1
|
Cui W, Liang X, Xiao W, Wang Y, Liu F, Chen S, Long J, Jin Y, Duan G, Yang H. The role and mechanism of efflux pump norB in biofilm formation of Staphylococcus aureus. Gene 2025; 936:149105. [PMID: 39566593 DOI: 10.1016/j.gene.2024.149105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/22/2024]
Abstract
Staphylococcus aureus (S. aureus) is one of the notorious bacteria responsible for community and hospital infections. It can attach to the indwelling medical devices to form biofilms, which increases resistance to antibiotics and causes frequent chronic or persistent infections. This study attempted to determine the contribution and mechanism between the efflux pump norB gene and biofilm development in S. aureus. The expression levels of norB gene were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The norB gene knockout strain USA300 ΔnorB was constructed by homologous recombination technology. Crystal violet staining was utilized to detect the biofilm formation ability. Differentially expressed genes between norB knockout strains and wild-type strains were screened by RNA-Seq technology and verified by qRT-PCR. In comparison to strains with weak biofilm development capacity, higher expression levels of the norB gene were detected in S. aureus strains that showed strong biofilm forming capabilities. The expression levels of norB were significantly up-regulated in biofilm bacteria in comparison to planktonic bacteria. The knockout of norB gene reduced the biofilm formation ability in S. aureus. The deletion of norB gene up-regulated the expression of genes related to biofilm formation including agrD, sdrC, sdrD, agrB, agrC, fnbB, nuc, lytS, lrgA, sdrE, agrA and saeS, and down-regulated the expression of genes related to biofilm formation including clfA, icaR, sarA and rot. In conclusion, the efflux pump norB gene serves as a crucial role in the production of biofilm, thus rendering it a promising avenue for biofilm suppression.
Collapse
Affiliation(s)
- Wenjing Cui
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Xuan Liang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Wenwei Xiao
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Ying Wang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Fang Liu
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Shuaiyin Chen
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Jinzhao Long
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yuefei Jin
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Guangcai Duan
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Watabe N, Subsomwong P, Yamane K, Asano K, Nakane A. Polygonum tinctorium extract suppresses the virulence of methicillin-resistant Staphylococcus aureus by disrupting its extracellular vesicles. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118933. [PMID: 39396717 DOI: 10.1016/j.jep.2024.118933] [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: 07/23/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Methicillin-resistant S. aureus (MRSA) is a significant global health concern, causing both hospital- and community-acquired infections. The extracellular vesicles released by S. aureus (SaEVs) contain essential factors related to the bacterial survival and pathogenicity. Polygonum tinctorium is traditionally used as a natural dye (indigo) and for treating various infectious diseases caused by microorganisms. However, the effect of P. tinctorium extract (Indigo Ex) and its mechanism on SaEVs is unknown. AIM OF THE STUDY We investigated the effect and mechanism of Indigo Ex on SaEVs, which could be used in controlling S. aureus, especially MRSA infection. MATERIALS AND METHODS Indigo Ex was prepared from pesticide-free P. tinctorium, which was dried, powdered, and extracted with d-limonene. SaEVs were isolated and purified from MRSA culture supernatant by step-gradient ultracentrifugation. The effect of Indigo Ex on SaEVs morphology was observed by both transmission and scanning electron microscopy after incubating the Indigo Ex and SaEVs under shaking conditions. The cytotoxicity of Indigo Ex was performed using mouse macrophage cell line, RAW 264.7. In addition, the ability of Indigo Ex-treated SaEVs to stimulate the immune response and cytotoxicity in RAW 264.7 cells were evaluated by ELISA and WST-1 assay, respectively. RESULTS SaEV particles were disrupted when treated with undiluted Indigo Ex in a time-dependent manner. For the cytotoxicity of Indigo Ex on RAW 264.7 cells, over 50% of the cell viability decreased when diluted Indigo Ex 1000-fold and no cytotoxic effect was observed at a 25,000-fold dilution of Indigo Ex. Interestingly, the Indigo Ex-treated SaEVs showed less cytotoxic effect than SaEVs alone. Similarly, SaEVs treated with Indigo Ex reduced stimulation of pro-inflammatory cytokines (TNF-α and IL-6) and anti-inflammatory cytokine (IL-10) in RAW 264.7 cells compared to untreated SaEVs. Our results indicate that Indigo Ex disrupted SaEV particles, resulting in reduced virulence and stimulation of immune response. CONCLUSIONS This study reveals that the low concentration of Indigo Ex can suppresses the virulence of SaEVs without causing cytotoxicity to the host cells. Therefore, Indigo Ex may have the potential to be used to control S. aureus infection.
Collapse
Affiliation(s)
- Naoko Watabe
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | - Phawinee Subsomwong
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | | | - Krisana Asano
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan; Department of Biopolymer and Health Science, Hirosaki University Graduate School of. Medicine, Hirosaki, Aomori, Japan.
| | - Akio Nakane
- Department of Biopolymer and Health Science, Hirosaki University Graduate School of. Medicine, Hirosaki, Aomori, Japan.
| |
Collapse
|
3
|
Yang X, Tang X, Yi S, Guo T, Liao Y, Wang Y, Zhang X. Maltodextrin-derived nanoparticles resensitize intracellular dormant Staphylococcus aureus to rifampicin. Carbohydr Polym 2025; 348:122843. [PMID: 39562116 DOI: 10.1016/j.carbpol.2024.122843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 11/21/2024]
Abstract
Intracellular bacteria are recognized as a crucial factor in the persistence and recurrence of infections. The efficacy of current antibiotic treatments faces substantial challenges due to the dormant state formation of intracellular bacteria. In this study, we devised a strategy aimed at reverting intracellular dormant bacteria to a metabolically active state, thereby increasing their vulnerability to antibiotics. We found that oligosaccharides, especially maltodextrin (MD), can be absorbed by dormant S. aureus, leading to their revival and restoration of sensitivity to rifampicin (Rif). We then synthesized a reactive oxygen species (ROS)-responsive MD-prodrug by covalently binding MD with 4-(hydroxymethyl) phenylboronic acid pinacol ester (MD-PBAP) and prepared a ROS-responsive nanoparticles (MDNP) using a nanoprecipitation and self-assembly method. Once internalized by host cells, MDNP was degraded to MD, reactivating dormant S. aureus, and enhancing their susceptibility to Rif. More importantly, MDNP treatment restored the sensitivity of intracellular persistent S. aureus to Rif in both a reservoir transfer model and whole-body infection model. Additionally, MDNP have demonstrated excellent biocompatibility in both in vitro and in vivo settings. These results offer a promising therapeutic avenue for managing persistent intracellular bacterial infections by reviving and resensitizing intracellular dormant bacteria to conventional antibiotics.
Collapse
Affiliation(s)
- Xiaodi Yang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Xiyu Tang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Sisi Yi
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Tao Guo
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Yue Liao
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Yan Wang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Xiangjun Zhang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China.
| |
Collapse
|
4
|
Song Y, Ma Q, Luo J, Yang Z, Li J, Zhao J. Liushen Wan alleviates the virulence and inflammation of Staphylococcus aureus via NLRP3 inflammasome and TLR2-NF-κB/p38 MAPK signaling pathways. Int Immunopharmacol 2025; 144:113633. [PMID: 39566390 DOI: 10.1016/j.intimp.2024.113633] [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: 05/30/2024] [Revised: 10/25/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
Infectious diseases have been a major threat to health worldwide, with bacterial infections being particularly prominent. Staphylococcus aureus (S. aureus) infections are associated with the most deaths. Inhibition of virulence factor and excessive inflammation induced by S. aureus has become a potential antibiotic alternative/synergistic therapy without causing greater survival pressure to prevent the emergence of "superbugs" in the future. Liushen Wan (LSW), a traditional Chinese medicine, used for multiple bacterial infectious diseases. In this work, we researched its therapeutic effect and explored the potential mechanism of LSW aiming at S. aureus in vivo and in vitro. Minimal inhibitory concentration (MIC) assay, hemolysis assay, invasion assay, staphyloxanthin assay and evolution of resistance assay were performed to show that LSW alleviated the virulence of S. aureus without suppressing S. aureus activity, and short-term use of LSW did not make bacteria resistant to it. Biofilm inhibition assay demonstrated that LSW inhibited the formation of biofilm and destroyed mature biofilm of S. aureus. In vitro experiments using RT-qPCR, ELISA and western blot analysis indicated LSW inhibited the inflammatory reaction triggered by HK-S. aureus and S. aureus through NLRP3 inflammasome and TLR2-NF-κB/p38 MAPK pathway. Moreover, LSW alleviated lung damage induced by S. aureus. Taken together, LSW is a promising antibacterial, anti-virulence and anti-inflammatory drug, which could provide the pharmacological basis on the traditional application of LSW for diseases associated with S. aureus infection in clinical.
Collapse
Affiliation(s)
- Yudi Song
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510000, China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510000, China
| | - Jincan Luo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510000, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510000, China; Guangzhou Laboratory, Guangzhou, Guangdong, 510000, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau (SAR), 519020, China.
| | - Jiqiang Li
- The Second Affiliated Hospital Of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510000, China.
| | - Jin Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510000, China.
| |
Collapse
|
5
|
Borah A, Srivastava A. Impact of extracellular enzymes on Staphylococcus aureus host tissue adaptation and infection. APMIS 2025; 133:e13502. [PMID: 39604200 DOI: 10.1111/apm.13502] [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: 08/02/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
Staphylococcus aureus is a multi-host pathogen that can colonize and infect both humans and livestock in a tissue-specific manner. This amazing feature of the pathogen is mainly facilitated by the surplus virulence agents produced upon necessity and favorable environmental factors. These factors are adept at damaging cellular barriers, manipulating host immune factors, and circumventing the host complement system. The delicate balance between the timely release of virulent factors and the regulation of their production underscores the significance of the exoenzyme network. Moreover, the intricate relationship between the pathogen and host tissue highlights the importance of understanding tissue-specific phenotypes for effective therapeutic strategies. Here, we provide a review on the diverse role played by the extracellular enzymes of S. aureus in tissue-specific infection and systemic colonization leading to distinctive diseased conditions. The article highlights the need to study the role of staphylococcal exoenzymes in various systemic invasions, their impact on the deterioration of host tissue, and the regulation of S. aureus virulence factors.
Collapse
Affiliation(s)
- Atlanta Borah
- Biotechnology Research Innovation Council-National Institute of Animal Biotechnology (BRIC-NIAB), Hyderabad, Telangana, India
| | - Anand Srivastava
- Biotechnology Research Innovation Council-National Institute of Animal Biotechnology (BRIC-NIAB), Hyderabad, Telangana, India
| |
Collapse
|
6
|
Nirenjen S, Narayanan J. Computational profiling and pharmacokinetic modelling of Febuxostat: Evaluating its potential as a therapeutic agent for diabetic wound healing. Biochim Biophys Acta Gen Subj 2025; 1869:130735. [PMID: 39653251 DOI: 10.1016/j.bbagen.2024.130735] [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: 09/12/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Diabetic wounds, a significant complication of Type 2 Diabetes Mellitus (T2DM), face delayed healing due to impaired inflammation, angiogenesis, and collagen synthesis. This study explores Febuxostat, a xanthine oxidase inhibitor for its therapeutic potential in wound healing. Combining computational approaches and in-vitro assays, the study evaluates its effects on key wound healing pathways, cell viability, migration. METHODOLOGY The potential of Febuxostat in diabetic wound healing was studied using in-silico tools for Molecular docking and ADMET profiling, alongside Molecular dynamics (MD) simulations. Toxicity was assessed with OSIRIS Explorer, and biological activity was predicted using the PASS tool. In-vitro MTT and scratch assays on L929 cells further validated cytotoxicity and wound healing efficacy. RESULTS Docking analysis revealed strong binding affinities to key wound healing targets, including VEGF (-9.11 kcal/mol) and NFKβ (-8.62 kcal/mol). Pharmacokinetic studies highlighted favorable skin permeability, supporting topical applications. Toxicity predictions indicated a safe profile. Molecular dynamics simulations demonstrated stable protein-ligand complexes, particularly with VEGF. Cytotoxicity studies on L929 cells revealed an IC50 of 6.08 μM and the scratch assay demonstrated significant wound healing activity, highlighting its effectiveness in promoting cell migration and closure. CONCLUSION Febuxostat shows remarkable potential in enhancing diabetic wound healing by promoting cell migration, targeting wound-healing proteins, as demonstrated through in-silico and in-vitro studies. This drug is poised to effectively treat diabetic wounds, accelerating healing and reducing complications. Rigorous pre-clinical and clinical evaluations are essential to validate its safety, efficacy, and therapeutic potential.
Collapse
Affiliation(s)
- S Nirenjen
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu 603203, Tamil Nadu, India
| | - J Narayanan
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu 603203, Tamil Nadu, India.
| |
Collapse
|
7
|
Chien YS, Tsai TT, Lin JH, Chang CC, Chen CF. One-step copper deposition-induced signal amplification for multiplex bacterial infection diagnosis on a lateral flow immunoassay device. Biosens Bioelectron 2025; 267:116849. [PMID: 39406071 DOI: 10.1016/j.bios.2024.116849] [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: 08/07/2024] [Revised: 09/19/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024]
Abstract
The lateral flow immunoassay (LFIA) is predominant in rapid diagnostic tests owing to its cost-effectiveness and operational simplicity. However, the conventional LFIA exhibits limited sensitivity and is susceptible to human variance for the result readout, impacting result interpretation. In this study, we introduced a novel one-step copper deposition-induced signal amplification lateral flow immunoassay (osa-LFIA) that markedly enhances the detection sensitivity for Staphylococcus aureus (protein A) and Pseudomonas aeruginosa (exotoxin A). Utilizing gold nanoparticles (AuNPs) as a catalyst, this approach employs ascorbic acid to reduce Cu2+ to Cu0, depositing on AuNPs at the test line and amplifying the signal. A user-friendly design features a three-dimensional paper structure incorporating pre-dried reagents, enabling a streamlined, efficient testing process. The osa-LFIA significantly lowers detection limits to 3 ng mL-1 for protein A and 10 ng mL-1 for exotoxin A, offering a tenfold improvement over conventional LFIA. Additionally, we developed a portable grayscale detection device, achieving less than 10% error in quantitative analysis compared to the data acquired and analyzed in the lab. This entire process, from detection to signal amplification, is completed in just 20 min. For the clinical trial, we utilized the osa-LFIA to test synovial fluid samples infected with Staphylococcus aureus. We also successfully detected different concentrations of the exotoxin A in parallel, with a recovery value of 96%-110%. Our findings demonstrate the osa-LFIA's potential as a rapid, highly sensitive, and simple-to-use diagnostic tool for detecting various pathogens, significantly advancing the field of rapid diagnostic testing.
Collapse
Affiliation(s)
- Yuh-Shiuan Chien
- Institute of Applied Mechanics, National Taiwan University, Taipei, 106, Taiwan
| | - Tsung-Ting Tsai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, 333, Taiwan
| | - Jia-Hui Lin
- Department of Applied Chemistry, National University of Kaohsiung, Kaohsiung, 811, Taiwan
| | - Chien-Cheng Chang
- Institute of Applied Mechanics, National Taiwan University, Taipei, 106, Taiwan.
| | - Chien-Fu Chen
- Institute of Applied Mechanics, National Taiwan University, Taipei, 106, Taiwan; Graduate School of Advanced Technology, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
8
|
Andersen-Ranberg E, Nymo IH, Jokelainen P, Emelyanova A, Jore S, Laird B, Davidson RK, Ostertag S, Bouchard E, Fagerholm F, Skinner K, Acquarone M, Tryland M, Dietz R, Abass K, Rautio A, Hammer S, Evengård B, Thierfelder T, Stimmelmayr R, Jenkins E, Sonne C. Environmental stressors and zoonoses in the Arctic: Learning from the past to prepare for the future. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:176869. [PMID: 39423885 DOI: 10.1016/j.scitotenv.2024.176869] [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: 06/04/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
The risk of zoonotic disease transmission from animals to humans is elevated for people in close contact with domestic and wild animals. About three-quarters of all known human infectious diseases are zoonotic, and potential health impacts of these diseases are higher where infectious disease surveillance and access to health care and public health services are limited. This is especially the case for remote circumarctic regions, where drivers for endemic, emerging, and re-emerging zoonotic diseases include anthropogenic influences, such as pollution by long-range transport of industrial chemicals, climate change, loss of biodiversity and ecosystem alterations. In addition to these, indirect effects including natural changes in food web dynamics, appearance of invasive species and thawing permafrost also affect the risk of zoonotic disease spill-over. In other words, the Arctic represents a changing world where pollution, loss of biodiversity and habitat, and maritime activity are likely driving forward occurrence of infectious diseases. As a broad international consortium with a wide range of expertise, we here describe a selection of case studies highlighting the importance of a One Health approach to zoonoses in the circumarctic, encompassing human health, animal health, and environmental health aspects. The cases highlight critical gaps in monitoring and current knowledge, focusing on environmental stressors and lifestyle factors, and they are examples of current occurrences in the Arctic that inform on critically needed actions to prepare us for the future. Through these presentations, we recommend measures to enhance awareness and management of existing and emerging zoonoses with epidemic and pandemic potential while also focusing on the impacts of various environmental stressors and lifestyle factors on zoonoses in the Arctic.
Collapse
Affiliation(s)
- Emilie Andersen-Ranberg
- University of Copenhagen, Faculty of Health and Medical Sciences, Department of Veterinary Clinical Sciences, Dyrlægevej 16, 1870 Frederiksberg, Denmark.
| | - Ingebjørg H Nymo
- Norwegian Veterinary Institute, Holtveien 66, 9016 Tromsø, Norway; Department of Arctic and Marine Biology, UiT - The Arctic University of Norway, Framstredet 39, Breivika, 9019 Tromsø, Norway
| | - Pikka Jokelainen
- Infectious Disease Preparedness, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark
| | - Anastasia Emelyanova
- Thule Institute, University of Oulu, Paavo Havaksen tie 3, 90570 Oulu, Finland; Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Paavo Havaksen tie 3 Linnanmaa, 90014, Finland
| | - Solveig Jore
- Department of Zoonotic, Food & Waterborne Infections, Norwegian Institute of Public Health, Postbox 222 Skøyen, 0213 Oslo, Norway
| | - Brian Laird
- School of Public Health Sciences, University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1, Canada
| | | | - Sonja Ostertag
- School of Public Health Sciences, University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1, Canada
| | - Emilie Bouchard
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, St Hyacinthe J2T 1B3, Canada; Department of Veterinary Microbiology, University of Saskatchewan, 52 Campus Drive, Saskatoon S7N 5B4, Canada
| | - Freja Fagerholm
- Department of Clinical Microbiology and the Arctic Center, Umeå University, Johan Bures Väg 5, 90187 Umeå, Sweden
| | - Kelly Skinner
- School of Public Health Sciences, University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1, Canada
| | - Mario Acquarone
- Arctic Monitoring and Assessment Programme, Hjalmar Johansens gate 14, 9007 Tromsø, Norway
| | - Morten Tryland
- Department of Forestry and Wildlife Management, Inland Norway University of Applied Sciences, Anne Evenstads Veg 80, 2480 Koppang, Norway
| | - Rune Dietz
- Aarhus University, Faculty of Technological Sciences, Department of Ecoscience, Frederiksborgvej 399, 4000 Roskilde, Denmark
| | - Khaled Abass
- Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Paavo Havaksen tie 3 Linnanmaa, 90014, Finland; Department of Environmental Health Sciences, College of Health Sciences, University of Sharjah, postbox 27272, United Arab Emirates
| | - Arja Rautio
- Thule Institute, University of Oulu, Paavo Havaksen tie 3, 90570 Oulu, Finland; Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Paavo Havaksen tie 3 Linnanmaa, 90014, Finland
| | - Sjúrður Hammer
- Faroese Environment Agency, Traðagøta 38, 165 Argir, Faroe Islands; University of the Faroe Islands, Vestara Bryggja 15, 100 Tórshavn, Faroe Islands
| | - Birgitta Evengård
- Department of Clinical Microbiology and the Arctic Center, Umeå University, Johan Bures Väg 5, 90187 Umeå, Sweden
| | - Tomas Thierfelder
- Department of Energy and Technology, Swedish University of Agricultural Sciences, postbox 75651, Uppsala, Sweden
| | - Raphaela Stimmelmayr
- Department of Wildlife management, North Slope Borough, postbox 69, 99723 Utqiagvik, AK, USA
| | - Emily Jenkins
- Department of Veterinary Microbiology, University of Saskatchewan, 52 Campus Drive, Saskatoon S7N 5B4, Canada
| | - Christian Sonne
- Aarhus University, Faculty of Technological Sciences, Department of Ecoscience, Frederiksborgvej 399, 4000 Roskilde, Denmark.
| |
Collapse
|
9
|
Amoli MS, Jin L, Rezapourdamanab S, Saadeh M, Singh Y, Ning L, Hwang B, Tomov ML, LaRock CN, Mahmoudi M, Bauser-Heaton H, Serpooshan V. Protocol for 3D bioprinting of nanoparticle-laden hydrogels to enhance antibacterial and imaging properties. STAR Protoc 2024; 5:103393. [PMID: 39412997 PMCID: PMC11530900 DOI: 10.1016/j.xpro.2024.103393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/22/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024] Open
Abstract
This protocol describes the preparation of a nanoparticle-encapsulated bioink capable of protecting tissue-engineered constructs against bacterial infections while also providing contrast for magnetic resonance (MR) imaging modalities. The report includes details of preparing the methacrylated gelatin-based bioinks and the incorporation of superparamagnetic iron oxide nanoparticles. A detailed protocol is presented for characterizing the bioink, evaluating cell response, and assessing its antibacterial effect. Overall, this article presents a robust approach for the development of antibacterial, MR-visible bioinks suitable for various tissue engineering applications. For complete details on the use and execution of this protocol, please refer to Theus et al.1.
Collapse
Affiliation(s)
- Mehdi Salar Amoli
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA.
| | - Linqi Jin
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Sarah Rezapourdamanab
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Maher Saadeh
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Yamini Singh
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; Department of Mechanical Engineering, Cleveland State University, Cleveland, OH 44115, USA
| | - Boeun Hwang
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Martin L Tomov
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christopher N LaRock
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA; Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, GA, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI, USA
| | - Holly Bauser-Heaton
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Sibley Heart Center at Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.
| |
Collapse
|
10
|
Egyir B, Owusu-Nyantakyi C, Bortey A, Rabbi Amuasi G, Owusu FA, Boateng W, Ahmed H, Danso JK, Oclu AAG, Mohktar Q, Tetteh-Ocloo G, Amegbletor H, Fosu K, Tetteh FKM, Asante-Sefa S, Deberu ON, Osei KM, Twasam J, Kodom S, Gyinae E, Sampah J, Dzifa Dayie N, Obeng-Nkrumah N, Mills-Pappoe WA, Boateng G, Nilsson P, Bonful HA, Adu B, Hendriksen RS. Whole genome sequencing revealed high proportions of ST152 MRSA among clinical Staphylococcus aureus isolates from ten hospitals in Ghana. mSphere 2024; 9:e0044624. [PMID: 39565128 DOI: 10.1128/msphere.00446-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
Previous studies in Ghana indicated low prevalence of methicillin-resistant Staphylococcus aureus (MRSA) and predominance of ST152 methicillin-susceptible S. aureus (MSSA) among clinical isolates. ST152 MRSA clones are associated with severe infections and epidemics. Using whole genome sequencing (WGS), 159 S. aureus isolated from clinical sources (wound, blood, urine, ear, abscess, umbilical cord, eye, vaginal samples, and others) from 10 hospitals across Ghana were investigated. mecA (gene for methicillin resistance) was detected in 38% of the isolates. Panton-Valentine leucocidin toxin (PVL) gene occurred in 65% isolates, with 84% of the MRSA's harboring the PVL gene. ST152 was the major clone, with 74% harboring the mecA gene. Other MRSA clones detected were ST5, ST5204, ST852, and ST1. MSSA clones included ST3249, ST152, ST5, ST1, and ST8. Twenty-three genes encoding resistance to 12 antimicrobial classes were observed with blaZ (97%) being the most prevalent. Other predominant resistance genes included tetK (46%), cat (42%), and dfrG (36%) encoding resistance for tetracyclines, phenicols, and diaminopyrimidine, respectively. Virulence genes for enterotoxins, biofilms, toxic-shock-syndrome toxins, hemolysins, and leukotoxins were also detected. Phylogenetic analysis revealed a shift in the dominant clone from MSSA ST152 to MRSA ST152 over the past decade. The study provides valuable insights into the genomic content of S. aureus from clinical sources in Ghana. The finding of ST152 MRSA in high numbers suggests a shifting epidemiological landscape of these pathogens and continuous surveillance using robust tools like WGS is needed to monitor the rise and spread of these epidemic clones in the country.IMPORTANCESince its emergence in 1959, MRSA has been a significant public health concern, causing infections in both clinical and community settings. Patients with MRSA-related infections experience higher mortality rates due to its ability to evade antimicrobials and immune defenses. In Ghana, understanding the molecular epidemiology of MRSA has been hindered by the lack of appropriate laboratory infrastructure and the limited capacity for molecular data analysis. This study, the largest genomic study of S. aureus in Ghana, addresses this gap by utilizing whole genome sequencing to examine the diversity of circulating S. aureus strains from 10 hospitals. Our findings highlight the predominance of pandemic clones, particularly ST152, and the notable transition of ST152 MSSA to ST152 MRSA over the past decade. The findings from this study supports AMR surveillance efforts in Ghana and emphasize the importance of implementing genomic surveillance using WGS to comprehensively monitor the rise and spread of multi-drug-resitant organisms such as MRSA in the country.
Collapse
Affiliation(s)
- Beverly Egyir
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Christian Owusu-Nyantakyi
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Alfred Bortey
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Grebstad Rabbi Amuasi
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Felicia Amoa Owusu
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - William Boateng
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Hawawu Ahmed
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Justice Kwesi Danso
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Agnes Akosua Gyamaah Oclu
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Quaneeta Mohktar
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | | | | | | | | | - Solomon Asante-Sefa
- Sekondi Public Health Laboratory, Effia Nkwanta Regional Hospital, Takoradi, Ghana
| | | | | | | | | | | | | | - Nicholas Dzifa Dayie
- Department of Medical Microbiology, University of Ghana Medical School, Korle-Bu, Ghana
| | - Noah Obeng-Nkrumah
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana
| | | | | | - Pernille Nilsson
- Research Group for Global Capacity Building, National Food Institute, WHO Collaborating Centre (WHO CC) for Antimicrobial Resistance in Foodborne Pathogens and Genomics, FAO Reference Laboratory (FAO RL) for Antimicrobial Resistance, European Union Reference Laboratory for Antimicrobial Resistance (EURL-AR), Technical University of Denmark, Kongens Lyngby, Denmark
| | - Harriet Affran Bonful
- Department of Epidemiology and Disease Control, School of Public Health, University of Ghana, Accra, Ghana
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Rene S Hendriksen
- Research Group for Global Capacity Building, National Food Institute, WHO Collaborating Centre (WHO CC) for Antimicrobial Resistance in Foodborne Pathogens and Genomics, FAO Reference Laboratory (FAO RL) for Antimicrobial Resistance, European Union Reference Laboratory for Antimicrobial Resistance (EURL-AR), Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
11
|
Freitas PR, Araújo ACJD, Araújo IM, Almeida RS, Oliveira Borges JAD, Lima CMG, Oliveira-Tintino CDM, Paulo CLR, Miranda GM, Araújo-Neto JBD, Almeida-Bezerra JW, Nascimento IJDS, Araújo-Júnior JXD, Silva-Júnior EFD, Aquino TMD, Mendonca Junior FJB, Marinho ES, Santos HSD, Menezes IRAD, Tintino SR, Coutinho HDM. Evaluating efflux pump inhibition in Staphylococcus aureus 1199B strain using thiadiazine-derived compounds: In vitro and in silico approaches. Biochimie 2024:S0300-9084(24)00313-4. [PMID: 39708974 DOI: 10.1016/j.biochi.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
Thiadiazines are heterocyclic compounds known for some pharmacological activities. However, the ability of these compounds and their derivatives to act as antibacterial agents and inhibitors of the efflux system in resistant bacteria remains unknown. This study aims to evaluate the antibacterial and NorA efflux pump inhibitory activities of thiadiazine-derived compounds (IJ14, IJ15, IJ16, IJ17, IJ18, IJ19, and IJ20) against the Staphylococcus aureus 1199B strain. Minimum Inhibitory Concentration (MIC) tests and antibacterial activity assessment through NorA efflux system inhibition were performed using microdilution assays in 96-well plates. Additionally, ethidium bromide (EtBr) fluorescence emission assays were conducted to evaluate efflux system inhibition. The methodology revealed that the IJ17 and IJ20 compounds presented MIC values of 256 and 597.3 μg/mL, respectively. The efflux pump inhibition assessment using the microdilution method showed significant results for all compounds, which also increased the fluorescence rates emitted by EtBr. Consequently, thiadiazine-derived compounds exhibit promising results in targeting a key bacterial resistance mechanism, underscoring the need for further studies, such as molecular tests, to evaluate their mechanism of action and clarify the feasibility and efficacy of these compounds as antibacterial agents.
Collapse
Affiliation(s)
| | | | - Isaac Moura Araújo
- Department of Biological Chemistry, Universidade Regional Do Cariri, Crato, CE, Brazil
| | - Ray Silva Almeida
- Department of Biological Chemistry, Universidade Regional Do Cariri, Crato, CE, Brazil
| | | | | | | | | | | | - José Bezerra de Araújo-Neto
- Postgraduate Program in Biological Sciences, Biosciences Center, Federal University of Pernambuco, Recife, PE, 50740-570, Brazil
| | | | - Igor José Dos Santos Nascimento
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, 57072-900, Maceió, AL, Brazil
| | - João Xavier de Araújo-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, 57072-900, Maceió, AL, Brazil
| | - Edeildo Ferreira da Silva-Júnior
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-900, Maceió, AL, Brazil
| | - Thiago Mendonça de Aquino
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-900, Maceió, AL, Brazil
| | | | | | | | | | - Saulo Relison Tintino
- Department of Biological Chemistry, Universidade Regional Do Cariri, Crato, CE, Brazil
| | | |
Collapse
|
12
|
Rios-Delgado G, McReynolds AKG, Pagella EA, Norambuena J, Briaud P, Zheng V, Munneke MJ, Kim J, Racine H, Carroll RK, Zelzion E, Skaar E, Bose JL, Parker D, Lalaouna D, Boyd JM. The Staphylococcus aureus non-coding RNA IsrR regulates TCA cycle activity and virulence. Nucleic Acids Res 2024:gkae1243. [PMID: 39704109 DOI: 10.1093/nar/gkae1243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/08/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
Staphylococcus aureus has evolved mechanisms to cope with low iron (Fe) availability in host tissues. Staphylococcus aureus uses the ferric uptake transcriptional regulator (Fur) to sense titers of cytosolic Fe. Upon Fe depletion, apo-Fur relieves transcriptional repression of genes utilized for Fe uptake. We demonstrate that an S. aureus Δfur mutant has decreased expression of acnA, which codes for the Fe-dependent enzyme aconitase. This prevents the Δfur mutant from growing with amino acids as sole carbon and energy sources. We used a suppressor screen to exploit this phenotype and determined that a mutation that decreases the transcription of isrR, which produces a regulatory RNA, increased acnA expression, thereby enabling growth. Directed mutation of bases predicted to facilitate the interaction between the acnA transcript and IsrR, decreased the ability of IsrR to control acnA expression in vivo and IsrR bound to the acnA transcript in vitro. IsrR also bound transcripts coding the alternate tricarboxylic acid cycle proteins sdhC, mqo, citZ and citM. Whole-cell metal analyses suggest that IsrR promotes Fe uptake and increases intracellular Fe not ligated by macromolecules. Lastly, we determined that Fur and IsrR promote infection using murine skin and acute pneumonia models.
Collapse
Affiliation(s)
- Gustavo Rios-Delgado
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, 76 Lipman Dr, New Brunswick, NJ 08901, USA
| | - Aubrey K G McReynolds
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Emma A Pagella
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Javiera Norambuena
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, 76 Lipman Dr, New Brunswick, NJ 08901, USA
| | - Paul Briaud
- Department of Biological Sciences, Ohio University, 7 Depot St, Athens, OH 45701, USA
| | - Vincent Zheng
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, 76 Lipman Dr, New Brunswick, NJ 08901, USA
| | - Matthew J Munneke
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, USA
| | - Jisun Kim
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Hugo Racine
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR9002, 15 rue René Descartes, Strasbourg 67000, France
| | - Ronan K Carroll
- Department of Biological Sciences, Ohio University, 7 Depot St, Athens, OH 45701, USA
| | - Ehud Zelzion
- Office of Advanced Research Computing, Rutgers University, 96 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Eric Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, USA
| | - Jeffrey L Bose
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - David Lalaouna
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR9002, 15 rue René Descartes, Strasbourg 67000, France
| | - Jeffrey M Boyd
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, 76 Lipman Dr, New Brunswick, NJ 08901, USA
| |
Collapse
|
13
|
Zomorodi AR, Motamedifar M, Rahmanian K, Shakeri M, Hajikhani B, Heidari H, Mansury D, Jahromi AS. Investigation of integron classes 1, 2, and 3 among multi-drug resistant Staphylococcus aureus isolates in Iran: a multi-center study. BMC Infect Dis 2024; 24:1430. [PMID: 39696000 DOI: 10.1186/s12879-024-10311-5] [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: 09/01/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Rising methicillin-resistant Staphylococcus aureus (MRSA) poses a global health threat, contributing to serious infections with high mortality rates. Integrons are recognized as significant genetic elements in disseminating multidrug-resistant (MDR) strains. This study focuses on assessing the prevalence of integron classes 1, 2, and 3 in S. aureus strains from four major cities in Iran. METHODS This cross-sectional study analyzed 183 S. aureus isolates from Shiraz, Tehran, Isfahan, and Yazd in Iran. The isolates were identified using specific biochemical and molecullar tests. The Kirby-Bauer disc diffusion method and microbroth dilution method were employed to determine the susceptibility of the isolates to relevant antibiotics and vancomycin, respectively. The macrolide-lincosamide-streptogramin B (MLSB) resistance phenotype was also evaluated using the D-test. All isolates were sought for presence of the intI1, intI2, and intI3 genes. RESULTS Among 183 S. aureus isolates, high resistance rates were noted: 86.3% for erythromycin, 66.1% for ciprofloxacin, and 61.7% for clindamycin, while all isolates were susceptible to linezolid and vancomycin. Of the 183 isolates, 59.6% were identified as MRSA and 78.1% as MDR. According to the D-test results, 112/183 (61.2%), 29/183 (15.8%), 25/183 (13.7%), and 17/183 (9.7%) of S. aureus isolates showed constitutive resistance-MLSB, inducible resistance-MLSB, sensitive, and resistance to macrolide-streptogramin B (MS) phenotypes, respectively. The intI1 gene was found in 14 out of 183 S. aureus isolates (7.6%), while none were positive for the intI2 or intI3 genes. Notably, 11/14 (78.5%) and 13/14 (92.8%) intI1-positive isolates were MRSA and MDR, respectively. CONCLUSIONS The distribution of MRSA and MDR S. aureus isolates in Iran seems concerning. Although the prevalence of intI1 was not as high as in prior studies, almost all S. aureus harbored the intI1 gene were MRSA and MDR.
Collapse
Affiliation(s)
- Abolfazl Rafati Zomorodi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Motamedifar
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Masihollah Shakeri
- Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Bahareh Hajikhani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Heidari
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Davood Mansury
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
14
|
Ko EM, Kim S. Genome sequences of Staphylococcus aureus RN4220. Microbiol Resour Announc 2024:e0046224. [PMID: 39679716 DOI: 10.1128/mra.00462-24] [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: 05/03/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
In this study, we present whole genome sequences of Staphylococcus aureus RN4220. Using PacBio DNA sequencing and de novo assembly, we obtained a single contig of 2,697,647 bp, which includes 2,652 predicted genes comprising protein-coding sequences, tRNAs, and rRNAs.
Collapse
Affiliation(s)
- Eon-Min Ko
- Division of Bacterial Disease Research, Center for Infectious Disease Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, South Korea
| | - Seonghan Kim
- Division of Bacterial Disease Research, Center for Infectious Disease Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, South Korea
| |
Collapse
|
15
|
Ayoub N, Upadhyay A, Tête A, Pietrancosta N, Munier-Lehmann H, O'Sullivan TP. Synthesis, evaluation and mechanistic insights of novel IMPDH inhibitors targeting ESKAPEE bacteria. Eur J Med Chem 2024; 280:116920. [PMID: 39369481 DOI: 10.1016/j.ejmech.2024.116920] [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: 08/11/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/08/2024]
Abstract
Antimicrobial resistance poses a significant threat to global health, necessitating the development of novel therapeutic agents with unique mechanisms of action. Inosine 5'-monophosphate dehydrogenase (IMPDH), an essential enzyme in guanine nucleotide biosynthesis, is a promising target for the discovery of new antimicrobial agents. High-throughput screening studies have previously identified several urea-based leads as potential inhibitors, although many of these are characterised by reduced chemical stability. In this work, we describe the design and synthesis of a series of heteroaryl-susbtituted analogues and the evaluation of their inhibitory potency against IMPDHs. Our screening targets ESKAPEE pathogens, including Pseudomonas aeruginosa, Staphylococcus aureus and Escherichia coli. Several analogues with submicromolar inhibitory potency are identified and show no inhibitory potency on human IMPDH nor cytotoxic effects on human cells. Kinetic studies revealed that these molecules act as noncompetitive inhibitors with respect to the substrates and ligand virtual docking simulations provided insights into the binding interactions at the interface of the NAD+ and IMP binding sites on IMPDH.
Collapse
Affiliation(s)
- Nour Ayoub
- Université Paris Cité, INSERM UMRS-1124, Institut Pasteur, Structural Biology and Chemistry Department, F-75006, Paris, France
| | - Amit Upadhyay
- School of Chemistry, University College Cork, Cork, Ireland; School of Pharmacy, University College Cork, Cork, Ireland; Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland
| | - Arnaud Tête
- Université Paris Cité, INSERM UMRS-1124, F-75006, Paris, France
| | - Nicolas Pietrancosta
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, F-75005, Paris, France; Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), F-75005, Paris, France
| | - Hélène Munier-Lehmann
- Université Paris Cité, INSERM UMRS-1124, Institut Pasteur, Structural Biology and Chemistry Department, F-75006, Paris, France.
| | - Timothy P O'Sullivan
- School of Chemistry, University College Cork, Cork, Ireland; School of Pharmacy, University College Cork, Cork, Ireland; Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland.
| |
Collapse
|
16
|
Wang Z, An Z, Richel A, Huang M, Gou X, Xu D, Zhang M, Mo H, Hu L, Zhou X. Ferrous sulfate remodels the properties of sodium alginate-based hydrogel and facilitates the healing of wound infection caused by MRSA. Carbohydr Polym 2024; 346:122554. [PMID: 39245535 DOI: 10.1016/j.carbpol.2024.122554] [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: 05/13/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 09/10/2024]
Abstract
Frequent occurrence of wound infection caused by multiple-resistant bacteria (MRB) has posed a serious challenge to the current healthcare system relying on antibiotics. The development of novel antimicrobial materials with high safety and efficacy to heal wound infection is of great importance in combating this crisis. Herein, we prepared a promising antibacterial hydrogel by cross-linking ferrous ions (Fe2+) with the deprotonated carboxyl anion in sodium alginate (Na-ALG) to cure wound infections caused by methicillin-resistant Staphylococcus aureus (MRSA). Interestingly, ferrous-modified Na-ALG (Fe-ALG) hydrogel demonstrated better properties compared to the traditional Na-ALG-based hydrogels, including injectability, self-healing, appropriate fluidity, high-water retention, potent MRSA-killing efficacy, and excellent biocompatibility. Importantly, the addition of Fe2+ enhances the antibacterial efficacy of the Na-ALG hydrogel, enabling it to effectively eliminate MRSA and accelerate the healing of antibiotic-resistant bacterial-infected wounds in a remarkably short period (10 days). This modification not only facilitates wound closure and fur generation, but also mitigates systemic inflammation, thereby effectively impeding the spread of MRSA to the lungs. Taken together, Fe-ALG hydrogel is a promising therapeutic material for treating wound infections by Staphylococcus aureus, especially by antibiotic-resistant strains like MRSA.
Collapse
Affiliation(s)
- Zhen Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China; Laboratory of Biomass and Green Technologies, University of Liege, Belgium
| | - Zinuo An
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Aurore Richel
- Laboratory of Biomass and Green Technologies, University of Liege, Belgium
| | - Minmin Huang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Dan Xu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Min Zhang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Haizhen Mo
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Liangbin Hu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China.
| | - Xiaohui Zhou
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
17
|
Lin J, Wang S, Cao M, Pan Y, Dai Z, Wang Y, Jin Z, Xu Y, Wu J, Liu J, Yuan Z, Zhang Y, Gui L. Photoresponsive nitric oxide photocage/photodynamic integrated prodrug for advanced management of drug-resistant bacteria-infected wound therapy. Bioorg Chem 2024; 154:108062. [PMID: 39705939 DOI: 10.1016/j.bioorg.2024.108062] [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: 08/14/2024] [Revised: 11/24/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
Single treatment methods for wound infections caused by drug-resistant bacteria often fail to simultaneously achieve high antimicrobial efficacy and wound healing efficiency. Therefore, it is crucial to develop a small molecule prodrug that can achieve both goals, in this context, we have engineered a green-light-activated nitric oxide photocage/photodynamic therapeutic prodrug, designed to release NO, which not only potentiates antimicrobial efficacy but also facilitates collagen accumulation at the wound interface, thereby expediting the wound healing process. Additionally, it mitigates tissue inflammation by suppressing the NF-κB signaling pathway. In addition, the introduction of the photosensitiser rhodamine can be used for antimicrobial photodynamic therapy (aPDT) against Methicillin-resistant Staphylococcus aureus (MRSA). aPDT exhibits enhanced antimicrobial efficacy by specifically targeting and disrupting bacterial cell membranes. Notably, this treatment significantly reduced bacterial viability in vitro and accelerated healing of MRSA-induced wound infections even at low concentrations. The developed NO photocage/photodynamic prodrug RhB-NO-1 is biocompatible and has demonstrated the ability to significantly accelerate the healing process of diabetic foot ulcers (DFU), making it a promising next-generation pharmacological treatment for chronic wound management.
Collapse
Affiliation(s)
- Jingjing Lin
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, 93 East Jichuan Road, Hailing District, Taizhou, Jiangsu 225300, China; Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China
| | - Shiya Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China
| | - Mingyi Cao
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China
| | - Yuhan Pan
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China
| | - Zhiyue Dai
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China
| | - Yuxin Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China
| | - Zhiyuan Jin
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China
| | - Yue Xu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China
| | - Jianbing Wu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China
| | - Ji Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Yanliang Zhang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210006, China.
| | - Lijuan Gui
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, 93 East Jichuan Road, Hailing District, Taizhou, Jiangsu 225300, China.
| |
Collapse
|
18
|
Vanbaelen T, Manoharan-Basil SS, Kenyon C. Stop classifying Neisseria gonorrhoeae as an obligate pathogen in men who have sex with men: A viewpoint. Int J STD AIDS 2024:9564624241306600. [PMID: 39661376 DOI: 10.1177/09564624241306600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Affiliation(s)
- Thibaut Vanbaelen
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | | | - Chris Kenyon
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- University of Cape Town, Cape Town, South Africa
| |
Collapse
|
19
|
Salikin NH, Keong LC, Azemin WA, Philip N, Yusuf N, Daud SA, Rashid SA. Combating multidrug-resistant (MDR) Staphylococcus aureus infection using terpene and its derivative. World J Microbiol Biotechnol 2024; 40:402. [PMID: 39627623 DOI: 10.1007/s11274-024-04190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024]
Abstract
Multidrug-resistant (MDR) Staphylococcus aureus represents a major global health issue resulting in a wide range of debilitating infections and fatalities. The slow progression of new antibiotics, limited choices for treatment, and scarcity of new drug approvals create immense obstacles in new drug line development. S. aureus poses a significant public health risk, due to the emergence of methicillin-resistant (MRSA) and vancomycin-resistant strains (VRSA), necessitating novel antibiotics for effective control management. Current studies are delving into the terpenes' potential as an antimicrobial agent, indicating positive prospects as promising substitutes or complementary to conventional antibiotics. Concurrent reactions of terpenes with conventional antibiotics create synergistic effects that significantly enhance antibiotic efficacy. Accumulated evidence has shown that while efflux pump (e.g., NorA, TetK, and MepA) is revealed as an essential defense of S. aureus against antibiotics, terpene and its derivative act as its potent inhibitor, suggesting the promising potential of terpenes in combating those infectious pathogens. Furthermore, pronounced cell membrane disruptive activity and antibiofilm properties by terpenes have been exerted, signifying their significance as promising prevention against microbial pathogenesis and antimicrobial resistance. This review provides an overview of the potential of terpenes and their derivatives in combating S. aureus infections, highlighting their potential mechanisms of action (MOA), synergistic effects with conventional antibiotics, and challenges in clinical translation. The unique properties of terpenes offer an opportunity for their use in developing an exceptional defense strategy against antibiotic-resistant S. aureus.
Collapse
Affiliation(s)
- Nor Hawani Salikin
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Lee Chee Keong
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Wan-Atirah Azemin
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Noraini Philip
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Nurhaida Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Syiah Kuala University, Aceh, Indonesia
| | - Siti Aisyah Daud
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Syarifah Ab Rashid
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia.
| |
Collapse
|
20
|
Saporta R, Nielsen EI, Menetrey A, Cameron DR, Nicolas-Metral V, Friberg LE. Model-based translation of results from in vitro to in vivo experiments for afabicin activity against Staphylococcus aureus. J Antimicrob Chemother 2024; 79:3150-3159. [PMID: 39315768 PMCID: PMC11638087 DOI: 10.1093/jac/dkae334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Translation of experimental data on antibiotic activity typically relies on pharmacokinetic/pharmacodynamic (PK/PD) indices. Model-based approaches, considering the full antibiotic killing time course, could be an alternative. OBJECTIVES To develop a mechanism-based modelling framework to assess the in vitro and in vivo activity of the FabI inhibitor antibiotic afabicin, and explore the ability of a model built on in vitro data to predict in vivo outcome. METHODS A PK/PD model was built to describe bacterial counts from 162 static in vitro time-kill curves evaluating the effect of afabicin desphosphono, the active moiety of the prodrug afabicin, against 21 Staphylococcus aureus strains. Combined with a mouse PK model, outcomes of afabicin doses of 0.011-190 mg/kg q6h against nine S. aureus strains in a murine thigh infection model were predicted, and thereafter refined by estimating PD parameters. RESULTS A sigmoid Emax model, with EC50 scaled by the MIC described the afabicin desphosphono killing in vitro. This model predicted, without parameter re-estimation, the in vivo bacterial counts at 24 h within a ±1 log margin for most dosing groups. When parameters were allowed to be estimated, EC50 was 38%-45% lower in vivo, compared with in vitro, within the studied MIC range. CONCLUSIONS The developed PK/PD model described the time course of afabicin activity across experimental conditions and bacterial strains. This model showed translational capacity as parameters estimated on in vitro time-kill data could well predict the in vivo outcome for a wide variety of doses in a mouse thigh infection model.
Collapse
Affiliation(s)
| | | | - Annick Menetrey
- Translational Medicine Department, Debiopharm International SA, Lausanne, Switzerland
| | - David R Cameron
- Translational Medicine Department, Debiopharm International SA, Lausanne, Switzerland
| | | | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
21
|
Li JG, Zhang CJ, Liang LY, Lu TY, Zhong LG, Zhong WC, Niu CY, Sun J, Liao XP, Zhou YF. Assessment of anti-MRSA activity of auranofin and florfenicol combination: a PK/PD analysis. J Appl Microbiol 2024; 135:lxae299. [PMID: 39694699 DOI: 10.1093/jambio/lxae299] [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: 07/07/2024] [Revised: 11/27/2024] [Accepted: 12/17/2024] [Indexed: 12/20/2024]
Abstract
AIMS Methicillin-resistant Staphylococcus aureus (MRSA) is an important zoonotic pathogen with multidrug-resistant phenotypes increasingly prevalent in both human and veterinary clinics. This study evaluated the potential of auranofin (AF) as an antibiotic adjuvant to enhance the anti-MRSA activity of florfenicol (FFC) and established a pharmacokinetic/pharmacodynamic (PK/PD) model to compare the efficacy of FFC alone or in combination with AF against MRSA. METHODS AND RESULTS We observed an increased susceptibility and significant synergistic effects of MRSA to FFC in the presence of AF. The combination treatment of FFC and AF significantly inhibited MRSA biofilm formation and decreased the metabolic activity of mature biofilms. Importantly, AF fully restored the efficacy of FFC in both Galleria mellonella larvae and murine models. PK/PD studies demonstrated that the AUC24h/MIC targets required to achieve the bacteriostatic and bactericidal effects were significantly lower with the combination therapy compared to florfenicol monotherapy. CONCLUSIONS These results reveal the potential of AF as a novel adjuvant to improve the efficacy of FFC in treating MRSA invasive infections and provide valuable PK/PD insights for designing effective combination therapies.
Collapse
Affiliation(s)
- Jian-Guo Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Chuan-Jian Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Liu-Yan Liang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Ting-Yin Lu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Long-Gen Zhong
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Wei-Cheng Zhong
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Chao-Yan Niu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, China
| | - Xiao-Ping Liao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, China
| | - Yu-Feng Zhou
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, China
| |
Collapse
|
22
|
Khan BA, Roy S, Tahsin N, Baidya K, Das KC, Islam MS, Ahsan N, Salam A. Antibiotic resistance of bioaerosols in particulate matter from indoor environments of the hospitals in Dhaka Bangladesh. Sci Rep 2024; 14:29884. [PMID: 39623045 PMCID: PMC11612278 DOI: 10.1038/s41598-024-81376-0] [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: 06/09/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
The emergence and spread of antibiotic resistance in microorganisms pose significant challenges to public health, especially in hospitals. This study investigated the existence or occurrence of bacterial bioaerosol and their antibiotic resistance patterns in particulate matter (PM) collected from hospitals in the greater Dhaka region, Bangladesh. The real-time particulate matter concentrations (PM1.0, PM2.5, and PM10) were measured in four hospitals and two ambient locations. Air sampling was conducted using a filter-based method with a low-volume air sampler, while AEROCET-531 S (USA) was employed to monitor particulate matter concentrations. Bacterial bioaerosol concentration was determined using a culture-based method, and eleven bacterial species, including nine individual species, i.e., Staphylococcus aureus, Pseudomonas aeruginosa, P. stutzeri, Bacillus cereus, Acinetobacter schindleri, Proteus vulgaris, B. subtilis, Escherichia coli, and B. aerius, were isolated. Antibiotic susceptibility testing was conducted using the Kirby-Bauer disk diffusion method with 21 antibiotics. Bacterial isolates were detected using partial sequencing of the 16 S rRNA gene. Bioaerosol concentration ranged from 194.65 ± 22.48 CFU/m3 to 948.39 ± 84.14 CFU/m3, showing significant correlations with PM1.0 and PM2.5 concentrations (R2 = 0.80 and 0.85, respectively). All bacterial isolates collected from the hospitals exhibited resistance against four or more antibiotics, indicating multidrug resistance (MDR). Notably, the bacterial isolates displayed the highest resistance rate against ampicillin (90.90%), azithromycin (81.81%), erythromycin (81.81%), cefixime (81.81%), and cotrimoxazole (54.54%), among the tested antibiotics. Except B. aerius, all other bacterial isolates were associated with hospital-acquired infections (HAIs). These findings highlight the high rates of antibiotic resistance, underscoring the pressing requirement for infection control measures and continuous surveillance strategies in hospital settings. These findings emphasize the necessity for global hospital infection control strategies focusing airborne multidrug-resistant microorganisms.
Collapse
Affiliation(s)
- Badhon Ali Khan
- Department of Chemistry, Faculty of Science, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Shatabdi Roy
- Department of Chemistry, Faculty of Science, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Nishat Tahsin
- Department of Chemistry, Faculty of Science, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Kalpana Baidya
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Keshob Chandra Das
- Molecular Biotechnology Division, National Institute of Biotechnology (NIB), Ganakbari, Savar, Dhaka, 1349, Ashulia, Bangladesh
| | - Md Safiqul Islam
- Department of Chemistry, Faculty of Science, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Nazmul Ahsan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Abdus Salam
- Department of Chemistry, Faculty of Science, University of Dhaka, Dhaka, 1000, Bangladesh.
| |
Collapse
|
23
|
McReynolds AKG, Pagella EA, Ridder MJ, Rippee O, Clark Z, Rekowski MJ, Pritchard MT, Bose JL. YjbH contributes to Staphylococcus aureus skin pathology and immune response through Agr-mediated α-toxin regulation. Virulence 2024; 15:2399798. [PMID: 39229975 PMCID: PMC11404607 DOI: 10.1080/21505594.2024.2399798] [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: 06/06/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/05/2024] Open
Abstract
Staphylococcus aureus is the most common cause of skin and soft tissue infections (SSTIs) with Methicillin-Resistant S. aureus (MRSA) strains being a major contributor in both community and hospital settings. S. aureus relies on metabolic diversity and a large repertoire of virulence factors to cause disease. This includes α-hemolysin (Hla), an integral player in tissue damage found in various models, including SSTIs. Previously, we identified a role for the Spx adapter protein, YjbH, in the regulation of several virulence factors and as an inhibitor of pathogenesis in a sepsis model. In this study, we found that YjbH is critical for tissue damage during SSTI, and its absence leads to decreased proinflammatory chemokines and cytokines in the skin. We identified no contribution of YjbI, encoded on the same transcript as YjbH. Using a combination of reporters and quantitative hemolysis assays, we demonstrated that YjbH impacts Hla expression and activity both in vitro and in vivo. Additionally, expression of Hla from a non-native promoter reversed the tissue damage phenotype of the ΔyjbIH mutant. Lastly, we identified reduced Agr activity as the likely cause for reduced Hla production in the ΔyjbH mutant. This work continues to define the importance of YjbH in the pathogenesis of S. aureus infection as well as identify a new pathway important for Hla production.
Collapse
Affiliation(s)
- Aubrey K. G. McReynolds
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Emma A. Pagella
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Miranda J. Ridder
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Olivia Rippee
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Zachary Clark
- The Mass Spectrometry and Proteomics Core, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michaella J. Rekowski
- The Mass Spectrometry and Proteomics Core, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michele T. Pritchard
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeffrey L. Bose
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
24
|
Sivori F, Cavallo I, Truglio M, De Maio F, Sanguinetti M, Fabrizio G, Licursi V, Francalancia M, Fraticelli F, La Greca I, Lucantoni F, Camera E, Mariano M, Ascenzioni F, Cristaudo A, Pimpinelli F, Di Domenico EG. Staphylococcus aureus colonizing the skin microbiota of adults with severe atopic dermatitis exhibits genomic diversity and convergence in biofilm traits. Biofilm 2024; 8:100222. [PMID: 39381779 PMCID: PMC11460521 DOI: 10.1016/j.bioflm.2024.100222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder exacerbated by Staphylococcus aureus colonization. The specific factors that drive S. aureus overgrowth and persistence in AD remain poorly understood. This study analyzed skin barrier functions and microbiome diversity in lesional (LE) and non-lesional (NL) forearm sites of individuals with severe AD compared to healthy control subjects (HS). Notable differences were found in transepidermal water loss, stratum corneum hydration, and microbiome composition. Cutibacterium was more prevalent in HS, while S. aureus and S. lugdunensis were predominantly found in AD LE skin. The results highlighted that microbial balance depends on inter-species competition. Specifically, network analysis at the genus level demonstrated that overall bacterial correlations were higher in HS, indicating a more stable microbial community. Notably, network analysis at the species level revealed that S. aureus engaged in competitive interactions in NL and LE but not in HS. Whole-genome sequencing (WGS) showed considerable genetic diversity among S. aureus strains from AD. Despite this variability, the isolates exhibited convergence in key phenotypic traits such as adhesion and biofilm formation, which are crucial for microbial persistence. These common phenotypes suggest an adaptive evolution, driven by competition in the AD skin microenvironment, of S. aureus and underscoring the interplay between genetic diversity and phenotypic convergence in microbial adaptation.
Collapse
Affiliation(s)
- Francesca Sivori
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Ilaria Cavallo
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Mauro Truglio
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Flavio De Maio
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli” IRCSS, Rome, Italy
| | - Maurizio Sanguinetti
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli” IRCSS, Rome, Italy
| | - Giorgia Fabrizio
- Department of Biology and Biotechnology “C. Darwin” Sapienza University of Rome, Rome, Italy
| | - Valerio Licursi
- Institute of Molecular Biology and Pathology, National Research Council of Italy, Rome, Italy
| | - Massimo Francalancia
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Fulvia Fraticelli
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Ilenia La Greca
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Federica Lucantoni
- Department of Biology and Biotechnology “C. Darwin” Sapienza University of Rome, Rome, Italy
| | - Emanuela Camera
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Maria Mariano
- Clinical Dermatology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Fiorentina Ascenzioni
- Department of Biology and Biotechnology “C. Darwin” Sapienza University of Rome, Rome, Italy
| | - Antonio Cristaudo
- Clinical Dermatology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Fulvia Pimpinelli
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Enea Gino Di Domenico
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| |
Collapse
|
25
|
Straub C, Taylor W, French NP, Murdoch DR, Priest P, Anderson T, Scott P. Zoonotic transmission of asymptomatic carriage Staphylococcus aureus on dairy farms in Canterbury, New Zealand. Microb Genom 2024; 10:001318. [PMID: 39630492 PMCID: PMC11616781 DOI: 10.1099/mgen.0.001318] [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: 04/17/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024] Open
Abstract
Zoonotic pathogen transmission is of growing concern globally, with agricultural intensification facilitating interactions between humans, livestock and wild animals. Staphylococcus aureus is a major human pathogen, but it also causes mastitis in dairy cattle, leading to an economic burden on the dairy industry. Here, we investigated transmission within and between cattle and humans, including potential zoonotic transmission of S. aureus isolated from cattle and humans from three dairy farms and an associated primary school in New Zealand. Nasal swabs (N=170) were taken from healthy humans. Inguinal and combined nasal/inguinal swabs were taken from healthy cattle (N=1163). Whole-genome sequencing was performed for 96 S. aureus isolates (44 human and 52 cattle). Multilocus sequence typing and assessments of antimicrobial resistance and virulence were carried out. Potential within- and across-species transmission events were determined based on single nucleotide polymorphisms (SNPs). Thirteen potential transmission clusters were detected, with 12 clusters restricted to within-species and one potential zoonotic transmission cluster (ST5). Potential transmission among cattle was mostly limited to single age groups, likely because different age groups are managed separately on farms. While the prevalence of antimicrobial resistance (AMR) was low among both bovine and human isolates, the discovery of an extended-spectrum beta-lactamase gene (bla TEM-116) in a bovine isolate was concerning. This study provides evidence around frequency and patterns of potential transmission of S. aureus on dairy farms and highlights the AMR and virulence profile of asymptomatic carriage S. aureus isolates.
Collapse
Affiliation(s)
- Christina Straub
- The Institute of Environmental Science and Research, Auckland, New Zealand
- Genomics Aotearoa, Dunedin, New Zealand
- Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - William Taylor
- The Institute of Environmental Science and Research, Christchurch, New Zealand
| | - Nigel P. French
- Tāwharau Ora, School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - David R. Murdoch
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Patricia Priest
- Department of Preventive and Social Medicine, University of Otago, Dunedin, New Zealand
| | - Trevor Anderson
- Microbiology Department, Canterbury Health Laboratories, Te Whatu Ora – Health New Zealand Waitaha, Christchurch, New Zealand
| | - Pippa Scott
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
26
|
Jian Y, Chen T, Yang Z, Xiang G, Xu K, Wang Y, Zhao N, He L, Liu Q, Li M. Small regulatory RNA RSaX28 promotes virulence by reinforcing the stability of RNAIII in community-associated ST398 clonotype Staphylococcus aureus. Emerg Microbes Infect 2024; 13:2341972. [PMID: 38597192 PMCID: PMC11034457 DOI: 10.1080/22221751.2024.2341972] [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: 12/28/2023] [Accepted: 04/06/2024] [Indexed: 04/11/2024]
Abstract
Staphylococcus aureus (S. aureus) is a notorious pathogen that cause metastatic or complicated infections. Hypervirulent ST398 clonotype strains, remarkably increased in recent years, dominated Community-associated S. aureus (CA-SA) infections in the past decade in China. Small RNAs like RNAIII have been demonstrated to play important roles in regulating the virulence of S. aureus, however, the regulatory roles played by many of these sRNAs in the ST398 clonotype strains are still unclear. Through transcriptome screening and combined with knockout phenotype analysis, we have identified a highly transcribed sRNA, RSaX28, in the ST398 clonotype strains. Sequence analysis revealed that RSaX28 is highly conserved in the most epidemic clonotypes of S. aureus, but its high transcription level is particularly prominent in the ST398 clonotype strains. Characterization of RSaX28 through RACE and Northern blot revealed its length to be 533nt. RSaX28 is capable of promoting the hemolytic ability, reducing biofilm formation capacity, and enhancing virulence of S. aureus in the in vivo murine infection model. Through IntaRNA prediction and EMSA validation, we found that RSaX28 can specifically interact with RNAIII, promoting its stability and positively regulating the translation of downstream alpha-toxin while inhibiting the translation of Sbi, thereby regulating the virulence and biofilm formation capacity of the ST398 clonotype strains. RSaX28 is an important virulence regulatory factor in the ST398 clonotype S. aureus and represents a potential important target for future treatment and immune intervention against S. aureus infections.
Collapse
Affiliation(s)
- Ying Jian
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Tianchi Chen
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Ziyu Yang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Guoxiu Xiang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Kai Xu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yanan Wang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Na Zhao
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Lei He
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qian Liu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
27
|
Evans DC, Khamas AB, Payne-Dwyer A, Wollman AJ, Rasmussen KS, Klitgaard JK, Kallipolitis B, Leake MC, Meyer RL. Cooperation between coagulase and von willebrand factor binding protein in Staphylococcus aureus fibrin pseudocapsule formation. Biofilm 2024; 8:100233. [PMID: 39555140 PMCID: PMC11564979 DOI: 10.1016/j.bioflm.2024.100233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/27/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024] Open
Abstract
The major human pathogen Staphylococcus aureus forms biofilms comprising of a fibrin network that increases attachment to surfaces and shields bacteria from the immune system. It secretes two coagulases, Coagulase (Coa) and von Willebrand factor binding protein (vWbp), which hijack the host coagulation cascade and trigger the formation of this fibrin clot. However, it is unclear how Coa and vWbp contribute differently to the localisation and dynamics of clot assembly in growing biofilms. Here, we address this question using high-precision time-resolved confocal microscopy of fluorescent fibrin to establish the spatiotemporal dynamics of fibrin clot formation in functional biofilms. We also use fluorescent fusion proteins to visualise the locations of Coa and vWbp in biofilms using both confocal laser scanning and high resolution highly inclined and laminated optical sheet microscopy. We visualise and quantify the spatiotemporal dynamics of fibrin production during initiation of biofilms in plasma amended with fluorescently labelled fibrinogen. We find that human serum stimulates coagulase production, and that Coa and vWbp loosely associate to the bacterial cell surface. Coa localises to cell surfaces to produce a surface-attached fibrin pseudocapsule but can diffuse from cells to produce matrix-associated fibrin. vWbp produces matrix-associated fibrin in the absence of Coa, and furthermore accelerates pseudocapsule production when Coa is present. Finally, we observe that fibrin production varies across the biofilm. A sub-population of non-dividing cells does not produce any pseudocapsule but remains within the protective extended fibrin network, which could be important for the persistence of S. aureus biofilm infections as antibiotics are more effective against actively growing cells. Our findings indicate a more cooperative role between Coa and vWbp in building fibrin networks than previously thought, and a bet-hedging cell strategy where some cells produce biofilm matrix while others do not, but instead assume a dormant phenotype that could be associated with antibiotic tolerance.
Collapse
Affiliation(s)
- Dominique C.S. Evans
- School of Physics, Engineering and Technology, University of York, York, UK
- Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus, Denmark
| | - Amanda B. Khamas
- Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus, Denmark
| | - Alex Payne-Dwyer
- School of Physics, Engineering and Technology, University of York, York, UK
| | - Adam J.M. Wollman
- School of Physics, Engineering and Technology, University of York, York, UK
- Department of Biology, University of York, York, UK
| | - Kristian S. Rasmussen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Janne K. Klitgaard
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Birgitte Kallipolitis
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Mark C. Leake
- School of Physics, Engineering and Technology, University of York, York, UK
- Department of Biology, University of York, York, UK
| | - Rikke L. Meyer
- Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus, Denmark
| |
Collapse
|
28
|
Larsen TG, Samaniego Castruita JA, Worning P, Westh H, Bartels MD. Within-host genomic evolution of methicillin-resistant Staphylococcus aureus in long-term carriers. Appl Microbiol Biotechnol 2024; 108:95. [PMID: 38212970 PMCID: PMC10784349 DOI: 10.1007/s00253-023-12932-3] [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: 08/01/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 01/13/2024]
Abstract
Assessing the genomic evolution of Staphylococcus aureus can help us understand how the bacteria adapt to its environment. In this study, we aimed to assess the mutation rate within 144 methicillin-resistant Staphylococcus aureus (MRSA) carriers with a carriage time from 4 to 11 years, including some carriers who belonged to the same households. We found that 23 of the 144 individuals had completely different MRSA types over time and were therefore not long-term carriers of the same MRSA. From the remaining 121 individuals, we performed whole-genome sequencing (WGS) on 424 isolates and then compared these pairwise using core genome multilocus sequence typing (cgMLST) and single-nucleotide polymorphism (SNP) analyses. We found a median within-host mutation rate in long-term MRSA carriers of 4.9 (3.4-6.9) SNPs/genome/year and 2.7 (1.8-4.2) allelic differences/genome/year, when excluding presumed recombination. Furthermore, we stratified the cohort into subgroups and found no significant difference between the median mutation rate of members of households, individuals with presumed continued exposure, e.g., from travel and persons without known continued exposure. Finally, we found that SNPs occurred at random within the genes in our cohort. KEY POINTS: • Median mutation rate within long-term MRSA carriers of 4.9 (3.4-6.9) SNPs/genome/year • Similar median mutation rates in subgroups (households, travelers) • No hotspots for SNPs within the genome.
Collapse
Affiliation(s)
- Tine Graakjær Larsen
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | | | - Peder Worning
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Henrik Westh
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mette Damkjær Bartels
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
29
|
Dao TL, Hoang VT, Gautret P. Respiratory Carriage of Methicillin-Resistant Staphylococcus aureus-Encoding Gene in Hajj Pilgrims. J Epidemiol Glob Health 2024; 14:1662-1667. [PMID: 39470975 DOI: 10.1007/s44197-024-00322-5] [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: 10/02/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024] Open
Abstract
OBJECTIVES To assess the carriage of methicillin-resistant Staphylococcus aureus-encoding genes (MRSA) among French Hajj pilgrim cohorts. METHODS A prospective cohort study was conducted on pilgrims from Marseille during the 2014 to 2018 Hajj. Respiratory samples were collected before and after the pilgrimage. S. aureus and then MRSA-encoding genes were identified using real-time PCR. RESULTS A total of 606 pilgrims were included with a sex ratio of 1:1.3 with a median age of 61 years (interquartile = 56-66 years, range = 21-88 years). A total of 511/606 (84.3%) pilgrims presented at least one respiratory symptom during their pilgrimage. Cough was the most frequent symptom occurring in 76.2% of pilgrims, followed by sore throat (57.6%), rhinitis (54.6%), and voice failure (36.3%). 87 (14.4%) were positive with S. aureus before travelling. On return, 130/606 (21.4%) participants were positive. The acquisition rate of S. aureus was 13.0% (79/606). The prevalence of MRSA pre- and post-travel and acquisition rate was 4.1% (25/606), 10.6% (62/606), and 8.2% (50/606), respectively. All MRSA were positive with mecA gene. No case was positive with mecC. CONCLUSION Our study highlights the importance of surveillance and infection control measures during mass gatherings such as the Hajj to mitigate the spread of infectious pathogens, including antimicrobial-resistant bacteria like MRSA. Further research is warranted to elucidate the specific factors contributing to S. aureus and MRSA transmission during the pilgrimage and to inform targeted interventions aimed at reducing the burden of MRSA infection among pilgrims.
Collapse
Affiliation(s)
- Thi Loi Dao
- Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam
| | - Van Thuan Hoang
- Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam.
| | - Philippe Gautret
- Aix Marseille Univ, AP-HM, SSA, RITMES, Marseille, France.
- IHU-Méditerranée Infection, Marseille, France.
| |
Collapse
|
30
|
Alhamdi HW, Anazi HK, Mokhtar FA, Elhawary SS, Elbehairi SEI, Alfaifi MY, Shati AA, Fahmy LI, Elekhnawy E, Hassan A, Negm WA, Fahmy SA, Selim N. Panicum maximum Jacq. mediated green synthesis of silver nanoparticles: synthesis, characterization, and biological activities supported by molecular docking. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:411-425. [PMID: 39193730 DOI: 10.1080/21691401.2024.2395811] [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: 04/15/2024] [Revised: 05/28/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024]
Abstract
This study uses the aerial parts of Panicum maximum total extract (PMTE) to synthesize silver nanoparticles (AgNPs) in an environmentally friendly manner. TEM, SEM, FTIR, X-ray powder diffraction (XRD), Zeta potential, UV, and FTIR were used to characterize the green silver nanoparticles (PM-AgNPs). PM-AgNPs were evaluated as anticancer agents compared to (PMTE) against breast (MCF-7), lung (A549), and ovary adenocarcinoma (SKOV3) human tumour cells. The antibacterial activity of AgNPs was assessed against Staphylococcus aureus isolates. The PM-AgNPs had an absorbance of 418 nm, particle size of 15.18 nm, and zeta potential of -22.4 mV, ensuring the nanosilver's stability. XRD evaluated the crystallography nature of the formed PM-AgNPs. The cytotoxic properties of PM-AgNPs on MCF-7 and SKOV 3 were the strongest, with IC50s of 0.13 ± 0.015 and 3.5 ± 0.5 g/ml, respectively, as compared to A549 (13 ± 3.2 µg/mL). The increase in the apoptotic cells was 97.79 ± 1.61 and 96.6 ± 1.91% for MCF-7 and SKOV3 cell lines, respectively. PM-AgNPs were found to affect the membrane integrity and membrane permeability of 50 and 43.75% of the tested isolates, respectively. Also, PM-AgNPs have recorded a reduction in the biofilm formation of S. aurues. These results suggest using PM-AgNPs to treat breast and ovarian cancers.
Collapse
Affiliation(s)
- Heba W Alhamdi
- Department of Biology, College of Sciences, King Khalid University, Abha, Saudi Arabia
| | - Hanan Khalaf Anazi
- Department of Biology, Collage of Science, Tabuk University, Tabuk, Saudi Arabia
| | - Fatma Alzahraa Mokhtar
- Fujairah Research Centre, Fujairah, United Arab Emirates
- Department of Pharmacognosy, Faculty of Pharmacy, El Saleheya El Gadida University, Sharkia, Egypt
| | - Seham S Elhawary
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Serag Eldin I Elbehairi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y Alfaifi
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha, Saudi Arabia
| | - Ali A Shati
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Lamiaa I Fahmy
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Engy Elekhnawy
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Afnan Hassan
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Cairo, Egypt
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo, Egypt
| | - Nabil Selim
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
31
|
Dewan D, Basu A, Dolai D, Pal S. Biological and Biophysical Methods for Evaluation of Inhibitors of Sortase A in Staphylococcus aureus: An Overview. Cell Biochem Funct 2024; 42:e70002. [PMID: 39470102 DOI: 10.1002/cbf.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Staphylococcus aureus, one of the most notorious pathogens, develops antibiotic resistance by the formation of a thick layer of exopolysaccharides known as biofilms. Sortase A, a transpeptidase responsible for biofilm formation and attachment to the host surface, has emerged as an important drug target for development of anti-virulence agents. A number of sortase A inhibitors, both peptide and non-peptides are reported which involved the use of several experiments which may provide insights regarding binding affinity, specificity, safety, and efficacy of ligands. In this review, we focus on the principles, pros and cons, and the type of information obtained from biophysical (FRET assay, Microscale Thermophoresis, Surface Plasmon Resonance, CD spectroscopy etc.) and biological (cell viability assay, biofilm formation assay, CLSM, western blot analysis, in vivo characterization on mice etc.) methods for estimation of probable sortase A inhibitors, which might be helpful to the researchers who might be interested to delve into the development of sortase A inhibitors as a drug, to address the burning question of antimicrobial resistance (AMR).
Collapse
|
32
|
González-García S, Hamdan-Partida A, Pérez-Ramos J, Aguirre-Garrido JF, Bustos-Hamdan A, Bustos-Martínez J. Comparison of the bacterial microbiome in the pharynx and nasal cavity of persistent, intermittent carriers and non-carriers of Staphylococcus aureus. J Med Microbiol 2024; 73:001940. [PMID: 39629792 PMCID: PMC11616445 DOI: 10.1099/jmm.0.001940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/13/2024] [Indexed: 12/08/2024] Open
Abstract
Introduction. Staphylococcus aureus is a bacterium that colonizes various human sites. The pharynx has been considered as a site of little clinical relevance and little studied. Recently, it has been reported that S. aureus can colonize more the pharynx than the nose. In addition, S. aureus can persist in these sites for prolonged periods of time.Hypothesis. The composition of the pharyngeal and nasal microbiome will differ between persistent, intermittent carriers and non-carriers of S. aureus.Aim. Determine whether the pharyngeal and nasal microbiome is different between carriers and non-carriers of S. aureus.Methodology. S. aureus carriers were monitored by means of pharyngeal and nasal exudates of apparently healthy adult university students for 3 months. Samples from individuals of the same carrier type were pooled, and DNA was extracted and the 16S rRNA was sequenced. The sequences were analysed in MOTHUR v.1.48.0 software, by analysing the percentages of relative abundance in the STAMP 2.1.3 program, in addition to the predictive analysis of metabolic pathways in PICRUSt2.Results. A greater colonization of S. aureus was found in the pharynx than in the nose. The microbiomes of S. aureus carriers and non-carriers do not show significant differences. The main microbiome difference found was between pharyngeal and nasal microbiomes. No significant differences were found in the abundance of the genus Staphylococcus in pharyngeal and nasal S. aureus carriers and non-carriers. The nasal microbiome was found to have more variation compared to the pharyngeal microbiome, which appears to be more stable between individuals and pools. Predictive analysis of metabolic pathways showed a greater presence of Staphylococcus-associated pathways in the nose than in the pharynx.Conclusion. S. aureus can colonize and persist in the pharynx in equal or greater proportion than in the nose. No statistically significant differences were found in the microbiome of the pharyngeal and nasal carriers and non-carriers of S. aureus, but the pharyngeal and nasal microbiomes are different independent of the type of S. aureus carrier or non-carrier. Therefore, the microbiome apparently does not influence the persistence of S. aureus.
Collapse
Affiliation(s)
- Samuel González-García
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Aida Hamdan-Partida
- Departamento de Atención a la Salud, UAM Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, Alcaldía Coyoacán, C.P. 04960, CDMX, Mexico
| | - Julia Pérez-Ramos
- Departamento de Sistemas Biológicos, UAM Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, Alcaldía Coyoacán, C.P. 04960, CDMX, Mexico
| | - José Félix Aguirre-Garrido
- Departamento de Ciencias Ambientales, UAM Lerma, Av. de las Garzas 10E, l Panteón 52005, Municipio Lerma de Villada, Estado de México, Mexico
| | - Anaíd Bustos-Hamdan
- Departamento de Atención a la Salud, UAM Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, Alcaldía Coyoacán, C.P. 04960, CDMX, Mexico
| | - Jaime Bustos-Martínez
- Departamento de Atención a la Salud, UAM Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, Alcaldía Coyoacán, C.P. 04960, CDMX, Mexico
| |
Collapse
|
33
|
Aarestrup FM, Hansen EB, Kumburu HH, Mzee T, Otani S. Improved ability to utilize lactose and grow in milk as a potential explanation for emergence of the novel bovine Staphylococcus aureus ST5477. Int J Med Microbiol 2024; 317:151637. [PMID: 39442481 DOI: 10.1016/j.ijmm.2024.151637] [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: 08/05/2024] [Revised: 08/23/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
Staphyloccous aureus belonging to sequence type 5477 have recently been identified as a predominant clone causing bovine mastitis in Rwanda and Tanzania. We compared nine S. aureus ST5477 to 17 isolates belonging to other sequence types by their biochemical profile and ability to acidify milk and grow in minimum media containing lactose. We found that ST5477 isolates all were positive in ONPG (o-nitrophenyl-β-D-galactopyranoside) test and negative for mannitol fermentation potentially challenging the correct identification of this sequence type as S. aureus. In addition, ST5477 isolates were all much faster in acidifying milk and grew faster in minimal media with lactose compared to other strains suggesting an increased lactose utilization and thereby adaptation to the bovine udder environment as a possible reason for the recent successful emergence. Comparison of the lac gene region of the genome of a recently sequenced ST5477 and that of S. aureus reference genome showed that both strains contained the known lacABCD genes involved in the lactose degradation, but that ST5477 had a 12 amino-acid deletion and two amino-acid differences in the lac gene transcription regulator, suggesting that increased transcription might play a role. In conclusion, these preliminary data suggests that improved lactose utilization and the ability to grow faster in milk may have been a key feature for the recent success of ST5477 as a bovine adapted clone.
Collapse
Affiliation(s)
| | | | - Happiness H Kumburu
- Kilimanjaro Clinical Research Institute, Moshi, Tanzania; Kilimanjaro Christian Medical University College, Tanzania; Kilimanjaro Christian Medical Centre, Tanzania
| | - Tutu Mzee
- Ifakara Health Institute, Bagamoyo Branch, Bagamoyo, Tanzania
| | | |
Collapse
|
34
|
Huang J, Xu Z, He P, Lin Z, Peng R, Yu Z, Li P, Deng Q, Liu X. Repurposing TAK-285 as An Antibacterial Agent against Multidrug-Resistant Staphylococcus aureus by Targeting Cell Membrane. Curr Microbiol 2024; 82:8. [PMID: 39585416 DOI: 10.1007/s00284-024-04001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024]
Abstract
Infections and antimicrobial resistance are becoming serious global public health crises. Multidrug-resistant Staphylococcus aureus (S. aureus) infections necessitate novel antimicrobial development. In this study, we demonstrated TAK-285, a novel dual HER2/EGFR inhibitor, exerted antibacterial activity against 17 clinical methicillin-resistant S. aureus (MRSA) and 15 methicillin sensitive S. aureus (MSSA) isolates in vitro, with a minimum inhibitory concentration (MIC) of 13.7 μg/mL. At 1 × MIC, TAK-285 completely inhibited the growth of S. aureus bacterial planktonic cells, and at 2 × MIC, it exhibited a superior inhibitory effect on intracellular S. aureus SA113-GFP compared to linezolid. Moreover, TAK-285 effectively inhibited biofilm formation at sub-MIC, eradicated mature biofilm and eliminated bacteria within biofilms, as confirmed by CLSM. Furthermore, the disruption of cell membrane permeability and potential was found by TAK-285 on S. aureus, suggesting its targeting of cell membrane integrity. Global proteomic analysis demonstrated that TAK-285 disturbed the metabolic processes of S. aureus, interfered with biofilm-related gene expression, and disrupted membrane-associated proteins. Conclusively, we repurposed TAK-285 as an antimicrobial with anti-biofilm properties against S. aureus by targeting cell membrane. This study provided strong evidence for the potential of TAK-285 as a promising antimicrobial agent against S. aureus.
Collapse
Affiliation(s)
- Jinlian Huang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China
- 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
| | - Zhichao Xu
- 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
| | - Peikun He
- 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
| | - Zhiwei Lin
- 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
| | - Renhai Peng
- 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
| | - 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.
| | - Qiwen Deng
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China.
- 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.
| | - Xiaoju Liu
- 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
|
Shi J, Shen L, Xiao Y, Wan C, Wang B, Zhou P, Zhang J, Han W, Hu R, Yu F, Wang H. Identification and validation of diagnostic biomarkers and immune cell abundance characteristics in Staphylococcus aureus bloodstream infection by integrative bioinformatics analysis. Front Immunol 2024; 15:1450782. [PMID: 39654884 PMCID: PMC11626409 DOI: 10.3389/fimmu.2024.1450782] [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: 06/18/2024] [Accepted: 08/01/2024] [Indexed: 12/12/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is an opportunistic pathogen that could cause life-threatening bloodstream infections. The objective of this study was to identify potential diagnostic biomarkers of S. aureus bloodstream infection. Gene expression dataset GSE33341 was optimized as the discovery dataset, which contained samples from human and mice. GSE65088 dataset was utilized as a validation dataset. First, after overlapping the differentially expressed genes (DEGs) in S. aureus infection samples from GSE33341-human and GSE33341-mice samples, we detected 63 overlapping genes. Subsequently, the hub genes including DRAM1, PSTPIP2, and UPP1 were identified via three machine-learning algorithms: random forest, support vector machine-recursive feature elimination, and least absolute shrinkage and selection operator. Additionally, the receiver operating characteristic curve was leveraged to verify the efficacy of the hub genes. DRAM1 (AUC=1), PSTPIP2 (AUC=1), and UPP1 (AUC=1) were investigated and demonstrated significant expression differences (all P < 0.05) and diagnostic efficacy in the training and validation datasets. Furthermore, the relationship between the diagnostic markers and the abundance of immune cells was assessed using cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT). These three diagnostic indicators also correlated with multiple immune cells to varying degrees. The expression of DRAM1 was significantly positively correlated with B cell naive and mast cell activation, and negatively correlated with NK cells and CD4/CD8+ T cells. The expression of PSTPIP2 was significantly positively correlated with macrophage M0, macrophage M1, B cell naive, and dendritic cell activation, while the expression of PSTPIP2 was negatively correlated with NK cells and CD4/CD8+ T cells. Significant negative correlations between UPP1 expression and T cell CD4 memory rest and neutrophils were also observed. Finally, we established a mouse model of S. aureus bloodstream infection and collected the blood samples for RNA-Seq analysis and RT-qPCR experiments. The analysis results in RNA-Seq and RT-qPCR experiments further confirmed the significant expression differences (all P < 0.05) of these three genes. Overall, three candidate hub genes (DRAM1, PSTPIP2, and UPP1) were identified initially for S. aureus bloodstream infection diagnosis. Our study could provide potential diagnostic biomarkers for S. aureus bloodstream infection patients.
Collapse
Affiliation(s)
- Junhong Shi
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cailing Wan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peiyao Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Han
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rongrong Hu
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hongxiu Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
36
|
Konwar AN, Basak S, Gurumayum S, Borah JC, Thakur D. Exploring northeast India's culturable soil Actinomycetia for potent antibacterial agents against gram-positive bacterial pathogens of clinical importance. Sci Rep 2024; 14:28640. [PMID: 39562678 PMCID: PMC11576755 DOI: 10.1038/s41598-024-77644-8] [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: 06/20/2024] [Accepted: 10/24/2024] [Indexed: 11/21/2024] Open
Abstract
This study investigated the isolation and bioactivity of Actinomycetia from the soil of Northeast India, a region rich in microbial diversity. A total of 187 presumptive Actinomycetia isolates were obtained and 53 were found to exhibit antimicrobial properties. Phylogenetic analysis based on 16 S rRNA gene sequencing revealed that the isolates were predominantly from the genus Streptomyces. Among these, the strain Streptomyces sp. NP14 (ANP14ARS) demonstrated specific and significant antimicrobial activity against gram-positive bacterial pathogens. The strain was further assayed against a panel of clinically important bacterial pathogens including Methicillin-resistant Staphylococcus aureus (MRSA). Its antimicrobial activity was confirmed using disc diffusion and membrane disruption assay, and its minimum inhibitory concentration was determined to be ≥ 3.12 ± 0.5 µg/ml against MRSA. Chemical analyses using FTIR and GC-MS identified key bioactive compounds, including Pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-3-(2-methylpropyl)- and Phenol 3,5-bis(1,1-dimethylethyl), known for their antimicrobial properties. Toxicity studies against animal liver cell lines indicated that the ethyl acetate extract was non-toxic at concentrations below 125 µg/mL, compared to Vancomycin, which was found to be hepatotoxic at similar concentrations. These findings highlight the potential of ANP14ARS as a source of bioactive natural products that is specific towards priority pathogens such as MRSA.
Collapse
Affiliation(s)
- Aditya Narayan Konwar
- Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Studies in Science and Technology, Guwahati, 781035, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Surajit Basak
- Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Studies in Science and Technology, Guwahati, 781035, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shalini Gurumayum
- Chemical Biology Laboratory - 1, Life Sciences Division, Institute of Advanced Studies in Science and Technology, Guwahati, 781035, Assam, India
| | - Jagat Chandra Borah
- Chemical Biology Laboratory - 1, Life Sciences Division, Institute of Advanced Studies in Science and Technology, Guwahati, 781035, Assam, India
| | - Debajit Thakur
- Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Studies in Science and Technology, Guwahati, 781035, Assam, India.
| |
Collapse
|
37
|
Scherr F, Darisipudi MN, Börner FR, Austermeier S, Hoffmann F, Eberhardt M, Abdurrahman G, Saade C, von Eggeling F, Kasper L, Holtfreter S, Bröker BM, Kiehntopf M. Alpha-1-antitrypsin as novel substrate for S. aureus' Spl proteases - implications for virulence. Front Immunol 2024; 15:1481181. [PMID: 39628483 PMCID: PMC11611844 DOI: 10.3389/fimmu.2024.1481181] [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: 08/15/2024] [Accepted: 10/22/2024] [Indexed: 12/06/2024] Open
Abstract
Background The serine protease like (Spl) proteases of Staphylococcus aureus are a family of six proteases whose function and impact on virulence are poorly understood. Here we propose alpha-1-antitrypsin (AAT), an important immunomodulatory serine protease inhibitor as target of SplD, E and F. AAT is an acute phase protein, interacting with many proteases and crucial for prevention of excess tissue damage by neutrophil elastase during the innate immune response to infections. Methods We used MALDI-TOF-MS to identify the cleavage site of Spl proteases within AAT's reactive center loop (RCL) and LC-MS/MS to quantify the resulting peptide cleavage product in in vitro digestions of AAT and heterologous expressed proteases or culture supernatants from different S. aureus strains. We further confirmed proteolytic cleavage and formation of a covalent complex with Western Blots, investigated AAT's inhibitory potential against Spls and examined the NETosis inhibitory activity of AAT-Spl-digestions. Results SplD, E and F, but not A or B, cleave AAT in its RCL, resulting in the release of a peptide consisting of AAT's C-terminal 36 amino acids (C36). Synthetic C36, as well as AAT-SplD/E/F-digestions exhibit NETosis inhibition. Only SplE, but not D or F, was partly inhibited by AAT, forming a covalent complex. Conclusion We unraveled a new virulence trait of S. aureus, where SplD/E/F cleave and inactivate AAT while the cleavage product C36 inhibits NETosis.
Collapse
Affiliation(s)
- Franziska Scherr
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | | | - Friedemann R. Börner
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | - Sophie Austermeier
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology -Hans Knoell Institute Jena (HKI), Jena, Germany
| | - Franziska Hoffmann
- Department of Otorhinolaryngology, Matrix-assisted Laser Desorption/Ionization (MALDI) Imaging and Clinical Biophotonics, Jena University Hospital, Jena, Germany
| | - Martin Eberhardt
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | - Goran Abdurrahman
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Christopher Saade
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Ferdinand von Eggeling
- Department of Otorhinolaryngology, Matrix-assisted Laser Desorption/Ionization (MALDI) Imaging and Clinical Biophotonics, Jena University Hospital, Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology -Hans Knoell Institute Jena (HKI), Jena, Germany
- Institute of Novel and Emerging Infectious Diseases (INNT), Friedrich-Loeffler-Institut, Greifswald, Germany
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Kiehntopf
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| |
Collapse
|
38
|
Sundar Das D, Dey A, Das G, Naik S. Staphylococcus aureus Dissemination Presenting With Encephalopathy and Epidural Abscess. Case Rep Infect Dis 2024; 2024:6889110. [PMID: 39606097 PMCID: PMC11599475 DOI: 10.1155/crdi/6889110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/09/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Staphylococcal infection is a common bacterial disease with common clinical features. Untreated infection, especially in immunosenescence cases, can affect other organs. This can lead to multiorgan dysfunction and cause increased morbidity and mortality. Unlike commonly presented features of pneumonia, dissemination of infection can pose diagnostic and therapeutic enigma. Therefore, any such presentation in common clinical practice can yield a conundrum of diagnoses. Case Report: A 69-year-old elderly male presented to the Emergency Department with acute onset encephalopathy. Historically, cues were limited, and evaluation was negated for acute cerebrovascular event or seizure. Laboratory findings were suggestive of a severe sepsis. While clinical medicine workup and diagnostic dilemma were ongoing, possible sources of the sepsis were thoroughly sought including range of infectious causes. This patient's presentation was one of its kind: staphylococcal bacteremia seeding to cause pneumonia and unusual epidural abscess in due course of illness. Conclusion: The health outcome of the critically ill especially elderly patients depends mostly on the importance of clinical medicine to address the diagnostic enigma and virtue of supportive care delivered. Staphylococcus aureus infections are capable of developing distant infectious foci, as highlighted in this case, and that the clinician should be alert to this possibility. This particular case firmly posits an admonition for clinicians and the importance of clinical medicine for critical reasoning to improve the patient outcome.
Collapse
Affiliation(s)
- Dhriti Sundar Das
- Department of General Medicine, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Anupam Dey
- Department of General Medicine, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Gurudip Das
- Department of Orthopedics, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Suprava Naik
- Department of Radiodiagnosis, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| |
Collapse
|
39
|
Golban M, Charostad J, Kazemian H, Heidari H. Phage-Derived Endolysins Against Resistant Staphylococcus spp.: A Review of Features, Antibacterial Activities, and Recent Applications. Infect Dis Ther 2024:10.1007/s40121-024-01069-z. [PMID: 39549153 DOI: 10.1007/s40121-024-01069-z] [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: 08/19/2024] [Accepted: 10/22/2024] [Indexed: 11/18/2024] Open
Abstract
Antimicrobial resistance is a significant global public health issue, and the dissemination of antibiotic resistance in Gram-positive bacterial pathogens has significantly increased morbidity, mortality rates, and healthcare costs. Among them, Staphylococcus, especially methicillin-resistant Staphylococcus aureus (MRSA), causes a wide range of diseases due to its diverse pathogenic factors and infection strategies. These bacteria also present significant issues in veterinary medicine and food safety. Effectively managing staphylococci-related problems necessitates a concerted effort to implement preventive measures, rapidly detect the pathogen, and develop new and safe antimicrobial therapies. In recent years, there has been growing interest in using endolysins to combat bacterial infections. These enzymes, which are also referred to as lysins, are a unique class of hydrolytic enzymes synthesized by double-stranded DNA bacteriophages. They possess glycosidase, lytic transglycosylase, amidase, and endopeptidase activities, effectively destroying the peptidoglycan layer and resulting in bacterial lysis. This unique property makes endolysins powerful antimicrobial agents, particularly against Gram-positive organisms with more accessible peptidoglycan layers. Therefore, considering the potential benefits of endolysins compared to conventional antibiotics, we have endeavored to gather and review the characteristics and uses of endolysins derived from staphylococcal bacteriophages, as well as their antibacterial effectiveness against Staphylococcus spp. based on conducted experiments and trials.
Collapse
Affiliation(s)
- Mina Golban
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Javad Charostad
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Kazemian
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hamid Heidari
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
40
|
Zhao Y, Xiong M, Ho K, Rao Y, Huang Y, Cao J, Yue Y, Wang J, Wen G, Li J. Bioaerosol emission and exposure risk from a wastewater treatment plant in winter and spring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117294. [PMID: 39504877 DOI: 10.1016/j.ecoenv.2024.117294] [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: 07/30/2024] [Revised: 11/02/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
The potential health risks posed by bioaerosols containing pathogens originating from wastewater treatment plants (WWTPs) have gaining intensive attention. This study designated sampling locations within a WWTP situated in Xi'an, a major city in northwest China. The airborne bacterial concentration, taxonomic composition, and the associated health risks were analyzed in the aeration tanks with bottom microporous aeration system. The Anaerobic-Anoxic-Oxic (AAO) tank emitted significantly higher culturable bacteria (1.58×104 CFU m-3 in spring, 6.69×103 CFU m-3 in winter) compared to Double-ditch (DE) oxidation ditch and aerated grit chamber (AGC) chamber, aligning with 16S rDNA quantification results. The bacterial concentrations are higher in spring than that in winter, with the AAO tank posing the highest exposure risk during the spring season. The dominant genera in the air samples include Cutibacterium, Lawsonella, Acinetobacter, Pseudomonas, and Aeromonas. Among the identified genus, 139 bacterial genera were identified as potential human pathogens like Neisseria, Moraxella, Haemophilus, Escherichia-Shigella and Streptococcus. These pathogens further elevate exposure risks from WWTP bioaerosols. This study provides relevant information on the seasonal health risk variations tied to bioaerosol emissions from diverse aeration tanks with bottom microporous aeration system in the mega city of northwest China, emphasizing the imperative to enhance the management and control measures for bioaerosols originating from the AAO tank.
Collapse
Affiliation(s)
- Yulei Zhao
- Key Laboratory of Aerosol Chemistry & Physics, State Key Laboratory of Loess and Quaternary Geology (SKLLQG), Institute of Earth Environment, Chinese Academy of Sciences (CAS), Xi'an 710061, PR China; Xi'an Institute for Innovative Earth Environment Research, Xi'an 710061, PR China
| | - Mingyu Xiong
- Key Laboratory of Aerosol Chemistry & Physics, State Key Laboratory of Loess and Quaternary Geology (SKLLQG), Institute of Earth Environment, Chinese Academy of Sciences (CAS), Xi'an 710061, PR China; Xi'an Institute for Innovative Earth Environment Research, Xi'an 710061, PR China
| | - Kinfai Ho
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Yongfang Rao
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yu Huang
- Key Laboratory of Aerosol Chemistry & Physics, State Key Laboratory of Loess and Quaternary Geology (SKLLQG), Institute of Earth Environment, Chinese Academy of Sciences (CAS), Xi'an 710061, PR China.
| | - Junji Cao
- Institute of Atmospheric Physics, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Yang Yue
- Institute of Environmental Engineering, ETH Zurich, Zürich, Switzerland
| | - Jing Wang
- Institute of Environmental Engineering, ETH Zurich, Zürich, Switzerland; Laboratory of Advanced Analytical Technologies, Empa, Dübendorf, Switzerland
| | - Gang Wen
- Key Laboratory of Northwest Water Resource, Environment and Ecology, MOE, Xi'an University of Architecture and Technology, Xi'an 710055, PR China
| | - Juntang Li
- Research Centre for Occupation and Environment Medicine, Collaborative Innovation Centre for Medical Equipment, Key Laboratory of Biological Damage Effect and Protection, Luoyang 471031, PR China
| |
Collapse
|
41
|
Ifedinezi OV, Nnaji ND, Anumudu CK, Ekwueme CT, Uhegwu CC, Ihenetu FC, Obioha P, Simon BO, Ezechukwu PS, Onyeaka H. Environmental Antimicrobial Resistance: Implications for Food Safety and Public Health. Antibiotics (Basel) 2024; 13:1087. [PMID: 39596781 PMCID: PMC11591122 DOI: 10.3390/antibiotics13111087] [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: 10/15/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Antimicrobial resistance (AMR) is a serious global health issue, aggravated by antibiotic overuse and misuse in human medicine, animal care, and agriculture. This study looks at the different mechanisms that drive AMR, such as environmental contamination, horizontal gene transfer, and selective pressure, as well as the severe implications of AMR for human and animal health. This study demonstrates the need for concerted efforts across the scientific, healthcare, agricultural, and policy sectors to control the emergence of AMR. Some crucial strategies discussed include developing antimicrobial stewardship (AMS) programs, encouraging targeted narrow-spectrum antibiotic use, and emphasizing the significance of strict regulatory frameworks and surveillance systems, like the Global Antimicrobial Resistance and Use Surveillance System (GLASS) and the Access, Watch, and Reserve (AWaRe) classification. This study also emphasizes the need for national and international action plans in combating AMR and promotes the One Health strategy, which unifies environmental, animal, and human health. This study concludes that preventing the spread of AMR and maintaining the effectiveness of antibiotics for future generations requires a comprehensive, multidisciplinary, and internationally coordinated strategy.
Collapse
Affiliation(s)
| | - Nnabueze Darlington Nnaji
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK
- Department of Microbiology, University of Nigeria, Nsukka 410001, Nigeria
| | | | | | | | | | - Promiselynda Obioha
- Microbiology Research Unit, School of Human Sciences, London Metropolitan University, 166-220 Holloway Road, London N7 8DB, UK
| | - Blessing Oteta Simon
- Department of Public Health Sciences, National Open University of Nigeria, Abuja 900108, Nigeria
| | | | - Helen Onyeaka
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
42
|
Mady MS, Elsayed HE, Tawfik NF, Moharram FA. Volatiles extracted from Melaleuca Rugulosa (Link) Craven leaves: comparative profiling, bioactivity screening, and metabolomic analysis. BMC Complement Med Ther 2024; 24:394. [PMID: 39538246 PMCID: PMC11562704 DOI: 10.1186/s12906-024-04683-z] [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: 04/09/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Melaleuca species (family Myrtaceae) are characterized by their wide-ranging applications as antimicrobials and in skin-related conditions. Herein, we estimated the volatile profile and biological significance of M. rugulosa (Link) leaves for the first time supported by a dereplication protocol. MATERIALS AND METHODS Volatile components were extracted using hydrodistillation (HD), supercritical fluid (SF), and headspace (HS) techniques and identified using GC/MS. The variations among the three extracts were assessed using principal component analysis and orthogonal partial least square discriminant analysis (OPLS-DA). The extracted volatiles were tested for radical scavenging activity, anti-aging, and anti-hyperpigmentation potential. Finally, disc diffusion and broth microdilution assays were implemented to explore the antibacterial capacity against Streptococcus pyogenes, Staphylococcus aureus, Clostridium perfringens, and Pseudomonas aeruginosa. RESULTS The yield of the SF technique (0.8%) was three times higher than HD. GC/MS analysis revealed that the oxygenated compounds are the most proponents in the three extracts being 95.93% (HD), 80.94% (HS), and 48.4% (SF). Moreover, eucalyptol (1,8-cineol) represents the major component in the HD-EO (89.60%) and HS (73.13%) volatiles, while dl-α-tocopherol (16.27%) and α-terpineol (11.89%) represent the highest percentage in SF extract. Regarding the bioactivity profile, the HD-EO and SF-extract showed antioxidant potential in terms of oxygen radical absorbance capacity, and β- carotene assays, while exerting weak activity towards DPPH. In addition, they displayed potent anti-elastase and moderate anti-collagenase activities. The HD-EO exhibited potent anti-tyrosinase activity, while the SF extract showed a moderate level compared to tested controls. OPLS-DA and dereplication studies predicted that the selective antibacterial activity of HD-EO to S. aureus was related to eucalyptol, while SF extract to C. perfringens was related to α-tocopherol. CONCLUSIONS M. rugulosa leaves are considered a vital source of bioactive volatile components that are promoted for controlling skin aging and infection. However, further safety and clinical studies are recommended.
Collapse
Affiliation(s)
- Mohamed S Mady
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt.
| | - Heba E Elsayed
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Nashwa F Tawfik
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Fatma A Moharram
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| |
Collapse
|
43
|
Gonsalves LJ, Tran A, Gardiner T, Freeman T, Dutta A, Miller CJ, McNamara S, Waalkes A, Long DR, Salipante SJ, Hoffman LR, Wolter DJ. Mechanisms of Staphylococcus aureus survival of trimethoprim-sulfamethoxazole-induced thymineless death. mBio 2024; 15:e0163424. [PMID: 39445807 PMCID: PMC11559000 DOI: 10.1128/mbio.01634-24] [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: 05/29/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Trimethoprim-sulfamethoxazole (SXT) is commonly used to treat diverse Staphylococcus aureus infections, including those associated with cystic fibrosis (CF) pulmonary disease. Studies with Escherichia coli found that SXT impairs tetrahydrofolate production, leading to DNA damage, stress response induction, and accumulation of reactive oxygen species (ROS) in a process known as thymineless death (TLD). TLD survival can occur through the uptake of exogenous thymidine, countering the effects of SXT; however, a growing body of research has implicated central metabolism as another potentially important determinant of bacterial survival of SXT and other antibiotics. Here, we conducted studies to better understand the mechanisms of TLD survival in S. aureus. We found that thymidine abundances in CF sputum were insufficient to prevent TLD of S. aureus, highlighting the importance of alternative survival mechanisms in vivo. In S. aureus cultured in vitro with SXT and low thymidine, we frequently identified adaptive mutations in genes encoding carbohydrate, nucleotide, and amino acid metabolism, supporting reduced metabolism as a common survival mechanism. Although intracellular ROS levels rose with SXT treatment in vitro, survival was not improved in the presence of ROS scavengers, unlike in E. coli. SXT challenge induced the SOS response, which was alleviated by added thymidine. Finally, an inactivating mutation in the phosphotransferase gene ptsI conferred both limitation in cellular ATP and improved survival against TLD. Collectively, these results suggest that alterations in core metabolic functions, particularly those that reduce ATP levels, predominantly confer S. aureus survival and persistence during SXT treatment, potentially identifying novel targets for co-treatment.IMPORTANCEStaphylococcus aureus is a ubiquitous organism and one of the leading causes of human infections, many of which are difficult to treat due to persistence, antibiotic resistance, or antibiotic tolerance. As our arsenal of effective antibiotics dwindles, the need for improved treatments becomes increasingly urgent, necessitating a better understanding of the precise mechanisms by which pathogens evade our most critical antimicrobial agents. Here, we report a systematic characterization of the mechanisms of S. aureus survival to treatment with the first-line antistaphylococcal antibiotic trimethoprim-sulfamethoxazole, identifying pathways and candidate targets for enhancing the efficacy of available antimicrobial agents.
Collapse
Affiliation(s)
- Lauren J. Gonsalves
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Allyson Tran
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Tessa Gardiner
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Tiia Freeman
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Angshita Dutta
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Carson J. Miller
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Sharon McNamara
- Pulmonary Division, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Lucas R. Hoffman
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Pulmonary Division, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Daniel J. Wolter
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Pulmonary Division, Seattle Children’s Hospital, Seattle, Washington, USA
| |
Collapse
|
44
|
Cuellar-Gaviria TZ, Rincon-Benavides MA, Halipci Topsakal HN, Salazar-Puerta AI, Jaramillo-Garrido S, Kordowski M, Vasquez-Martinez CA, Nguyen KT, Rima XY, Rana PSJB, Combita-Heredia O, Deng B, Dathathreya K, McComb DW, Reategui E, Wozniak D, Higuita-Castro N, Gallego-Perez D. Tissue nano-transfection of antimicrobial genes drives bacterial biofilm killing in wounds and is potentially mediated by extracellular vesicles. J Control Release 2024; 376:1300-1315. [PMID: 39491627 DOI: 10.1016/j.jconrel.2024.10.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/06/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The emergence of bacteria that are resistant to antibiotics is on track to become a major global health crisis. Therefore, there is an urgent need for new treatment options. Here, we studied the implementation of tissue-nanotransfection (TNT) to treat Staphylococcus aureus-infected wounds by delivering gene cargos that boost the levels of naturally produced antimicrobial peptides. The Cathelicidin Antimicrobial Peptide gene (CAMP), which produces the antimicrobial peptide LL-37, was used as model gene cargo. In vitro evaluation showed successful transfection and an increase in the transcription and translation of CAMP-coding plasmid in mouse primary epithelial cells. Moreover, we found that the extracellular vesicles (EVs) derived from the transfected cells (in vitro and in vivo) carried significantly higher concentrations of CAMP transcripts and LL-37 peptide compared to control EVs, possibly mediating the trafficking of the antimicrobial contents to other neighboring cells. The TNT platform was then used in vivo on an excisional wound model in mice to nanotransfect the CAMP-coding plasmid on the edge of infected wounds. After 4 days of daily treatment, we observed a significant decrease in the bacterial load in the CAMP-treated group compared to the sham group. Moreover, histological analysis and bacterial load quantification also revealed that TNT of CAMP on S. aureus-infected wounds was effective in treating biofilm progression by reducing the bacterial load. Lastly, we observed a significant increase in macrophage recruitment to the infected tissue, a robust increase in vascularization, as well as and an increased expression of IL10 and Fli1. Our results demonstrate that TNT-based delivery of gene cargos coding for antimicrobial compounds to the wound is a promising approach for combating biofilm infections in wounds.
Collapse
Affiliation(s)
- Tatiana Z Cuellar-Gaviria
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Maria Angelica Rincon-Benavides
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Hatice Nur Halipci Topsakal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Istanbul Atlas University, Istanbul 34408, Turkiye
| | | | | | - Mia Kordowski
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Carlos A Vasquez-Martinez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; CONACYT - Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca 68020, Mexico
| | - Kim Truc Nguyen
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Xilal Y Rima
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Pranav S J B Rana
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | | | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH 43210, USA
| | - Kavya Dathathreya
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH 43210, USA; Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Eduardo Reategui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Wozniak
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA; Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
45
|
Kongari R, Ray MD, Lehman SM, Plaut RD, Hinton DM, Stibitz S. The Transcriptional Program of Staphylococcus aureus Phage K Is Affected by a Host rpoC Mutation That Confers Phage K Resistance. Viruses 2024; 16:1773. [PMID: 39599887 PMCID: PMC11598898 DOI: 10.3390/v16111773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
To better understand host-phage interactions and the genetic bases of phage resistance in a model system relevant to potential phage therapy, we isolated several spontaneous mutants of the USA300 S. aureus clinical isolate NRS384 that were resistant to phage K. Six of these had a single missense mutation in the host rpoC gene, which encodes the RNA polymerase β' subunit. To examine the hypothesis that mutations in the host RNA polymerase affect the transcription of phage genes, we performed RNA-seq analysis on total RNA samples collected from NRS384 wild-type (WT) and rpoCG17D mutant cultures infected with phage K, at different timepoints after infection. Infection of the WT host led to a steady increase of phage transcription relative to the host. Our analysis allowed us to define 53 transcriptional units and to categorize genes based on their temporal expression patterns. Predicted promoter sequences defined by conserved -35, -10, and, in some cases, extended -10 elements, were found upstream of early and middle genes. However, in many cases, sequences upstream of late genes did not contain clear, complete, canonical promoter sequences, suggesting that factors in addition to host RNA polymerase are required for their expression. Infection of the rpoCG17D mutant host led to a transcriptional pattern that was similar to that of the WT at early timepoints. However, beginning at 20 min after infection, transcription of late genes (such as phage structural genes and host lysis genes) was severely reduced. Our data indicate that the rpoCG17D mutation prevents the expression of phage late genes, resulting in a failed infection cycle for phage K. In addition to illuminating the global transcriptional landscape of phage K throughout the infection cycle, this study will inform our investigations into the basis of phage K's control of its transcriptional program as well as mechanisms of phage resistance.
Collapse
Affiliation(s)
- Rohit Kongari
- Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Melissa D. Ray
- Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Susan M. Lehman
- Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Roger D. Plaut
- Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Deborah M. Hinton
- Gene Expression and Regulation Section, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Scott Stibitz
- Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
46
|
Kilani AM, Alabi ED, Adeleke OE. Coexistence of the blaZ gene and selected virulence determinants in multidrug-resistant Staphylococcus aureus: insights from three Nigerian tertiary hospitals. BMC Infect Dis 2024; 24:1269. [PMID: 39528974 PMCID: PMC11552187 DOI: 10.1186/s12879-024-10171-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND AND PURPOSE Infections caused by β-lactamase-producing strains of Staphylococcus aureus have become increasingly difficult to treat due to the expression of multiple virulence factors. This has heightened concerns about managing S. aureus-related infections. This study was conducted to characterize the blaZ gene and selected virulence determinants in β-lactam resistant S. aureus from human sources in three Nigerian tertiary hospitals. MATERIALS AND METHODS Three hundred and sixty samples were collected for the study. S. aureus was isolated and characterized following standard microbiological protocols and nuc gene amplification. Antibiotic susceptibility and minimum inhibitory concentration tests were performed using the disk diffusion method and E-tests, respectively. Biofilm formation and β-lactamase production were assessed using Congo red agar and nitrocefin kits, while the blaZ gene was examined using conventional PCR. Capsular polysaccharide genotyping, accessory gene regulator (agr) detection, Panton-valentine leucocidin (PVL), and PVL proteins were performed using PCR and Western blotting. RESULTS S. aureus was recovered from 145 samples, 50 (34.5%) of these isolates exhibited multidrug resistance, with MICs ranging from 0.125 to 1.00 µg/mL, and showed significant resistance to aminoglycosides, fluoroquinolones, and β-lactams. Of these, 31 strains produced β-lactamases, 30 of which carried the blaZ gene in combination with cap8 (80%) or cap5 (20%). Biofilm formation and PVL gene were observed in 85% of the 20 randomly selected blaZ-positive multidrug-resistant (MDR) strains. The agr2 allele was predominant, found in 70% of the selected MDR strains. No significant difference in the occurrence of the blaZ gene was found among the three clinical sources (p ≤ α0.05). CONCLUSION The co-occurrence of the blaZ gene with PVL, capsular polysaccharide genes, and agr alleles is associated with biofilm formation, indicating a high risk of β-lactam-resistant S. aureus infections. Our findings highlight the need for continuous molecular surveillance to enhance infection management, treatment options, and patient outcomes in the study locality. A limitation of this study is the random selection of MDR isolates, which may affect the comprehensiveness of the analyses.
Collapse
Affiliation(s)
- Adetunji Misbau Kilani
- Department of Microbiology, Federal University Dutsin-Ma, Dutsin-Ma, Katsina State, Nigeria
| | - Emmanuel Dayo Alabi
- Department of Microbiology, Federal University Dutsin-Ma, Dutsin-Ma, Katsina State, Nigeria.
| | | |
Collapse
|
47
|
Tan LT, Salleh NF. Marine Cyanobacteria: A Rich Source of Structurally Unique Anti-Infectives for Drug Development. Molecules 2024; 29:5307. [PMID: 39598696 PMCID: PMC11596561 DOI: 10.3390/molecules29225307] [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: 09/30/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
Marine cyanobacteria represent a promising yet underexplored source of novel natural products with potent biological activities. Historically, the focus has been on isolating cytotoxic compounds from marine cyanobacteria, but a substantial number of these photosynthetic microorganisms also produce diverse specialized molecules with significant anti-infective properties. Given the global pressing need for new anti-infective lead compounds, this review provides a concise yet comprehensive overview of the current knowledge on anti-infective secondary metabolites derived from marine cyanobacteria. A majority of these molecules were isolated from free-living filamentous cyanobacteria, while several examples were derived from marine cyanobacterial symbionts. In addition, SAR studies and potent synthetic analogs based on selected molecules will be featured. With more than 200 molecules, this review presents their antibacterial, antifungal, antiviral, antiprotozoal, and molluscicidal activities, with the chemical and biological information covered in the literature up to September 2024.
Collapse
Affiliation(s)
- Lik Tong Tan
- Natural Sciences and Science Education, National Institute of Education, Nanyang Technological University, 1 Nanyang Walk, Singapore 637616, Singapore;
| | | |
Collapse
|
48
|
Rahmanian N, Moulavi P, Ashrafi F, Sharifi A, Asadi S. Surface-functionalized UIO-66-NH 2 for dual-drug delivery of vancomycin and amikacin against vancomycin-resistant Staphylococcus aureus. BMC Microbiol 2024; 24:462. [PMID: 39516717 PMCID: PMC11546402 DOI: 10.1186/s12866-024-03615-8] [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: 07/26/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Conventional antibacterial compounds can inhibit the growth of microorganisms, but their adverse effects and the development of drug limit their widespread use. The current study aimed to synthesize PEG-coated UIO-66-NH2 nanoparticles loaded with vancomycin and amikacin (VAN/AMK-UIO-66-NH2@PEG) and evaluate their antibacterial and anti-biofilm activities against vancomycin-resistant Staphylococcus aureus (VRSA) clinical isolates. METHODS The VAN/AMK-UIO-66-NH2@PEG were characterized using scanning electron microscopy (SEM), transmission electron microscopy (TEM), and dynamic light scattering (DLS) to determine their size, polydispersity index (PDI), encapsulation efficiency (EE%), zeta-potential, drug release profile, and physical stability. Antibacterial activity was evaluated using minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), and time-kill assays. Biofilm formation by VRSA was assessed using the crystal violet (CV) and minimum biofilm eradication concentration (MBEC) assays. The effect of sub-MIC concentrations of the formulations on the expression of biofilm-related genes (icaA, icaD) and resistance-related genes (mecA, vanA) was investigated using quantitative real-time polymerase chain reaction (RT-qPCR). RESULTS As demonstrated by MIC, MBC and time-kill assay, the VAN/AMK-UIO-66-NH2@PEG nanoparticles exhibited enhanced antibacterial activity against VRSA isolates compared to free drugs and prepared formulations. Furthermore, CV and MBEC tests indicated that the VAN/AMK-UIO-66@NH2/PEG can reduce biofilm formation dramatically compared to VAN/AMK and VAN/AMK-UIO-66@NH2, due to its great drug release properties. This study also found that the expression level of the mecA, vanA, icaA, and icaD genes in VAN/AMK-UIO-66@NH2/PEG treated VRSA isolates was substantially decreased compared to other groups. CONCLUSIONS These findings highlighted the efficiency of VAN/AMK-UIO-66@NH2/PEG in combating antimicrobial resistance and biofilm formation in VRSA isolates. Future studies, particularly in vivo models, are necessary to evaluate the safety, efficacy, and clinical applicability of these nanoparticles for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Nazanin Rahmanian
- Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pooria Moulavi
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Ashrafi
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Aram Sharifi
- Department of Animal Science, Faculty of Agriculture, University of Kurdistan, Sanandaj, Kurdistan, Iran
| | - Sepideh Asadi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
49
|
Ge R, Zhao H, Tang Q, Chandarajoti K, Bai H, Wang X, Zhang K, Ye W, Han X, Wang C, Zhou W. A novel α-mangostin derivative synergistic to antibiotics against MRSA with unique mechanisms. Microbiol Spectr 2024; 12:e0163124. [PMID: 39508612 PMCID: PMC11619392 DOI: 10.1128/spectrum.01631-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) remains a leading cause of hospital-acquired infections, often linked to complicated treatments, increased mortality risk, and significant cost burdens. Several antibacterial agents have been developed to address MRSA resistance. In this study, potential agents to combat MRSA resistance were explored, with the antibacterial activity of synthesized α-mangostin (α-MG) derivatives being evaluated alongside investigations into their cellular mechanisms against MRSA2. α-MG-4, featuring an allyl group at C3 of the lead compound α-MG, restored the sensitivity of MRSA2 to penicillin, enrofloxacin, and gentamicin, while also demonstrating improved safety profiles. Although α-MG-4 alone was ineffective against MRSA2, it exhibited an optimal synergistic ratio in vitro when combined with these antibiotics. This significant synergistic antibacterial effect was further confirmed in vivo using a mouse skin abscess model. Additionally, the synergistic mechanisms revealed that α-MG-4 was associated with changes in membrane permeability and inhibition of the MepA and NorA genes, which encode the efflux pumps of MRSA2. α-MG-4 also inhibited PBP2a expression, potentially by occupying a crucial binding site in a dose-dependent manner.IMPORTANCEMethicillin-resistant Staphylococcus aureus (MRSA)'s resistance to multiple antibiotics poses significant health and safety concerns. A novel α-mangostin (α-MG) derivative, α-MG-4, was first identified as a xanthone-based PBP2a inhibitor that reverses MRSA2 resistance to penicillin. The synergistic antibacterial effects of α-MG-4 were linked to increased cell membrane permeability and the inhibition of genes involved in efflux pump function.
Collapse
Affiliation(s)
- Rile Ge
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Haiyan Zhao
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Qun Tang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Kasemsiri Chandarajoti
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, Thailand
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla, Thailand
| | - Han Bai
- Department of Clinical Pharmacy, College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Xiaoyang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Keyu Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Wenchong Ye
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Chunmei Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
50
|
Garbo V, Venuti L, Albano C, Caruana C, Cuccia A, Condemi A, Boncori G, Polara VF, Cascio A, Salerno S, Colomba C. Investigating Osteomyelitis as a Rare Adverse Effect of Vaccination in the Pediatric Population. Pathogens 2024; 13:972. [PMID: 39599525 PMCID: PMC11597684 DOI: 10.3390/pathogens13110972] [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: 08/07/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Immunization is a preventive measure of crucial importance. As with any other medication, side effects are a possibility and include the rare occurrence of severe infections, such as osteomyelitis. We report an unusual case of pediatric osteomyelitis following vaccination and provide a review of similar reports submitted to the Vaccine Adverse Event Report System (VAERS), aiming to explore the association between the vaccination procedure and the occurrence of osteomyelitis in childhood. A previously healthy infant, with no history of trauma or infection, presented with hyperpyrexia, swelling, and functional impairment in the left leg and was eventually diagnosed with osteomyelitis of the left femur. An edema was noted at the site of the injection that he received days before for immunization purposes. The infection required surgical drainage and a four-week-long intravenous antibiotic treatment, and the patient was discharged upon showing improved clinical conditions. Forty-seven reports of similar cases submitted to VAERS between 1994 and 2023 were collected, and several cases from the literature, including a case of femoral osteomyelitis in a newborn vaccinated against Hepatitis B, attributed to improper injection technique. Another case was reported in a 15-year-old girl, which aligned with six similar cases of osteomyelitis in adolescents following HPV vaccines collected from VAERS. Despite the small sample number, the findings that in 77% of cases the infection was localized in the vaccinated limb and that symptoms appeared on average 4.3 days (IQR 1.0-5.7 days) post-vaccination suggest a possible link to the injection procedure and highlight the need to adhere to recommendations regarding skin preparation and the selection of the appropriate needle length and injection site.
Collapse
Affiliation(s)
- Valeria Garbo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (V.G.)
| | - Laura Venuti
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (V.G.)
| | - Chiara Albano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (V.G.)
| | - Costanza Caruana
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (V.G.)
| | - Alessandra Cuccia
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (V.G.)
| | - Anna Condemi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (V.G.)
| | - Giovanni Boncori
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (V.G.)
| | - Valentina Frasca Polara
- Division of Paediatric Infectious Disease, “G. Di Cristina” Hospital, ARNAS Civico Di Cristina Benfratelli, 90127 Palermo, Italy
| | - Antonio Cascio
- Infectious and Tropical Disease Unit, Sicilian Regional Reference Center for the Fight Against AIDS, AOU Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Sergio Salerno
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| | - Claudia Colomba
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (V.G.)
- Division of Paediatric Infectious Disease, “G. Di Cristina” Hospital, ARNAS Civico Di Cristina Benfratelli, 90127 Palermo, Italy
| |
Collapse
|