affiliation(s)
- Sylvia Ighem Chi
- Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Annika Flint
- Listeriosis Reference Centre, Microbiology Research Division, Bureau of Microbial Hazards, Food Directorate, Health Canada, Ottawa, Canada
- Microbiology Research Division, Bureau of Microbial Hazards, Food Directorate, Health Canada, Ottawa, Canada
| | - Kelly Weedmark
- Listeriosis Reference Centre, Microbiology Research Division, Bureau of Microbial Hazards, Food Directorate, Health Canada, Ottawa, Canada
- Microbiology Research Division, Bureau of Microbial Hazards, Food Directorate, Health Canada, Ottawa, Canada
| | - Franco Pagotto
- Listeriosis Reference Centre, Microbiology Research Division, Bureau of Microbial Hazards, Food Directorate, Health Canada, Ottawa, Canada
- Microbiology Research Division, Bureau of Microbial Hazards, Food Directorate, Health Canada, Ottawa, Canada
| | - Sandra Ramirez-Arcos
- Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
6
|
Monecke S, Boswihi S, Braun SD, Diezel C, Müller E, Reinicke M, Udo E, Ehricht R. Sequencing a CC239-MRSA-III with a novel composite SCC mec element from Kuwait. Eur J Clin Microbiol Infect Dis 2024; 43:1761-1775. [PMID: 38990431 DOI: 10.1007/s10096-024-04891-y] [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: 02/21/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024]
Abstract
Staphylococcus aureus CC239-MRSA-III is an ancient pandemic strain of hospital-associated, methicillin-resistant S. aureus that spread globally for decades and that still can be found in some parts of the world. In Kuwait, microarray-based surveillance identified from 2019 to 2022 a series of isolates of a hitherto unknown variant of this strain that carried a second set of recombinase genes, ccrA/B-2. To elucidate the structure of its SCCmec element, two isolates were subjected to nanopore sequencing. This revealed, in addition to ccrA/B-2, several SCC-associated genes including speG (spermidine N acetyltransferase) and a gene encoding a large "E-domain containing protein" (dubbed as edcP-SCC). This gene contained three regions consisting of multiple repeating units. In terms of sequence and structure it was similar but not identical to the biofilm-related aap gene from S. epidermidis. A review of published sequences identified edcP-SCC in eighteen genome sequences of S. aureus, S. epidermidis and S. capitis, and frequently it appears in a similar cluster of genes as in the strains sequenced herein. Isolates also carried a prophage with the adhesion factor sasX/sesI and aminoglycoside resistance genes. This is consistent with an affiliation to the "South-East Asian" Clade of CC239. The emergence of edcP-SCC and sasX-positive CC239 strain shows that, against a global trend towards community-associated MRSA, the ancient pandemic CC239 hospital strain still continues to evolve and to cause outbreaks.
Collapse
Affiliation(s)
- Stefan Monecke
- Leibniz Institute of Photonic Technology (IPHT), Leibniz Center for Photonics in Infection Research (LPI), Jena, Germany.
- InfectoGnostics Research Campus, Jena, Germany.
| | - Samar Boswihi
- Faculty of Medicine, Department of Microbiology, Kuwait University, Kuwait City, Kuwait
| | - Sascha D Braun
- Leibniz Institute of Photonic Technology (IPHT), Leibniz Center for Photonics in Infection Research (LPI), Jena, Germany
- InfectoGnostics Research Campus, Jena, Germany
| | - Celia Diezel
- Leibniz Institute of Photonic Technology (IPHT), Leibniz Center for Photonics in Infection Research (LPI), Jena, Germany
- InfectoGnostics Research Campus, Jena, Germany
| | - Elke Müller
- Leibniz Institute of Photonic Technology (IPHT), Leibniz Center for Photonics in Infection Research (LPI), Jena, Germany
- InfectoGnostics Research Campus, Jena, Germany
| | - Martin Reinicke
- Leibniz Institute of Photonic Technology (IPHT), Leibniz Center for Photonics in Infection Research (LPI), Jena, Germany
- InfectoGnostics Research Campus, Jena, Germany
| | - Edet Udo
- Faculty of Medicine, Department of Microbiology, Kuwait University, Kuwait City, Kuwait
| | - Ralf Ehricht
- Leibniz Institute of Photonic Technology (IPHT), Leibniz Center for Photonics in Infection Research (LPI), Jena, Germany
- InfectoGnostics Research Campus, Jena, Germany
- Institute of Physical Chemistry, Friedrich-Schiller University, Jena, Germany
| |
Collapse
|
7
|
Bwanga F, Mukashyaka C, Kateete DP, Tumuhamye J, Okeng A, Aboce E, Namugga O, Kwizera R, Sommerfelt H, Nankabirwa V. Vaginal colonization with virulent and methicillin resistant Staphylococcus aureus among Ugandan women in Labour. BMC Microbiol 2024; 24:307. [PMID: 39155368 PMCID: PMC11331675 DOI: 10.1186/s12866-024-03460-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/12/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND Staphylococcus aureus (S. aureus) often colonizes the human skin, upper respiratory and genital tracts. In the female genital tract, it can be passed on to the newborn during vaginal delivery leading to either ordinary colonization, or neonatal infections notably umbilical stump sepsis, scalded skin syndrome, arthritis, or bacteraemia/sepsis. These infections are mediated by staphylococcal virulence factors such as (i) Staphylococcal Enterotoxins A, B, C, D, and E encoded by the sea, seb, sec, sed, see genes, (ii) Exfoliative Toxins A and B encoded by the eta and etb genes, (iii) Toxic Shock Syndrome Toxin 1 (TSST-1) encoded by the tst gene, (iv) Panton-Valentine Leukocidin (PVL) encoded by the pvl gene, and (v) Hemolysins alpha and delta encoded by the hla and hld genes, respectively. We determined the prevalence of S. aureus possessing one or more virulence factor genes and of methicillin resistant Staphylococcus aureus (MRSA) in this population. METHODS This was a cross-sectional study, which used 85 S. aureus isolates from the Chlorohexidine (CHX) clinical trial study in Uganda. The isolates had been obtained by culturing vaginal swabs (VS) from 1472 women in labour, frozen at minus 80oC, then thawed, sub-cultured, and tested for the selected virulence genes sea, seb, sec, sed, see eta, etb, tst, pvl, hla and hld, and for the methicillin resistance determining gene (mecA). Data were analyzed using SPSS version 20. RESULTS Of the 85 S. aureus isolates 13 (15.3%) were positive for one or more virulence factor genes, as follows: pvl 9/85 (10.6%), hld 5/85 (5.9%), sea 1/85 (1.2%) and seb genes 1/85 (1.2%). The other virulence genes (sec, sed, see, eta, etb, hla and tst) were not detected in any of the isolates. MRSA was detected in 55.3% (47/85) of the isolates, but only two of these carried the pvl virulence gene. CONCLUSION This study demonstrated that 15% of the S. aureus colonizing the female lower genital tract of mothers in labour in central Uganda carried one or more virulence genes, mostly pvl, indicating potential for newborn infection with S. aureus acquired in the maternal birth canal. More than half of the isolates were MRSA.
Collapse
Affiliation(s)
- Freddie Bwanga
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, P. O Box 7072, Kampala, Uganda
| | - Claudine Mukashyaka
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, P. O Box 7072, Kampala, Uganda.
| | - David Patrick Kateete
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, P. O Box 7072, Kampala, Uganda
| | - Josephine Tumuhamye
- Centre for Intervention Science in Maternal and Child Health, University of Bergen, Bergen, Norway
| | | | | | - Olive Namugga
- School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Richard Kwizera
- Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Halvor Sommerfelt
- Centre for Intervention Science in Maternal and Child Health, University of Bergen, Bergen, Norway
| | - Victoria Nankabirwa
- Centre for Intervention Science in Maternal and Child Health, University of Bergen, Bergen, Norway
- School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| |
Collapse
|
8
|
Henríquez L, Martín C, Echeverz M, Lasa Í, Ezpeleta C, Portillo ME. Evaluation of the use of sonication combined with enzymatic treatment for biofilm removal in the microbiological diagnosis of prosthetic joint infection. Microbiol Spectr 2024; 12:e0002024. [PMID: 38916322 PMCID: PMC11302281 DOI: 10.1128/spectrum.00020-24] [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: 01/04/2024] [Accepted: 05/14/2024] [Indexed: 06/26/2024] Open
Abstract
Sonicating explanted prosthetic implants to physically remove biofilms is a recognized method for improving the microbiological diagnosis of prosthetic joint infection (PJI); however, chemical and enzymatic treatments have been investigated as alternative biofilm removal methods. We compared the biofilm dislodging efficacy of sonication followed by the addition of enzyme cocktails with different activity spectra in the diagnosis of PJI with that of the sonication of fluid cultures alone. Consecutive patients who underwent prosthesis explantation due to infection at our institution were prospectively enrolled for 1 year. The diagnostic procedure included the collection of five intraoperative tissue cultures, sonication of the removed devices, and conventional culture of the sonication fluid. The resulting sonication fluid was also treated with an enzyme cocktail consisting of homemade dispersin B (0.04 µg/mL) and proteinase K (Sigma; 100 µg/mL) for 45 minutes at 37°C. The resulting sonication (S) and sonication with subsequent enzymatic treatment (SE) fluids were plated for aerobic and anaerobic culture broth for 7 days (aerobic) or 14 days (anaerobic). Identification was performed by matrix-assisted laser desorption ionization-time-of-flight mass spectrometry (Bruker). We included 107 patients from whom a prosthetic implant had been removed, among which PJI was diagnosed in 36 (34%). The sensitivity of S alone was significantly greater than that of SE alone (82% vs 71%; P < 0.05). Four patients with PJI were positive after sonication alone but negative after sonication plus enzymatic treatment. The four microorganisms missed after the addition of the enzyme cocktail were Staphylococcus aureus, two coagulase-negative Staphylococci, and Cutibacterium acnes. In conclusion, sonication alone was more sensitive than sonication followed by enzymatic treatment. The combination of these two methods had no synergistic effect; in contrast, the results suggest that the combination of both dislodgment methods affects the viability of gram-positive microorganisms. IMPORTANCE While the potential of sonication and enzymes as biofilm dispersal agents has been previously described, the originality of our work resides in the combination of both methods, which is hypothesized to enhance the ability to remove biofilm and, therefore, improve the microbiological diagnosis of PJI.
Collapse
Affiliation(s)
- Lucía Henríquez
- Department of Clinical Microbiology, University Hospital of Navarra, Institute of Healthcare Research of Navarra (IdiSNa), Pamplona, Spain
| | - Carmen Martín
- Department of Clinical Microbiology, University Hospital of Navarra, Institute of Healthcare Research of Navarra (IdiSNa), Pamplona, Spain
| | - Maite Echeverz
- Laboratory of Microbial Pathogenesis, Navarrabiomed, Public University of Navarra (UPNA), Institute of Healthcare Research of Navarra (IdiSNa), Pamplona, Spain
| | - Íñigo Lasa
- Laboratory of Microbial Pathogenesis, Navarrabiomed, Public University of Navarra (UPNA), Institute of Healthcare Research of Navarra (IdiSNa), Pamplona, Spain
| | - Carmen Ezpeleta
- Department of Clinical Microbiology, University Hospital of Navarra, Institute of Healthcare Research of Navarra (IdiSNa), Pamplona, Spain
| | - María Eugenia Portillo
- Department of Clinical Microbiology, University Hospital of Navarra, Institute of Healthcare Research of Navarra (IdiSNa), Pamplona, Spain
- CIBER, Epidemiología y Salud Pública, (CIBERESP), Madrid, Spain
| |
Collapse
|
9
|
Yang J, Gould TJ, Jeon B, Ji Y. Broad-Spectrum Antibacterial Activity of Antioxidant Octyl Gallate and Its Impact on Gut Microbiome. Antibiotics (Basel) 2024; 13:731. [PMID: 39200031 PMCID: PMC11350663 DOI: 10.3390/antibiotics13080731] [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: 07/04/2024] [Revised: 08/01/2024] [Accepted: 08/03/2024] [Indexed: 09/01/2024] Open
Abstract
In this study, we investigated the antibacterial activity of octyl gallate (OG), an antioxidant food additive, against both Gram-positive and Gram-negative bacterial pathogens. OG demonstrated robust bactericidal activity against Gram-positive bacterial pathogens with minimum inhibitory concentrations (MIC) of 4 to 8 µg/mL and minimum bactericidal concentrations (MBC) of 8 to 16 µg/mL in vitro. However, OG exhibited limited antibacterial activity against Gram-negative bacteria, including E. coli, although it could inhibit bacterial growth in vitro. Importantly, OG administration in mice altered the fecal microbiome, significantly reducing microbial diversity, modifying community structure, and increasing the abundance of beneficial bacteria. Additionally, OG displayed low cytotoxicity and hemolytic activity. These findings suggest that OG could be developed as a novel antibacterial agent, particularly against multi-drug-resistant MRSA. Our results provide new insights into the therapeutic potential of OG in modulating the gut microbiome, especially in conditions associated with microbial imbalance, while ensuring food safety.
Collapse
Affiliation(s)
- Junshu Yang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | - Trevor J. Gould
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Byeonghwa Jeon
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Saint Paul, MN 55108, USA
| | - Yinduo Ji
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
10
|
Suzuki Y, Kawada-Matsuo M, Le MNT, Eng S, Hisatsune J, Sugai M, Sakaguchi T, Komatsuzawa H. The two-component regulatory systems GraRS and SrrAB mediate Staphylococcus aureus susceptibility to Pep5 produced by clinical isolate of Staphylococcus epidermidis. Appl Environ Microbiol 2024; 90:e0030024. [PMID: 38832774 PMCID: PMC11267926 DOI: 10.1128/aem.00300-24] [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: 02/26/2024] [Accepted: 05/08/2024] [Indexed: 06/05/2024] Open
Abstract
Staphylococcus aureus is a common bacterium on the skin and in the nose that sometimes causes severe illness. Bacteriocins, antimicrobial peptides, or proteins produced by bacteria are candidates for the treatment of S. aureus infection. In this study, we found that a clinical Staphylococcus epidermidis strain, KSE112, produced the lantibiotic Pep5, which showed anti-S. aureus activity. The complete nucleotide sequence of the Pep5-encoding plasmid was determined. Several S. aureus two-component regulatory systems (TCSs) are known to be involved in bacteriocin susceptibility. Therefore, susceptibility tests were performed using TCS-inactivated S. aureus mutants to determine which TCS is responsible for Pep5 susceptibility; the ΔgraRS mutant exhibited increased susceptibility to Pep5, while the ΔsrrAB mutant exhibited decreased susceptibility. GraRS is known to regulate dltABCD and mprF in concert with vraFG, and Pep5 susceptibility was significantly increased in the ΔdltABCD, ΔmprF, and ΔvraFG mutants. Regarding the ΔsrrAB mutant, cross-resistance to aminoglycosides was observed. As aminoglycoside activity is known to be affected by aerobic respiration, we focused on qoxABCD and cydAB, which are quinol oxidase genes that are necessary for aerobic respiration and have downregulated the expression in the ΔsrrAB mutant. We constructed ΔqoxABCD and ΔcydAB mutants and found that qoxABCD inactivation decreased susceptibility to Pep5 and aminoglycosides. These results indicate that reduced aerobic respiration due to the reduced qoxABCD expression in the ΔsrrAB mutant decreased Pep5 activity.IMPORTANCEThe emergence of drug-resistant bacteria, including MRSA, is a severe health problem worldwide. Thus, the development of novel antimicrobial agents, including bacteriocins, is needed. In this report, we found a Pep5-producing strain with anti-S. aureus activity. We determined the complete sequence of the Pep5-encoding plasmid for the first time. However, in S. aureus, GraRS and its effectors conferred decreased susceptibility to Pep5. We also revealed that another TCS, SrrAB, affects susceptibility Pep5 and other lantibiotics by controlling aerobic respiration. In our study, we investigated the efficacy of Pep5 against S. aureus and other Gram-positive bacteria and revealed that respiratory constancy regulated by TCS is required for the antimicrobial activity of nisin, nukacin, and Pep5. These findings provide important information for the clinical application of bacteriocins and suggest that they have different properties among similar pore-forming lantibiotics.
Collapse
Affiliation(s)
- Yujin Suzuki
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Department of Virology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashi Murayama, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Project Research Centre for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan
| | - Mi Nguyen-Tra Le
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Project Research Centre for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan
| | - Sopongselamuny Eng
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Junzo Hisatsune
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashi Murayama, Japan
- Project Research Centre for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan
| | - Motoyuki Sugai
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashi Murayama, Japan
- Project Research Centre for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan
| | - Takemasa Sakaguchi
- Department of Virology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Project Research Centre for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
11
|
Motomura Y, Miyazaki M, Kamada M, Morimoto S, Nakamura Y, Satho T, Takata T, Kashige N. Genotypic Shift and Diversification of MRSA Blood Stream Isolates in a University Hospital Setting: Evidence from a 12-Year Observational Study. Antibiotics (Basel) 2024; 13:670. [PMID: 39061352 PMCID: PMC11273934 DOI: 10.3390/antibiotics13070670] [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: 06/29/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
There have been few reports regarding the long-term trends in the genotypes of methicillin-resistant Staphylococcus aureus (MRSA) bloodstream isolates. Therefore, this study was performed to investigate the longitudinal trends in the genotypes of MRSA bloodstream isolates obtained from hospitalized patients during a 12-year study period from 2010 to 2021 at a tertiary care university hospital. Over the 12-year period from 2010 to 2021, we conducted a genetic investigation focusing on 245 MRSA strains isolated from the blood of hospitalized patients. The genotypes of the MRSA bloodstream isolates were determined by Staphylococcal Cassette Chromosome mec (SCCmec) typing, accessory gene regulator (agr) typing, PCR-based ORF typing (POT), and multilocus sequence typing (MLST). Strains with the same POT type detected in two or more isolates were designated as epidemic clones, while strains without a common POT type were classified as sporadic clones. Until 2015, isolates with SCCmec II/agr II were prevalent, but isolates with SCCmec IV/agr III increased from 2016. A total of 128 strains (52%) were identified as epidemic clones, while 117 strains (48%) were classified as sporadic clones. The detection rate of sporadic clones increased significantly since 2016 (p < 0.05). The epidemic clones were classified into three clusters, with MRSA of clonal complex (CC) 1 being prominent after 2016. This study showed that the genotypes of MRSA bloodstream isolates underwent a shift from SCCmec II/agr II type to SCCmec IV/agr III type, with a notable increase in MRSA of CC1, after 2016. There was a significant increase in the proportion of sporadic strains among the isolates, suggesting the diversification of genotypes.
Collapse
Affiliation(s)
- Yuka Motomura
- Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (Y.M.); (M.M.); (T.S.); (N.K.)
| | - Motoyasu Miyazaki
- Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (Y.M.); (M.M.); (T.S.); (N.K.)
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan
| | - Mitsuhiro Kamada
- Department of Pharmacy, Fukuoka University Hospital, Fukuoka 814-0180, Japan;
| | - Shinichi Morimoto
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan; (S.M.); (Y.N.)
| | - Yoshihiko Nakamura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan; (S.M.); (Y.N.)
| | - Tomomitsu Satho
- Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (Y.M.); (M.M.); (T.S.); (N.K.)
| | - Tohru Takata
- Department of Oncology, Hematology, and Infectious Diseases, Fukuoka University Hospital, Fukuoka 814-0180, Japan
- Department of Infection Control, Fukuoka University Hospital, Fukuoka 814-0180, Japan
| | - Nobuhiro Kashige
- Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (Y.M.); (M.M.); (T.S.); (N.K.)
| |
Collapse
|
12
|
Katahira K, Gotoh Y, Kasama K, Yoshimura D, Itoh T, Shimauchi C, Tajiri A, Hayashi T. Mobile genetic element-driven genomic changes in a community-associated methicillin-resistant Staphylococcus aureus clone during its transmission in a regional community outbreak in Japan. Microb Genom 2024; 10:001272. [PMID: 39017043 PMCID: PMC11316552 DOI: 10.1099/mgen.0.001272] [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: 03/25/2024] [Accepted: 06/26/2024] [Indexed: 07/18/2024] Open
Abstract
Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) infections are now a public health concern in both community and healthcare settings worldwide. We previously identified a suspected case of a maternity clinic-centred outbreak of CA-MRSA skin infection in a regional community in Japan by PFGE-based analysis. In this study, we performed genome sequence-based analyses of 151 CA-MRSA isolates, which included not only outbreak-related isolates that we previously defined based on identical or similar PFGE patterns but also other isolates obtained during the same period in the same region. Our analysis accurately defined 133 isolates as outbreak-related isolates, collectively called the TDC clone. They belonged to a CA-MRSA lineage in clonal complex (CC) 30, known as the South West Pacific (SWP) clone. A high-resolution phylogenetic analysis of these isolates combined with their epidemiological data revealed that the TDC clone was already present and circulating in the region before the outbreak was recognized, and only the isolates belonging to two sublineages (named SL4 and SL5) were directly involved in the outbreak. Long persistence in patients/carriers and frequent intrahousehold transmission of the TDC clone were also revealed by this analysis. Moreover, by systematic analyses of the genome changes that occurred in this CA-MRSA clone during transmission in the community, we revealed that most variations were associated with mobile genetic elements (MGEs). Variant PFGE types were generated by alterations of prophages and genomic islands or insertion sequence (IS)-mediated insertion of a plasmid or a sequence of unknown origin. Dynamic changes in plasmid content, which were linked to changes in antimicrobial resistance profiles in specific isolates, were generated by frequent gain and loss of plasmids, most of which were self-transmissible or mobilizable. The introduction of IS256 by a plasmid (named pTDC02) into sublineage SL5 led to SL5-specific amplification of IS256, and amplified IS256 copies were involved in some of the structural changes of chromosomes and plasmids and generated variations in the repertoire of virulence-related genes in limited isolates. These data revealed how CA-MRSA genomes change during transmission in the community and how MGEs are involved in this process.
Collapse
Affiliation(s)
- Katsuyuki Katahira
- Department of Bacteriology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Respiratory Medicine, NHO Omuta Hospital, Tachibana, Omuta City 837-0911, Japan
| | - Yasuhiro Gotoh
- Department of Bacteriology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kentaro Kasama
- Department of Bacteriology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Dai Yoshimura
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Meguro-ku, Tokyo 152-8550, Japan
| | - Takehiko Itoh
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Meguro-ku, Tokyo 152-8550, Japan
| | - Chieko Shimauchi
- Department of Nursing Humanics I, Miyazaki Prefectural Nursing University, Manabino, Miyazaki 880-0929, Japan
| | - Akihiko Tajiri
- Tajiri Dermatology Clinic, Kiyotake, Miyazaki 889-1067, Japan
| | - Tetsuya Hayashi
- Department of Bacteriology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
13
|
Podkowik M, Perault AI, Putzel G, Pountain A, Kim J, DuMont AL, Zwack EE, Ulrich RJ, Karagounis TK, Zhou C, Haag AF, Shenderovich J, Wasserman GA, Kwon J, Chen J, Richardson AR, Weiser JN, Nowosad CR, Lun DS, Parker D, Pironti A, Zhao X, Drlica K, Yanai I, Torres VJ, Shopsin B. Quorum-sensing agr system of Staphylococcus aureus primes gene expression for protection from lethal oxidative stress. eLife 2024; 12:RP89098. [PMID: 38687677 PMCID: PMC11060713 DOI: 10.7554/elife.89098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
The agr quorum-sensing system links Staphylococcus aureus metabolism to virulence, in part by increasing bacterial survival during exposure to lethal concentrations of H2O2, a crucial host defense against S. aureus. We now report that protection by agr surprisingly extends beyond post-exponential growth to the exit from stationary phase when the agr system is no longer turned on. Thus, agr can be considered a constitutive protective factor. Deletion of agr resulted in decreased ATP levels and growth, despite increased rates of respiration or fermentation at appropriate oxygen tensions, suggesting that Δagr cells undergo a shift towards a hyperactive metabolic state in response to diminished metabolic efficiency. As expected from increased respiratory gene expression, reactive oxygen species (ROS) accumulated more in the agr mutant than in wild-type cells, thereby explaining elevated susceptibility of Δagr strains to lethal H2O2 doses. Increased survival of wild-type agr cells during H2O2 exposure required sodA, which detoxifies superoxide. Additionally, pretreatment of S. aureus with respiration-reducing menadione protected Δagr cells from killing by H2O2. Thus, genetic deletion and pharmacologic experiments indicate that agr helps control endogenous ROS, thereby providing resilience against exogenous ROS. The long-lived 'memory' of agr-mediated protection, which is uncoupled from agr activation kinetics, increased hematogenous dissemination to certain tissues during sepsis in ROS-producing, wild-type mice but not ROS-deficient (Cybb-/-) mice. These results demonstrate the importance of protection that anticipates impending ROS-mediated immune attack. The ubiquity of quorum sensing suggests that it protects many bacterial species from oxidative damage.
Collapse
Affiliation(s)
- Magdalena Podkowik
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
| | - Andrew I Perault
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Gregory Putzel
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
- Microbial Computational Genomic Core Lab, NYU Grossman School of MedicineNew YorkUnited States
| | - Andrew Pountain
- Institute for Systems Genetics; NYU Grossman School of MedicineNew YorkUnited States
| | - Jisun Kim
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical SchoolNewarkUnited States
| | - Ashley L DuMont
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
| | - Erin E Zwack
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Robert J Ulrich
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
| | - Theodora K Karagounis
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Ronald O. Perelman Department of Dermatology; NYU Grossman School of MedicineNew YorkUnited States
| | - Chunyi Zhou
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
| | - Andreas F Haag
- School of Medicine, University of St AndrewsSt AndrewsUnited Kingdom
| | - Julia Shenderovich
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Gregory A Wasserman
- Department of Surgery, Northwell Health Lenox Hill HospitalNew YorkUnited States
| | - Junbeom Kwon
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
| | - John Chen
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Anthony R Richardson
- Department of Microbiology and Molecular Genetics, University of PittsburghPittsburghUnited States
| | - Jeffrey N Weiser
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Carla R Nowosad
- Department of Pathology, NYU Grossman School of MedicineNew YorkUnited States
| | - Desmond S Lun
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers UniversityCamdenUnited States
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical SchoolNewarkUnited States
| | - Alejandro Pironti
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
- Microbial Computational Genomic Core Lab, NYU Grossman School of MedicineNew YorkUnited States
| | - Xilin Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen UniversityXiamenChina
| | - Karl Drlica
- Public Health Research Institute, New Jersey Medical School, Rutgers UniversityNew YprkUnited States
- Department of Microbiology, Biochemistry & Molecular Genetics, New Jersey Medical School, Rutgers UniversityNewarkUnited States
| | - Itai Yanai
- Institute for Systems Genetics; NYU Grossman School of MedicineNew YorkUnited States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of MedicineNew YorkUnited States
| | - Victor J Torres
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Bo Shopsin
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
14
|
Crosby HA, Keim K, Kwiecinski JM, Langouët-Astrié CJ, Oshima K, LaRivière WB, Schmidt EP, Horswill AR. Host-derived protease promotes aggregation of Staphylococcus aureus by cleaving the surface protein SasG. mBio 2024; 15:e0348323. [PMID: 38511930 PMCID: PMC11005337 DOI: 10.1128/mbio.03483-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Staphylococcus aureus is one of the leading causes of hospital-acquired infections, many of which begin following attachment and accumulation on indwelling medical devices or diseased tissue. These infections are often linked to the establishment of biofilms, but another often overlooked key characteristic allowing S. aureus to establish persistent infection is the formation of planktonic aggregates. Such aggregates are physiologically similar to biofilms and protect pathogens from innate immune clearance and increase antibiotic tolerance. The cell-wall-associated protein SasG has been implicated in biofilm formation via mechanisms of intercellular aggregation but the mechanism in the context of disease is largely unknown. We have previously shown that the expression of cell-wall-anchored proteins involved in biofilm formation is controlled by the ArlRS-MgrA regulatory cascade. In this work, we demonstrate that the ArlRS two-component system controls aggregation, by repressing the expression of sasG by activation of the global regulator MgrA. We also demonstrate that SasG must be proteolytically processed by a non-staphylococcal protease to induce aggregation and that strains expressing functional full-length sasG aggregate significantly upon proteolysis by a mucosal-derived host protease found in human saliva. We used fractionation and N-terminal sequencing to demonstrate that human trypsin within saliva cleaves within the A domain of SasG to expose the B domain and induce aggregation. Finally, we demonstrated that SasG is involved in virulence during mouse lung infection. Together, our data point to SasG, its processing by host proteases, and SasG-driven aggregation as important elements of S. aureus adaptation to the host environment.IMPORTANCEHere, we demonstrate that the Staphylococcus aureus surface protein SasG is important for cell-cell aggregation in the presence of host proteases. We show that the ArlRS two-component regulatory system controls SasG levels through the cytoplasmic regulator MgrA. We identified human trypsin as the dominant protease triggering SasG-dependent aggregation and demonstrated that SasG is important for S. aureus lung infection. The discovery that host proteases can induce S. aureus aggregation contributes to our understanding of how this pathogen establishes persistent infections. The observations in this study demonstrate the need to strengthen our knowledge of S. aureus surface adhesin function and processing, regulation of adhesin expression, and the mechanisms that promote biofilm formation to develop strategies for preventing chronic infections.
Collapse
Affiliation(s)
- Heidi A. Crosby
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Klara Keim
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jakub M. Kwiecinski
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Christophe J. Langouët-Astrié
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kaori Oshima
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Wells B. LaRivière
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Eric P. Schmidt
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
15
|
Iturbe P, Martín AS, Hamamoto H, Marcet-Houben M, Galbaldón T, Solano C, Lasa I. Noncontiguous operon atlas for the Staphylococcus aureus genome. MICROLIFE 2024; 5:uqae007. [PMID: 38651166 PMCID: PMC11034616 DOI: 10.1093/femsml/uqae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/20/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
Bacteria synchronize the expression of genes with related functions by organizing genes into operons so that they are cotranscribed together in a single polycistronic messenger RNA. However, some cellular processes may benefit if the simultaneous production of the operon proteins coincides with the inhibition of the expression of an antagonist gene. To coordinate such situations, bacteria have evolved noncontiguous operons (NcOs), a subtype of operons that contain one or more genes that are transcribed in the opposite direction to the other operon genes. This structure results in overlapping transcripts whose expression is mutually repressed. The presence of NcOs cannot be predicted computationally and their identification requires a detailed knowledge of the bacterial transcriptome. In this study, we used direct RNA sequencing methodology to determine the NcOs map in the Staphylococcus aureus genome. We detected the presence of 18 NcOs in the genome of S. aureus and four in the genome of the lysogenic prophage 80α. The identified NcOs comprise genes involved in energy metabolism, metal acquisition and transport, toxin-antitoxin systems, and control of the phage life cycle. Using the menaquinone operon as a proof of concept, we show that disarrangement of the NcO architecture results in a reduction of bacterial fitness due to an increase in menaquinone levels and a decrease in the rate of oxygen consumption. Our study demonstrates the significance of NcO structures in bacterial physiology and emphasizes the importance of combining operon maps with transcriptomic data to uncover previously unnoticed functional relationships between neighbouring genes.
Collapse
Affiliation(s)
- Pablo Iturbe
- Laboratory of Microbial Pathogenesis, Navarrabiomed-Universidad Pública de Navarra (UPNA)-Hospital Universitario de Navarra (HUN), IdiSNA, Irunlarrea 3, Pamplona, 31008 Navarra, Spain
| | - Alvaro San Martín
- Laboratory of Microbial Pathogenesis, Navarrabiomed-Universidad Pública de Navarra (UPNA)-Hospital Universitario de Navarra (HUN), IdiSNA, Irunlarrea 3, Pamplona, 31008 Navarra, Spain
| | - Hiroshi Hamamoto
- Faculty of Medicine, Department of Infectious diseases, Yamagata University, 2-2-2 Lida-Nishi, 990-9585 Yamagata, Japan
| | - Marina Marcet-Houben
- Barcelona Supercomputing Centre (BSC-CNS). Plaça Eusebi Güell, 1-3, 08034 Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Toni Galbaldón
- Barcelona Supercomputing Centre (BSC-CNS). Plaça Eusebi Güell, 1-3, 08034 Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina Solano
- Laboratory of Microbial Pathogenesis, Navarrabiomed-Universidad Pública de Navarra (UPNA)-Hospital Universitario de Navarra (HUN), IdiSNA, Irunlarrea 3, Pamplona, 31008 Navarra, Spain
| | - Iñigo Lasa
- Laboratory of Microbial Pathogenesis, Navarrabiomed-Universidad Pública de Navarra (UPNA)-Hospital Universitario de Navarra (HUN), IdiSNA, Irunlarrea 3, Pamplona, 31008 Navarra, Spain
| |
Collapse
|
16
|
Sato'o Y, Hisatsune J, Aziz F, Tatsukawa N, Shibata-Nakagawa M, Ono HK, Naito I, Omoe K, Sugai M. Coordination of prophage and global regulator leads to high enterotoxin production in staphylococcal food poisoning-associated lineage. Microbiol Spectr 2024; 12:e0292723. [PMID: 38319074 PMCID: PMC10913437 DOI: 10.1128/spectrum.02927-23] [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: 07/25/2023] [Accepted: 01/05/2024] [Indexed: 02/07/2024] Open
Abstract
Staphylococcus species in food produce Staphylococcal enterotoxins (SEs) that cause Staphylococcal food poisoning (SFP). More than 20 SE types have been reported, among which Staphylococcal enterotoxin A (SEA) has been recognized as one of the most important SEs associated with SFP. However, the regulatory mechanisms underlying its production remain unclear. Previously, we identified a major SFP clone in Japan, CC81 subtype-1, which exhibits high SEA production. In this study, we attempted to identify the factors contributing to this phenomenon. Thus, we demonstrated that the attenuation of the activity of endogenous regulator, Staphylococcal accessory regulator S (SarS), and the lysogenization of a high SEA-producing phage contributed to this phenomenon in CC81 subtype-1. Furthermore, our results indicated that SarS could directly bind to the promoter upstream of the sea gene and suppress SEA expression; this low SarS repression activity was identified as one of the reasons for the high SEA production observed. Therefore, we revealed that both exogenous and endogenous factors may probably contribute to the high SEA production. Our results confirmed that SE production is a fundamental and critical factor in SFP and clarified the associated production mechanism while enhancing our understanding as to why a specific clone frequently causes SFP. IMPORTANCE The importance of this study lies in its unveiling of a molecular regulatory mechanism associated with the most important food poisoning toxin and the evolution of Staphylococcal food poisoning (SFP)-associated clone. SFP is primarily caused by Staphylococcus aureus, with Staphylococcal enterotoxin A (SEA) being commonly involved in many cases. Thus, SEA has been recognized as a major toxin type. However, despite almost a century since its discovery, the complete mechanism of SEA production is as yet unknown. In this study, we analyzed an SEA-producing SFP clone isolated in East Asia and discovered that this strain, besides acquiring the high SEA-producing phage, exhibits remarkably high SEA production due to the low activity of SarS, an intrinsic regulatory factor. This is the first report documenting the evolution of the SFP clone through the coordinated action of exogenous mobile genetic factors and endogenous regulators on this notorious toxin.
Collapse
Affiliation(s)
- Yusuke Sato'o
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Junzo Hisatsune
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | - Fatkhanuddin Aziz
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Nobuyuki Tatsukawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Mari Shibata-Nakagawa
- Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka city, Japan
| | - Hisaya K. Ono
- Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka city, Japan
- Laboratory of Zoonoses, Kitasato University School of Veterinary Medicine, Towada city, Japan
| | - Ikunori Naito
- Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka city, Japan
| | - Katsuhiko Omoe
- Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka city, Japan
| | - Motoyuki Sugai
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| |
Collapse
|
17
|
Podkowik M, Perault AI, Putzel G, Pountain A, Kim J, Dumont A, Zwack E, Ulrich RJ, Karagounis TK, Zhou C, Haag AF, Shenderovich J, Wasserman GA, Kwon J, Chen J, Richardson AR, Weiser JN, Nowosad CR, Lun DS, Parker D, Pironti A, Zhao X, Drlica K, Yanai I, Torres VJ, Shopsin B. Quorum-sensing agr system of Staphylococcus aureus primes gene expression for protection from lethal oxidative stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.08.544038. [PMID: 37333372 PMCID: PMC10274873 DOI: 10.1101/2023.06.08.544038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The agr quorum-sensing system links Staphylococcus aureus metabolism to virulence, in part by increasing bacterial survival during exposure to lethal concentrations of H2O2, a crucial host defense against S. aureus. We now report that protection by agr surprisingly extends beyond post-exponential growth to the exit from stationary phase when the agr system is no longer turned on. Thus, agr can be considered a constitutive protective factor. Deletion of agr increased both respiration and fermentation but decreased ATP levels and growth, suggesting that Δagr cells assume a hyperactive metabolic state in response to reduced metabolic efficiency. As expected from increased respiratory gene expression, reactive oxygen species (ROS) accumulated more in the agr mutant than in wild-type cells, thereby explaining elevated susceptibility of Δagr strains to lethal H2O2 doses. Increased survival of wild-type agr cells during H2O2 exposure required sodA, which detoxifies superoxide. Additionally, pretreatment of S. aureus with respiration-reducing menadione protected Δagr cells from killing by H2O2. Thus, genetic deletion and pharmacologic experiments indicate that agr helps control endogenous ROS, thereby providing resilience against exogenous ROS. The long-lived "memory" of agr-mediated protection, which is uncoupled from agr activation kinetics, increased hematogenous dissemination to certain tissues during sepsis in ROS-producing, wild-type mice but not ROS-deficient (Nox2-/-) mice. These results demonstrate the importance of protection that anticipates impending ROS-mediated immune attack. The ubiquity of quorum sensing suggests that it protects many bacterial species from oxidative damage.
Collapse
Affiliation(s)
- Magdalena Podkowik
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
| | - Andrew I. Perault
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Gregory Putzel
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
- Microbial Computational Genomic Core Lab, NYU Grossman School of Medicine, New York, NY, USA
| | - Andrew Pountain
- Institute for Systems Genetics; NYU Grossman School of Medicine, New York, NY, USA
| | - Jisun Kim
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School Cancer Center, Newark, NJ, USA
| | - Ashley Dumont
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Erin Zwack
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Robert J. Ulrich
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
| | - Theodora K. Karagounis
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Ronald O. Perelman Department of Dermatology; NYU Grossman School of Medicine, New York, NY, USA
| | - Chunyi Zhou
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
| | - Andreas F. Haag
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Julia Shenderovich
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Junbeom Kwon
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
| | - John Chen
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anthony R. Richardson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey N. Weiser
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Carla R. Nowosad
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Desmond S. Lun
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School Cancer Center, Newark, NJ, USA
| | - Alejandro Pironti
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
- Microbial Computational Genomic Core Lab, NYU Grossman School of Medicine, New York, NY, USA
| | - Xilin Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian Province, China
| | - Karl Drlica
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, USA
- Department of Microbiology, Biochemistry & Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Itai Yanai
- Institute for Systems Genetics; NYU Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Victor J. Torres
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Bo Shopsin
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
18
|
Sychla A, Stach CS, Roach SN, Hayward AN, Langlois RA, Smanski MJ. High-throughput investigation of genetic design constraints in domesticated Influenza A Virus for transient gene delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580300. [PMID: 38405907 PMCID: PMC10888799 DOI: 10.1101/2024.02.14.580300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Replication-incompetent single cycle infectious Influenza A Virus (sciIAV) has demonstrated utility as a research and vaccination platform. Protein-based therapeutics are increasingly attractive due to their high selectivity and potent efficacy but still suffer from low bioavailability and high manufacturing cost. Transient RNA-mediated delivery is a safe alternative that allows for expression of protein-based therapeutics within the target cells or tissues but is limited by delivery efficiency. Here, we develop recombinant sciIAV as a platform for transient gene delivery in vivo and in vitro for therapeutic, research, and manufacturing applications (in vivo antimicrobial production, cell culture contamination clearance, and production of antiviral proteins in vitro). While adapting the system to deliver new protein cargo we discovered expression differences presumably resulting from genetic context effects. We applied a high-throughput screen to map these within the 3'-untranslated and coding regions of the hemagglutinin-encoding segment 4. This screen revealed permissible mutations in the 3'-UTR and depletion of RNA level motifs in the N-terminal coding region.
Collapse
Affiliation(s)
- Adam Sychla
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Saint Paul, MN 55108
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108
| | - Christopher S Stach
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Saint Paul, MN 55108
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108
| | - Shanley N Roach
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Saint Paul, MN 55108
- Department of Microbiology and Immunology, University of Minnesota, Saint Paul, MN 55108
| | - Amanda N Hayward
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Saint Paul, MN 55108
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108
| | - Ryan A Langlois
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Saint Paul, MN 55108
- Department of Microbiology and Immunology, University of Minnesota, Saint Paul, MN 55108
| | - Michael J Smanski
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Saint Paul, MN 55108
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108
| |
Collapse
|
19
|
Che Hamzah AM, Chew CH, Al-Trad EI, Puah SM, Chua KH, A Rahman NI, Ismail S, Maeda T, Palittapongarnpim P, Yeo CC. Whole genome sequencing of methicillin-resistant Staphylococcus aureus clinical isolates from Terengganu, Malaysia, indicates the predominance of the EMRSA-15 (ST22-SCCmec IV) clone. Sci Rep 2024; 14:3485. [PMID: 38347106 PMCID: PMC10861583 DOI: 10.1038/s41598-024-54182-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/09/2024] [Indexed: 02/15/2024] Open
Abstract
Despite the importance of methicillin-resistant Staphylococcus aureus (MRSA) as a priority nosocomial pathogen, the genome sequences of Malaysian MRSA isolates are currently limited to a small pool of samples. Here, we present the genome sequence analyses of 88 clinical MRSA isolates obtained from the main tertiary hospital in Terengganu, Malaysia in 2016-2020, to obtain in-depth insights into their characteristics. The EMRSA-15 (ST22-SCCmec IV) clone of the clonal complex 22 (CC22) lineage was predominant with a total of 61 (69.3%) isolates. Earlier reports from other Malaysian hospitals indicated the predominance of the ST239 clone, but only two (2.3%) isolates were identified in this study. Two Indian-origin clones, the Bengal Bay clone ST772-SCCmec V (n = 2) and ST672 (n = 10) were also detected, with most of the ST672 isolates obtained in 2020 (n = 7). Two new STs were found, with one isolate each, and were designated ST7879 and ST7883. From the core genome phylogenetic tree, the HSNZ MRSA isolates could be grouped into seven clades. Antimicrobial phenotype-genotype concordance was high (> 95%), indicating the accuracy of WGS in predicting most resistances. Majority of the MRSA isolates were found to harbor more than 10 virulence genes, demonstrating their pathogenic nature.
Collapse
Affiliation(s)
- Ainal Mardziah Che Hamzah
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, 21300, Kuala Nerus, Terengganu, Malaysia
| | - Ching Hoong Chew
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, 21300, Kuala Nerus, Terengganu, Malaysia.
| | - Esra'a Ibrahim Al-Trad
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, 20400, Kuala Terengganu, Terengganu, Malaysia
- Faculty of Allied Medical Sciences, Jadara University, Irbid, Jordan
| | - Suat Moi Puah
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nor Iza A Rahman
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, 20400, Kuala Terengganu, Terengganu, Malaysia
| | - Salwani Ismail
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, 20400, Kuala Terengganu, Terengganu, Malaysia
| | - Toshinari Maeda
- Department of Biological Functions and Engineering, Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu-Ku, Kitakyushu, 808-0196, Japan
| | - Prasit Palittapongarnpim
- Pornchai Matangkasombut Center for Microbial Genomics (CENMIG), Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Chew Chieng Yeo
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, 20400, Kuala Terengganu, Terengganu, Malaysia.
| |
Collapse
|
20
|
Mihai MM, Popa MI, Holban AM, Gheorghe-Barbu I, Popa LG, Chifiriuc MC, Giurcăneanu C, Bleotu C, Cucu CI, Lazăr V. Clinical and microbiological features of host-bacterial interplay in chronic venous ulcers versus other types of chronic skin ulcers. Front Microbiol 2024; 14:1326904. [PMID: 38375067 PMCID: PMC10875999 DOI: 10.3389/fmicb.2023.1326904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/19/2023] [Indexed: 02/21/2024] Open
Abstract
Introduction Chronic venous ulcers of the lower limbs develop in the context of advanced venous disease and have a significant impact on the patient's quality of life, being associated with depression and worrisome suicide rates, as well as with an economic burden caused by increased medical care costs and high epidemiological risks of healthcare associated infections and emergence of strains resistant to multiple classes of antibiotics and/ or antiseptics. Although numerous studies have investigated the composition of the chronic wounds microbiome, either by culture-dependent or independent methods, there are no data on the association between virulence and resistance profiles of strains isolated from venous ulcers and the clinical picture of this pathology. The elucidation of pathogenic mechanisms, at both phenotypic and molecular level, is crucial in the fight against these important human microbial agents, in order to develop novel biomarkers and discover new therapeutic targets. Methods In this study we aimed to characterize the phenotypic virulence profiles (including the ability to develop biofilms) of microorganisms isolated from chronic skin wounds and to correlate them with the clinical symptomatology. Considering the high incidence of Staphylococcus aureus infections in chronic ulcers, but also the ability of this species to develop multi-drug resistance, we performed an more in-depth study of the phenotypic and genotypic virulence profiles of methicillin-resistant Staphylococcus. Results The study revealed important differences regarding the clinical evolution and virulence profiles of microorganisms isolated from lower limb wounds, as well as between patients diagnosed with chronic venous ulcers and those with lesions of different etiology.
Collapse
Affiliation(s)
- Mara Mădălina Mihai
- Department of Oncologic Dermatology–“Elias” University Emergency Hospital, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Department of Botany-Microbiology, Faculty of Biology, Research Institute of the University of Bucharest, University of Bucharest, Bucharest, Romania
| | - Mircea Ioan Popa
- Department of Microbiology—“Cantacuzino” Institute, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Alina Maria Holban
- Department of Botany-Microbiology, Faculty of Biology, Research Institute of the University of Bucharest, University of Bucharest, Bucharest, Romania
| | - Irina Gheorghe-Barbu
- Department of Botany-Microbiology, Faculty of Biology, Research Institute of the University of Bucharest, University of Bucharest, Bucharest, Romania
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology–“Elias” University Emergency Hospital, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Department of Botany-Microbiology, Faculty of Biology, Research Institute of the University of Bucharest, University of Bucharest, Bucharest, Romania
| | - Călin Giurcăneanu
- Department of Oncologic Dermatology–“Elias” University Emergency Hospital, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Coralia Bleotu
- Department of Botany-Microbiology, Faculty of Biology, Research Institute of the University of Bucharest, University of Bucharest, Bucharest, Romania
- Cellular and Molecular Department, “Ştefan S. Nicolau” Institute of Virology, Bucharest, Romania
| | - Corina Ioana Cucu
- Department of Oncologic Dermatology–“Elias” University Emergency Hospital, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Veronica Lazăr
- Department of Botany-Microbiology, Faculty of Biology, Research Institute of the University of Bucharest, University of Bucharest, Bucharest, Romania
| |
Collapse
|
21
|
Vasconcelos PC, Leite EL, Saraiva MMS, Ferrari RG, Cibulski SP, Silva NMV, Freitas Neto OC, Givisiez PEN, Vieira RFC, Oliveira CJB. Genomic Analysis of a Community-Acquired Methicillin-Resistant Staphylococcus aureus Sequence Type 1 Associated with Caprine Mastitis. Pathogens 2023; 13:23. [PMID: 38251331 PMCID: PMC10819347 DOI: 10.3390/pathogens13010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/11/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
This study aimed to investigate the genomic and epidemiological features of a methicillin-resistant Staphylococcus aureus sequence type 1 (MRSA ST1) strain associated with caprine subclinical mastitis. An S. aureus strain was isolated from goat's milk with subclinical mastitis in Paraiba, Northeastern Brazil, by means of aseptic procedures and tested for antimicrobial susceptibility using the disk-diffusion method. Whole genome sequencing was performed using the Illumina MiSeq platform. After genome assembly and annotation, in silico analyses, including multilocus sequence typing (MLST), antimicrobial resistance and stress-response genes, virulence factors, and plasmids detection were performed. A comparative SNP-based phylogenetic analysis was performed using publicly available MRSA genomes. The strain showed phenotypic resistance to cefoxitin, penicillin, and tetracycline and was identified as sequence type 1 (ST1) and spa type 128 (t128). It harbored the SCCmec type IVa (2B), as well as the lukF-PV and lukS-PV genes. The strain was phylogenetically related to six community-acquired MRSA isolates (CA-MRSA) strains associated with human clinical disease in North America, Europe, and Australia. This is the first report of a CA-MRSA strain associated with milk in the Americas. The structural and epidemiologic features reported in the MRSA ST1 carrying a mecA-SCCmec type IVa suggest highly complex mechanisms of horizontal gene transfer in MRSA. The SNP-based phylogenetic analysis suggests a zooanthroponotic transmission, i.e., a strain of human origin.
Collapse
Affiliation(s)
- Priscylla C. Vasconcelos
- Department of Animal Science, College for Agricultural Sciences, Federal University of Paraiba (CCA/UFPB), Areia 58051-900, PB, Brazil; (P.C.V.); (E.L.L.); (M.M.S.S.); (R.G.F.); (P.E.N.G.)
| | - Elma L. Leite
- Department of Animal Science, College for Agricultural Sciences, Federal University of Paraiba (CCA/UFPB), Areia 58051-900, PB, Brazil; (P.C.V.); (E.L.L.); (M.M.S.S.); (R.G.F.); (P.E.N.G.)
| | - Mauro M. S. Saraiva
- Department of Animal Science, College for Agricultural Sciences, Federal University of Paraiba (CCA/UFPB), Areia 58051-900, PB, Brazil; (P.C.V.); (E.L.L.); (M.M.S.S.); (R.G.F.); (P.E.N.G.)
- School of Agricultural and Veterinarian Sciences, Department of Pathology, Reproduction, and One Health, São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil
| | - Rafaela G. Ferrari
- Department of Animal Science, College for Agricultural Sciences, Federal University of Paraiba (CCA/UFPB), Areia 58051-900, PB, Brazil; (P.C.V.); (E.L.L.); (M.M.S.S.); (R.G.F.); (P.E.N.G.)
| | - Samuel P. Cibulski
- Center for Biotechnology (CBiotec), Federal University of Paraiba (CBiotec/UFPB), João Pessoa 58051-900, PB, Brazil;
| | - Nubia M. V. Silva
- Animal Production Center, National Institute of Semiarid (INSA), Campina Grande 58434-700, PB, Brazil;
| | - Oliveiro C. Freitas Neto
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil;
| | - Patrícia E. N. Givisiez
- Department of Animal Science, College for Agricultural Sciences, Federal University of Paraiba (CCA/UFPB), Areia 58051-900, PB, Brazil; (P.C.V.); (E.L.L.); (M.M.S.S.); (R.G.F.); (P.E.N.G.)
| | - Rafael F. C. Vieira
- Department of Public Health Sciences, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
- Center for Computational Intelligence to Predict Health and Environmental Risks (CIPHER), The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Celso J. B. Oliveira
- Department of Animal Science, College for Agricultural Sciences, Federal University of Paraiba (CCA/UFPB), Areia 58051-900, PB, Brazil; (P.C.V.); (E.L.L.); (M.M.S.S.); (R.G.F.); (P.E.N.G.)
| |
Collapse
|
22
|
Jiang JH, Cameron DR, Nethercott C, Aires-de-Sousa M, Peleg AY. Virulence attributes of successful methicillin-resistant Staphylococcus aureus lineages. Clin Microbiol Rev 2023; 36:e0014822. [PMID: 37982596 PMCID: PMC10732075 DOI: 10.1128/cmr.00148-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of severe and often fatal infections. MRSA epidemics have occurred in waves, whereby a previously successful lineage has been replaced by a more fit and better adapted lineage. Selection pressures in both hospital and community settings are not uniform across the globe, which has resulted in geographically distinct epidemiology. This review focuses on the mechanisms that trigger the establishment and maintenance of current, dominant MRSA lineages across the globe. While the important role of antibiotic resistance will be mentioned throughout, factors which influence the capacity of S. aureus to colonize and cause disease within a host will be the primary focus of this review. We show that while MRSA possesses a diverse arsenal of toxins including alpha-toxin, the success of a lineage involves more than just producing toxins that damage the host. Success is often attributed to the acquisition or loss of genetic elements involved in colonization and niche adaptation such as the arginine catabolic mobile element, as well as the activity of regulatory systems, and shift metabolism accordingly (e.g., the accessory genome regulator, agr). Understanding exactly how specific MRSA clones cause prolonged epidemics may reveal targets for therapies, whereby both core (e.g., the alpha toxin) and acquired virulence factors (e.g., the Panton-Valentine leukocidin) may be nullified using anti-virulence strategies.
Collapse
Affiliation(s)
- Jhih-Hang Jiang
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - David R Cameron
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Cara Nethercott
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Marta Aires-de-Sousa
- Laboratory of Molecular Genetics, Institutode Tecnologia Químicae Biológica António Xavier (ITQB-NOVA), Universidade Nova de Lisboa, Oeiras, Portugal
- Escola Superior de Saúde da Cruz Vermelha Portuguesa-Lisboa (ESSCVP-Lisboa), Lisbon, Portugal
| | - Anton Y Peleg
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Centre to Impact Antimicrobial Resistance, Monash University, Clayton, Melbourne, Victoria, Australia
| |
Collapse
|
23
|
Vergoz D, Le H, Bernay B, Schaumann A, Barreau M, Nilly F, Desriac F, Tahrioui A, Giard JC, Lesouhaitier O, Chevalier S, Brunel JM, Muller C, Dé E. Antibiofilm and Antivirulence Properties of 6-Polyaminosteroid Derivatives against Antibiotic-Resistant Bacteria. Antibiotics (Basel) 2023; 13:8. [PMID: 38275318 PMCID: PMC10812528 DOI: 10.3390/antibiotics13010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The emergence of multi-drug resistant pathogens is a major public health problem, leading us to rethink and innovate our bacterial control strategies. Here, we explore the antibiofilm and antivirulence activities of nineteen 6-polyaminosterol derivatives (squalamine-based), presenting a modulation of their polyamine side chain on four major pathogens, i.e., carbapenem-resistant A. baumannii (CRAB) and P. aeruginosa (CRPA), methicillin-resistant S. aureus (MRSA), and vancomycin-resistant E. faecium (VRE) strains. We screened the effect of these derivatives on biofilm formation and eradication. Derivatives 4e (for CRAB, VRE, and MRSA) and 4f (for all the strains) were the most potent ones and displayed activities as good as those of conventional antibiotics. We also identified 11 compounds able to decrease by more than 40% the production of pyocyanin, a major virulence factor of P. aeruginosa. We demonstrated that 4f treatment acts against bacterial infections in Galleria mellonella and significantly prolonged larvae survival (from 50% to 80%) after 24 h of CRAB, VRE, and MRSA infections. As shown by proteomic studies, 4f triggered distinct cellular responses depending on the bacterial species but essentially linked to cell envelope. Its interesting antibiofilm and antivirulence properties make it a promising a candidate for use in therapeutics.
Collapse
Affiliation(s)
- Delphine Vergoz
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000 Rouen, France; (D.V.); (H.L.); (A.S.)
| | - Hung Le
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000 Rouen, France; (D.V.); (H.L.); (A.S.)
| | - Benoit Bernay
- Univ Caen Normandie, Proteogen Platform, US EMERODE, F-14000 Caen, France;
| | - Annick Schaumann
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000 Rouen, France; (D.V.); (H.L.); (A.S.)
| | - Magalie Barreau
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Flore Nilly
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Florie Desriac
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Ali Tahrioui
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | | | - Olivier Lesouhaitier
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Sylvie Chevalier
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | | | - Cécile Muller
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, F-76000 Rouen, France; (M.B.); (F.N.); (F.D.); (A.T.); (O.L.); (S.C.)
| | - Emmanuelle Dé
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000 Rouen, France; (D.V.); (H.L.); (A.S.)
| |
Collapse
|
24
|
Keim KC, Horswill AR. Staphylococcus aureus. Trends Microbiol 2023; 31:1300-1301. [PMID: 37487767 DOI: 10.1016/j.tim.2023.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/26/2023]
Affiliation(s)
- Klara C Keim
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA; Department of Veterans Affairs, Eastern Colorado Healthcare System, Aurora, CO, USA.
| |
Collapse
|
25
|
Agyirifo DS, Mensah TA, Senya ASY, Hounkpe A, Dornyoh CD, Otwe EP. Dynamics of antimicrobial resistance and virulence of staphylococcal species isolated from foods traded in the Cape Coast metropolitan and Elmina municipality of Ghana. Heliyon 2023; 9:e21584. [PMID: 38027608 PMCID: PMC10663863 DOI: 10.1016/j.heliyon.2023.e21584] [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: 04/03/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
The impact of staphylococci on food poisoning and infections could be higher than previously reported. In this study, we characterised the occurrence and coexistence of antimicrobial resistance and virulence genes of staphylococci isolates in foods. Staphylococci were isolated from 236 samples of selected street-vended foods and identified. The pattern of antimicrobial resistance and virulence genes in the staphylococci were assessed using disc diffusion, PCR and analysis of next-generation sequencing data. The food samples (70.76 %) showed a high prevalence of staphylococci and differed among the food categories. Forty-five Staphylococcus species were identified and comprised coagulase-negative and positive species. Staphylococcus sciuri (now Mammaliicoccus sciuri), S. aureus, S. kloosii, S. xylosus, S. saprophyticus, S. haemolyticus and S. succinus were the most abundant species. The staphylococcal isolates exhibited resistance to tetracycline, levofloxacin, ciprofloxacin, norfloxacin, gentamicin and amikacin and susceptibility to nitrofurantoin. Antimicrobial susceptibilities were also reported for cefoperazone, ceftriaxone, cefotaxime, nalidixic acid and piperacillin-tazobactam. The antimicrobial resistance and virulence genes commonly detected consisted of tet, arl, macB, van, gyr, nor, optrA, bcrA, blaZ, taeA and S. aureus lmrS. The isolates frequently exhibited multiple resistance (30.42 %) of up to eight antimicrobial drug classes. The isolates predominantly harboured genes that express efflux pump proteins (50.53 %) for antibiotic resistance compared with inactivation (10.05 %), target alteration (26.72 %), protection (7.67 %) and replacement (3.17 %). The virulence determinants comprised genes of pyrogenic toxin superantigens (eta, etb, tst), adhesions (clf, fnbA, fnbB, cna, map, ebp, spA, vWbp, coa) and genes that express exoproteins (nuclease, metalloprotease, γ-hemolysin, hyaluronate lyase). There was a statistically significant difference in the prevalence of staphylococci isolates and their antimicrobial resistance and virulence profile as revealed by the phenotypic, PCR and next-generation sequencing techniques. The findings suggest a higher health risk for consumers. We recommend a critical need for awareness and antimicrobial susceptibility and anti-virulence strategies to ensure food safety and counteract the spread of this clinically relevant genus.
Collapse
Affiliation(s)
- Daniel Sakyi Agyirifo
- Department of Molecular Biology and Biotechnology, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Theophilus Abonyi Mensah
- Department of Molecular Biology and Biotechnology, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Andrews Senyenam Yao Senya
- Department of Molecular Biology and Biotechnology, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Alphonse Hounkpe
- Department of Molecular Biology and Biotechnology, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Cindy Deladem Dornyoh
- Department of Molecular Biology and Biotechnology, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Emmanuel Plas Otwe
- Department of Molecular Biology and Biotechnology, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
26
|
Campbell AE, McCready-Vangi AR, Uberoi A, Murga-Garrido SM, Lovins VM, White EK, Pan JTC, Knight SAB, Morgenstern AR, Bianco C, Planet PJ, Gardner SE, Grice EA. Variable staphyloxanthin production by Staphylococcus aureus drives strain-dependent effects on diabetic wound-healing outcomes. Cell Rep 2023; 42:113281. [PMID: 37858460 PMCID: PMC10680119 DOI: 10.1016/j.celrep.2023.113281] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 08/24/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023] Open
Abstract
Strain-level variation in Staphylococcus aureus is a factor that contributes to disease burden and clinical outcomes in skin disorders and chronic wounds. However, the microbial mechanisms that drive these variable host responses are poorly understood. To identify mechanisms underlying strain-specific outcomes, we perform high-throughput phenotyping screens on S. aureus isolates cultured from diabetic foot ulcers. Isolates from non-healing wounds produce more staphyloxanthin, a cell membrane pigment. In murine diabetic wounds, staphyloxanthin-producing isolates delay wound closure significantly compared with staphyloxanthin-deficient isolates. Staphyloxanthin promotes resistance to oxidative stress and enhances bacterial survival in neutrophils. Comparative genomic and transcriptomic analysis of genetically similar clinical isolates with disparate staphyloxanthin phenotypes reveals a mutation in the sigma B operon, resulting in marked differences in stress response gene expression. Our work illustrates a framework to identify traits that underlie strain-level variation in disease burden and suggests more precise targets for therapeutic intervention in S. aureus-positive wounds.
Collapse
Affiliation(s)
- Amy E Campbell
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amelia R McCready-Vangi
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aayushi Uberoi
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sofía M Murga-Garrido
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Victoria M Lovins
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ellen K White
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jamie Ting-Chun Pan
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Simon A B Knight
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexis R Morgenstern
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Colleen Bianco
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul J Planet
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Departments of Pediatrics and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sue E Gardner
- College of Nursing, University of Iowa, Iowa City, IA 52242, USA
| | - Elizabeth A Grice
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Chen Q, Zhao G, Yang W, Chen F, Qi Y, Lou Z. Investigation into the prevalence of enterotoxin genes and genetic background of Staphylococcus aureus isolates from retain foods in Hangzhou, China. BMC Microbiol 2023; 23:294. [PMID: 37848808 PMCID: PMC10580612 DOI: 10.1186/s12866-023-03027-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Staphylococcus aureus expresses numerous toxins, many of which are strongly believed to be responsible for specific symptoms and even diseases, making it significant in the pathogenesis of human health. Enterotoxins, which are vital toxins, are associated with foodborne illnesses that manifest through symptoms like vomiting and diarrhea. In the present study, 264 S. aureus isolates obtained from various retail foods in Hangzhou, China were further investigated the profiles of enterotoxin genes and genetic backgrounds. RESULTS Approximately, 64.02% of the isolates from diverse sources contained at least one Staphylococcal Enterotoxin (SE) genes, displaying a total of 36 distinct combinations. Enterotoxin gene cluster (egc) encoded enterotoxin genes, normally designated by seg, sei, sem, sen, seo and selu, plus with sep were more frequently detected (33.73%, each). In contrast, see, ses and set were absent in any of the isolates tested. A total of 44 sequence types (STs), 20 clonal complexes (CCs) and 66 different staphylococcal protein A (spa) types (including six novel types) were identified among those 169 SE-positive isolates. Moreover, nineteen methicillin-resistant Staphylococcus aureus (MRSA) isolates were identified. The majority of those isolates belonged to the CC59-Sccmec IVa cluster and carried the seb-sek-seq gene cluster. The egc cluster, either coexisting with or without other enterotoxin genes, was observed in all isolates allocated into CC5, CC9, CC20, CC25, CC72 and ST672. Irrespective of the spa types and origins of the food, it appeared that seh was a distinct genetic element present in isolates belonging to the CC1 clonal lineage. CONCLUSIONS The results not only proposed a suspected relationship between distribution of enterotoxigenic strains and genetic backgrounds, but also attributed the presence of novel enterotoxins to potential hazards in food safety.
Collapse
Affiliation(s)
- Qi Chen
- Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, 310000, Hangzhou, China.
| | - Gang Zhao
- Hangzhou Center for Disease Control and Prevention, 310021, Hangzhou, China
| | - Wei Yang
- Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, 310000, Hangzhou, China
| | - Fuhong Chen
- Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, 310000, Hangzhou, China
| | - Yan Qi
- Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, 310000, Hangzhou, China
| | - Zhengqing Lou
- Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, 310000, Hangzhou, China.
| |
Collapse
|
28
|
Deusenbery C, Carneiro O, Oberkfell C, Shukla A. Synergy of Antibiotics and Antibiofilm Agents against Methicillin-Resistant Staphylococcus aureus Biofilms. ACS Infect Dis 2023; 9:1949-1963. [PMID: 37646612 DOI: 10.1021/acsinfecdis.3c00239] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections are some of the most common antibiotic-resistant infections, often exacerbated by the formation of biofilms. Here, we evaluated six compounds, three common antibiotics used against MRSA and three antibiofilm compounds, in nine combinations to investigate the mechanisms of synergistic eradication of MRSA biofilms. Using metabolic assessment, colony enumeration, confocal fluorescence microscopy, and scanning electron microscopy, we identified two promising combinations of antibiotics with antibiofilm agents against preformed MRSA biofilms. The broad-spectrum protease, proteinase K, and membrane-targeting antibiotic, daptomycin, worked in synergy against MRSA biofilms by manipulating the protein content, increasing access to the cell membrane of biofilm bacteria. We also found that the combination of cationic peptide, IDR-1018, with the cell wall cross-linking inhibitor, vancomycin, exhibited synergy against MRSA biofilms by causing bacterial damage and preventing repair. Our findings identify synergistic combinations of antibiotics and antibiofilm agents, providing insight into mechanisms that may be explored further for the development of effective treatments against MRSA biofilm.
Collapse
Affiliation(s)
- Carly Deusenbery
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Olivia Carneiro
- Therapeutic Sciences Graduate Program, Division of Biology and Medicine, Brown University, Providence, Rhode Island 02912, United States
| | - Carleigh Oberkfell
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Anita Shukla
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
29
|
Hachem AA, Filkins LM, Kidane YH, Raj P, Tareen NG, Arana CA, Muthukrishnan G, Copley LA. Staphylococcus aureus isolates from children with clinically differentiated osteomyelitis exhibit distinct transcriptomic signatures. PLoS One 2023; 18:e0288758. [PMID: 37561761 PMCID: PMC10414669 DOI: 10.1371/journal.pone.0288758] [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: 01/10/2023] [Accepted: 07/04/2023] [Indexed: 08/12/2023] Open
Abstract
There is substantial genomic heterogeneity among Staphylococcus aureus isolates of children with acute hematogenous osteomyelitis (AHO) but transcriptional behavior of clinically differentiated strains has not been previously described. This study evaluates transcriptional activity of S. aureus isolates of children with AHO that may regulate metabolism, biosynthesis, or virulence during bacterial growth and pathogenesis. In vitro growth kinetics were compared between three S. aureus clinical isolates from children with AHO who had mild, moderate, and severe illness. Total RNA sequencing was performed for each isolate at six separate time points throughout the logarithmic phase of growth. The NASA RNA-Sequencing Consensus Pipeline was used to identify differentially expressed genes allowing for 54 comparisons between the three isolates during growth. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathways were used to evaluate transcriptional variation in metabolism, biosynthesis pathways and virulence potential of the isolates. The S. aureus isolates demonstrated differing growth kinetics under standardized conditions with the mild isolate having higher optical densities with earlier and higher peak rates of growth than that of the other isolates (p<0.001). Enrichment pathway analysis established distinct transcriptional signatures according to both sampling time and clinical severity. Moderate and severe isolates demonstrated pathways of bacterial invasion, S. aureus infection, quorum sensing and two component systems. In comparison, the mild strain favored biosynthesis and metabolism. These findings suggest that transcriptional regulation during the growth of S. aureus may impact the pathogenetic mechanisms involved in the progression of severity of illness in childhood osteomyelitis. The clinical isolates studied demonstrated a tradeoff between growth and virulence. Further investigation is needed to evaluate these transcriptional pathways in an animal model or during active clinical infections of children with AHO.
Collapse
Affiliation(s)
- Ahmad A. Hachem
- Department of Pediatrics, University of Florida College of Medicine –Jacksonville, Jacksonville, FL, United States of America
| | - Laura M. Filkins
- Department of Microbiology, University of Texas Southwestern, Children’s Health System of Texas, Dallas, TX, United States of America
| | - Yared H. Kidane
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, United States of America
| | - Prithvi Raj
- Microbiome Research Laboratory, University of Texas Southwestern, Dallas, TX, United States of America
| | - Naureen G. Tareen
- Department of Pediatric Orthopaedic Surgery, Children’s Health System of Texas, Dallas, TX, United States of America
| | - Carlos A. Arana
- Genomics Core, University of Texas Southwestern, Dallas, TX, United States of America
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Lawson A. Copley
- Department of Pediatric Orthopaedic Surgery, Children’s Health System of Texas, Dallas, TX, United States of America
- Department of Pediatric Orthopaedic Surgery, University of Texas Southwestern, Dallas, TX, United States of America
| |
Collapse
|
30
|
Chee MSJ, Serrano E, Chiang YN, Harling-Lee J, Man R, Bacigalupe R, Fitzgerald JR, Penadés JR, Chen J. Dual pathogenicity island transfer by piggybacking lateral transduction. Cell 2023; 186:3414-3426.e16. [PMID: 37541198 DOI: 10.1016/j.cell.2023.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 03/30/2023] [Accepted: 07/03/2023] [Indexed: 08/06/2023]
Abstract
Lateral transduction (LT) is the process by which temperate phages mobilize large sections of bacterial genomes. Despite its importance, LT has only been observed during prophage induction. Here, we report that superantigen-carrying staphylococcal pathogenicity islands (SaPIs) employ a related but more versatile and complex mechanism of gene transfer to drive chromosomal hypermobility while self-transferring with additional virulence genes from the host. We found that after phage infection or prophage induction, activated SaPIs form concatamers in the bacterial chromosome by switching between parallel genomic tracks in replication bubbles. This dynamic life cycle enables SaPIbov1 to piggyback its LT of staphylococcal pathogenicity island vSaα, which encodes an array of genes involved in host-pathogen interactions, allowing both islands to be mobilized intact and transferred in a single infective particle. Our findings highlight previously unknown roles of pathogenicity islands in bacterial virulence and show that their evolutionary impact extends beyond the genes they carry.
Collapse
Affiliation(s)
- Melissa Su Juan Chee
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| | - Ester Serrano
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Yin Ning Chiang
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| | - Joshua Harling-Lee
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh EH259RG, UK
| | - Rebecca Man
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh EH259RG, UK
| | - Rodrigo Bacigalupe
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh EH259RG, UK
| | - J Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh EH259RG, UK
| | - José R Penadés
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; Departamento de Ciencias Biomédicas, Universidad CEU Cardenal Herrera, 46113 Moncada, Spain; Centre for Bacterial Resistance Biology, Imperial College London, London SW7 2AZ, UK.
| | - John Chen
- Infectious Diseases Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.
| |
Collapse
|
31
|
Chan LC, Lee HK, Wang L, Chaili S, Xiong YQ, Bayer AS, Proctor RA, Yeaman MR. Diflunisal and Analogue Pharmacophores Mediating Suppression of Virulence Phenotypes in Staphylococcus aureus. Antibiotics (Basel) 2023; 12:1180. [PMID: 37508276 PMCID: PMC10376238 DOI: 10.3390/antibiotics12071180] [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: 05/21/2023] [Revised: 06/26/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Invasive methicillin-resistant Staphylococcus aureus (MRSA) infections are leading causes of morbidity and mortality that are complicated by increasing resistance to conventional antibiotics. Thus, minimizing virulence and enhancing antibiotic efficacy against MRSA is a public health imperative. We originally demonstrated that diflunisal (DIF; [2-hydroxy-5-(2,4-difluorophenyl) benzoic acid]) inhibits S. aureus virulence factor expression. To investigate pharmacophores that are active in this function, we evaluated a library of structural analogues for their efficacy to modulate virulence phenotypes in a panel of clinically relevant S. aureus isolates in vitro. Overall, the positions of the phenyl, hydroxyl, and carboxylic moieties and the presence or type of halogen (F vs. Cl) influenced the efficacy of compounds in suppressing hemolysis, proteolysis, and biofilm virulence phenotypes. Analogues lacking halogens inhibited proteolysis to an extent similar to DIF but were ineffective at reducing hemolysis or biofilm production. In contrast, most analogues lacking the hydroxyl or carboxylic acid groups did not suppress proteolysis but did mitigate hemolysis and biofilm production to an extent similar to DIF. Interestingly, chirality and the substitution of fluorine with chlorine resulted in a differential reduction in virulence phenotypes. Together, this pattern of data suggests virulence-suppressing pharmacophores of DIF and structural analogues integrate halogen, hydroxyl, and carboxylic acid moiety stereochemistry. The anti-virulence effects of DIF were achieved using concentrations that are safe in humans, do not impair platelet antimicrobial functions, do not affect S. aureus growth, and do not alter the efficacy of conventional antibiotics. These results offer proof of concept for using novel anti-virulence strategies as adjuvants to antibiotic therapy to address the challenge of MRSA infection.
Collapse
Affiliation(s)
- Liana C. Chan
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.); (L.W.)
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.); (A.S.B.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Hong K. Lee
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.); (L.W.)
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Ling Wang
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.); (L.W.)
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Siyang Chaili
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, 2311 Pierce Ave., Nashville, TN 37232, USA;
| | - Yan Q. Xiong
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.); (A.S.B.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Arnold S. Bayer
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.); (A.S.B.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Richard A. Proctor
- Departments of Medical Microbiology & Immunology and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Michael R. Yeaman
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.); (L.W.)
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.); (A.S.B.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| |
Collapse
|
32
|
Cavaiuolo M, Lefebvre D, Mutel I, Vingadassalon N, Merda D, Hennekinne JA, Nia Y. First report of enterotoxigenic Staphylococcus argenteus as a foodborne pathogen. Int J Food Microbiol 2023; 394:110182. [PMID: 36965358 DOI: 10.1016/j.ijfoodmicro.2023.110182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023]
Abstract
Staphylococcal enterotoxins preformed in food are the causative agents of staphylococcal food poisoning outbreaks (SFPO). In this study we characterised in depth two coagulase-positive non-pigmented staphylococci involved in two independent outbreaks that occurred in France. While indistinguishable from Staphylococcus aureus using PCR methods and growth phenotype comparisons, both isolates were identified as Staphylococcus argenteus by whole genome sequencing. The genomes were analysed for the presence of enterotoxin genes, whose expression was determined in laboratory medium and, for the first time, in artificially-contaminated milk samples by using liquid chromatography-mass spectrometry and ELISA methods. The concentration measured for the SEB toxin in milk (0.67 ng/ml) was comparable to concentrations reported for other types of enterotoxins behind SFPO. From a collection of publicly available genomes, we performed an unprecedented systematic investigation of the enterotoxin gene set of S. argenteus, including variants and pseudogenes. The most prevalent genes were sex, followed by sel26, sel27 and sey. The egc cluster was less frequent and most of the time carried a dysfunctional seg gene. Our results shed light on the enterotoxigenic properties of S. argenteus, and emphasize the importance in monitoring of S. argenteus as an emerging foodborne pathogen.
Collapse
Affiliation(s)
- Marina Cavaiuolo
- University Paris Est, ANSES, Laboratory for Food Safety, SBCL Unit, Maisons-Alfort location, F-94701 Maisons-Alfort, France.
| | - Donatien Lefebvre
- University Paris Est, ANSES, Laboratory for Food Safety, SBCL Unit, Maisons-Alfort location, F-94701 Maisons-Alfort, France
| | - Isabelle Mutel
- University Paris Est, ANSES, Laboratory for Food Safety, SBCL Unit, Maisons-Alfort location, F-94701 Maisons-Alfort, France
| | - Noémie Vingadassalon
- University Paris Est, ANSES, Laboratory for Food Safety, SBCL Unit, Maisons-Alfort location, F-94701 Maisons-Alfort, France
| | - Déborah Merda
- University Paris Est, ANSES, SPAAD unit, Maisons-Alfort location, F-94701 Maisons-Alfort, France
| | - Jacques-Antoine Hennekinne
- University Paris Est, ANSES, Laboratory for Food Safety, SBCL Unit, Maisons-Alfort location, F-94701 Maisons-Alfort, France
| | - Yacine Nia
- University Paris Est, ANSES, Laboratory for Food Safety, SBCL Unit, Maisons-Alfort location, F-94701 Maisons-Alfort, France
| |
Collapse
|
33
|
Fernández-Fernández R, Lozano C, Reuben RC, Ruiz-Ripa L, Zarazaga M, Torres C. Comprehensive Approaches for the Search and Characterization of Staphylococcins. Microorganisms 2023; 11:1329. [PMID: 37317303 PMCID: PMC10221470 DOI: 10.3390/microorganisms11051329] [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: 04/04/2023] [Revised: 05/02/2023] [Accepted: 05/09/2023] [Indexed: 06/16/2023] Open
Abstract
Novel and sustainable approaches are required to curb the increasing threat of antimicrobial resistance (AMR). Within the last decades, antimicrobial peptides, especially bacteriocins, have received increased attention and are being explored as suitable alternatives to antibiotics. Bacteriocins are ribosomally synthesized antimicrobial peptides produced by bacteria as a self-preservation method against competitors. Bacteriocins produced by Staphylococcus, also referred to as staphylococcins, have steadily shown great antimicrobial potential and are currently being considered promising candidates to mitigate the AMR menace. Moreover, several bacteriocin-producing Staphylococcus isolates of different species, especially coagulase-negative staphylococci (CoNS), have been described and are being targeted as a good alternative. This revision aims to help researchers in the search and characterization of staphylococcins, so we provide an up-to-date list of bacteriocin produced by Staphylococcus. Moreover, a universal nucleotide and amino acid-based phylogeny system of the well-characterized staphylococcins is proposed that could be of interest in the classification and search for these promising antimicrobials. Finally, we discuss the state of art of the staphylococcin applications and an overview of the emerging concerns.
Collapse
Affiliation(s)
| | - Carmen Lozano
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain
| | | | | | | | | |
Collapse
|
34
|
Chan LC, Park M, Lee HK, Chaili S, Xiong YQ, Bayer AS, Proctor RA, Yeaman MR. Diflunisal Attenuates Virulence Factor Gene Regulation and Phenotypes in Staphylococcus aureus. Antibiotics (Basel) 2023; 12:902. [PMID: 37237805 PMCID: PMC10215304 DOI: 10.3390/antibiotics12050902] [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: 04/05/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Virulence factor expression is integral to pathogenicity of Staphylococcus aureus. We previously demonstrated that aspirin, through its major metabolite, salicylic acid (SAL), modulates S. aureus virulence phenotypes in vitro and in vivo. We compared salicylate metabolites and a structural analogue for their ability to modulate S. aureus virulence factor expression and phenotypes: (i) acetylsalicylic acid (ASA, aspirin); (ii) ASA metabolites, salicylic acid (SAL), gentisic acid (GTA) and salicyluric acid (SUA); or (iii) diflunisal (DIF), a SAL structural analogue. None of these compounds altered the growth rate of any strain tested. ASA and its metabolites SAL, GTA and SUA moderately impaired hemolysis and proteolysis phenotypes in multiple S. aureus strain backgrounds and their respective deletion mutants. Only DIF significantly inhibited these virulence phenotypes in all strains. The kinetic profiles of ASA, SAL or DIF on expression of hla (alpha hemolysin), sspA (V8 protease) and their regulators (sigB, sarA, agr (RNAIII)) were assessed in two prototypic strain backgrounds: SH1000 (methicillin-sensitive S. aureus; MSSA) and LAC-USA300 (methicillin-resistant S. aureus; MRSA). DIF induced sigB expression which is coincident with the significant inhibition of RNAIII expression in both strains and precedes significant reductions in hla and sspA expression. The inhibited expression of these genes within 2 h resulted in the durable suppression of hemolysis and proteolysis phenotypes. These results indicate that DIF modulates the expression of key virulence factors in S. aureus via a coordinated impact on their relevant regulons and target effector genes. This strategy may hold opportunities to develop novel antivirulence strategies to address the ongoing challenge of antibiotic-resistant S. aureus.
Collapse
Affiliation(s)
- Liana C. Chan
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.)
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mihyun Park
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.)
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Hong K. Lee
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.)
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Siyang Chaili
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, 2311 Pierce Ave., Nashville, TN 37232, USA
| | - Yan Q. Xiong
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Arnold S. Bayer
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Richard A. Proctor
- Departments of Medical Microbiology/Immunology and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Michael R. Yeaman
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.)
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| |
Collapse
|
35
|
Vergoz D, Nilly F, Desriac F, Barreau M, Géry A, Lepetit C, Sichel F, Jeannot K, Giard JC, Garon D, Chevalier S, Muller C, Dé E, Brunel JM. 6-Polyaminosteroid Squalamine Analogues Display Antibacterial Activity against Resistant Pathogens. Int J Mol Sci 2023; 24:ijms24108568. [PMID: 37239913 DOI: 10.3390/ijms24108568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
A series of 6-polyaminosteroid analogues of squalamine were synthesized with moderate to good yields and evaluated for their in vitro antimicrobial properties against both susceptible and resistant Gram-positive (vancomycin-resistant Enterococcus faecium and methicillin-resistant Staphylococcus aureus) and Gram-negative (carbapenem-resistant Acinetobacter baumannii and Pseudomonas aeruginosa) bacterial strains. Minimum inhibitory concentrations against Gram-positive bacteria ranged from 4 to 16 µg/mL for the most effective compounds, 4k and 4n, and showed an additive or synergistic effect with vancomycin or oxacillin. On the other hand, the derivative 4f, which carries a spermine moiety like that of the natural trodusquemine molecule, was found to be the most active derivative against all the resistant Gram-negative bacteria tested, with an MIC value of 16 µg/mL. Our results suggest that 6-polyaminosteroid analogues of squalamine are interesting candidates for Gram-positive bacterial infection treatments, as well as potent adjuvants to fight Gram-negative bacterial resistance.
Collapse
Affiliation(s)
- Delphine Vergoz
- Polymers, Biopolymers, Surfaces Laboratory, University of Rouen Normandie, INSA Rouen, CNRS, UMR 6270, 76000 Rouen, France
| | - Flore Nilly
- Communication Bactérienne et Stratégies Anti-Infectieuses, University of Rouen Normandie, CBSA, 27000 Evreux, France
| | - Florie Desriac
- Communication Bactérienne et Stratégies Anti-Infectieuses, UNICAEN, Normandie University, UR4312, CBSA, 14032 Caen, France
| | - Magalie Barreau
- Communication Bactérienne et Stratégies Anti-Infectieuses, University of Rouen Normandie, CBSA, 27000 Evreux, France
| | - Antoine Géry
- UNICAEN, Normandie University, ABTE UR4651 and Centre François Baclesse, 14032 Caen, France
| | - Charlie Lepetit
- UNICAEN, Normandie University, ABTE UR4651 and Centre François Baclesse, 14032 Caen, France
| | - François Sichel
- UNICAEN, Normandie University, ABTE UR4651 and Centre François Baclesse, 14032 Caen, France
| | - Katy Jeannot
- UMR 6249 Chrono-Environnement, CNRS-Université de Bourgogne/Franche-Comté, 25000 Besançon, France
| | - Jean-Christophe Giard
- UNICAEN, University of Rouen Normandie, INSERM, DYNAMICURE UMR 1311 F, 14000 Caen, France
| | - David Garon
- UNICAEN, Normandie University, ABTE UR4651 and Centre François Baclesse, 14032 Caen, France
| | - Sylvie Chevalier
- Communication Bactérienne et Stratégies Anti-Infectieuses, University of Rouen Normandie, CBSA, 27000 Evreux, France
| | - Cécile Muller
- Communication Bactérienne et Stratégies Anti-Infectieuses, UNICAEN, Normandie University, UR4312, CBSA, 14032 Caen, France
| | - Emmanuelle Dé
- Polymers, Biopolymers, Surfaces Laboratory, University of Rouen Normandie, INSA Rouen, CNRS, UMR 6270, 76000 Rouen, France
| | | |
Collapse
|
36
|
Buckley PT, Chan R, Fernandez J, Luo J, Lacey KA, DuMont AL, O'Malley A, Brezski RJ, Zheng S, Malia T, Whitaker B, Zwolak A, Payne A, Clark D, Sigg M, Lacy ER, Kornilova A, Kwok D, McCarthy S, Wu B, Morrow B, Nemeth-Seay J, Petley T, Wu S, Strohl WR, Lynch AS, Torres VJ. Multivalent human antibody-centyrin fusion protein to prevent and treat Staphylococcus aureus infections. Cell Host Microbe 2023; 31:751-765.e11. [PMID: 37098341 DOI: 10.1016/j.chom.2023.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 02/12/2023] [Accepted: 04/03/2023] [Indexed: 04/27/2023]
Abstract
Treating and preventing infections by antimicrobial-resistant bacterial pathogens is a worldwide problem. Pathogens such as Staphylococcus aureus produce an array of virulence determinants, making it difficult to identify single targets for the development of vaccines or monoclonal therapies. We described a human-derived anti-S. aureus monoclonal antibody (mAb)-centyrin fusion protein ("mAbtyrin") that simultaneously targets multiple bacterial adhesins, resists proteolysis by bacterial protease GluV8, avoids Fc engagement by S. aureus IgG-binding proteins SpA and Sbi, and neutralizes pore-forming leukocidins via fusion with anti-toxin centyrins, while maintaining Fc- and complement-mediated functions. Compared with the parental mAb, mAbtyrin protected human phagocytes and boosted phagocyte-mediated killing. The mAbtyrin also reduced pathology, reduced bacterial burden, and protected from different types of infections in preclinical animal models. Finally, mAbtyrin synergized with vancomycin, enhancing pathogen clearance in an animal model of bacteremia. Altogether, these data establish the potential of multivalent mAbs for treating and preventing S. aureus diseases.
Collapse
Affiliation(s)
- Peter T Buckley
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA.
| | - Rita Chan
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Jeffrey Fernandez
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Jinquan Luo
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Ashley L DuMont
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Aidan O'Malley
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Randall J Brezski
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Songmao Zheng
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Thomas Malia
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Brian Whitaker
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Adam Zwolak
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Angela Payne
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Desmond Clark
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Martin Sigg
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Eilyn R Lacy
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Anna Kornilova
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Debra Kwok
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Steve McCarthy
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Bingyuan Wu
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Brian Morrow
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | | | - Ted Petley
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Sam Wu
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - William R Strohl
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | | | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Langone Health, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA.
| |
Collapse
|
37
|
Al-Trad EI, Chew CH, Che Hamzah AM, Suhaili Z, Rahman NIA, Ismail S, Puah SM, Chua KH, Kwong SM, Yeo CC. The Plasmidomic Landscape of Clinical Methicillin-Resistant Staphylococcus aureus Isolates from Malaysia. Antibiotics (Basel) 2023; 12:antibiotics12040733. [PMID: 37107095 PMCID: PMC10135026 DOI: 10.3390/antibiotics12040733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a priority nosocomial pathogen with plasmids playing a crucial role in its genetic adaptability, particularly in the acquisition and spread of antimicrobial resistance. In this study, the genome sequences of 79 MSRA clinical isolates from Terengganu, Malaysia, (obtained between 2016 and 2020) along with an additional 15 Malaysian MRSA genomes from GenBank were analyzed for their plasmid content. The majority (90%, 85/94) of the Malaysian MRSA isolates harbored 1-4 plasmids each. In total, 189 plasmid sequences were identified ranging in size from 2.3 kb to ca. 58 kb, spanning all seven distinctive plasmid replication initiator (replicase) types. Resistance genes (either to antimicrobials, heavy metals, and/or biocides) were found in 74% (140/189) of these plasmids. Small plasmids (<5 kb) were predominant (63.5%, 120/189) with a RepL replicase plasmid harboring the ermC gene that confers resistance to macrolides, lincosamides, and streptogramin B (MLSB) identified in 63 MRSA isolates. A low carriage of conjugative plasmids was observed (n = 2), but the majority (64.5%, 122/189) of the non-conjugative plasmids have mobilizable potential. The results obtained enabled us to gain a rare view of the plasmidomic landscape of Malaysian MRSA isolates and reinforces their importance in the evolution of this pathogen.
Collapse
Affiliation(s)
- Esra'a I Al-Trad
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| | - Ching Hoong Chew
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Kuala Nerus 21300, Malaysia
| | | | - Zarizal Suhaili
- Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, Besut 22200, Malaysia
| | - Nor Iza A Rahman
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| | - Salwani Ismail
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| | - Suat Moi Puah
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Stephen M Kwong
- Infectious Diseases & Microbiology, School of Medicine, Western Sydney University, Campbelltown 2560, Australia
| | - Chew Chieng Yeo
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| |
Collapse
|
38
|
André C, Schrank CL, Cheng Jaramillo AV, Mylonakis E, Wuest WM, Gilmore MS, Kim W, Bispo PJM. Antimicrobial activity of a new class of synthetic retinoid antibiotics and comparator agents against ocular staphylococci. FRONTIERS IN ANTIBIOTICS 2023; 2:1101450. [PMID: 39816665 PMCID: PMC11732056 DOI: 10.3389/frabi.2023.1101450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/14/2023] [Indexed: 01/18/2025]
Abstract
Objectives Antimicrobial resistance is global pandemic that poses a major threat to vision health as ocular pathogens, especially staphylococcal species, are becoming increasingly resistant to first-line therapies. Here we evaluated the antimicrobial activity of a new class of synthetic retinoids in comparison to currently used antibiotics against clinically relevant ocular staphylococcal isolates. Methods Antimicrobial susceptibility testing was performed by broth microdilution for 3 novel synthetic retinoids (CD1530, CD437, and a CD437 analogue) and 7 comparator antibiotics, against a collection of 216 clinical isolates. Results CD437 MIC50 and MIC90 were 2 µg/mL for Staphylococcus aureus, and 1 µg/mL and 2 µg/mL respectively, for coagulase-negative staphylococci (CoNS). CD1530 (MIC50 = 2 µg/mL for all species) also displayed good activity with an in vitro potency slightly lower (2-fold) for S. aureus (MIC90 = 4 µg/mL) when compared to CD437. A CD437 analogue also demonstrated good in vitro activity (MIC50 = 2 µg/mL for all species) and potency (MIC90 = 2 µg/mL for MRSA and 4 µg/mL for MSSA and CoNS). In vitro potencies were similar or higher than that of comparator agents, and were not impacted by multidrug resistance phenotypes. Conclusion Our results demonstrate that synthetic retinoids display potent in vitro activity against ocular staphylococcal species, including multidrug-resistant isolates.
Collapse
Affiliation(s)
- Camille André
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
- Infectious Disease Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | - Eleftherios Mylonakis
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - William M. Wuest
- Department of Chemistry, Emory University, Atlanta, GA, United States
| | - Michael S. Gilmore
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
- Infectious Disease Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, United States
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Paulo J. M. Bispo
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
- Infectious Disease Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
39
|
Oh J, Warner M, Ambler JE, Schuch R. The Lysin Exebacase Has a Low Propensity for Resistance Development in Staphylococcus aureus and Suppresses the Emergence of Resistance to Antistaphylococcal Antibiotics. Microbiol Spectr 2023; 11:e0526122. [PMID: 36862002 PMCID: PMC10100934 DOI: 10.1128/spectrum.05261-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/15/2023] [Indexed: 03/03/2023] Open
Abstract
Exebacase (CF-301) belongs to a novel class of protein-based antibacterial agents, called lysins (peptidoglycan hydrolases). Exebacase exhibits potent antistaphylococcal activity and is the first lysin to initiate clinical trials in the United States. To support clinical development, the potential for resistance development to exebacase was assessed over 28 days of serial daily subculture in the presence of increasing concentrations of the lysin performed in its reference broth medium. Exebacase MICs remained unchanged over serial subculture for three replicates each of methicillin-susceptible Staphylococcus aureus (MSSA) strain ATCC 29213 and methicillin-resistant S. aureus (MRSA) strain MW2. For comparator antibiotics also tested, oxacillin MICs increased by 32-fold with ATCC 29213 and daptomycin and vancomycin MICs increased by 16- and 8-fold, respectively, with MW2. Serial passage was also used to examine the capacity of exebacase to suppress selection for increased oxacillin, daptomycin, and vancomycin MICs when used together in combination, wherein daily exposures to increasing concentrations of antibiotic were performed over 28 days with the added presence of fixed sub-MIC amounts of exebacase. Exebacase suppressed increases in antibiotic MICs over this period. These findings are consistent with a low propensity for resistance to exebacase and an added benefit of reducing the potential for development of antibiotic resistance. IMPORTANCE To guide development of an investigational new antibacterial drug, microbiological data are required to understand the potential for development of resistance to the drug in the target organism(s). Exebacase is a lysin (peptidoglycan hydrolase) that represents a novel antimicrobial modality based on degradation of the cell wall of Staphylococcus aureus. Exebacase resistance was examined here using an in vitro serial passage method that assesses the impact of daily exposures to increasing concentrations of exebacase over 28 days in medium approved for use in exebacase antimicrobial susceptibility testing (AST) by the Clinical and Laboratory Standards Institute (CLSI). No changes in susceptibility to exebacase were observed over the 28-day period for multiple replicates of two S. aureus strains, indicating a low propensity for resistance development. Interestingly, while high-level resistance to commonly used antistaphylococcal antibiotics was readily obtained using the same method, the added presence of exebacase acted to suppress antibiotic resistance development.
Collapse
Affiliation(s)
- Jun Oh
- ContraFect Corporation, Yonkers, New York, USA
| | | | | | | |
Collapse
|
40
|
Dzyhovskyi V, Stokowa-Sołtys K. Divalent metal ion binding to Staphylococcus aureus FeoB transporter regions. J Inorg Biochem 2023; 244:112203. [PMID: 37018851 DOI: 10.1016/j.jinorgbio.2023.112203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
Transition metal ions such as iron, copper, zinc, manganese or, nickel are essential in many biological processes. Bacteria have developed a number of mechanisms for their acquisition and transport, in which numerous of proteins and smaller molecules are involved. One of the representatives of these proteins is FeoB, which belongs to the Feo (ferrous ion transporter) family. Although ferrous iron transport system is widespread among microorganisms, it is still poorly described in Gram-positive pathogens, such as Staphylococcus aureus. In this work, combined potentiometric and spectroscopic studies (UV-Vis, CD and EPR) were carried out to determine Cu(II), Fe(II) and Zn(II) binding modes to FeoB fragments (Ac-IDYHKLMK-NH2, Ac-ETSHDKY-NH2, and Ac-SFLHMVGS-NH2). For the first time iron(II) complexes with peptides were characterized by potentiometry. All studied ligands are able to form a variety of thermodynamically stable complexes with transition metal ions. It was concluded that among the studied systems, the most effective metal ion binding is observed for the Ac-ETSHDKY-NH2 peptide. Moreover, comparing preferences of all ligands towards different metal ions, copper(II) complexes are the most stable ones at physiological pH.
Collapse
|
41
|
Monecke S, Bedewy AK, Müller E, Braun SD, Diezel C, Elsheredy A, Kader O, Reinicke M, Ghazal A, Rezk S, Ehricht R. Characterisation of Methicillin-Resistant Staphylococcus aureus from Alexandria, Egypt. Antibiotics (Basel) 2023; 12:78. [PMID: 36671279 PMCID: PMC9855118 DOI: 10.3390/antibiotics12010078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/28/2022] [Accepted: 12/09/2022] [Indexed: 01/03/2023] Open
Abstract
The present study aims to characterise clinical MRSA isolates from a tertiary care centre in Egypt's second-largest city, Alexandria. Thirty isolates collected in 2020 were genotypically characterised by microarray to detect their resistance and virulence genes and assign them to clonal complexes (CC) and strains. Isolates belonged to 11 different CCs and 14 different strains. CC15-MRSA-[V+fus] (n = 6), CC1-MRSA-[V+fus+tir+ccrA/B-1] (PVL+) (n = 5) as well as CC1-MRSA-[V+fus+tir+ccrA/B-1] and CC1153-MRSA-[V+fus] (PVL+) (both with n = 3) were the most common strains. Most isolates (83%) harboured variant or composite SCCmec V or VI elements that included the fusidic acid resistance gene fusC. The SCCmec [V+fus+tir+ccrA/B-1] element of one of the CC1 isolates was sequenced, revealing a presence not only of fusC but also of blaZ, aacA-aphD and other resistance genes. PVL genes were also common (40%). The hospital-acquired MRSA CC239-III strain was only found twice. A comparison to data from a study on strains collected in 2015 (Montelongo et al., 2022) showed an increase in fusC and PVL carriage and a decreasing prevalence of the CC239 strain. These observations indicate a diffusion of community-acquired strains into hospital settings. The beta-lactam use in hospitals and the widespread fusidic acid consumption in the community might pose a selective pressure that favours MRSA strains with composite SCCmec elements comprising mecA and fusC. This is an unsettling trend, but more MRSA typing data from Egypt are required.
Collapse
Affiliation(s)
- Stefan Monecke
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany
- InfectoGnostics Research Campus, 07743 Jena, Germany
- Institute for Medical Microbiology and Virology, Dresden University Hospital, 01307 Dresden, Germany
| | - Amira K. Bedewy
- Department of Microbiology, Medical Research Institute, Alexandria University, Alexandria 5424041, Egypt
| | - Elke Müller
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany
- InfectoGnostics Research Campus, 07743 Jena, Germany
| | - Sascha D. Braun
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany
- InfectoGnostics Research Campus, 07743 Jena, Germany
| | - Celia Diezel
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany
- InfectoGnostics Research Campus, 07743 Jena, Germany
| | - Amel Elsheredy
- Department of Microbiology, Medical Research Institute, Alexandria University, Alexandria 5424041, Egypt
| | - Ola Kader
- Department of Microbiology, Medical Research Institute, Alexandria University, Alexandria 5424041, Egypt
| | - Martin Reinicke
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany
- InfectoGnostics Research Campus, 07743 Jena, Germany
| | - Abeer Ghazal
- Department of Microbiology, Medical Research Institute, Alexandria University, Alexandria 5424041, Egypt
| | - Shahinda Rezk
- Department of Microbiology, Medical Research Institute, Alexandria University, Alexandria 5424041, Egypt
| | - Ralf Ehricht
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany
- InfectoGnostics Research Campus, 07743 Jena, Germany
- Institute of Physical Chemistry, Friedrich-Schiller University, 07743 Jena, Germany
| |
Collapse
|
42
|
Molendijk MM, Phan MVT, Bode LGM, Strepis N, Prasad DK, Worp N, Nieuwenhuijse DF, Schapendonk CME, Boekema BKHL, Verbon A, Koopmans MPG, de Graaf M, van Wamel WJB. Microcalorimetry: A Novel Application to Measure In Vitro Phage Susceptibility of Staphylococcus aureus in Human Serum. Viruses 2022; 15:14. [PMID: 36680055 PMCID: PMC9865112 DOI: 10.3390/v15010014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Infections involving antibiotic resistant Staphylococcus aureus (S. aureus) represent a major challenge to successful treatment. Further, although bacteriophages (phages) could be an alternative to antibiotics, there exists a lack of correlation in phage susceptibility results between conventional in vitro and in vivo assays. This discrepancy may hinder the potential implementation of bacteriophage therapy. In this study, the susceptibility of twelve S. aureus strains to three commercial phage cocktails and two single phages was assessed. These S. aureus strains (including ten clinical isolates, five of which were methicillin-resistant) were compared using four assays: the spot test, efficiency of plating (EOP), the optical density assay (all in culture media) and microcalorimetry in human serum. In the spot test, EOP and optical density assay, all cocktails and single phages lysed both methicillin susceptible and methicillin resistant S. aureus strains. However, there was an absence of phage-mediated lysis in high concentrations of human serum as measured using microcalorimetry. As this microcalorimetry-based assay more closely resembles in vivo conditions, we propose that microcalorimetry could be included as a useful addition to conventional assays, thereby facilitating more accurate predictions of the in vivo susceptibility of S. aureus to phages during phage selection for therapeutic purposes.
Collapse
Affiliation(s)
- Michèle M. Molendijk
- Department Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 Rotterdam, The Netherlands
- Department of Viroscience, Erasmus MC, 3015 Rotterdam, The Netherlands
| | - My V. T. Phan
- Department of Viroscience, Erasmus MC, 3015 Rotterdam, The Netherlands
- Medical Research Council/Uganda Virus Research Institute, London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe P.O. Box 49, Uganda
| | - Lonneke G. M. Bode
- Department Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 Rotterdam, The Netherlands
| | - Nikolas Strepis
- Department Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 Rotterdam, The Netherlands
| | - Divyae K. Prasad
- Department of Viroscience, Erasmus MC, 3015 Rotterdam, The Netherlands
| | - Nathalie Worp
- Department of Viroscience, Erasmus MC, 3015 Rotterdam, The Netherlands
| | | | | | | | - Annelies Verbon
- Department Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 Rotterdam, The Netherlands
| | | | - Miranda de Graaf
- Department of Viroscience, Erasmus MC, 3015 Rotterdam, The Netherlands
| | - Willem J. B. van Wamel
- Department Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 Rotterdam, The Netherlands
| |
Collapse
|
43
|
In Silico Genome-Scale Analysis of Molecular Mechanisms Contributing to the Development of a Persistent Infection with Methicillin-Resistant Staphylococcus aureus (MRSA) ST239. Int J Mol Sci 2022; 23:ijms232416086. [PMID: 36555727 PMCID: PMC9781258 DOI: 10.3390/ijms232416086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/05/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
The increasing frequency of isolation of methicillin-resistant Staphylococcus aureus (MRSA) limits the chances for the effective antibacterial therapy of staphylococcal diseases and results in the development of persistent infection such as bacteremia and osteomyelitis. The aim of this study was to identify features of the MRSAST239 0943-1505-2016 (SA943) genome that contribute to the formation of both acute and chronic musculoskeletal infections. The analysis was performed using comparative genomics data of the dominant epidemic S. aureus lineages, namely ST1, ST8, ST30, ST36, and ST239. The SA943 genome encodes proteins that provide resistance to the host's immune system, suppress immunological memory, and form biofilms. The molecular mechanisms of adaptation responsible for the development of persistent infection were as follows: amino acid substitution in PBP2 and PBP2a, providing resistance to ceftaroline; loss of a large part of prophage DNA and restoration of the nucleotide sequence of beta-hemolysin, that greatly facilitates the escape of phagocytosed bacteria from the phagosome and formation of biofilms; dysfunction of the AgrA system due to the presence of psm-mec and several amino acid substitutions in the AgrC; partial deletion of the nucleotide sequence in genomic island vSAβ resulting in the loss of two proteases of Spl-operon; and deletion of SD repeats in the SdrE amino acid sequence.
Collapse
|
44
|
Study of SarA by DNA Affinity Capture Assay (DACA) Employing Three Promoters of Key Virulence and Resistance Genes in Methicillin-Resistant Staphylococcus aureus. Antibiotics (Basel) 2022; 11:antibiotics11121714. [PMID: 36551372 PMCID: PMC9774152 DOI: 10.3390/antibiotics11121714] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), one of the most well-known human pathogens, houses many virulence factors and regulatory proteins that confer resistance to diverse antibiotics. Although they have been investigated intensively, the correlations among virulence factors, regulatory proteins and antibiotic resistance are still elusive. We aimed to identify the most significant global MRSA regulator by concurrently analyzing protein-binding and several promoters under same conditions and at the same time point. DNA affinity capture assay (DACA) was performed with the promoters of mecA, sarA, and sarR, all of which significantly impact survival of MRSA. Here, we show that SarA protein binds to all three promoters. Consistent with the previous reports, ΔsarA mutant exhibited weakened antibiotic resistance to oxacillin and reduced biofilm formation. Additionally, production and activity of many virulence factors such as phenol-soluble modulins (PSM), α-hemolysin, motility, staphyloxanthin, and other related proteins were decreased. Comparing the sequence of SarA with that of clinical strains of various lineages showed that all sequences were highly conserved, in contrast to that observed for AgrA, another major regulator of virulence and resistance in MRSA. We have demonstrated that SarA regulates antibiotic resistance and the expression of various virulence factors. Our results warrant that SarA could be a leading target for developing therapeutic agents against MRSA infections.
Collapse
|
45
|
First Genome-Based Characterisation and Staphylococcal Enterotoxin Production Ability of Methicillin-Susceptible and Methicillin-Resistant Staphylococcus aureus Strains Isolated from Ready-to-Eat Foods in Algiers (Algeria). Toxins (Basel) 2022; 14:toxins14110731. [PMID: 36355981 PMCID: PMC9694651 DOI: 10.3390/toxins14110731] [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: 09/02/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 01/26/2023] Open
Abstract
Staphylococcus aureus is a pathogenic microorganism of humans and animals, able to cause foodborne intoxication due to the production of staphylococcal enterotoxins (SEs) and to resist antibiotic treatment as in the case of methicillin-resistant S. aureus (MRSA). In this study, we performed a genomic characterisation of 12 genetically diverse S. aureus strains isolated from ready-to-eat foods in Algiers (Algeria). Moreover, their ability to produce some classical and new staphylococcal enterotoxins (SEs) was investigated. The 12 S. aureus strains resulted to belong to nine known sequence types (STs) and to the novel ST7199 and ST7200. Furthermore, S. aureus SA46 was assigned to the European clone MRSA-ST80-SCCmec-IV. The 12 strains showed a wide endowment of se and sel (staphylococcal enterotoxin-like toxin) genes (sea, seb, sed, seg, seh, sei, selj, sek, sem, sen, seo, seq, ser, selu2, selw, selx, sey, sel30; ψent1-ψent2), including variants and pseudogenes, and harboured the enterotoxin gene cluster (egc) types 1 and 5. Additionally, they produced various amounts of SEA (64.54-345.02 ng/mL), SEB (2871.28-14739.17 ng/mL), SED (322.70-398.94 ng/mL), SEH (not detectable-239.48 ng/mL), and SER (36,720.10-63,176.06 ng/mL) depending on their genotypes. The genetic determinants related to their phenotypic resistance to β-lactams (blaZ, mecA), ofloxacin (gyrA-S84L), erythromycin (ermB), lincomycin (lmrS), kanamycin (aph(3')-III, ant(6)-I), and tetracyclin (tet(L), tet(38)) were also detected. A plethora of virulence-related genes, including major virulence genes such as the tst gene, determinant for the toxic shock syndrome toxin-1, and the lukF-PV and lukS-PV genes, encoding the panton-valentine leukocidin (PVL), were present in the S. aureus strains, highlighting their pathogenic potential. Furthermore, a phylogenomic reconstruction including worldwide foodborne S. aureus showed a clear clustering based on ST and geographical origin rather than the source of isolation.
Collapse
|
46
|
Characterization of MroQ-Dependent Maturation and Export of the Staphylococcus aureus Accessory Gene Regulatory System Autoinducing Peptide. Infect Immun 2022; 90:e0026322. [PMID: 36073934 PMCID: PMC9584314 DOI: 10.1128/iai.00263-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Gram-positive bacteria produce small autoinducing peptides (AIPs), which act to regulate expression of genes that promote adaptive traits, including virulence. The Gram-positive pathogen Staphylococcus aureus generates a cyclic AIP that controls expression of virulence factors via the accessory gene regulatory (Agr) system. S. aureus strains belong to one of four Agr groups (Agr-I, -II, -III, and -IV); each group harbors allelic variants of AgrD, the precursor of AIP. In a prior screen for S. aureus virulence factors, we identified MroQ, a putative peptidase. A ΔmroQ mutant closely resembled a Δagr mutant and had significant defects in AIP production in an Agr-I strain. Here, we show that expression of AgrD-I in a ΔmroQ mutant leads to accumulation of an AIP processing intermediate at the membrane that coincides with a loss of secreted mature AIP, indicating that MroQ promotes maturation of AgrD-I. MroQ is conserved in all Agr sequence variants, suggesting either identical function among all Agr types or activity specific to Agr-I strains. Our data indicate that MroQ is required for AIP maturation and activity in Agr-I, -II, and -IV strains irrespective of background. However, MroQ is not required for Agr-III activity despite an identifiable role in peptide maturation. Isogenic Δagr and Δagr ΔmroQ strains complemented with Agr-I to -IV validated the critical role of MroQ in the generation of active AIP-I, -II, and -IV but not AIP-III. These findings were reinforced by skin infection studies with mice. Our data substantiate the prevailing model that MroQ is a mediator of cyclic peptide maturation.
Collapse
|
47
|
Antibiotic combinations reduce Staphylococcus aureus clearance. Nature 2022; 610:540-546. [PMID: 36198788 PMCID: PMC9533972 DOI: 10.1038/s41586-022-05260-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 08/22/2022] [Indexed: 12/17/2022]
Abstract
The spread of antibiotic resistance is attracting increased attention to combination-based treatments. Although drug combinations have been studied extensively for their effects on bacterial growth1–11, much less is known about their effects on bacterial long-term clearance, especially at cidal, clinically relevant concentrations12–14. Here, using en masse microplating and automated image analysis, we systematically quantify Staphylococcus aureus survival during prolonged exposure to pairwise and higher-order cidal drug combinations. By quantifying growth inhibition, early killing and longer-term population clearance by all pairs of 14 antibiotics, we find that clearance interactions are qualitatively different, often showing reciprocal suppression whereby the efficacy of the drug mixture is weaker than any of the individual drugs alone. Furthermore, in contrast to growth inhibition6–10 and early killing, clearance efficacy decreases rather than increases as more drugs are added. However, specific drugs targeting non-growing persisters15–17 circumvent these suppressive effects. Competition experiments show that reciprocal suppressive drug combinations select against resistance to any of the individual drugs, even counteracting methicillin-resistant Staphylococcus aureus both in vitro and in a Galleria mellonella larva model. As a consequence, adding a β-lactamase inhibitor that is commonly used to potentiate treatment against β-lactam-resistant strains can reduce rather than increase treatment efficacy. Together, these results underscore the importance of systematic mapping the long-term clearance efficacy of drug combinations for designing more-effective, resistance-proof multidrug regimes. Different pairs of antibiotics show qualitatively different bacterial clearance interactions—some pairs show reciprocal suppression whereby the drug mixture efficacy is weaker than the individual drugs alone, and the clearance efficacy decreases as more drugs are added.
Collapse
|
48
|
Chong CE, Bengtsson RJ, Horsburgh MJ. Comparative genomics of Staphylococcus capitis reveals species determinants. Front Microbiol 2022; 13:1005949. [PMID: 36246238 PMCID: PMC9563023 DOI: 10.3389/fmicb.2022.1005949] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/05/2022] [Indexed: 11/27/2022] Open
Abstract
Staphylococcus capitis is primarily described as a human skin commensal but is now emergent as an opportunistic pathogen isolated from the bloodstream and prosthetic joint infections, and neonatal intensive care unit (NICU)-associated sepsis. We used comparative genomic analyses of S. capitis to provide new insights into commensal scalp isolates from varying skin states (healthy, dandruff lesional, and non-lesional), and to expand our current knowledge of the species populations (scalp isolates, n = 59; other skin isolates, n = 7; publicly available isolates, n = 120). A highly recombinogenic population structure was revealed, with genomes including the presence of a range of previously described staphylococcal virulence factors, cell wall-associated proteins, and two-component systems. Genomic differences between the two described S. capitis subspecies were explored, which revealed the determinants associated exclusively with each subspecies. The subspecies ureolyticus was distinguished from subspecies capitis based on the differences in antimicrobial resistance genes, β-lactam resistance genes, and β-class phenol soluble modulins and gene clusters linked to biofilm formation and survival on skin. This study will aid further research into the classification of S. capitis and virulence-linked phylogroups to monitor the spread and evolution of S. capitis.
Collapse
|
49
|
Pseudomonas aeruginosa Production of Hydrogen Cyanide Leads to Airborne Control of Staphylococcus aureus Growth in Biofilm and In Vivo Lung Environments. mBio 2022; 13:e0215422. [PMID: 36129311 DOI: 10.1128/mbio.02154-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Diverse bacterial volatile compounds alter bacterial stress responses and physiology, but their contribution to population dynamics in polymicrobial communities is not well known. In this study, we showed that airborne volatile hydrogen cyanide (HCN) produced by a wide range of Pseudomonas aeruginosa clinical strains leads to at-a-distance in vitro inhibition of the growth of a wide array of Staphylococcus aureus strains. We determined that low-oxygen environments not only enhance P. aeruginosa HCN production but also increase S. aureus sensitivity to HCN, which impacts P. aeruginosa-S. aureus competition in microaerobic in vitro mixed biofilms as well as in an in vitro cystic fibrosis lung sputum medium. Consistently, we demonstrated that production of HCN by P. aeruginosa controls S. aureus growth in a mouse model of airways coinfected by P. aeruginosa and S. aureus. Our study therefore demonstrates that P. aeruginosa HCN contributes to local and distant airborne competition against S. aureus and potentially other HCN-sensitive bacteria in contexts relevant to cystic fibrosis and other polymicrobial infectious diseases. IMPORTANCE Airborne volatile compounds produced by bacteria are often only considered attractive or repulsive scents, but they also directly contribute to bacterial physiology. Here, we showed that volatile hydrogen cyanide (HCN) released by a wide range of Pseudomonas aeruginosa strains controls Staphylococcus aureus growth in low-oxygen in vitro biofilms or aggregates and in vivo lung environments. These results are of pathophysiological relevance, since lungs of cystic fibrosis patients are known to present microaerobic areas and to be commonly associated with the presence of S. aureus and P. aeruginosa in polymicrobial communities. Our study therefore provides insights into how a bacterial volatile compound can contribute to the exclusion of S. aureus and other HCN-sensitive competitors from P. aeruginosa ecological niches. It opens new perspectives for the management or monitoring of P. aeruginosa infections in lower-lung airway infections and other polymicrobial disease contexts.
Collapse
|
50
|
Douglas EJA, Alkhzem AH, Wonfor T, Li S, Woodman TJ, Blagbrough IS, Laabei M. Antibacterial activity of novel linear polyamines against Staphylococcus aureus. Front Microbiol 2022; 13:948343. [PMID: 36071957 PMCID: PMC9441809 DOI: 10.3389/fmicb.2022.948343] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/04/2022] [Indexed: 01/11/2023] Open
Abstract
New therapeutic options are urgently required for the treatment of Staphylococcus aureus infections. Accordingly, we sought to exploit the vulnerability of S. aureus to naturally occurring polyamines. We have developed and tested the anti-staphylococcal activity of three novel linear polyamines based on spermine and norspermine. Using a panel of genetically distinct and clinically relevant multidrug resistant S. aureus isolates, including the polyamine resistant USA300 strain LAC, compound AHA-1394 showed a greater than 128-fold increase in inhibition against specific S. aureus strains compared to the most active natural polyamine. Furthermore, we show that AHA-1394 has superior biofilm prevention and biofilm dispersal properties compared to natural polyamines while maintaining minimal toxicity toward human HepG2 cells. We examined the potential of S. aureus to gain resistance to AHA-1394 following in vitro serial passage. Whole genome sequencing of two stable resistant mutants identified a gain of function mutation (S337L) in the phosphatidylglycerol lysyltransferase mprF gene. Inactivation of mutant mprF confirmed the importance of this allele to AHA-1394 resistance. Importantly, AHA-1394 resistant mutants showed a marked decrease in relative fitness and increased generation time. Intriguingly, mprF::S337L contributed to altered surface charge only in the USA300 background whereas increased cell wall thickness was observed in both USA300 and SH1000. Lastly, we show that AHA-1394 displays a particular proclivity for antibiotic potentiation, restoring sensitivity of MRSA and VRSA isolates to daptomycin, oxacillin and vancomycin. Together this study shows that polyamine derivatives are impressive drug candidates that warrant further investigation.
Collapse
Affiliation(s)
- Edward J. A. Douglas
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Abdulaziz H. Alkhzem
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Toska Wonfor
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Shuxian Li
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Timothy J. Woodman
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Ian S. Blagbrough
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Maisem Laabei
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
- *Correspondence: Maisem Laabei,
| |
Collapse
|