1
|
Jeznach A, Sidor-Dzitkowska K, Bandyszewska M, Grzanka M, Popławski P, Marszalik A, Domagała-Kulawik J, Stachowiak R, Hoser G, Skirecki T. Sepsis-induced inflammasome impairment facilitates development of secondary A. baumannii pneumonia. Emerg Microbes Infect 2025; 14:2492206. [PMID: 40202049 PMCID: PMC12016274 DOI: 10.1080/22221751.2025.2492206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/07/2025] [Accepted: 04/07/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Acinetobacter baumannii has become one of the most critical pathogens causing nosocomial pneumonia. Existing animal models of A. baumannii pneumonia are not relevant to the majority of critical care patients. We aimed to develop a novel model of secondary A. baumannii pneumonia in post-sepsis mice. METHODS A two-hit model of sepsis induced by cecal ligation and puncture followed by A. baumannii pneumonia on day 5 was established. In addition, the two-hit model was established in humanized mice. A period of 2 h of mechanical ventilation followed by observation was used in additional experiments. Lung histopathology, bacterial cultures, and cellular infiltration were analysed as well as markers of the inflammasome activity in vivo and ex vivo. RESULTS A. baumannii infection caused mortality and loss of body weight and temperature in post-sepsis mice. Increased lung bacterial burden and dissemination together with signs of enhanced inflammatory injury were observed in post-sepsis mice but not control mice that were challenged with A. baumannii. Post-sepsis mice were unable to mount inflammasome activation in response to secondary pneumonia to the level of control mice. Transfer of wild-type but not capsase-1 KO alveolar macrophages was able to restore the pulmonary protection against A. baumannii. Mechanical ventilation exacerbated the pathological response to pneumonia in post-sepsis mice but enhanced inflammasome signalling in non-sepsis mice with pneumonia. CONCLUSIONS We established a novel model of A. baumannii pneumonia that revealed sepsis-induced impairment of inflammasome activation in alveolar macrophages is critical for the control of secondary A. baumannii pneumonia.
Collapse
Affiliation(s)
- Aldona Jeznach
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Karolina Sidor-Dzitkowska
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Magdalena Bandyszewska
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Małgorzata Grzanka
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Piotr Popławski
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Anna Marszalik
- Department of Bacterial Physiology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | | - Radosław Stachowiak
- Department of Bacterial Physiology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Grażyna Hoser
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
2
|
Samir A, Mosallam T, Aboul-Ella H, Ali A, Samir O, Hegab M, Erian M, Youssef F, Zaher H. Zoonotic relevance of multidrug-resistant bacteria in parrots with respiratory illness. Vet Res Commun 2025; 49:194. [PMID: 40338404 PMCID: PMC12062053 DOI: 10.1007/s11259-025-10752-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025]
Abstract
Nowadays, research attention is paid to the investigation of bacterial pathogens in the cloaca of parrots rather than the nasal niche, which is largely ignored. Therefore, this study aimed to investigate the nasal carriage of multidrug-resistant bacteria with zoonotic potential in parrots suffering from respiratory illness. Nasal swabs were collected from 75 sick parrots with respiratory illness, and they were subjected to microbiological isolation and identification, followed by antimicrobial susceptibility testing. Escherichia coli, Klebsiella pneumoniae, Proteus mirabilis, and Staphylococcus aureus were isolated with a prevalence rate of 36%, 32%, 26.7%, and 9.3%, respectively, while one isolate (1.3%) of Staphylococcus pseudointermedius, Staphylococcus simulans, Staphylococcus sciuri, and Enterococcus faecalis was identified. E. coli, K. pneumoniae, and P. mirabilis were investigated for ESBL genes, Staphylococcus species for the mecA gene, followed by SCCmec typing, and E. faecalis for the vanA and vanB genes. Regarding beta-lactamase-encoding genes, blaTEM (97.6%), blaSHV (48.8%), and blaCTX-M (39%) gene families were detected, while blaOXA was not found. Sequencing of blaCTX-M in one strain of E. coli, K. pneumoniae, and P. mirabilis revealed blaCTX-M-15. The mecA was determined in three S. aureus and one S. sciuri strain, and the SCCmec typing of three MRSA isolates yielded type V, whereas type I in S. sciuri. Only the vanA gene was recognized in the E. faecalis strain. Moreover, 67.1% of bacterial isolates exhibited multidrug resistance. These findings highlight the potential role of parrots in the transmission of multidrug-resistant zoonotic bacteria, which may pose a threat to human contacts.
Collapse
Affiliation(s)
- Ahmed Samir
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | | | - Hassan Aboul-Ella
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Aisha Ali
- LeptoVet Veterinary Laboratory, Cairo, Egypt
| | - Ojena Samir
- LeptoVet Veterinary Laboratory, Cairo, Egypt
| | | | | | | | - Hala Zaher
- Department of Zoonoses, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
3
|
Predtechenskaya M, Arbizzani CJ, Shomento SR, Borgogna TR, Voyich JM. Lung surfactant reduces Staphylococcus aureus cytotoxicity and protects host immune cells from membrane damage. Microbiol Spectr 2025:e0138624. [PMID: 40237467 DOI: 10.1128/spectrum.01386-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/20/2025] [Indexed: 04/18/2025] Open
Abstract
In this study, we identify that lung surfactant significantly reduces the cytotoxicity of Staphylococcus aureus (S. aureus) membrane-damaging toxins. Data demonstrate that natural surfactants from mice and rats and commercially available surfactant, Infasurf, protect human primary cells (neutrophils and peripheral blood mononuclear cells) from cytolytic activity caused by S. aureus supernatants. Supernatants from S. aureus grown in surfactant showed a significant reduction in plasma membrane damage against primary human cells as compared to supernatants grown without surfactant. This reduction was not due to a direct bactericidal effect of the surfactants on S. aureus growth. Rat and mouse surfactants downregulated the gene expression of saeR, the response regulator of the S. aureus two-component system SaeR/S that is responsible for the production of virulence factors which are important during lung infection and cause membrane damage in host cells. Rat and lung surfactants also reduced transcript abundance of SaeR/S-regulated genes lukF-PV, hla, and hlgA. Interestingly, the commercially available surfactant Infasurf did not recapitulate the effect of natural surfactants and did not decrease gene transcription of the virulence genes tested. These data suggest that components of natural surfactants protect lungs from S. aureus by suppressing S. aureus virulence factors and have implications for the role of surfactants in host defense against S. aureus.IMPORTANCEThis study explored the influence of lung surfactants on membrane-damaging Staphylococcus aureus (S. aureus) toxins. We demonstrate that natural and commercially available lung surfactants minimize the cytolytic capacity of S. aureus supernatants against primary human cells. Data indicate that cytolytic reduction by mouse and rat surfactants was partially due to surfactants reducing transcript abundance of virulence factors. This work identifies a novel role for surfactants and suggests their importance in modulating the severity of S. aureus lung infections.
Collapse
Affiliation(s)
- Maria Predtechenskaya
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Corbin J Arbizzani
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Sofia R Shomento
- University of Washington, School of Medicine, Seattle, Washington, USA
| | - Timothy R Borgogna
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Jovanka M Voyich
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
4
|
Bartsch B, Ackerschott A, Al Zaidi M, Jamin RN, Nazir MLF, Altrogge M, Fester L, Lambertz J, Coburn M, Nickenig G, Parcina M, Zimmer S, Weisheit CK. A novel approach to studying infective endocarditis: Ultrasound-guided wire injury and bacterial challenge in mice. PLoS One 2025; 20:e0318955. [PMID: 40193365 PMCID: PMC11975138 DOI: 10.1371/journal.pone.0318955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/23/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Infective endocarditis (IE) is frequently caused by Staphylococcus aureus (S. aureus) and most commonly affects the aortic valve. Early diagnosis and treatment initiation are challenging because the involved immunological processes are poorly understood due to a lack of suitable in vivo models. OBJECTIVES To establish a novel reproducible murine IE model, based on ultrasound-guided wire injury (WI) induced endothelial damage. METHODS IE was established by inducing endothelial damage via ultrasound-guided wire injury followed by bacterial challenge with S. aureus using 104-6 colony-forming units (CFU) 24h to 72h after wire injury. Cross-sections of valvular leaflets were prepared for scanning electron microscopy (SEM) and immunofluorescence microscopy to visualize valvular invasion of macrophages, neutrophils, and S. aureus. Bacterial cultivation was carried out from blood and valve samples. Systemic immune response was assessed using flow cytometry. RESULTS Wire injury induced endothelial damage was observed in all mice after wire-injury in SEM imaging. We reliably induced IE using 105 (85%) and 106 (91%) CFU S. aureus after wire injury. Aortic regurgitation was more prevalent in wire injury mice after bacterial challenge. Mice undergoing bacterial challenge responded with significant neutrophilia and elevated pro-inflammatory cytokines in the blood. Immunofluorescence staining revealed significantly increased immune cell accumulations using our proposed model compared to controls. CONCLUSION Echocardiography and ex vivo histological staining demonstrated consistent infective endocarditis induction in our new model, combining a wire injury-induced endothelial damage and S. aureus administration. Further exploration of the initial immune cell response and biomarker expression could potentially identify indicators for early IE diagnosis and novel treatment targets.
Collapse
Affiliation(s)
- Benedikt Bartsch
- Department of Internal Medicine-II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Ansgar Ackerschott
- Department of Internal Medicine-II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Muntadher Al Zaidi
- Department of Internal Medicine-II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Raul Nicolas Jamin
- Department of Internal Medicine-II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Mariam Louis Fathy Nazir
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Moritz Altrogge
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Lars Fester
- Institute of Neuroanatomy of the University of Bonn, University Bonn, Bonn, Germany
| | - Jessica Lambertz
- Institute of Neuroanatomy of the University of Bonn, University Bonn, Bonn, Germany
| | - Mark Coburn
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Georg Nickenig
- Department of Internal Medicine-II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Marijo Parcina
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Sebastian Zimmer
- Department of Internal Medicine-II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
5
|
Chen S, Mondile Q, Du X, Wang C, Mukim M, Wrenger C, Dömling ASS, Tastan Bishop Ö, Groves MR. Exploring Aspartate Transcarbamoylase: A Promising Broad-Spectrum Target for Drug Development. Chembiochem 2025; 26:e202401009. [PMID: 39937588 PMCID: PMC12002100 DOI: 10.1002/cbic.202401009] [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: 12/09/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/13/2025]
Abstract
Pyrimidine nucleotides are essential for a wide variety of cellular processes and are synthesized either via a salvage pathway or through de novo biosynthesis. The latter is particularly important in proliferating cells, such as infectious diseases and cancer cells. Aspartate transcarbamoylase (ATCase) catalyzes the first committed and rate-limiting step in the de novo pyrimidine biosynthesis pathway, making it an attractive therapeutic target for various diseases. This review summarizes the development of a series of allosteric ATCase inhibitors, advancing them as potential candidates for malarial, tuberculosis and cancer therapies. Furthermore, it explores the potential for these compounds to be expanded into drugs targeting neglected tropical diseases, antimicrobial-resistant infections caused by the ESKAPE pathogens, and their possible application as herbicides. We identify the likely equivalent allosteric pocket in these systems and perform a structure and sequence-based analysis of the residues comprising it, providing a rationale for continued exploration of this compound series as both specific and broad-range inhibitors. The review concludes by emphasizing the importance of continued research into ATCase inhibitors, given their potential broad applicability in treating diverse diseases to enhance both human health and agricultural practices.
Collapse
Affiliation(s)
- Siyao Chen
- Department of Chemical and Pharmaceutical BiologyUniversity of GroningenAntonius Deusinglaan 19731AVGroningenThe Netherlands
| | - Queenie Mondile
- Research Unit in Bioinformatics (RUBi)Department of BiochemistryMicrobiology and BiochemistryRhodes University
| | - XiaoChen Du
- Department of Chemical and Pharmaceutical BiologyUniversity of GroningenAntonius Deusinglaan 19731AVGroningenThe Netherlands
| | - Chao Wang
- NeurobiologyMRC-Laboratory of Molecular Biology Cambridge Biomedical CampusFrancis Crick Ave, TrumpingtonCambridgeCB2 0QH
| | - Mayur Mukim
- Czech Advanced Technology and Research Institute (CATRIN)and Institute of Molecular and Translational Medicine (IMTMFaculty of Medicine and DentistryPalacky UniversityŠlechtitelů 27779 00OlomoucCzech Republic
| | - Carsten Wrenger
- Unit for Drug DiscoveryDepartment of ParasitologyInstitute of Biomedical SciencesUniversity of São PauloAvenida Professor Lineu Prestes 137405508-000São Paulo-SPBrazil
| | - Alexander S. S. Dömling
- Czech Advanced Technology and Research Institute (CATRIN)and Institute of Molecular and Translational Medicine (IMTMFaculty of Medicine and DentistryPalacky UniversityŠlechtitelů 27779 00OlomoucCzech Republic
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi)Department of BiochemistryMicrobiology and BiochemistryRhodes University
- National Institute for Theoretical and Computational Sciences (NITheCS)South Africa
- Genomics for Health in Africa (GHA)Africa-Europe Cluster of Research Excellence (CoRE)
| | - Matthew R. Groves
- Department of Chemical and Pharmaceutical BiologyUniversity of GroningenAntonius Deusinglaan 19731AVGroningenThe Netherlands
- Genomics for Health in Africa (GHA)Africa-Europe Cluster of Research Excellence (CoRE)
| |
Collapse
|
6
|
Khrisna DF, Ambreen J, Wei TY, Riduan NAH, Daud NM, Zain NM, Abdullah F, Nik Malek NAN, Ulum MF, Saidin S. Immobilization of Antibacterial Chlorhexidine on Biodegradable Polycaprolactone/Estradiol Electrospun Nanofibrous Membrane for Bone Regeneration. J Biomed Mater Res B Appl Biomater 2025; 113:e35575. [PMID: 40167028 DOI: 10.1002/jbm.b.35575] [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: 02/22/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Abstract
Membrane-based scaffold for bone regeneration is vastly being explored to address issues that persist in defective bone regeneration, associated with infection and inflammation. This study focused on incorporating estradiol (E2) into biodegradable polycaprolactone (PCL) electrospun nanofibrous membrane, followed by the immobilization with antibacterial chlorhexidine (CHX) through the aid of a polydopamine (PDA) grafting layer. Several analyses including field emission scanning electron microscopy (FESEM), Fourier transform infrared spectroscopy (FTIR), wettability, biodegradation, drug release, antibacterial, and cytotoxicity analyses were conducted to study the physicochemical and biological properties of the membranes. The nanofibers were constructed with an average diameter of 1.32-1.33 μm and a porosity of 51%-53%, which is accommodating bone regeneration. The grafting of PDA was not only able to improve the surface wettability, which in turn allowed controllable degradability and sustained the release of E2 and CHX from the nanofibrous membranes. The immobilization of CHX on the PCL/E2 nanofibers has greatly retarded Gram-negative Escherichia coli compared to Gram-positive Staphylococcus aureus. The in vitro cytotoxicity assay statistically depicted the ability of the fabricated nanofibrous membranes to support cell proliferation without cytotoxic effects at the cell viability above 70%. These cumulative results indicate the potential development of CHX-immobilized PCL/E2 membrane as an alternative strategy to resolve bone regeneration issues.
Collapse
Affiliation(s)
- Dida Faadihilah Khrisna
- Department of Biomedical Engineering & Health Sciences, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | - Jaweria Ambreen
- IJN-UTM Cardiovascular Engineering Center, Institute of Human Centered Engineering, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
- Department of Chemistry, COMSATS University Islamabad, Islamabad, Pakistan
| | - Tan Yu Wei
- Department of Biomedical Engineering & Health Sciences, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | - Nurul Aina Haziqah Riduan
- Department of Biomedical Engineering & Health Sciences, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | - Nurizzati Mohd Daud
- Department of Biomedical Engineering & Health Sciences, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | - Norhidayu Muhamad Zain
- Academy of Islamic Civilization, Faculty of Social Sciences and Humanities, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | - Faizuan Abdullah
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Malaysia
| | - Nik Ahmad Nizam Nik Malek
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Malaysia
- Centre for Sustainable Nanomaterials (CSNano), Ibnu Sina Institute for Scientific and Industrial Research (ISI-SIR), Universiti Teknologi Malaysia, Malaysia
| | - Mokhamad Fakhrul Ulum
- Division of Reproduction, Obstetrics and Gynaecology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, Indonesia
| | - Syafiqah Saidin
- Department of Biomedical Engineering & Health Sciences, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
- IJN-UTM Cardiovascular Engineering Center, Institute of Human Centered Engineering, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| |
Collapse
|
7
|
Xu Y, Wang L, Guo D, Wang Y, Liu X, Sun Y, Wang R, Sun L, Jiang P, Liu Q, Wang B, Yan M, Zhao Y. Baohuoside I targets SaeR as an antivirulence strategy to disrupt MRSA biofilm formation and pathogenicity. NPJ Biofilms Microbiomes 2025; 11:45. [PMID: 40102466 PMCID: PMC11920273 DOI: 10.1038/s41522-025-00681-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 03/08/2025] [Indexed: 03/20/2025] Open
Abstract
The emergence of methicillin-resistant Staphylococcus aureus (MRSA) represents a critical global health challenge, making the SaeRS two-component system (TCS), a key regulator of S. aureus virulence, an ideal target for novel therapeutic approaches. In this study, virtual screening and thermal shift assays identified Baohuoside I (BI), a flavonol glycoside, as a potent inhibitor of the SaeR response regulator. BI significantly attenuated S. aureus pathogenicity without bactericidal effects, suppressing the expression of key virulence factors, such as hemolysin A (Hla) and Panton-Valentine leukocidin (PVL), while modulating immune evasion pathways. Additionally, BI disrupted biofilm formation, promoting the development of porous, less structured biofilms. Biochemical assays, including EMSA, CETSA, fluorescence quenching, and SPR, confirmed strong binding interactions between SaeR and BI. In vivo, BI demonstrated therapeutic efficacy in Galleria mellonella and rat MRSA models. These findings establish BI as a promising lead for nonbactericidal therapies to combat MRSA infections and mitigate resistance.
Collapse
Affiliation(s)
- Yueshan Xu
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Li Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Dongbin Guo
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yueying Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xinyao Liu
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yun Sun
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Rong Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Luanbiao Sun
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Peitong Jiang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Quan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun, China
| | - Bingmei Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China.
| | - Ming Yan
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China.
| | - Yicheng Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun, China.
- Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| |
Collapse
|
8
|
Barua N, Chan BCL, Lau CBS, Leung PC, Fung KP, Ip M. Antivirulence Properties of Kuraridin Against Methicillin-Resistant Staphylococcus aureus (MRSA). Biomedicines 2025; 13:564. [PMID: 40149540 PMCID: PMC11940505 DOI: 10.3390/biomedicines13030564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/15/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Methicillin-resistant Staphylococcus aureus (MRSA) is a major human opportunistic pathogen that causes a wide range of infections. The vast arsenal of virulence factors expressed remains the biggest challenge in treating MRSA with conventional antibiotic therapy. Methods: We investigated the effects of Kuraridin at subinhibitory minimum inhibition concentrations (MICs) of 1/8, 1/16, and 1/32 (concentrations that did not inhibit bacterial growth) on adhesion to fibrinogen, adhesion, internalization into HaCaT cells, and biofilm production in three MRSA strains representing the clonal types USA300, ST30, and ST239. Results: All three MRSA strains exhibited a significant decrease (p < 0.001) in adhesion to fibrinogen upon treatment with 1/8 and 1/16 MICs of Kuraridin. The adhesion and internalization of all the MRSA strains to HaCaT cells were decreased significantly (p < 0.001) upon treatment with the three subinhibitory concentrations of Kuraridin. The biofilm formation of USA300 (p < 0.001), ST30 (p < 0.001), and ST239 (p < 0.01) was significantly reduced at a 1/8 MIC. A significant decrease in biofilm formation at a 1/16 MIC was observed for USA300 (p < 0.001) and ST30 (p < 0.05). Confocal laser scanning microscopy (CSLM) analysis of the biofilms revealed a reduction in biofilm formation in the MRSA strain when treated with Kuraridin. In the in vivo Caenorhabditis elegans model, Kuraridin offered a sizable degree of protection against MRSA infection without being toxic to the nematode. Conclusions: Our findings reveal that Kuraridin has the potential to be an alternative antivirulence option for reducing MRSA pathogenicity.
Collapse
Affiliation(s)
- Nilakshi Barua
- Department of Microbiology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China
| | - Ben Chung Lap Chan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China; (B.C.L.C.)
| | - Clara Bik-San Lau
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China; (B.C.L.C.)
| | - Ping-Chung Leung
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China; (B.C.L.C.)
| | - Kwok Pui Fung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China
| | - Margaret Ip
- Department of Microbiology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China
| |
Collapse
|
9
|
Giormezis N, Rechenioti A, Doumanas K, Sotiropoulos C, Paliogianni F, Kolonitsiou F. Bacteriophage Resistance, Adhesin's and Toxin's Genes Profile of Staphylococcus aureus Causing Infections in Children and Adolescents. Microorganisms 2025; 13:484. [PMID: 40142376 PMCID: PMC11946024 DOI: 10.3390/microorganisms13030484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Staphylococcus aureus is a common pathogen, often recovered from children's infections. Βiofilm formation, antimicrobial resistance and production of adhesins and toxins contribute to its virulence. As resistance to antimicrobials rises worldwide, alternative therapies like bacteriophages (among them the well-studied Bacteriophage K) can be helpful. The aim of this study was to determine the bacteriophage and antimicrobial susceptibility and the presence of virulence genes among S. aureus from infections in children and adolescents. Eighty S. aureus isolates were tested for biofilm formation and antimicrobial susceptibility. The presence of two genes of the ica operon (icaA, icaD), adhesin's (fnbA, fnbB, sasG) and toxin's genes (PVL, tst, eta, etb) was tested by PCRs. Susceptibility to Bacteriophage K was determined using a spot assay. Thirteen isolates were methicillin-resistant (MRSA) and 41 were multi-resistant. Twenty-five S. aureus (31.3%) were resistant to Bacteriophage K, mostly from ocular and ear infections. Twelve S. aureus (15%) were PVL-positive, seven (8.8%) positive for tst, 18 (22.5%) were eta-positive and 46 were (57.5%) etb-positive. A total of 66 (82.5%) isolates carried fnbA, 16 (20%) fnbB and 26 (32.5%) sasG. PVL, tst and sasG carriage were more frequent in MRSA. Bacteriophage-susceptible isolates carried more frequently eta (32.7%) and etb (69.1%) compared to phage-resistant S. aureus (0% and 32%, respectively). Although mainly methicillin-sensitive, S. aureus from pediatric infections exhibited high antimicrobial resistance and carriage of virulence genes (especially for exfoliative toxins and fnbA). MRSA was associated with PVL, tst and sasG carriage, whereas Bacteriophage susceptibility was associated with eta and etb. The high level of Bacteriophage K susceptibility highlights its potential use against staphylococcal infections.
Collapse
Affiliation(s)
- Nikolaos Giormezis
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Assimina Rechenioti
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Konstantinos Doumanas
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| | | | - Fotini Paliogianni
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Fevronia Kolonitsiou
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
10
|
Jahangiri A, Dahaghin S, Malekara E, Halabian R, Mahboobi M, Behzadi E, Sedighian H. Highly sensitive detection of Staphylococcus aureus α-hemolysin protein (Hla or α-toxin) by apta-qPCR. J Microbiol Methods 2025; 229:107084. [PMID: 39742925 DOI: 10.1016/j.mimet.2024.107084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Alpha-toxin of Staphylococcus aureus belongs to the pore-forming toxin (PFT) family, which can lyse red and white blood cells. In addition to the existence of the hla gene in the majority of S. aureus strains (about 95 %), higher expression exhibits enhanced pathogenicity to the bacteria. Various methods, such as antibodies and aptamers, could serve to detect this toxin. In the current study, for the first time, an improved sandwich aptamer-antibody-based method was developed using specific murine polyclonal antibodies and a specific aptamer to detect a wide range of α-toxin levels. Denatured recombinant α-toxin was administered to mice to trigger the production of specific antibodies, which were subsequently purified from immune sera. These antibodies served as capturers in the designed apta-qPCR assay, with an aptamer employed as a detector. The results showed that spiked α-toxin in the undiluted serum samples could detect α-toxin between 300 and 0.5 ng/mL with no cross-reactivity. The coefficient of variation (CV) percent of intra- and inter-assays were 0.84 and 1.06, respectively. Since in the apta-qPCR assay, a combination of specific polyclonal antibodies as capture and specific aptamer along with real-time PCR (qPCR) sensitivity is used, this robust method could be used in diagnostic laboratories to detect various levels of the toxin in human serum samples.
Collapse
Affiliation(s)
- Abolfazl Jahangiri
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Samira Dahaghin
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ehsan Malekara
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahdieh Mahboobi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- The Academy of Medical Sciences of Iran, Tehran, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Lin YT, Bui NN, Cheng YS, Lin CW, Lee CL, Lee TF, Hsueh PR. High hemolytic activity in Staphylococcus aureus t1081/ST45 due to increased hla protein production and potential RNAIII-independent regulation. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025; 58:70-76. [PMID: 39322509 DOI: 10.1016/j.jmii.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND α-Hemolysin, encoded by hla, is a major virulence factor of Staphylococcus aureus. Sequence type (ST) 45 is a globally spread clone with increasing clinical prevalence in Taiwan. Our previous study showed that among the CC45 isolates, the spa type t1081 isolates presented greater hemolytic activity. MATERIALS AND METHODS The hemolytic activity of 67 CC45 isolates (44 t1081 and 23 non-t1081) from clinical blood cultures was assessed using rabbit red blood cells. The sequences of hla and its upstream regulatory regions and RNAIII were compared between the two groups. The expression of hla and its regulators RNAIII, mgrA, and saeR was analyzed via qRT‒PCR, while Hla protein levels were measured via Western blotting. RESULTS Compared with non-t1081 isolates, t1081 isolates presented increased hemolytic activity. No significant differences in hla sequences, upstream regulatory regions, or gene expression levels were detected between the two groups. The expression of the transcriptional regulators mgrA and saeR was also similar between the two groups. Western blotting revealed increased Hla protein in the t1081 isolates. However, neither the sequence or expression of RNAIII, a regulator of hla at both the transcriptional and posttranscriptional levels, differed between the groups. CONCLUSION Our study revealed that, compared with other CC45 isolates, the t1081/ST45 isolates presented greater hemolytic activity. This heightened activity was due mainly to increased Hla protein levels. Moreover, the higher translation levels may be independent of the known regulator RNAIII, indicating a potential RNAIII-independent mechanism for Hla regulation.
Collapse
Affiliation(s)
- Yu-Tzu Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan.
| | - Ngoc-Niem Bui
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan; Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Viet Nam
| | - Yu-Syuan Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Cheng-Wen Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Chun-Li Lee
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tai-Fen Lee
- Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Po-Ren Hsueh
- Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Laboratory Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan; Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
12
|
Faraj R, Ramadan H, Bentum KE, Alkaraghulli B, Woube Y, Hassan Z, Samuel T, Adesiyun A, Jackson CR, Abebe W. Antimicrobial Resistance, Virulence Gene Profiling, and Spa Typing of Staphylococcus aureus Isolated from Retail Chicken Meat in Alabama, USA. Pathogens 2025; 14:107. [PMID: 40005484 PMCID: PMC11858072 DOI: 10.3390/pathogens14020107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Antibiotic-resistant Staphylococcus aureus (S. aureus) in retail meat poses a public health threat requiring continuous surveillance. This study investigated the frequency of isolation, toxin genes, and antibiotic resistance profile of S. aureus recovered from retail poultry meat samples and presented results beneficial to public health interventions. Of 200 samples collected, 16% (32/200) tested positive for S. aureus, and these were recovered from thigh 37.5% (12/32), wing 34.4% (11/32), gizzard (15.6% (5/32), and liver 12.5% (4/32) samples. Findings of spa typing analysis revealed that 68.8% (22/32), 18.8% (6/32), 9.4% (3/32), and 3.0% (1/32) of the isolates belonged to the spa types t267, t160, t548, and t008, respectively. For antibiotic susceptibility testing, 12.5% (4/32) of the isolates were resistant to only penicillin, but one isolate (1/32; 3%) showed resistance to the antibiotics penicillin, erythromycin, ampicillin, and oxacillin. PCR analysis revealed that 9.4% (3/32) of the isolates carried the mecA gene associated with methicillin-resistant Staphylococcus aureus (MRSA) isolates. One MRSA isolate was identified as a t008 spa type, and harbored a 26,974 bp-sized plasmid, which was the source of its resistance to penicillin, ampicillin, erythromycin, and oxacillin. The staphylococcal enterotoxin (SE) genes seg, sei, sek, seb, selm, and seln were also identified among the isolates, and mostly the antimicrobial and enterotoxin genes were carried on plasmids of the isolates. This study raises awareness on the continuous circulation of pathogenic microbes like S. aureus in retail poultry meat.
Collapse
Affiliation(s)
- Rawah Faraj
- Center for Food Animal Health, Food Safety and Defense, Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA; (R.F.); (K.E.B.); (B.A.); (Y.W.); (Z.H.); (T.S.)
| | - Hazem Ramadan
- Hygiene and Zoonoses Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Kingsley E. Bentum
- Center for Food Animal Health, Food Safety and Defense, Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA; (R.F.); (K.E.B.); (B.A.); (Y.W.); (Z.H.); (T.S.)
| | - Bilal Alkaraghulli
- Center for Food Animal Health, Food Safety and Defense, Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA; (R.F.); (K.E.B.); (B.A.); (Y.W.); (Z.H.); (T.S.)
| | - Yilkal Woube
- Center for Food Animal Health, Food Safety and Defense, Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA; (R.F.); (K.E.B.); (B.A.); (Y.W.); (Z.H.); (T.S.)
| | - Zakaria Hassan
- Center for Food Animal Health, Food Safety and Defense, Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA; (R.F.); (K.E.B.); (B.A.); (Y.W.); (Z.H.); (T.S.)
- Department of Food Hygiene and Control, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32511, Egypt
| | - Temesgen Samuel
- Center for Food Animal Health, Food Safety and Defense, Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA; (R.F.); (K.E.B.); (B.A.); (Y.W.); (Z.H.); (T.S.)
| | - Abiodun Adesiyun
- Faculty of Medical Sciences, School of Veterinary Medicine, University of the West Indies, St. Augustine 999183, Trinidad and Tobago;
| | - Charlene R. Jackson
- Poultry Microbiological Safety and Processing Research Unit USDA-ARS, U.S. National Poultry Research Center, Athens, GA 30605, USA;
| | - Woubit Abebe
- Center for Food Animal Health, Food Safety and Defense, Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA; (R.F.); (K.E.B.); (B.A.); (Y.W.); (Z.H.); (T.S.)
| |
Collapse
|
13
|
González-Arjona M, Sobrino G, Cussó L, Guembe M, Calle D, Díaz Crespo F, Bouza E, Muñoz P, Desco M, Salinas B. 99mTc-DTPA-Collagen Radiotracer for the Noninvasive Detection of Infective Endocarditis. ACS Infect Dis 2025; 11:121-130. [PMID: 39645608 PMCID: PMC11731287 DOI: 10.1021/acsinfecdis.4c00460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 12/09/2024]
Abstract
Infective endocarditis (IE) represents a significant concern among hospital-acquired infections, frequently caused by the Gram-positive bacterium Staphylococcus aureus. Nuclear imaging is emerging as a noninvasive and precise diagnostic tool. However, the gold standard radiotracer [18F]-FDG cannot distinguish between infection and inflammation, resulting in false positives. Based on the presence of collagen-binding proteins in the cell wall of S. aureus, we propose the radiolabeling of collagen for its evaluation in IE animal models by single-photon emission computed tomography (SPECT) imaging. We radiolabeled rat tail collagen I using DTPA chelator and [99mTc]NaTcO4. Selectivity was evaluated in vitro using 3 Gram-positive bacteria, 1 Gram-negative bacteria and 1 yeast. In vivo SPECT/computed tomography (CT) imaging was conducted on 8 SD rat models of IE and 8 sterile sham model as controls. Ex vivo biodistribution and autoradiography were performed following imaging. Diagnosis of IE was confirmed through microbiological studies and H&E histopathology. [99mTc]-DTPA-Collagen was synthesized successfully with a yield of 42.86 ± 6.35%, a purity of 95.84 ± 1.85% and a stability higher than 90% after 50 h postincubation. In vitro uptake demonstrated the selectivity for Gram-positive bacteria (63.85 ± 15.15%). Ex vivo analysis confirmed hepato-splenic excretion. In vivo SPECT/CT imaging revealed highly localized uptake within the aortic valve with a sensitivity of 62.5% and specificity of 87.5%. We successfully synthesized and characterized a new SPECT radiotracer based on [99mTc]Tc-radiolabeled collagen. In vitro studies demonstrated the selectivity of the radiotracer for Gram-positive bacteria. In vivo SPECT/CT-based assessment in an IE model confirmed the potential of this approach to detect active IE.
Collapse
Affiliation(s)
- Mario González-Arjona
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Gorka Sobrino
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Lorena Cussó
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad
de Imagen Avanzada, Centro Nacional de Investigaciones
Cardiovasculares (CNIC), Madrid 28029, Spain
- Centro
de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid 28029, Spain
| | - María Guembe
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Servicio
de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid 28007, Spain
| | - Daniel Calle
- Unidad
de Imagen Avanzada, Centro Nacional de Investigaciones
Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Francisco Díaz Crespo
- Servicio
de Anatomía Patológica, Hospital
General Universitario Gregorio Marañón, Madrid 28007, Spain
| | - Emilio Bouza
- Servicio
de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid 28007, Spain
- Departamento
de Medicina, Facultad de Medicina, Universidad
Complutense de Madrid, Madrid 28040, Spain
- Centro
de Investigación Biomédica en Red de Enfermedades Respiratorias
(CIBERES), Madrid 28029, Spain
| | - Patricia Muñoz
- Servicio
de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid 28007, Spain
- Departamento
de Medicina, Facultad de Medicina, Universidad
Complutense de Madrid, Madrid 28040, Spain
- Centro
de Investigación Biomédica en Red de Enfermedades Respiratorias
(CIBERES), Madrid 28029, Spain
| | - Manuel Desco
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad
de Imagen Avanzada, Centro Nacional de Investigaciones
Cardiovasculares (CNIC), Madrid 28029, Spain
- Centro
de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid 28029, Spain
- Departamento
de Bioingeniería, Universidad Carlos
III de Madrid, Madrid 28911, Spain
| | - Beatriz Salinas
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad
de Imagen Avanzada, Centro Nacional de Investigaciones
Cardiovasculares (CNIC), Madrid 28029, Spain
- Centro
de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid 28029, Spain
- Departamento
de Bioingeniería, Universidad Carlos
III de Madrid, Madrid 28911, Spain
| |
Collapse
|
14
|
Mantovam VB, Dos Santos DF, Giola Junior LC, Landgraf M, Pinto UM, Todorov SD. Listeria monocytogenes, Salmonella spp., and Staphylococcus aureus: Threats to the Food Industry and Public Health. Foodborne Pathog Dis 2025. [PMID: 39761068 DOI: 10.1089/fpd.2024.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
Foodborne pathogens have always been of public health concern and represent safety issues for food processors. These pathogens develop new ways to overcome antibiotics, survive in different environmental conditions, and the ability to reproduce in many hostile environments configure them as serious health hazards. Considering the huge number of microorganisms, three bacterial representatives were selected to provide a better knowledge about the question of which one is the worst enemy for humans, from the food industry point of view, taking into consideration their multiplication specificity, virulence, and resistance. As we constantly are exposed to these pathogens in our nutritional habits, this overview aims to summarize the most relevant characteristics associated with the pathogenicity, clinical symptoms and most importantly, how deadly Listeria monocytogenes, Salmonella spp., and Staphylococcus aureus can be in the hospital and the food industry, by comparing among them. Overall, the microbiological knowledge clearly suggests that while all three pathogens are dangerous, L. monocytogenes presents the highest risk of death due to their ability to cause severe complications in vulnerable populations as it presents a range of virulence factors that facilitate evasion of the immune system and cytological effects. Additionally, it shows great resistance to standard food processing and preservation techniques, making it one of the most difficult pathogens to control. Understanding the risks and characteristics of these foodborne pathogens is essential for implementing effective control measures to prevent their occurrence in food products and to promote public health.
Collapse
Affiliation(s)
- Vinicius B Mantovam
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - David F Dos Santos
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Luis C Giola Junior
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Mariza Landgraf
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Uelinton M Pinto
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Svetoslav D Todorov
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos e Nutrição Experimental, FoRC, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Ingmer H, Leisner JJ, Fulaz S. Forssman and the staphylococcal hemolysins. APMIS 2025; 133:e13459. [PMID: 39188243 PMCID: PMC11669744 DOI: 10.1111/apm.13459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
Forssman was a Swedish pathologist and microbiologist who, in the 1920s and 1930s conducted a long series of experiments that led to unique insights into surface antigens of blood cells, as well as added to the discrimination of toxins produced by staphylococci that lyse red blood cells. This review takes offset in the studies published by Forssman in APMIS addressing the hemolytic properties of staphylococcal toxins displayed against erythrocytes of animal and human origin. In light of current knowledge, we will discuss the insights we now have and how they may pave the way for curing infections with pathogenic staphylococci, including Staphylococcus aureus.
Collapse
Affiliation(s)
- Hanne Ingmer
- Department of Veterinary and Animal ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Jørgen J. Leisner
- Department of Veterinary and Animal ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Stephanie Fulaz
- Department of Veterinary and Animal ScienceUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
16
|
Prem P, Naveenkumar S, Kamaraj C, Ragavendran C, Priyadharsan A, Manimaran K, Alharbi NS, Rarokar N, Cherian T, Sugumar V, Thiruvengadam M, Kumarasamy V, Subramaniyan V. Valeriana jatamansi
root extract a potent source for biosynthesis of silver nanoparticles and their biomedical applications, and photocatalytic decomposition. GREEN CHEMISTRY LETTERS AND REVIEWS 2024; 17. [DOI: 10.1080/17518253.2024.2305142] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Affiliation(s)
- Pradisha Prem
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology (SRMIST), Chennai, India
| | - Selvam Naveenkumar
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology (SRMIST), Chennai, India
| | - Chinnaperumal Kamaraj
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology (SRMIST), Chennai, India
- Interdisciplinary Institute of Indian System of Medicine, Directorate of Research, SRM Institute of Science and Technology, Chennai, India
| | - Chinnasamy Ragavendran
- Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Arumugam Priyadharsan
- Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- Research Center for Advanced Materials – National Research and Innovation Agency (BRIN), Banten, Indonesia
| | - Kumar Manimaran
- Research Center for Applied Microbiology, National Research and Innovation Agency (BRIN), Cibinong, Indonesia
| | - Naify S Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | | | - Tijo Cherian
- School of Biosciences, Mar Athanasios College for Advanced Studies Tiruvalla (MACFAST), Tiruvalla, India
| | - Vimal Sugumar
- Department of Biochemistry, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Sciences, Konkuk University, Seoul, Republic of Korea
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
17
|
Santos INDS, Lilenbaum W, Moreno DSA, Bastos MDCDF. Production of bacteriocin-like inhibitory substance (BLIS) by Staphylococcus spp. isolates from dogs. Braz J Microbiol 2024; 55:3885-3897. [PMID: 39467939 PMCID: PMC11711726 DOI: 10.1007/s42770-024-01532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
In the present study, 39 canine isolates of Staphylococcus spp. were tested for antimicrobial substance (AMS) production. Seven AMS producers were identified, whose products exhibited a non-acidic character and a proteinaceous nature, therefore being considered bacteriocin-like inhibitory substances (BLIS). The producer strains of BLIS P1, P16 and I3 showed a broad spectrum of antimicrobial activity. Human, veterinary and plant pathogens, such as Listeria monocytogenes, Klebsiella pneumoniae, Staphylococcus spp. and Clavibacter michiganensis, were among the inhibited micro-organisms, suggesting the potential biotechnological application of these peptides. MALDI-TOF mass spectrometry and 16 S rDNA sequencing identified the producer strains of BLIS P1, P16 and I3 as Staphylococcus pseudintermedius P1, Staphylococcus schleiferi P16 and Staphylococcus pseudintermedius I3. The plasmid profile of these strains suggests that the BLIS production is linked to biosynthetic genes located on plasmids. PCR analyses revealed that BLIS P1, P16 and I3 are different from 11 staphylococcins already described in the literature and that their genomic DNAs do not carry the most prevalent staphylococcal enterotoxin genes. The highest levels of BLIS production were achieved after 18-24 h of growth of the producer strains in TSB medium. Moreover, BLIS P1 and I3 exhibited high resistance to temperature and pH variations, and BLIS P16 maintained 100% of its activity in almost all conditions tested. The characteristics associated with BLIS P1, P16 and I3 described in this work encourage further investigation of these substances, in addition to this study being the first report of BLIS production by a strain of S. schleiferi.
Collapse
Affiliation(s)
- Ilana Nascimento de Sousa Santos
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes - UFRJ, Av. Carlos Chagas Filho, 373 CCS - Bloco I - Cidade Universitária, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Walter Lilenbaum
- Departamento de Microbiologia e Parasitologia 2, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Daniela Sales Alviano Moreno
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes - UFRJ, Av. Carlos Chagas Filho, 373 CCS - Bloco I - Cidade Universitária, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Maria do Carmo de Freire Bastos
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes - UFRJ, Av. Carlos Chagas Filho, 373 CCS - Bloco I - Cidade Universitária, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
18
|
Lin YT, Lee CL, Lin CY, Lee TF, Hsueh PR. High hemolytic activity of the Staphylococcus aureus spa t1081 among clonal complex 45 in Taiwan. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:906-915. [PMID: 39266361 DOI: 10.1016/j.jmii.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Methicillin-resistant Staphylococcus aureus (MRSA) sequence type (ST) 45 was first reported in Taiwan in 2006. Since then, the prevalence of ST45 MRSA in clinical isolates has increased. This study was carried out to understand the changes in the proportions, evolutionary relationships, and infection advantages of ST45 and its related clones. MATERIALS AND METHODS S. aureus including MRSA and MSSA (methicillin-sensitive S. aureus), and clonal complex (CC) 45 blood isolates were collected in 2000, 2005, and from January 2010 to August 2014. Molecular typing, multiple-locus variable-number tandem repeat analysis (MLVA) and single nucleotide polymorphism (SNP)-based phylogenetic analysis were performed. Fitness and virulence analyses were used to understand the infection advantages of the isolates. RESULTS Among the 67 CC45 isolates, only MSSA ST508 isolates were found in 2000 and 2005. Since 2010, the prevalence of MRSA has increased, t1081/ST45 has become dominant, and MRSA ST508 has been found. Phylogenetic analysis indicated that most of the ST45 isolates were located in a cluster distinct from those of ST508 and ST929. However, the t026 isolates clustered with the ST508 isolates rather than with the other ST45 isolates. Moreover, fitness and virulence analyses revealed that the t1081 isolates had higher hemolytic activity than the t026 and ST508 isolates did. CONCLUSION Our findings indicated that the increased prevalence of ST45 MRSA isolates from blood cultures in Taiwan was due to the t1081 isolates, and their high hemolytic activity may provide an infection advantage.
Collapse
Affiliation(s)
- Yu-Tzu Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan.
| | - Chun-Li Lee
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chin-Yun Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Tai-Fen Lee
- Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Po-Ren Hsueh
- Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Laboratory Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan; Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
19
|
Scull G, Aligwekwe A, Rey Y, Koch D, Nellenbach K, Sheridan A, Pandit S, Sollinger J, Pierce JG, Flick MJ, Gilbertie J, Schnabel L, Brown AC. Fighting fibrin with fibrin: Vancomycin delivery into coagulase-mediated Staphylococcus aureus biofilms via fibrin-based nanoparticle binding. J Biomed Mater Res A 2024; 112:2071-2085. [PMID: 38874363 PMCID: PMC11464197 DOI: 10.1002/jbm.a.37760] [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: 02/12/2024] [Revised: 05/02/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Staphylococcus aureus skin and soft tissue infection is a common ailment placing a large burden upon global healthcare infrastructure. These bacteria are growing increasingly recalcitrant to frontline antimicrobial therapeutics like vancomycin due to the prevalence of variant populations such as methicillin-resistant and vancomycin-resistant strains, and there is currently a dearth of novel antibiotics in production. Additionally, S. aureus has the capacity to hijack the host clotting machinery to generate fibrin-based biofilms that confer protection from host antimicrobial mechanisms and antibiotic-based therapies, enabling immune system evasion and significantly reducing antimicrobial efficacy. Emphasis is being placed on improving the effectiveness of therapeutics that are already commercially available through various means. Fibrin-based nanoparticles (FBNs) were developed and found to interact with S. aureus through the clumping factor A (ClfA) fibrinogen receptor and directly integrate into the biofilm matrix. FBNs loaded with antimicrobials such as vancomycin enabled a targeted and sustained release of antibiotic that increased drug contact time and reduced the therapeutic dose required for eradicating the bacteria, both in vitro and in vivo. Collectively, these findings suggest that FBN-antibiotic delivery may be a novel and potent therapeutic tool for the treatment of S. aureus biofilm infections.
Collapse
Affiliation(s)
- Grant Scull
- Joint Department of Biomedical Engineering, NC State University and UNC-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
| | - Adrian Aligwekwe
- Joint Department of Biomedical Engineering, NC State University and UNC-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
| | - Ysabel Rey
- Joint Department of Biomedical Engineering, NC State University and UNC-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
| | - Drew Koch
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
- College of Veterinary Medicine, NC State University, Raleigh, North Carolina, USA
| | - Kimberly Nellenbach
- Joint Department of Biomedical Engineering, NC State University and UNC-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
| | - Ana Sheridan
- Joint Department of Biomedical Engineering, NC State University and UNC-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
| | - Sanika Pandit
- Joint Department of Biomedical Engineering, NC State University and UNC-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
| | - Jennifer Sollinger
- Joint Department of Biomedical Engineering, NC State University and UNC-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
| | - Joshua G Pierce
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
- Department of Chemistry, NC State University, Raleigh, North Carolina, USA
| | - Matthew J Flick
- UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
- Blood Research Center, UNC-Chapel Hill, Chapel Hill, NC, USA
| | - Jessica Gilbertie
- Department of Microbiology and Immunology, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia, USA
| | - Lauren Schnabel
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
- College of Veterinary Medicine, NC State University, Raleigh, North Carolina, USA
| | - Ashley C Brown
- Joint Department of Biomedical Engineering, NC State University and UNC-Chapel Hill, Raleigh, North Carolina, USA
- Comparative Medicine Institute, NC State University, Raleigh, North Carolina, USA
- Blood Research Center, UNC-Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
20
|
McGahon J, Woods S, D'Elia R, Roberts CW. Non-ionic surfactant vesicles exert anti-inflammatory effects through inhibition of NFκB. J Inflamm (Lond) 2024; 21:49. [PMID: 39593021 PMCID: PMC11590361 DOI: 10.1186/s12950-024-00419-5] [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: 02/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Inflammation can be an unwanted consequence or cause of debilitating diseases of infectious and non-infectious aetiologies. Current anti-inflammatory medications have several deficiencies including lack of specificity and undesirable side effects. Herein, the potential of non-ionic surfactant vesicles (NISV) comprised of monopalmityol glycerol, dicetyl phosphate and cholesterol) as an anti-inflammatory drug and their mode of action is investigated. NISV were able to inhibit LPS-induced IL-6 from BMD macrophages. The individual components of NISV, monopalmityol glycerol, dicetyl phosphate and cholesterol did not affect LPS induced IL-6 levels, proving that formulation of NISV is essential for their anti-inflammatory effects. Transcriptomic analyses showed NISV mediated down-regulation of transcripts for inflammatory mediators in LPS stimulated macrophages. Notably, NISV downregulate NF-κB transcripts in LPS stimulated macrophages. Measurement of inflammatory mediators by cytometric bead array validated a number of transcriptomic findings as NISV were found to inhibit LPS induced IL-6, IL-12, and multiple chemokines. Further investigation demonstrated that NISV inhibited Poly(I:C) or Pam3csk4 induced inflammatory mediators. This indicates that the effects of NISV are distal to both MyD88 and TRIF signalling. Overall, the data generated highlights the potential of NISV as an anti-inflammatory therapeutic.
Collapse
Affiliation(s)
- Jonathan McGahon
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Stuart Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
- School of Health and Life Sciences, University of West Scotland, Stephenson Place, Glasgow, Lanarkshire, G72 0LH, UK
| | - Riccardo D'Elia
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
- Chemical Biological and Radiological Division, Dstl, Porton Down, Salisbury, SP4 0JQ, UK
| | - Craig W Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK.
| |
Collapse
|
21
|
Wang Y, Liu Y, Xiang G, Jian Y, Yang Z, Chen T, Ma X, Zhao N, Dai Y, Lv Y, Wang H, He L, Shi B, Liu Q, Liu Y, Otto M, Li M. Post-translational toxin modification by lactate controls Staphylococcus aureus virulence. Nat Commun 2024; 15:9835. [PMID: 39537625 PMCID: PMC11561239 DOI: 10.1038/s41467-024-53979-8] [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: 07/09/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Diverse post-translational modifications have been shown to play important roles in regulating protein function in eukaryotes. By contrast, the roles of post-translational modifications in bacteria are not so well understood, particularly as they relate to pathogenesis. Here, we demonstrate post-translational protein modification by covalent addition of lactate to lysine residues (lactylation) in the human pathogen Staphylococcus aureus. Lactylation is dependent on lactate concentration and specifically affects alpha-toxin, in which a single lactylated lysine is required for full activity and virulence in infection models. Given that lactate levels typically increase during infection, our results suggest that the pathogen can use protein lactylation as a mechanism to increase toxin-mediated virulence during infection.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanfeng Liu
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guoxiu Xiang
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Jian
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ziyu Yang
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tianchi Chen
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowei Ma
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Na Zhao
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingxin Dai
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Lv
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Wang
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei He
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bisheng Shi
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Liu
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yao Liu
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Min Li
- Department of Laboratory Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Faculty of Medical Laboratory Science, College of Health Science and Technology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
22
|
Ke S, Kil H, Roggy C, Shields T, Quinn Z, Quinn AP, Small JM, Towne FD, Brooks AE, Brooks BD. Potential Therapeutic Targets for Combination Antibody Therapy Against Staphylococcus aureus Infections. Antibiotics (Basel) 2024; 13:1046. [PMID: 39596740 PMCID: PMC11591076 DOI: 10.3390/antibiotics13111046] [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: 10/08/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024] Open
Abstract
Despite the significant advances in antibiotic treatments and therapeutics, Staphylococcus aureus (S. aureus) remains a formidable pathogen, primarily due to its rapid acquisition of antibiotic resistance. Known for its array of virulence factors, including surface proteins that promote adhesion to host tissues, enzymes that break down host barriers, and toxins that contribute to immune evasion and tissue destruction, S. aureus poses a serious health threat. Both the Centers for Disease Control and Prevention (CDC) and the World Health Organization (WHO) classify S. aureus as an ESKAPE pathogen, recognizing it as a critical threat to global health. The increasing prevalence of drug-resistant S. aureus underscores the need for new therapeutic strategies. This review discusses a promising approach that combines monoclonal antibodies targeting multiple S. aureus epitopes, offering synergistic efficacy in treating infections. Such strategies aim to reduce the capacity of the pathogen to develop resistance, presenting a potent adjunct or alternative to conventional antibiotic treatments.
Collapse
Affiliation(s)
- Sharon Ke
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Hyein Kil
- Department of Surgery, Virtua Health, Camden, NJ 08103, USA
| | - Conner Roggy
- Department of Orthopaedic Surgery, Community Memorial Healthcare, Ventura, CA 93003, USA
| | - Ty Shields
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Zachary Quinn
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Alyssa P. Quinn
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - James M. Small
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Francina D. Towne
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Amanda E. Brooks
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Benjamin D. Brooks
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| |
Collapse
|
23
|
Bhattacharya M, Spencer BL, Kwiecinski JM, Podkowik M, Putzel G, Pironti A, Shopsin B, Doran KS, Horswill AR. Collagen binding adhesin restricts Staphylococcus aureus skin infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621145. [PMID: 39554114 PMCID: PMC11565922 DOI: 10.1101/2024.11.01.621145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Staphylococcus aureus causes approximately 80% of skin and soft tissue infections (SSTIs). Collagen is the most abundant human extracellular matrix protein with critical roles in wound healing, and S. aureus encodes a collagen binding adhesin (Cna). The role of this protein during skin infections is unknown. Here we report that inability to bind collagen results in worsened pathology of intradermal Δcna S. aureus infection. WT/Cna+ S. aureus showed reduced infection severity, aggregate formation, and significantly improved clearance of bacteria. Cna binds to the collagen-like domain of serum C1q protein to reduce its opsonophagocytic functions. We demonstrate that infection of C1qKO mice with WT bacteria show results similar to the Δcna group. Conversely, inability to bind collagen resulted in an amplified inflammatory response caused in part by macrophage and neutrophil small molecule mediators released at the infection site (MMP-9, MMP-12, LTB4), resulting in increased immune cell infiltration and death.
Collapse
Affiliation(s)
- Mohini Bhattacharya
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora CO, USA
| | - Brady L. Spencer
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora CO, USA
| | - Jakub M. Kwiecinski
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Podkowik
- Department of Medicine, Division of Infectious Diseases and Immunology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Gregory Putzel
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases and Immunology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Kelly S. Doran
- 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, Denver, CO, USA
- Lead author
| |
Collapse
|
24
|
Wang X, Lee JC. Staphylococcus aureus membrane vesicles: an evolving story. Trends Microbiol 2024; 32:1096-1105. [PMID: 38677977 PMCID: PMC11511790 DOI: 10.1016/j.tim.2024.04.003] [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: 12/15/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024]
Abstract
Staphylococcus aureus is an important bacterial pathogen that causes a wide variety of human diseases in community and hospital settings. S. aureus employs a diverse array of virulence factors, both surface-associated and secreted, to promote colonization, infection, and immune evasion. Over the past decade, a growing body of research has shown that S. aureus generates extracellular membrane vesicles (MVs) that package a variety of bacterial components, many of which are virulence factors. In this review, we summarize recent advances in our understanding of S. aureus MVs and highlight their biogenesis, cargo, and potential role in the pathogenesis of staphylococcal infections. Lastly, we present some emerging questions in the field.
Collapse
Affiliation(s)
- Xiaogang Wang
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA.
| | - Jean C Lee
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
25
|
Sujith S, Naresh R, Srivisanth BU, Sajeevan A, Rajaramon S, David H, Solomon AP. Aptamers: precision tools for diagnosing and treating infectious diseases. Front Cell Infect Microbiol 2024; 14:1402932. [PMID: 39386170 PMCID: PMC11461471 DOI: 10.3389/fcimb.2024.1402932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Infectious diseases represent a significant global health challenge, with bacteria, fungi, viruses, and parasitic protozoa being significant causative agents. The shared symptoms among diseases and the emergence of new pathogen variations make diagnosis and treatment complex. Conventional diagnostic methods are laborious and intricate, underscoring the need for rapid, accurate techniques. Aptamer-based technologies offer a promising solution, as they are cost-effective, sensitive, specific, and convenient for molecular disease diagnosis. Aptamers, which are single-stranded RNA or DNA sequences, serve as nucleotide equivalents of monoclonal antibodies, displaying high specificity and affinity for target molecules. They are structurally robust, allowing for long-term storage without substantial activity loss. Aptamers find applications in diverse fields such as drug screening, material science, and environmental monitoring. In biomedicine, they are extensively studied for biomarker detection, diagnostics, imaging, and targeted therapy. This comprehensive review focuses on the utility of aptamers in managing infectious diseases, particularly in the realms of diagnostics and therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
26
|
Fang L, Cosgriff C, Alonzo F. Determinants of maturation of the Staphylococcus aureus autoinducing peptide. J Bacteriol 2024; 206:e0019524. [PMID: 39177535 PMCID: PMC11412329 DOI: 10.1128/jb.00195-24] [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: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
The accessory gene regulatory (Agr) system is required for virulence factor gene expression and pathogenesis of Staphylococcus aureus. The Agr system is activated in response to the accumulation of a cyclic autoinducing peptide (AIP), which is matured and secreted by the bacterium. The precursor of AIP, AgrD, consists of the AIP flanked by an N-terminal [Formula: see text]-helical Leader and a charged C-terminal tail. AgrD is matured to AIP by the action of two proteases, AgrB and MroQ. AgrB cleaves the C-terminal tail and promotes the formation of a thiolactone ring, whereas MroQ cleaves the N-terminal Leader in a manner that depends on the four-amino acid linker immediately following a conserved IG helix breaker motif. However, the attributes of AgrD that dictate the sequence of events in peptide maturation are not fully defined. Here, we used engineered AgrD peptide intermediates to ascertain the sufficiency of MroQ for N-terminal peptide cleavage, peptide export, and generation of mature AIP. We found that MroQ promotes the removal of the N-terminal Leader peptide from both linear and cyclic peptide intermediates, while peptide cyclization remained essential for signaling. The expression of the Leader peptide in isolation was sufficient for MroQ-dependent cleavage proximal to the four-amino-acid linker. In addition, active site mutations within AgrB destabilized full-length AgrD and thiolactone-containing intermediates and prevented the release of the Leader peptide. Altogether, our data support a tandem peptide maturation event involving both MroQ and AgrB that appears to couple protease activity and export of bioactive AIP.IMPORTANCEThe accessory gene regulatory (Agr) system is important for S. aureus pathogenesis. Activation of the Agr system requires recognition of a cyclic peptide pheromone, which must be fully matured to exert its biological activity. The complete events in cyclic peptide maturation and export from the bacterial cell remain to be fully defined. We and others recently discovered that the membrane peptidase MroQ is required for pheromone maturation. This study builds off the identification of MroQ and considers the attributes of the pheromone pro-peptide that are required for MroQ-mediated processing as well as uncovers features important for peptide stability and export. Overall, the findings in this study have implications for understanding bacterial pheromone maturation and virulence.
Collapse
Affiliation(s)
- Liwei Fang
- Department of
Microbiology and Immunology, University of
Illinois, Chicago,
Illinois, USA
| | - Chance Cosgriff
- Department of
Microbiology and Immunology, Loyola University Chicago Stritch School of
Medicine, Maywood,
Illinois, USA
| | - Francis Alonzo
- Department of
Microbiology and Immunology, University of
Illinois, Chicago,
Illinois, USA
| |
Collapse
|
27
|
Piechowicz L, Kosznik-Kwaśnicka K, Jarzembowski T, Daca A, Necel A, Bonawenturczak A, Werbowy O, Stasiłojć M, Pałubicka A. Staphylococcus aureus Co-Infection in COVID-19 Patients: Virulence Genes and Their Influence on Respiratory Epithelial Cells in Light of Risk of Severe Secondary Infection. Int J Mol Sci 2024; 25:10050. [PMID: 39337536 PMCID: PMC11431965 DOI: 10.3390/ijms251810050] [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: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Pandemics from viral respiratory tract infections in the 20th and early 21st centuries were associated with high mortality, which was not always caused by a primary viral infection. It has been observed that severe course of infection, complications and mortality were often the result of co-infection with other pathogens, especially Staphylococcus aureus. During the COVID-19 pandemic, it was also noticed that patients infected with S. aureus had a significantly higher mortality rate (61.7%) compared to patients infected with SARS-CoV-2 alone. Our previous studies have shown that S. aureus strains isolated from patients with COVID-19 had a different protein profile than the strains in non-COVID-19 patients. Therefore, this study aims to analyze S. aureus strains isolated from COVID-19 patients in terms of their pathogenicity by analyzing their virulence genes, adhesion, cytotoxicity and penetration to the human pulmonary epithelial cell line A549. We have observed that half of the tested S. aureus strains isolated from patients with COVID-19 had a necrotizing effect on the A549 cells. The strains also showed greater variability in terms of their adhesion to the human cells than their non-COVID-19 counterparts.
Collapse
Affiliation(s)
- Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Katarzyna Kosznik-Kwaśnicka
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Tomasz Jarzembowski
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Agnieszka Daca
- Department of Physiopathology, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Agnieszka Necel
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Ada Bonawenturczak
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Olesia Werbowy
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Małgorzata Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland
| | - Anna Pałubicka
- Specialist Hospital in Koscierzyna Sp. z o.o., Department of Laboratory and Microbiological Diagnostics, Koscierzyna, Alojzego Piechowskiego 36, 83-400 Koscierzyna, Poland
| |
Collapse
|
28
|
Crepin DM, Chavignon M, Verhoeven PO, Laurent F, Josse J, Butin M. Staphylococcus capitis: insights into epidemiology, virulence, and antimicrobial resistance of a clinically relevant bacterial species. Clin Microbiol Rev 2024; 37:e0011823. [PMID: 38899876 PMCID: PMC11391707 DOI: 10.1128/cmr.00118-23] [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] [Indexed: 06/21/2024] Open
Abstract
SUMMARYStaphylococcus capitis is divided into two subspecies, S. capitis subsp. ureolyticus (renamed urealyticus in 1992; ATCC 49326) and S. capitis subsp. capitis (ATCC 27840), and fits with the archetype of clinically relevant coagulase-negative staphylococci (CoNS). S. capitis is a commensal bacterium of the skin in humans, which must be considered an opportunistic pathogen of interest particularly as soon as it is identified in a clinically relevant specimen from an immunocompromised patient. Several studies have highlighted the potential determinants underlying S. capitis pathogenicity, resistance profiles, and virulence factors. In addition, mobile genetic element acquisitions and mutations contribute to S. capitis genome adaptation to its environment. Over the past decades, antibiotic resistance has been identified for S. capitis in almost all the families of the currently available antibiotics and is related to the emergence of multidrug-resistant clones of high clinical significance. The present review summarizes the current knowledge concerning the taxonomic position of S. capitis among staphylococci, the involvement of this species in human colonization and diseases, the virulence factors supporting its pathogenicity, and the phenotypic and genomic antimicrobial resistance profiles of this species.
Collapse
Affiliation(s)
- Deborah M Crepin
- CIRI, Centre International de Recherche en Infectiologie, Staphylococcal pathogenesis team, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR 5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Marie Chavignon
- CIRI, Centre International de Recherche en Infectiologie, Staphylococcal pathogenesis team, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR 5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Paul O Verhoeven
- CIRI, Centre International de Recherche en Infectiologie, GIMAP Team, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR 5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Faculté de Médecine, Université Jean Monnet, St-Etienne, France
- Service des agents infectieux et d'hygiène, Centre Hospitalier Universitaire de St-Etienne, St-Etienne, France
| | - Frédéric Laurent
- CIRI, Centre International de Recherche en Infectiologie, Staphylococcal pathogenesis team, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR 5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Josse
- CIRI, Centre International de Recherche en Infectiologie, Staphylococcal pathogenesis team, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR 5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Marine Butin
- CIRI, Centre International de Recherche en Infectiologie, Staphylococcal pathogenesis team, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR 5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Service de Néonatologie et Réanimation Néonatale, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| |
Collapse
|
29
|
Odunitan TT, Apanisile BT, Akinboade MW, Abdulazeez WO, Oyaronbi AO, Ajayi TM, Oyekola SA, Ibrahim NO, Nafiu T, Afolabi HO, Olayiwola DM, David OT, Adeyemo SF, Ayodeji OD, Akinade EM, Saibu OA. Microbial mysteries: Staphylococcus aureus and the enigma of carcinogenesis. Microb Pathog 2024; 194:106831. [PMID: 39089512 DOI: 10.1016/j.micpath.2024.106831] [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: 12/30/2023] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Staphylococcus aureus, a common human pathogen, has long been the focus of scientific investigation due to its association with various infections. However, recent research has unveiled a tantalizing enigma surrounding this bacterium and its potential involvement in carcinogenesis. Chronic S. aureus infections have been linked to an elevated risk of certain cancers, including skin cancer and oral cancer. This review explores the current state of knowledge regarding this connection, examining epidemiological evidence, pathogenic mechanisms, and biological interactions that suggest a correlation. Although initial studies point to a possible link, the precise mechanisms through which S. aureus may contribute to cancer development remain elusive. Emerging evidence suggests that the chronic inflammation induced by persistent S. aureus infections may create a tumor-promoting environment. This inflammation can lead to DNA damage, disrupt cellular signaling pathways, and generate an immunosuppressive microenvironment conducive to cancer progression. Additionally, S. aureus produces a variety of toxins and metabolites that can directly interact with host cells, potentially inducing oncogenic transformations. Despite these insights, significant gaps remain in our understanding of the exact biological processes involved. This review emphasizes the urgent need for more comprehensive research to clarify these microbiological mysteries. Understanding the role of S. aureus in cancer development could lead to novel strategies for cancer prevention and treatment, potentially transforming therapeutic approaches.
Collapse
Affiliation(s)
- Tope T Odunitan
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria; Microbiology Unit, Helix Biogen Institute, Ogbomosho, Oyo State, Nigeria; Ehigie's Biochemistry and Biocomputational Laboratory, Ogbomosho, Oyo State, Nigeria.
| | - Boluwatife T Apanisile
- Department of Nutrition and Dietetics, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Modinat W Akinboade
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Waliu O Abdulazeez
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Adegboye O Oyaronbi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Temitope M Ajayi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Samuel A Oyekola
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Najahtulahi O Ibrahim
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Tawakalitu Nafiu
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Hezekiah O Afolabi
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Dolapo M Olayiwola
- Department of Medical Laboratory Science, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Oladunni T David
- Microbiology Unit, Helix Biogen Institute, Ogbomosho, Oyo State, Nigeria
| | - Stephen F Adeyemo
- Department of Biological Sciences, First Technical University, Ibadan, Oyo State, Nigeria; Division of Medical Artificial Intelligence, Helix Biogen Institute, Ogbomosho, Oyo State, Nigeria
| | - Oluwatobi D Ayodeji
- Department of Nursing, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Esther M Akinade
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Oluwatosin A Saibu
- Department of Chemistry and Biochemistry, New Mexico State University, USA
| |
Collapse
|
30
|
Licht P, Dominelli N, Kleemann J, Pastore S, Müller ES, Haist M, Hartmann KS, Stege H, Bros M, Meissner M, Grabbe S, Heermann R, Mailänder V. The skin microbiome stratifies patients with cutaneous T cell lymphoma and determines event-free survival. NPJ Biofilms Microbiomes 2024; 10:74. [PMID: 39198450 PMCID: PMC11358159 DOI: 10.1038/s41522-024-00542-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
Mycosis fungoides (MF) is the most common entity of Cutaneous T cell lymphomas (CTCL) and is characterized by the presence of clonal malignant T cells in the skin. The role of the skin microbiome for MF development and progression are currently poorly understood. Using shotgun metagenomic profiling, real-time qPCR, and T cell receptor sequencing, we compared lesional and nonlesional skin of 20 MF patients with early and advanced MF. Additionally, we isolated Staphylococcus aureus and other bacteria from MF skin for functional profiling and to study the S. aureus virulence factor spa. We identified a subgroup of MF patients with substantial dysbiosis on MF lesions and concomitant outgrowth of S. aureus on plaque-staged lesions, while the other MF patients had a balanced microbiome on lesional skin. Dysbiosis and S. aureus outgrowth were accompanied by ectopic levels of cutaneous antimicrobial peptides (AMPs), including adaptation of the plaque-derived S. aureus strain. Furthermore, the plaque-derived S. aureus strain showed a reduced susceptibility towards antibiotics and an upregulation of the virulence factor spa, which may activate the NF-κB pathway. Remarkably, patients with dysbiosis on MF lesions had a restricted T cell receptor repertoire and significantly lower event-free survival. Our study highlights the potential for microbiome-modulating treatments targeting S. aureus to prevent MF progression.
Collapse
Affiliation(s)
- Philipp Licht
- University Medical Centre Mainz, Department of Dermatology, Mainz, Germany.
| | - Nazzareno Dominelli
- Johannes Gutenberg-University, Institute of Molecular Physiology (imP), Biocenter II, Microbiology and Biotechnology, Mainz, Germany
| | - Johannes Kleemann
- University Hospital Frankfurt, Department of Dermatology, Venerology and Allergology, Frankfurt am Main, Germany
| | - Stefan Pastore
- University Medical Centre Mainz, Institute of Human Genetics, Mainz, Germany
- Johannes Gutenberg-University, Institute of Pharmaceutical and Biomedical Sciences, Mainz, Germany
| | - Elena-Sophia Müller
- Johannes Gutenberg-University, Institute of Molecular Physiology (imP), Biocenter II, Microbiology and Biotechnology, Mainz, Germany
| | - Maximilian Haist
- University Medical Centre Mainz, Department of Dermatology, Mainz, Germany
| | | | - Henner Stege
- University Medical Centre Mainz, Department of Dermatology, Mainz, Germany
| | - Matthias Bros
- University Medical Centre Mainz, Department of Dermatology, Mainz, Germany
| | - Markus Meissner
- University Hospital Frankfurt, Department of Dermatology, Venerology and Allergology, Frankfurt am Main, Germany
| | - Stephan Grabbe
- University Medical Centre Mainz, Department of Dermatology, Mainz, Germany
| | - Ralf Heermann
- Johannes Gutenberg-University, Institute of Molecular Physiology (imP), Biocenter II, Microbiology and Biotechnology, Mainz, Germany
| | - Volker Mailänder
- University Medical Centre Mainz, Department of Dermatology, Mainz, Germany.
- Max Planck Institute for Polymer Research, Mainz, Germany.
| |
Collapse
|
31
|
Koc F, Magner C, Murphy K, Kelleher ST, Tan MH, O'Toole M, Jenkins D, Boyle J, Lavelle M, Maguire N, Ross PR, Stanton C, McMahon CJ. Gut Microbiome in Children with Congenital Heart Disease After Cardiopulmonary Bypass Surgery (GuMiBear Study). Pediatr Cardiol 2024:10.1007/s00246-024-03634-2. [PMID: 39174731 DOI: 10.1007/s00246-024-03634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024]
Abstract
The gut microbiome of infants with congenital heart disease (CHD) undergoing cardiopulmonary bypass surgery (CPB) is at risk of profound alteration. The aim of this study was to examine the gut microbiome pre- and post-bypass surgery to explore potential implications of altered gut biodiversity. A prospective cohort study involving infants with CHD who underwent CPB was performed. Faecal samples were collected from infants alongside the collection of demographic and clinical data in order to examine gut microbiome changes before and after surgery. 16S rRNA sequencing analysis was performed on DNA isolated from stool samples to determine changes in gut microbiome composition. Thirty-three patients were recruited, with samples from thirteen of these available for final analysis. Compared with healthy, matched controls, at a genus level, pre-operative samples for infants with CHD demonstrated a higher relative abundance of Escherichia-Shigella (31% vs 2-6%) and a lower relative abundance of Bifidobacterium (13% vs 40-60%). In post-operative samples, the relative abundance of Escherichia-Shigella (35%), Enterococcus (11%), Akkermansia (6%), and Staphylococcus (5%) were higher than pre-op samples. One infant developed post-operative necrotising-enterocolitis (NEC). They displayed a marked abundance of the Enterococcus (93%) genus pre-operatively. This study demonstrates that infants with CHD have an altered gut microbiome when compared with healthy controls and there might be a possible link between an abundance of virulent species and NEC.
Collapse
Affiliation(s)
- Fatma Koc
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Claire Magner
- School of Nursing, Midwifery and Health Systems, University College Dublin, Dublin, Ireland
| | - Kiera Murphy
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Ireland
| | - Sean T Kelleher
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Mong H Tan
- Paediatric Intensive Care Unit, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Molly O'Toole
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Dominic Jenkins
- Laboratory, Children's Health Ireland at Crumlin, Crumlin, Ireland
| | - Jordan Boyle
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Marie Lavelle
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Niamh Maguire
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Paul R Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Colin J McMahon
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland.
- School of Medicine, University College Dublin, Dublin, Ireland.
- School of Health Professions Education (SHE), Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
32
|
Hao L, Zhou J, Yang H, He C, Shu W, Song H, Liu Q. Anti-virulence potential of iclaprim, a novel folic acid synthesis inhibitor, against Staphylococcus aureus. Appl Microbiol Biotechnol 2024; 108:432. [PMID: 39102054 DOI: 10.1007/s00253-024-13268-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024]
Abstract
Infections caused by Staphylococcus aureus pose a significant global public problem. Therefore, new antibiotics and therapeutic strategies are needed to combat this pathogen. This investigation delves into the effects of iclaprim, a newly discovered inhibitor of folic acid synthesis, on S. aureus virulence. The phenotypic and genotypic effects of iclaprim were thoroughly examined in relation to virulence factors, biofilm formation, and dispersal, as well as partial virulence-encoding genes associated with exoproteins, adherence, and regulation in S. aureus MW2, N315, and ATCC 25923. Then, the in vivo effectiveness of iclaprim on S. aureus pathogenicity was explored by a Galleria mellonella larvae infection model. The use of iclaprim at sub-inhibitory concentrations (sub-MICs) resulted in a reduction of α-hemolysin (Hla) production and a differential effect on the activity of coagulase in S. aureus strains. The results of biofilm formation and eradication assay showed that iclaprim was highly effective in depolymerizing the mature biofilm of S. aureus strains at concentrations of 1 MIC or greater, however, inhibited the biofilm-forming ability of only strains N315 and ATCC 25923 at sub-MICs. Interestingly, treatment of strains with sub-MICs of iclaprim resulted in significant stimulation or suppression of most virulence-encoding genes expression. Iclaprim did not affect the production of δ-hemolysin or staphylococcal protein A (SpA), nor did it impact the total activity of proteases, nucleases, and lipases. In vivo testing showed that sub-MICs of iclaprim significantly improves infected larvae survival. The present study offered valuable insights towards a better understating of the influence of iclaprim on different strains of S. aureus. The findings suggest that iclaprim may have potential as an anti-virulence and antibiofilm agent, thus potentially mitigating the pathogenicity of S. aureus and improving clinical outcomes associated with infections caused by this pathogen. KEY POINTS: • Iclaprim effectively inhibits α-hemolysin production and biofilm formation in a strain-dependent manner and was an excellent depolymerizing agent of mature biofilm • Iclaprim affected the mRNA expression of virulence-encoding genes associated with exoproteins, adherence, and regulation • In vivo study in G. mellonella larvae challenged with S. aureus exhibited that iclaprim improves larvae survival.
Collapse
Affiliation(s)
- Lingyun Hao
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingwen Zhou
- Department of Clinical Laboratory, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Rd., Shanghai, 200071, China
| | - Han Yang
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chunyan He
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wen Shu
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haoyue Song
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qingzhong Liu
- Department of Clinical Laboratory, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Rd., Shanghai, 200071, China.
| |
Collapse
|
33
|
Kothe CI, Carøe C, Mazel F, Zilber D, Cruz-Morales P, Mohellibi N, Evans JD. Novel misos shape distinct microbial ecologies: opportunities for flavourful sustainable food innovation. Food Res Int 2024; 189:114490. [PMID: 38876584 DOI: 10.1016/j.foodres.2024.114490] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 06/16/2024]
Abstract
Fermentation is resurgent around the world as people seek healthier, more sustainable, and tasty food options. This study explores the microbial ecology of miso, a traditional Japanese fermented paste, made with novel regional substrates to develop new plant-based foods. Eight novel miso varieties were developed using different protein-rich substrates: yellow peas, Gotland lentils, and fava beans (each with two treatments: standard and nixtamalisation), as well as rye bread and soybeans. The misos were produced at Noma, a restaurant in Copenhagen, Denmark. Samples were analysed with biological and technical triplicates at the beginning and end of fermentation. We also incorporated in this study six samples of novel misos produced following the same recipe at Inua, a former affiliate restaurant of Noma in Tokyo, Japan. To analyse microbial community structure and diversity, metabarcoding (16S and ITS) and shotgun metagenomic analyses were performed. The misos contain a greater range of microbes than is currently described for miso in the literature. The composition of the novel yellow pea misos was notably similar to the traditional soybean ones, suggesting they are a good alternative, which supports our culinary collaborators' sensory conclusions. For bacteria, we found that overall substrate had the strongest effect, followed by time, treatment (nixtamalisation), and geography. For fungi, there was a slightly stronger effect of geography and a mild effect of substrate, and no significant effects for treatment or time. Based on an analysis of metagenome-assembled genomes (MAGs), strains of Staphylococccus epidermidis differentiated according to substrate. Carotenoid biosynthesis genes in these MAGs appeared in strains from Japan but not from Denmark, suggesting a possible gene-level geographical effect. The benign and possibly functional presence of S. epidermidis in these misos, a species typically associated with the human skin microbiome, suggests possible adaptation to the miso niche, and the flow of microbes between bodies and foods in certain fermentation as more common than is currently recognised. This study improves our understanding of miso ecology, highlights the potential for developing novel misos using diverse local ingredients, and suggests how fermentation innovation can contribute to studies of microbial ecology and evolution.
Collapse
Affiliation(s)
- Caroline Isabel Kothe
- Sustainable Food Innovation Group, The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark.
| | | | - Florent Mazel
- Department of Fundamental Microbiology, University of Lausanne, Switzerland
| | - David Zilber
- Novonesis, Hørsholm, Denmark; Restaurant Noma, Copenhagen, Denmark
| | - Pablo Cruz-Morales
- Yeast Natural Products, The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark
| | - Nacer Mohellibi
- Université Paris-Saclay, INRAE, Micalis Institute, Jouy-en-Josas, France
| | - Joshua D Evans
- Sustainable Food Innovation Group, The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark.
| |
Collapse
|
34
|
Wen Z, Chen C, Shang Y, Fan K, Li P, Li C, Zheng J, Deng Q, Yu Z. Baohuoside I inhibits virulence of multidrug-resistant Staphylococcus aureus by targeting the transcription Staphylococcus accessory regulator factor SarZ. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155590. [PMID: 38810547 DOI: 10.1016/j.phymed.2024.155590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Staphylococcus aureus is a versatile pathogen that can cause a wide range of infections in humans. Biofilms play a crucial role in the pathogenicity of S. aureus and contribute to its ability to cause persistent and chronic infections. Baohuoside I has garnered increasing recognition as a natural flavonol glycoside with a wide spectrum of health-related activities. PURPOSE The antibacterial and anti-biofilm properties of Baohuoside I have not been extensively investigated. Our study aimed to assess its inhibitory effects and the underlying mechanisms on biofilm formation and hemolytic capacity in S. aureus. STUDY DESIGN/METHODS The impact of Baohuoside I on the biofilm and virulence of S. aureus was evaluated through in vitro experiments and Galleria mellonella as an in vivo infection model. The mechanisms were explored by Drug affinity responsive target stability (DARTS) and validated in genetic knockout strain and through molecular biological experiments using DARTS, molecular docking, electrophoretic mobility shift assay (EMSA), and bio-layer interferometry (BLI). RESULTS Baohuoside I significantly inhibits the formation of S. aureus biofilms and hemolytic activity at 6.25 µM. Proteomics analysis revealed that treatment with Baohuoside I led to a reduction in the expression of quorum-sensing system agr-regulated genes. DARTS analysis identified Staphylococcus accessory regulator factor (SarZ), a key regulator involved in the expression of virulence factors in S. aureus by acting as activator of the agr quorum-sensing system, was the direct target of Baohuoside I. Molecular docking, DARTS, BLI and EMSA assays collectively confirmed the direct binding of Baohuoside I to SarZ, inhibiting its binding to downstream promoters. Furthermore, it is found through site-directed protein mutagenesis that the Tyr27 and Phe117 residues are key for Baohuoside I binding to SarZ. Additionally, the knockout of SarZ significantly diminished the hemolytic ability of S. aureus, underscoring its crucial role as a pivotal regulator of virulence. Lastly, in vivo tests utilizing the G. mellonella infection model demonstrated the efficacy of Baohuoside I. CONCLUSION This study provides valuable insights into the mechanism by which Baohuoside I inhibits the virulence of S. aureus through its interaction with SarZ. These findings highlight the significance of SarZ as an effective target against the virulence of S. aureus.
Collapse
Affiliation(s)
- Zewen Wen
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Chengchun Chen
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Yongpeng Shang
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Kewei Fan
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Peiyu Li
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Congcong Li
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Jinxin Zheng
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Qiwen Deng
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China.
| | - Zhijian Yu
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Huazhong University of Science and Technology Union Shenzhen Hospital, No. 89 Taoyuan Road, Nanshan District, Shenzhen 518052, China.
| |
Collapse
|
35
|
Barber MF, Fitzgerald JR. Mechanisms of host adaptation by bacterial pathogens. FEMS Microbiol Rev 2024; 48:fuae019. [PMID: 39003250 PMCID: PMC11308195 DOI: 10.1093/femsre/fuae019] [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: 04/15/2024] [Revised: 07/02/2024] [Accepted: 07/24/2024] [Indexed: 07/15/2024] Open
Abstract
The emergence of new infectious diseases poses a major threat to humans, animals, and broader ecosystems. Defining factors that govern the ability of pathogens to adapt to new host species is therefore a crucial research imperative. Pathogenic bacteria are of particular concern, given dwindling treatment options amid the continued expansion of antimicrobial resistance. In this review, we summarize recent advancements in the understanding of bacterial host species adaptation, with an emphasis on pathogens of humans and related mammals. We focus particularly on molecular mechanisms underlying key steps of bacterial host adaptation including colonization, nutrient acquisition, and immune evasion, as well as suggest key areas for future investigation. By developing a greater understanding of the mechanisms of host adaptation in pathogenic bacteria, we may uncover new strategies to target these microbes for the treatment and prevention of infectious diseases in humans, animals, and the broader environment.
Collapse
Affiliation(s)
- Matthew F Barber
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, United States
- Department of Biology, University of Oregon, Eugene, OR 97403, United States
| | - J Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, Midlothian, EH25 9RG, United Kingdom
| |
Collapse
|
36
|
Piewngam P, Otto M. Staphylococcus aureus colonisation and strategies for decolonisation. THE LANCET. MICROBE 2024; 5:e606-e618. [PMID: 38518792 PMCID: PMC11162333 DOI: 10.1016/s2666-5247(24)00040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 03/24/2024]
Abstract
Staphylococcus aureus is a leading cause of death by infectious diseases worldwide. Treatment of S aureus infections is difficult due to widespread antibiotic resistance, necessitating alternative approaches and measures for prevention of infection. Because S aureus infections commonly arise from asymptomatic colonisation, decolonisation is considered a key approach for their prevention. Current decolonisation procedures include antibiotic-based and antiseptic-based eradication of S aureus from the nose and skin. However, despite the widespread implementation and partial success of such measures, S aureus infection rates remain worrisome, and resistance to decolonisation agents is on the rise. In this Review we outline the epidemiology and mechanisms of S aureus colonisation, describe how colonisation underlies infection, and discuss current and novel approaches for S aureus decolonisation, with a focus on the latest findings on probiotic strategies and the intestinal S aureus colonisation site.
Collapse
Affiliation(s)
- Pipat Piewngam
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
37
|
Lichota A, Gwozdzinski K, Kowalczyk E, Kowalczyk M, Sienkiewicz M. Contribution of staphylococcal virulence factors in the pathogenesis of thrombosis. Microbiol Res 2024; 283:127703. [PMID: 38537329 DOI: 10.1016/j.micres.2024.127703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024]
Abstract
Staphylococci are responsible for many infections in humans, starting with skin and soft tissue infections and finishing with invasive diseases such as endocarditis, sepsis and pneumonia, which lead to high mortality. Patients with sepsis often demonstrate activated clotting pathways, decreased levels of anticoagulants, decreased fibrinolysis, activated endothelial surfaces and activated platelets. This results in disseminated intravascular coagulation and formation of a microthrombus, which can lead to a multiorgan failure. This review describes various staphylococcal virulence factors that contribute to vascular thrombosis, including deep vein thrombosis in infected patients. The article presents mechanisms of action of different factors released by bacteria in various host defense lines, which in turn can lead to formation of blood clots in the vessels.
Collapse
Affiliation(s)
- Anna Lichota
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland.
| | | | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Lodz, Poland
| | | | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
38
|
Chauhan R, Nate Z, Ike B, Kwabena Adu D, Alake J, Gill AAS, Miya L, Bachheti Thapliyal N, Karpoormath R. One pot fabrication of diamino naphthalene -AuNPs decorated graphene nanoplatform for the MRSA detection in the biological sample. Bioelectrochemistry 2024; 157:108674. [PMID: 38460467 DOI: 10.1016/j.bioelechem.2024.108674] [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: 10/23/2023] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 03/11/2024]
Abstract
Early monitoring of MRSA can effectively mitigate the disease risk by using Penicillin-binding protein 2a (PbP2a) biomarker. Diamino naphthalene-AuNPs decorated graphene (AuNPsGO-DN) nanocomposite was synthesized for a rapid and sensitive immunosensor detecting PbP2a. The synthesized AuNPsGO-DN nanocomposites were characterized by field emission scanning electron microscopy (FE-SEM), energy-dispersive X-ray spectroscopy (SEM-EDX), Fourier transform infrared spectroscopy (FT-IR), Raman spectroscopy, and X-ray diffraction spectroscopy (XRD). Electrochemical characterization done with cyclic voltammetry (CV), differential pulse voltammetry (DPV), and electrical impedance spectroscopy (EIS) techniques. Anti-PbP2a monoclonal antibodies immobilized at AuNPsGO-DN/GCE via covalent bonding. AuNPs enhanced the electrode surface area and the antibodies' loading. Mercaptopropionic acid (MPA) was a linker between the AuNPs and antibodies, orientated the antibodies as opposite to the PbP2a antigen, and improved the sensitivity and specificity. The antiPbP2a/MPA/AuNPsGO-DN/GCE electrode displayed sensitive and selective detection towards the PbP2a antigen in phosphate buffer saline (PBS pH 7.4). The broad linear range from 0.01 to 8000 pg/mL was obtained with LOD of 0.154 pg/mL and 0.0239 pg/mL, respectively. A label-free, simple, and sensitive immunosensor was developed with a 98-106 % recovery rate in spiked biological samples. It shows the potential applicability of the developed immunoelectrode.
Collapse
Affiliation(s)
- Ruchika Chauhan
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Zondi Nate
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Blessing Ike
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Darko Kwabena Adu
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - John Alake
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Atal A S Gill
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Lungelo Miya
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Neeta Bachheti Thapliyal
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Rajshekhar Karpoormath
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa.
| |
Collapse
|
39
|
Garrine M, Andrade M, Neves J, Mandomando I, Couto I, Costa SS. Exploring the virulence potential of Staphylococcus aureus CC121 and CC152 lineages related to paediatric community-acquired bacteraemia in Manhiça, Mozambique. Sci Rep 2024; 14:10758. [PMID: 38730020 PMCID: PMC11087594 DOI: 10.1038/s41598-024-61345-3] [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: 12/19/2023] [Accepted: 05/05/2024] [Indexed: 05/12/2024] Open
Abstract
Staphylococcus aureus is a frequent agent of bacteraemia. This bacterium has a variety of virulence traits that allow the establishment and maintenance of infection. This study explored the virulence profile of S. aureus strains causing paediatric bacteraemia (SAB) in Manhiça district, Mozambique. We analysed 336 S. aureus strains isolated from blood cultures of children younger than 5 years admitted to the Manhiça District Hospital between 2001 and 2019, previously characterized for antibiotic susceptibility and clonality. The strains virulence potential was evaluated by PCR detection of the Panton-Valentine leucocidin (PVL) encoding genes, lukS-PV/lukF-PV, assessment of the capacity for biofilm formation and pathogenicity assays in Galleria mellonella. The overall carriage of PVL-encoding genes was over 40%, although reaching ~ 70 to 100% in the last years (2014 to 2019), potentially linked to the emergence of CC152 lineage. Strong biofilm production was a frequent trait of CC152 strains. Representative CC152 and CC121 strains showed higher virulence potential in the G. mellonella model when compared to reference strains, with variations within and between CCs. Our results highlight the importance of monitoring the emergent CC152-MSSA-PVL+ and other lineages, as they display important virulence traits that may negatively impact the management of SAB paediatric patients in Manhiça district, Mozambique.
Collapse
Affiliation(s)
- Marcelino Garrine
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Mariana Andrade
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Joana Neves
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Inácio Mandomando
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
- Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
- ISGlobal-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Isabel Couto
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Sofia Santos Costa
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal.
| |
Collapse
|
40
|
Léguillier V, Heddi B, Vidic J. Recent Advances in Aptamer-Based Biosensors for Bacterial Detection. BIOSENSORS 2024; 14:210. [PMID: 38785684 PMCID: PMC11117931 DOI: 10.3390/bios14050210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024]
Abstract
The rapid and sensitive detection of pathogenic bacteria is becoming increasingly important for the timely prevention of contamination and the treatment of infections. Biosensors based on nucleic acid aptamers, integrated with optical, electrochemical, and mass-sensitive analytical techniques, have garnered intense interest because of their versatility, cost-efficiency, and ability to exhibit high affinity and specificity in binding bacterial biomarkers, toxins, and whole cells. This review highlights the development of aptamers, their structural characterization, and the chemical modifications enabling optimized recognition properties and enhanced stability in complex biological matrices. Furthermore, recent examples of aptasensors for the detection of bacterial cells, biomarkers, and toxins are discussed. Finally, we explore the barriers to and discuss perspectives on the application of aptamer-based bacterial detection.
Collapse
Affiliation(s)
- Vincent Léguillier
- INRAE, AgroParisTech, Micalis Institut, Université Paris-Saclay, UMR 1319, 78350 Jouy-en-Josas, France;
- ENS Paris-Saclay, Laboratoire de Biologie et Pharmacologie Appliquée (LBPA), UMR8113 CNRS, 91190 Gif-sur-Yvette, France
| | - Brahim Heddi
- ENS Paris-Saclay, Laboratoire de Biologie et Pharmacologie Appliquée (LBPA), UMR8113 CNRS, 91190 Gif-sur-Yvette, France
| | - Jasmina Vidic
- INRAE, AgroParisTech, Micalis Institut, Université Paris-Saclay, UMR 1319, 78350 Jouy-en-Josas, France;
| |
Collapse
|
41
|
González MI, González-Arjona M, Cussó L, Morcillo MÁ, Aguilera-Correa JJ, Esteban J, Kestler M, Calle D, Cerón C, Cortes-Canteli M, Muñoz P, Bouza E, Desco M, Salinas B. In Vivo Detection of Staphylococcus aureus Infections Using Radiolabeled Antibodies Specific for Bacterial Toxins. Int J Biomed Imaging 2024; 2024:3655327. [PMID: 38665417 PMCID: PMC11045290 DOI: 10.1155/2024/3655327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/02/2024] [Accepted: 01/13/2024] [Indexed: 04/28/2024] Open
Abstract
Purpose The Gram-positive Staphylococcus aureus bacterium is one of the leading causes of infection in humans. The lack of specific noninvasive techniques for diagnosis of staphylococcal infection together with the severity of its associated complications support the need for new specific and selective diagnostic tools. This work presents the successful synthesis of an immunotracer that targets the α-toxin released by S. aureus. Methods [89Zr]Zr-DFO-ToxAb was synthesized based on radiolabeling an anti-α-toxin antibody with zirconium-89. The physicochemical characterization of the immunotracer was performed by high-performance liquid chromatography (HPLC), radio-thin layer chromatography (radio-TLC), and electrophoretic analysis. Its diagnostic ability was evaluated in vivo by positron emission tomography/computed tomography (PET/CT) imaging in an animal model of local infection-inflammation (active S. aureus vs. heat-killed S. aureus) and infective osteoarthritis. Results Chemical characterization of the tracer established the high radiochemical yield and purity of the tracer while maintaining antibody integrity. In vivo PET/CT image confirmed the ability of the tracer to detect active foci of S. aureus. Those results were supported by ex vivo biodistribution studies, autoradiography, and histology, which confirmed the ability of [89Zr]Zr-DFO-ToxAb to detect staphylococcal infectious foci, avoiding false-positives derived from inflammatory processes. Conclusions We have developed an immuno-PET tracer capable of detecting S. aureus infections based on a radiolabeled antibody specific for the staphylococcal alpha toxins. The in vivo assessment of [89Zr]Zr-DFO-ToxAb confirmed its ability to selectively detect staphylococcal infectious foci, allowing us to discern between infectious and inflammatory processes.
Collapse
Affiliation(s)
- María Isabel González
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Mario González-Arjona
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Lorena Cussó
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Ángel Morcillo
- Unidad de Aplicaciones Médicas de las Radiaciones Ionizantes, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain
| | - John Jairo Aguilera-Correa
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Servicio de Microbiología Clínica Instituto de Investigación Sanitaria Fundación Jiménez Díaz, UAM, 28040 Madrid, Spain
- CIBERINFEC-CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Jaime Esteban
- Servicio de Microbiología Clínica Instituto de Investigación Sanitaria Fundación Jiménez Díaz, UAM, 28040 Madrid, Spain
- CIBERINFEC-CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Martha Kestler
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Daniel Calle
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Carlos Cerón
- Cardiovascular Risk Factors and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | - Marta Cortes-Canteli
- Cardiovascular Risk Factors and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), 28040 Madrid, Spain
| | - Patricia Muñoz
- CIBER de Salud Mental, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Emilio Bouza
- CIBER de Salud Mental, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Manuel Desco
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, 28911 Madrid, Spain
| | - Beatriz Salinas
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, 28911 Madrid, Spain
| |
Collapse
|
42
|
Lin MH, Liu CC, Lu CW, Shu JC. Staphylococcus aureus foldase PrsA contributes to the folding and secretion of protein A. BMC Microbiol 2024; 24:108. [PMID: 38566014 PMCID: PMC10986000 DOI: 10.1186/s12866-024-03268-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Staphylococcus aureus secretes a variety of proteins including virulence factors that cause diseases. PrsA, encoded by many Gram-positive bacteria, is a membrane-anchored lipoprotein that functions as a foldase to assist in post-translocational folding and helps maintain the stability of secreted proteins. Our earlier proteomic studies found that PrsA is required for the secretion of protein A, an immunoglobulin-binding protein that contributes to host immune evasion. This study aims to investigate how PrsA influences protein A secretion. RESULTS We found that in comparison with the parental strain HG001, the prsA-deletion mutant HG001ΔprsA secreted less protein A. Deleting prsA also decreased the stability of exported protein A. Pulldown assays indicated that PrsA interacts with protein A in vivo. The domains in PrsA that interact with protein A are mapped to both the N- and C-terminal regions (NC domains). Additionally, the NC domains are essential for promoting PrsA dimerization. Furthermore, an immunoglobulin-binding assay revealed that, compared to the parental strain HG001, fewer immunoglobulins bound to the surface of the mutant strain HG001ΔprsA. CONCLUSIONS This study demonstrates that PrsA is critical for the folding and secretion of protein A. The information derived from this study provides a better understanding of virulent protein export pathways that are crucial to the pathogenicity of S. aureus.
Collapse
Affiliation(s)
- Mei-Hui Lin
- Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan, 333, Taiwan.
- Department of Laboratory Medicine, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, 333, Taiwan.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao- Yuan, 333, Taiwan.
| | - Chao-Chin Liu
- Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan, 333, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao- Yuan, 333, Taiwan
| | - Chiao-Wen Lu
- Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan, 333, Taiwan
- Department of Laboratory Medicine, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, 333, Taiwan
| | - Jwu-Ching Shu
- Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan, 333, Taiwan.
- Department of Laboratory Medicine, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, 333, Taiwan.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao- Yuan, 333, Taiwan.
| |
Collapse
|
43
|
Li BL, Chen JY, Hu JJ, Fan YW, Ao ZY, Zhang WJ, Lian X, Liang HJ, Li QR, Guan XX, Wu JW, Yuan J, Jiang DX. Three stilbenes from pigeon pea with promising anti-methicillin-resistant Staphylococcus aureus biofilm formation activity. Int Microbiol 2024; 27:535-544. [PMID: 37505307 DOI: 10.1007/s10123-023-00413-6] [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: 05/21/2023] [Revised: 07/17/2023] [Accepted: 07/22/2023] [Indexed: 07/29/2023]
Abstract
Cajaninstilbene acid (CSA), longistylin A (LLA), and longistylin C (LLC) are three characteristic stilbenes isolated from pigeon pea. The objective of this study was to evaluate the antibacterial activity of these stilbenes against Staphylococcus aureus and even methicillin-resistant Staphylococcus aureus (MRSA) and test the possibility of inhibiting biofilm formation. The minimum inhibitory concentrations (MICs) and minimum bactericidal concentrations (MBCs) of these stilbenes were evaluated. And the results showed that LLA was most effective against tested strains with MIC and MBC values of 1.56 μg/mL followed by LLC with MIC and MBC values of 3.12 μg/mL and 6.25 μg/mL as well as CSA with MIC and MBC values of 6.25 μg/mL and 6.25-12.5 μg/mL. Through growth curve and cytotoxicity analysis, the concentrations of these stilbenes were determined to be set at their respective 1/4 MIC in the follow-up research. In an anti-biofilm formation assay, these stilbenes were found to be effectively inhibited bacterial proliferation, biofilm formation, and key gene expressions related to the adhesion and virulence of MRSA. It is the first time that the anti-S. aureus and MRSA activities of the three stilbenes have been systematically reported. Conclusively, these findings provide insight into the anti-MRSA mechanism of stilbenes from pigeon pea, indicating these compounds may be used as antimicrobial agents or additives for food with health functions, and contribute to the development as well as application of pigeon pea in food science.
Collapse
Affiliation(s)
- Bai-Lin Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Core Botanical Garden, South China Botanical Garden, Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Applied Botany, Guangzhou, 510650, P. R. China
| | - Jia-Yan Chen
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Juan-Juan Hu
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, Kannapolis, NC, 28081, USA
| | - Yu-Wen Fan
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Zhuo-Yi Ao
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Wei-Jie Zhang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Xin Lian
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Hui-Jun Liang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Qian-Ran Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Xiao-Xian Guan
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Jie-Wei Wu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Jie Yuan
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China.
| | - Dong-Xu Jiang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| |
Collapse
|
44
|
Yamazaki Y, Ito T, Tamai M, Nakagawa S, Nakamura Y. The role of Staphylococcus aureus quorum sensing in cutaneous and systemic infections. Inflamm Regen 2024; 44:9. [PMID: 38429810 PMCID: PMC10905890 DOI: 10.1186/s41232-024-00323-8] [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: 12/18/2023] [Accepted: 02/15/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of human bacterial infections worldwide. It is the most common causative agent of skin and soft tissue infections, and can also cause various other infections, including pneumonia, osteomyelitis, as well as life-threatening infections, such as sepsis and infective endocarditis. The pathogen can also asymptomatically colonize human skin, nasal cavity, and the intestine. S. aureus colonizes approximately 20-30% of human nostrils, being an opportunistic pathogen for subsequent infection. Its strong ability to silently spread via human contact makes it difficult to eradicate S. aureus. A major concern with S. aureus is its capacity to develop antibiotic resistance and adapt to diverse environmental conditions. The variability in the accessory gene regulator (Agr) region of the genome contributes to a spectrum of phenotypes within the bacterial population, enhancing the likelihood of survival in different environments. Agr functions as a central quorum sensing (QS) system in S. aureus, allowing bacteria to adjust gene expression in response to population density. Depending on Agr expression, S. aureus secretes various toxins, contributing to virulence in infectious diseases. Paradoxically, expressing Agr may be disadvantageous in certain situations, such as in hospitals, causing S. aureus to generate Agr mutants responsible for infections in healthcare settings. MAIN BODY This review aims to demonstrate the molecular mechanisms governing the diverse phenotypes of S. aureus, ranging from a harmless colonizer to an organism capable of infecting various human organs. Emphasis will be placed on QS and its role in orchestrating S. aureus behavior across different contexts. SHORT CONCLUSION The pathophysiology of S. aureus infection is substantially influenced by phenotypic changes resulting from factors beyond Agr. Future studies are expected to give the comprehensive understanding of S. aureus overall profile in various settings.
Collapse
Affiliation(s)
- Yuriko Yamazaki
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka, University, Osaka, 565-0871, Japan
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Tomoka Ito
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Masakazu Tamai
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Seitaro Nakagawa
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Yuumi Nakamura
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka, University, Osaka, 565-0871, Japan.
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan.
| |
Collapse
|
45
|
Savin A, Anderson EE, Dyzenhaus S, Podkowik M, Shopsin B, Pironti A, Torres VJ. Staphylococcus aureus senses human neutrophils via PerR to coordinate the expression of the toxin LukAB. Infect Immun 2024; 92:e0052623. [PMID: 38235972 PMCID: PMC10863418 DOI: 10.1128/iai.00526-23] [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: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/19/2024] Open
Abstract
Staphylococcus aureus is a gram-positive pathogen that poses a major health concern, in part due to its large array of virulence factors that allow infection and evasion of the immune system. One of these virulence factors is the bicomponent pore-forming leukocidin LukAB. The regulation of lukAB expression is not completely understood, especially in the presence of immune cells such as human polymorphonuclear neutrophils (hPMNs). Here, we screened for transcriptional regulators of lukAB during the infection of primary hPMNs. We uncovered that PerR, a peroxide sensor, is vital for hPMN-mediated induction of lukAB and that PerR upregulates cytotoxicity during the infection of hPMNs. Exposure of S. aureus to hydrogen peroxide (H2O2) alone also results in increased lukAB promoter activity, a phenotype dependent on PerR. Collectively, our data suggest that S. aureus uses PerR to sense the H2O2 produced by hPMNs to stimulate the expression of lukAB, allowing the bacteria to withstand these critical innate immune cells.IMPORTANCEStaphylococcus aureus utilizes a diverse set of virulence factors, such as leukocidins, to subvert human neutrophils, but how these toxins are regulated is incompletely defined. Here, we identified the peroxide-sensitive repressor, PerR, as a required protein involved in the induction of lukAB in the presence of primary human neutrophils, a phenotype directly linked to the ability of PerR to sense H2O2. Thus, we show that S. aureus coordinates sensing and resistance to oxidative stress with toxin production to promote pathogen survival.
Collapse
Affiliation(s)
- Avital Savin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Biology, New York University, New York, New York, USA
| | - Exene E. Anderson
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sophie Dyzenhaus
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Magdalena Podkowik
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
46
|
Vasudevan S, David H, Chanemougam L, Ramani J, Ramesh Sangeetha M, Solomon AP. Emergence of persister cells in Staphylococcus aureus: calculated or fortuitous move? Crit Rev Microbiol 2024; 50:64-75. [PMID: 36548910 DOI: 10.1080/1040841x.2022.2159319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
A stable but reversible phenotype switch from normal to persister state is advantageous to the intracellular pathogens to cause recurrent infections and to evade the host immune system. Staphylococcus aureus is a versatile opportunistic pathogen known to cause chronic infections with significant mortality. One of the notable features is the ability to switch to a per-sisters cell, which is found in planktonic and biofilm states. This phenotypic switch is always an open question to explore the hidden fundamental science that coheres with a calculated or fortuitous move. Toxin-antitoxin modules, nutrient stress, and an erroneous translation-enabled state of dormancy entail this persistent behaviour in S. aureus. It is paramount to get a clear picture of why the cell chooses to enter a persistent condition, as it would decide the course of treatment. Analyzing the exit from a persistent state to an active state and the subsequent repercussion of this transition is essential to determine its role in chronic infections. This review attempts to provide a constructed argument discussing the most widely accepted mechanisms and identifying the various attributes of persistence.
Collapse
Affiliation(s)
- Sahana Vasudevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Lakshmi Chanemougam
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Jayalakshmi Ramani
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Maanasa Ramesh Sangeetha
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
47
|
Alkuraythi DM, Alkhulaifi MM, Binjomah AZ, Alarwi M, Mujallad MI, Alharbi SA, Alshomrani M, Gojobori T, Alajel SM. Comparative genomic analysis of antibiotic resistance and virulence genes in Staphylococcus aureus isolates from patients and retail meat. Front Cell Infect Microbiol 2024; 13:1339339. [PMID: 38282615 PMCID: PMC10811269 DOI: 10.3389/fcimb.2023.1339339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 12/26/2023] [Indexed: 01/30/2024] Open
Abstract
Introduction Staphylococcus aureus is a significant human pathogen that poses a threat to public health due to its association with foodborne contamination and a variety of infections. The factors contributing to the pathogenicity of S. aureus include virulence, drug resistance, and toxin production, making it essential to monitor their prevalence and genetic profiles. This study investigated and compared the genomic characteristics of S. aureus isolates from retail meat and patients in Saudi Arabia. Methods A total of 136 S. aureus isolates were obtained between October 2021 and June 2022:84 from patients and 53 from meat samples in Riyadh, Saudi Arabia. S. aureus isolates were identified using conventional methods and MALDI-TOF MS, and methicillin-resistant S. aureus (MRSA) was identified using VITEK2 and BD Phoenix systems. MRSA was confirmed phenotypically using chromogenic agar, and genotypically by detecting mecA. Genomic data were analyzed using BactopiaV2 pipeline, local BLAST, and MLST databases. Results Antibiotic resistance genes were prevalent in both meat and patient S. aureus isolates, with high prevalence of tet38, blaZ, and fosB. Notably, all S. aureus isolates from patients carried multidrug-resistant (MDR) genes, and a high percentage of S. aureus isolates from meat also harbored MDR genes. Phenotypically, 43% of the S. aureus isolates from meat and 100% of the patients' isolates were MDR. Enterotoxin genes, including selX, sem, and sei, exhibited high compatibility between meat and patient S. aureus isolates. Virulence genes such as cap, hly/hla, sbi, and isd were found in all S. aureus isolates from both sources. Conclusion Our study established a genetic connection between S. aureus isolates from meat and patients, showing shared antibiotic resistance and virulence genes. The presence of these genes in meat derived isolates underscores its role as a reservoir. Genomic relatedness also suggests potential transmission of resistance between different settings. These findings emphasize the necessity for a comprehensive approach to monitor and control S. aureus infections in both animals and humans.
Collapse
Affiliation(s)
- Dalal M. Alkuraythi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Manal M. Alkhulaifi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdulwahab Z. Binjomah
- Microbiology Department, Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
- College of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| | - Mohammed Alarwi
- Computational Bioscience Research Center, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | | | - Saleh Ali Alharbi
- Microbiology Department, Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Mohammad Alshomrani
- Microbiology Department, Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Takashi Gojobori
- Computational Bioscience Research Center, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Sulaiman M. Alajel
- Reference Laboratory for Microbiology, Executive Department for Reference Laboratories, Research and Laboratories Sector, Food and Drug Authority, Riyadh, Saudi Arabia
| |
Collapse
|
48
|
Jiang T, Yuan D, Wang R, Zhao C, Xu Y, Liu Y, Song W, Su X, Wang B. Echinacoside, a promising sortase A inhibitor, combined with vancomycin against murine models of MRSA-induced pneumonia. Med Microbiol Immunol 2023; 212:421-435. [PMID: 37796314 DOI: 10.1007/s00430-023-00782-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a pathogenic bacterium responsible for a range of severe infections, such as skin infections, bacteremia, and pneumonia. Due to its antibiotic-resistant nature, current research focuses on targeting its virulence factors. Sortase A (SrtA) is a transpeptidase that anchors surface proteins to the bacterial cell wall and is involved in adhesion and invasion to host cells. Through fluorescence resonance energy transfer (FRET), we identified echinacoside (ECH), a natural polyphenol, as a potential SrtA inhibitor with an IC50 of 38.42 μM in vitro. It was demonstrated that ECH inhibited SrtA-mediated S. aureus fibrinogen binding, surface protein A anchoring, and biofilm formation. The fluorescence quenching assay determined the binding mode of ECH to SrtA and calculated the KA-binding constant of 3.09 × 105 L/mol, demonstrating the direct interaction between the two molecules. Molecular dynamics simulations revealed that ECH-SrtA interactions occurred primarily at the binding sites of A92G, A104G, V168A, G192A, and R197A. Importantly, the combination of ECH and vancomycin offered protection against murine models of MRSA-induced pneumonia. Therefore, ECH may serve as a potential antivirulence agent against S. aureus infections, either alone or in combination with vancomycin.
Collapse
Affiliation(s)
- Tao Jiang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Dai Yuan
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Rong Wang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Chunhui Zhao
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yangming Xu
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yinghui Liu
- Changchun University of Chinese Medicine, Changchun, 130117, China
- Jilin Provincial People's Hospital, Changchun, 130021, China
| | - Wu Song
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Xin Su
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Bingmei Wang
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
49
|
Campbell MJ, Beenken KE, Ramirez AM, Smeltzer MS. The major role of sarA in limiting Staphylococcus aureus extracellular protease production in vitro is correlated with decreased virulence in diverse clinical isolates in osteomyelitis. Virulence 2023; 14:2175496. [PMID: 36748843 PMCID: PMC9928472 DOI: 10.1080/21505594.2023.2175496] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
We previously demonstrated that MgrA, SarA, SarR, SarS, SarZ, and Rot bind at least three of the four promoters associated with genes encoding primary extracellular proteases in Staphylococcus aureus (Aur, ScpA, SspA/SspB, SplA-F). We also showed that mutation of sarA results in a greater increase in protease production, and decrease in biofilm formation, than mutation of the loci encoding any of these other proteins. However, these conclusions were based on in vitro studies. Thus, the goal of the experiments reported here was to determine the relative impact of the regulatory loci encoding these proteins in vivo. To this end, we compared the virulence of mgrA, sarA, sarR, sarS, sarZ, and rot mutants in a murine osteomyelitis model. Mutants were generated in the methicillin-resistant USA300 strain LAC and the methicillin-sensitive USA200 strain UAMS-1, which was isolated directly from the bone of an osteomyelitis patient during surgical debridement. Mutation of mgrA and rot limited virulence to a statistically significant extent in UAMS-1, but not in LAC, while the sarA mutant exhibited reduced virulence in both strains. The reduced virulence of the sarA mutant was correlated with reduced cytotoxicity for osteoblasts and osteoclasts, reduced biofilm formation, and reduced sensitivity to the antimicrobial peptide indolicidin, all of which were directly attributable to increased protease production in both LAC and UAMS-1. These results illustrate the importance of considering diverse clinical isolates when evaluating the impact of regulatory mutations on virulence and demonstrate the significance of SarA in limiting protease production in vivo in S. aureus.
Collapse
Affiliation(s)
- Mara J. Campbell
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Karen E. Beenken
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Aura M. Ramirez
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | |
Collapse
|
50
|
Yang P, Yang J, Lin T, Liu Q, Yin Y, Chen D, Yang S. Efficient Genome Editing in Most Staphylococcus aureus by Using the Restriction-Modification System Silent CRISPR-Cas9 Toolkit. ACS Synth Biol 2023; 12:3340-3351. [PMID: 37830328 DOI: 10.1021/acssynbio.3c00339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Staphylococcus aureus is a clinically important pathogen that threatens human health due to its strong pathogenicity and drug resistance, leading to meningitis, endocarditis, and skin and soft tissue infections. Genetic manipulation in S. aureus is a powerful approach for characterizing the molecular mechanisms of bacterial drug resistance, pathogenicity, and virulence. However, a strong restriction barrier presents a major obstacle to the extensive utilization of genetic manipulation tools in clinical isolates of S. aureus. Here, we constructed a restriction-modification (RM) system silent CRISPR-Cas9 toolkit that synonymously eliminated the type I RM targets of S. aureus from plasmids, downsized plasmids using minicircle technology, and combined with a plasmid artificial modification (PAM) method to circumvent the type II RM system. The RM-silent CRISPR-Cas9 toolkit enables a significant improvement in transformation (105-106 transformants per microgram plasmid in strains we tested) and high-success efficiency editing for gene deletion (knockout strain obtained in one-round electroporation) in a wide range of S. aureus species including clinical isolates of unknown genetic background. The RM-silent CRISPR-Cas9 toolkits could expedite the process of mutant construction in most S. aureus strains, and this approach could be applied to the design of other genetic toolkit plasmids for utilization in a wider range of S. aureus strains.
Collapse
Affiliation(s)
- Ping Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Junjie Yang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Ting Lin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qi Liu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Yin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Daijie Chen
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Sheng Yang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|