1
|
Schöpf C, Knapp M, Scheler J, Coraça-Huber DC, Romanelli A, Ladurner P, Seybold AC, Binder U, Würzner R, Marx F. The antibacterial activity and therapeutic potential of the amphibian-derived peptide TB_KKG6K. mSphere 2025:e0101624. [PMID: 40387366 DOI: 10.1128/msphere.01016-24] [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: 12/06/2024] [Accepted: 04/13/2025] [Indexed: 05/20/2025] Open
Abstract
Antimicrobial peptides (AMPs) have great potential to be developed as topical treatments for microbial infections of the skin, including those caused by the gram-positive human pathogen Staphylococcus aureus. Among the AMPs, temporin B (TB) is of particular interest. This 13-amino-acid-long cationic peptide is secreted by the granular glands of the European frog Rana temporaria and represents a primary line of defense against invading pathogens. The objective of this study was to investigate the antibacterial efficacy and the mode of action of the synthetic TB analog, TB_KKG6K, in a drug-resistant clinical isolate of S. aureus and assess the peptide's tolerance and curative potential in an in vitro infection model using three-dimensional human epidermis equivalents (HEEs). The results revealed a high bactericidal efficacy of TB_KKG6K at low micromolar concentrations. The peptide perturbed the bacterial cell membrane integrity by permeabilization and depolarization. TB_KKG6K showed no toxicity in the invertebrate mini-host model Galleria mellonella and a high level of tolerance when topically applied in HEEs. Importantly, the therapeutic potential of TB_KKG6K was confirmed in HEEs infected with S. aureus. The topical application of TB_KKG6K significantly reduced the bacterial load and lowered the pro-inflammatory response in the infected HEEs. These findings reinforce the antibacterial potential and therapeutic efficacy of TB_KKG6K against S. aureus infection, particularly in the context of a cutaneous infection.IMPORTANCEThe emergence of multidrug-resistant bacteria has rendered the exploration of novel therapeutic treatment strategies a pivotal area of research. Among the most promising candidates are amphibian-derived antimicrobial peptides (AMPs), which are ideal for the development of novel drugs due to their multifaceted mode of action. Extensive studies have been conducted on these peptides over the last decade, resulting in the development of temporin B (TB) peptide analogs that have undergone modifications to their primary sequence. These modified analogs have demonstrated enhanced antibacterial and antifungal efficacy, while exhibiting reduced hemolytic activity. TB_KKG6K has the potential to be a promising candidate for topical treatments due to its small size and high antimicrobial activity against pathogens of the human skin. In particular, it demonstrated efficacy against Staphylococcus aureus, a skin commensal that can become an opportunistic pathogen, causing a range of infections from minor skin infections to life-threatening diseases such as bacteremia and sepsis.
Collapse
Affiliation(s)
- Cristina Schöpf
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Knapp
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Jakob Scheler
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Débora C Coraça-Huber
- Research Laboratory for Implant Associated Infections (BIOFILM LAB), Experimental Orthopaedics, University Hospital for Orthopaedics and Traumatology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Peter Ladurner
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Anna C Seybold
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Ulrike Binder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Reinhard Würzner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
2
|
Shi J, Chen C, Kong P, Yu F, Lv Q, Wang Z, Liu Y. Non-Membrane Active Peptide Resensitizes MRSA to β-Lactam Antibiotics and Inhibits S. aureus Virulence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416260. [PMID: 39976117 PMCID: PMC12005801 DOI: 10.1002/advs.202416260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/03/2025] [Indexed: 02/21/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a serious global health threat due to its high morbidity and mortality rates, creating a dire need for novel therapeutic strategies. Antimicrobial peptides (AMPs), with broad-spectrum activity and low propensity for resistance development, show promise as effective antibiotic adjuvants to reverse multidrug-resistance in bacteria. Herein, it is uncovered that a potent and non-toxic AMP termed GN1 substantially resensitizes MRSA to multiple β-lactam antibiotics at low concentrations. Mechanistic studies indicate that GN1 functions by suppressing both the production and enzymatic activity of MRSA-associated resistance determinants, including penicillin-binding protein 2a (PBP2a) and β-lactamase. Additionally, GN1 exhibits a robust anti-virulence profile by inhibiting MRSA biofilm formation and staphyloxanthin production. Furthermore, GN1 induces bacterial metabolic perturbation, resulting in glutamate accumulation and oxidative damage. Importantly, the combination of GN1 with β-lactam antibiotics effectively mitigates MRSA-induced infections in the animal infection models. Collectively, these findings suggest that GN1 represents a potent β-lactam adjuvant and anti-virulence agent, offering a safe and versatile solution to combat MRSA infections.
Collapse
Affiliation(s)
- Jingru Shi
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
| | - Chen Chen
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
| | - Pan Kong
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
| | - Feiyu Yu
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
| | - Qingyan Lv
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
| | - Zhiqiang Wang
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safetythe Ministry of Education of ChinaYangzhou UniversityYangzhou225009China
- Institute of Comparative MedicineYangzhou UniversityYangzhou225009China
| | - Yuan Liu
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safetythe Ministry of Education of ChinaYangzhou UniversityYangzhou225009China
- Institute of Comparative MedicineYangzhou UniversityYangzhou225009China
| |
Collapse
|
3
|
Aghmiyuni ZF, Ahmadi MH, Saderi H. Relationship between the strength of biofilm production and the presence of pvl and mecA genes in Staphylococcus aureus isolated from skin and soft tissue infections. Heliyon 2024; 10:e40524. [PMID: 39654786 PMCID: PMC11625253 DOI: 10.1016/j.heliyon.2024.e40524] [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: 08/20/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
This research sought to investigate the association between the occurrence of the pvl and mecA genes and the strength of biofilm formation, as well as to assess the efficacy of vancomycin and ceftaroline against Staphylococcus aureus strains obtained from skin and soft tissue infections (SSTIs). A total of 134 S. aureus isolates were collected from SSTI patients and identified through standard microbiological techniques. Vancomycin and ceftaroline susceptibility testing were performed using the agar dilution and disc diffusion methods, respectively. PCR analysis was conducted to identify the nuc, mecA, and pvl genes. Biofilm production was measured using the tissue culture plate method. Methicillin-resistant S. aureus (MRSA) represented 58.2 % of the isolates. All isolates displayed biofilm-forming capability, with 10.4 % classified as high-grade biofilm producers, 85.7 % of which were positive for the mecA gene (P = 0.02). 16.4 % of the isolates had pvl gene and 59 % of PVL-positive strains identified as MRSA. Most of the low-grade biofilm producers had the pvl gene (P = 0.03). Vancomycin susceptibility was observed in 98.5 % of isolates, with an MIC₅₀ of 1 μg/mL in 51.4 % of cases. Among MRSA strains, 1.4 % exhibited intermediate resistance to vancomycin, with MICs between 4 and 8 μg/mL. No resistance to ceftaroline was found. The results demonstrate a significant association between biofilm production strength and the occurrence of the mecA and pvl genes; mecA correlated with increased biofilm production, while pvl was associated with lower biofilm levels. These findings offer valuable insights for future studies, suggesting that ceftaroline could be an effective alternative to vancomycin for treating MRSA-related SSTIs, particularly given the increasing resistance to vancomycin.
Collapse
Affiliation(s)
| | | | - Horieh Saderi
- Molecular Microbiology Research Center, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
4
|
Yang R, Cui L, Xu S, Zhong Y, Xu T, Liu J, Lan Z, Qin S, Guo Y. Membrane-Targeting Amphiphilic Honokiol Derivatives Containing an Oxazole Moiety as Potential Antibacterials against Methicillin-Resistant Staphylococcus aureus. J Med Chem 2024; 67:16858-16872. [PMID: 39259708 DOI: 10.1021/acs.jmedchem.4c01860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Infections with methicillin-resistant Staphylococcus aureus (MRSA) are becoming increasingly serious, making the development of novel antimicrobials urgent. Here, we synthesized some amphiphilic honokiol derivatives bearing an oxazole moiety and investigated their antibacterial and hemolytic activities. Bioactivity evaluation showed that E17 possessed significant in vitro antibacterial activity against S. aureus and MRSA, along with low hemolytic activity. Moreover, E17 exhibited rapid bactericidal properties and was not susceptible to resistance. Mechanistic studies indicated that E17 interacts with phosphatidylglycerol and cardiolipin of bacterial cell membranes, leading to changes in cell membrane permeability and polarization, increased intracellular ROS, and leakage of DNA and proteins, thus accelerating bacterial death. Transcriptome analysis further demonstrated that E17 has membrane-targeting effects, affecting the expression of genes related to cell membranes and ABC transporter proteins. Notably, in vivo activity showed that E17 has prominent anti-MRSA efficacy, comparable to vancomycin, and is expected to be a new anti-MRSA drug candidate.
Collapse
Affiliation(s)
- Ruige Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Liping Cui
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Shengnan Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
| | - Yan Zhong
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
| | - Ting Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
| | - Jifeng Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Zhenwei Lan
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Yong Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| |
Collapse
|
5
|
Heidtmann CV, Fejer AR, Stærk K, Pedersen M, Asmussen MG, Hertz FB, Prabhala BK, Frimodt-Møller N, Klitgaard JK, Andersen TE, Nielsen CU, Nielsen P. Hit-to-Lead Identification and Validation of a Triaromatic Pleuromutilin Antibiotic Candidate. J Med Chem 2024; 67:3692-3710. [PMID: 38385364 DOI: 10.1021/acs.jmedchem.3c02153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Herein, we report the hit-to-lead identification of a drug-like pleuromutilin conjugate 16, based on a triaromatic hit reported in 2020. The lead arose as the clear candidate from a hit-optimization campaign in which Gram-positive antibacterial activity, solubility, and P-gp affinity were optimized. Conjugate 16 was extensively evaluated for its in vitro ADMET performance which, apart from solubility, was overall on par with lefamulin. This evaluation included Caco-2 cell permeability, plasma protein binding, hERG inhibition, cytotoxicity, metabolism in microsomes and CYP3A4, resistance induction, and time-kill kinetics. Intravenous pharmacokinetics of 16 proved satisfactory in both mice and pigs; however, oral bioavailability was limited likely due to insufficient solubility. The in vivo efficacy was evaluated in mice, systemically infected with Staphylococcus aureus, where 16 showed rapid reduction in blood bacteriaemia. Through our comprehensive studies, lead 16 has emerged as a highly promising and safe antibiotic candidate for the treatment of Gram-positive bacterial infections.
Collapse
Affiliation(s)
- Christoffer V Heidtmann
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Andreas R Fejer
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Kristian Stærk
- Department of Clinical Research, Research Unit of Clinical Microbiology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Maria Pedersen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Marco G Asmussen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Frederik B Hertz
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Bala K Prabhala
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Niels Frimodt-Møller
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Janne K Klitgaard
- Department of Clinical Research, Research Unit of Clinical Microbiology, University of Southern Denmark, DK-5230 Odense M, Denmark
- Department of Biochemistry and Molecular Biology, Research Unit of Molecular Microbiology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Thomas E Andersen
- Department of Clinical Research, Research Unit of Clinical Microbiology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Carsten U Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Poul Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| |
Collapse
|
6
|
Abstract
HIV infection alters the skin microbiome and predisposes to a wide range of cutaneous infections, from atypical presentations of common skin infections to severe disseminated infections involving the skin that are AIDS-defining illnesses. Bacterial infection of the skin, most commonly caused by Staphylococcus aureus, occurs frequently and can result in bacteremia. Nontuberculous mycobacterial infections that are usually localized to the skin may disseminate, and guidance on the treatment of these infections is limited. Herpes simplex can be severe, and less common presentations such as herpetic sycosis and herpes vegetans have been reported. Severe herpes zoster, including disseminated infection, requires intravenous antiviral treatment. Viral warts can be particularly difficult to treat, and in atypical or treatment-resistant cases a biopsy should be considered. Superficial candidosis occurs very commonly in people living with HIV, and antifungal resistance is an increasing problem in non-albicans Candida species. Systemic infections carry a poor prognosis. In tropical settings the endemic mycoses including histoplasmosis are a problem for people living with HIV, and opportunistic infections can affect those with advanced HIV in all parts of the world. Most cutaneous infections can develop or worsen as a result of immune reconstitution in the weeks to months after starting antiretroviral therapy. Direct microscopic examination of clinical material can facilitate rapid diagnosis and treatment initiation, although culture is important to provide microbiological confirmation and guide treatment.
Collapse
Affiliation(s)
- David J Chandler
- Dermatology Department, University Hospitals Sussex NHS Foundation Trust, Brighton, UK; Department of Global Health & Infection, Brighton and Sussex Medical School, Brighton, UK.
| | - Stephen L Walker
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK; Hospital for Tropical Diseases and Department of Dermatology, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
7
|
Gulone L, Di Gregorio S, Morales M, Haim MS, García S, Perazzi B, Famiglietti A, Mollerach M. The Changing Epidemiology and Antimicrobial Susceptibility of Staphylococcus aureus Isolated from Blood Cultures in a University Hospital from Argentina. Microb Drug Resist 2024; 30:109-117. [PMID: 38133499 DOI: 10.1089/mdr.2023.0219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Staphylococcus aureus bacteremia (SAB) is one of the most common serious bacterial infections worldwide. In this study, we demonstrated changes in SAB epidemiology in an Argentinean University Hospital during an 8-year period (2009-2016). A total of 326 S. aureus clinical isolates were recovered in three periods: P1: 2009-2010, P2: 2012-2014, and P3: 2015-2016. Among these, 127 were methicillin-resistant S. aureus (MRSA) and were characterized by phenotypic and molecular methods. We hereby report a significant decline in multiple drug resistance among MRSA isolates associated with an increase in SCCmec IV between the three periods. A diversity of MRSA-IV clones (mainly ST30-MRSA-IV, ST5-MRSA-IV, and ST8-MRSA-IV) replaced between 2009 and 2016 the previous prevalent MRSA clone causing bloodstream infections at this hospital (ST5-MRSA-I). MRSA population structure continued to diversify between P2 and P3. Notably, ST8-MRSA-IV-t008 related to USA300 was first detected during P2, and ST8-MRSA-IV together with ST30-MRSA-IV related to the Southwest Pacific clone were the more prevalent MRSA genotypes circulating during P3.
Collapse
Affiliation(s)
- Lucía Gulone
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Sabrina Di Gregorio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Maia Morales
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Hospital de Clínicas José de San Martín, Laboratorio de Bacteriología Clínica, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Sol Haim
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Susana García
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Hospital de Clínicas José de San Martín, Laboratorio de Bacteriología Clínica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Beatriz Perazzi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Hospital de Clínicas José de San Martín, Laboratorio de Bacteriología Clínica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Angela Famiglietti
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Hospital de Clínicas José de San Martín, Laboratorio de Bacteriología Clínica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Marta Mollerach
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
8
|
Kawanishi M, Matsuda M, Abo H, Ozawa M, Hosoi Y, Hiraoka Y, Harada S, Kumakawa M, Sekiguchi H. Prevalence and Genetic Characterization of Methicillin-Resistant Staphylococcus aureus Isolated from Pigs in Japan. Antibiotics (Basel) 2024; 13:155. [PMID: 38391541 PMCID: PMC10885860 DOI: 10.3390/antibiotics13020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/24/2024] Open
Abstract
We investigated the prevalence of livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) in pig slaughterhouses from 2018 to 2022 in Japan and the isolates were examined for antimicrobial susceptibility and genetic characteristics by whole-genome analysis. Although the positive LA-MRSA rates on farms (29.6%) and samples (9.9%) in 2022 in Japan remained lower than those observed in European countries exhibiting extremely high rates of confirmed human LA-MRSA infections, these rates showed a gradually increasing trend over five years. The ST398/t034 strain was predominant, followed by ST5/t002, and differences were identified between ST398 and ST5 in terms of antimicrobial susceptibility and the resistance genes carried. Notably, LA-MRSA possessed resistance genes toward many antimicrobial classes, with 91.4% of the ST398 strains harboring zinc resistance genes. These findings indicate that the co-selection pressure associated with multidrug and zinc resistance may have contributed markedly to LA-MRSA persistence. SNP analysis revealed that ST398 and ST5 of swine origin were classified into a different cluster of MRSA from humans, showing the same ST in Japan and lacking the immune evasion genes (scn, sak, or chp). Although swine-origin LA-MRSA is currently unlikely to spread to humans and become a problem in current clinical practice, preventing its dissemination requires using antimicrobials prudently, limiting zinc utilization to the minimum required nutrient, and practicing fundamental hygiene measures.
Collapse
Affiliation(s)
- Michiko Kawanishi
- Veterinary AMR Center, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo 185-8511, Japan
| | - Mari Matsuda
- Veterinary AMR Center, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo 185-8511, Japan
| | - Hitoshi Abo
- Veterinary AMR Center, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo 185-8511, Japan
| | - Manao Ozawa
- Veterinary AMR Center, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo 185-8511, Japan
| | - Yuta Hosoi
- Veterinary AMR Center, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo 185-8511, Japan
| | - Yukari Hiraoka
- Veterinary AMR Center, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo 185-8511, Japan
| | - Saki Harada
- Veterinary AMR Center, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo 185-8511, Japan
| | - Mio Kumakawa
- Veterinary AMR Center, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo 185-8511, Japan
| | - Hideto Sekiguchi
- Veterinary AMR Center, National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo 185-8511, Japan
| |
Collapse
|
9
|
Ren J, Zhang QW, He XJ, Chen XY, Zhou ZD, Zeng ZL, Jin Z, Tang YZ. Design, synthesis, biological evaluation and molecular docking study of novel pleuromutilin derivatives containing substituted benzoxazole as antibacterial agents. J Enzyme Inhib Med Chem 2023; 38:2251712. [PMID: 37664987 PMCID: PMC10478630 DOI: 10.1080/14756366.2023.2251712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/21/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023] Open
Abstract
A series of pleuromutilin analogs containing substituted benzoxazole were designed, synthesised, and assessed for their antibacterial activity both in vivo and in vitro. The MIC of the synthesised derivatives was initially assessed using the broth dilution method against four strains of Staphylococcus aureus (MRSA ATCC 43300, S. aureus ATCC 29213, clinical isolation of S. aureus AD3 and S. aureus 144). Most of the synthesised derivatives displayed prominent in vitro activity (MIC ≤ 0.5 µg/mL). Compounds 50 and 57 exhibited the most effective antibacterial effect against MRSA (MIC = 0.125 µg/mL). Furthermore, the time-kill curves showed that compounds 50 and 57 had a certain inhibitory effect against MRSA in vitro. The in vivo antibacterial activity of compound 50 was evaluated further using a murine thigh model infected with MRSA (-1.24 log10CFU/mL). Compound 50 exhibited superior antibacterial efficacy to tiamulin. It was also found that compound 50 did not display significant inhibitory effect on the proliferation of RAW 264.7 cells. Molecular docking study revealed that compound 50 can effectively bind to the active site of the 50S ribosome (the binding free energy -7.50 kcal/mol).
Collapse
Affiliation(s)
- Jie Ren
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qi-Wen Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xian-Jin He
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiao-Ying Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zi-Dan Zhou
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhen-Ling Zeng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhen Jin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - You-Zhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
10
|
Rocha J, Silva V, Poeta P, Botelho J, Manaia CM. Searching for links between environmental and clinical mecA +Staphylococcus aureus: A comparative genomics study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 895:165078. [PMID: 37356759 DOI: 10.1016/j.scitotenv.2023.165078] [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: 04/17/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
Staphylococcus aureus integrate the list of highly virulent and antibiotic resistant pathogens, mainly due to the mecA gene, associated with methicillin resistance. Given the ubiquity of this species, the aim of this study was to investigate whether closely related mecA+S. aureus found in the environment can be also thrive as clinical isolates and if the respective accessory genome may suggest bacterial adaptation. The genomes of environmental (water, animal facilities, food products, n = 111) isolates were compared with closely related genomes of clinical origin (human patients, n = 103). These genomes, available in the public database NCBI, were analysed for phylogeny, accessory genome, and presence of selected clinically relevant genes (n = 104). The genomes of environmental isolates belonged to 18 multi-locus sequence types (MLSTs), 11 of which also included clinical genomes, a result confirmed based on core-genome analysis. Genes significantly (p ≤ 0.05) more frequent among environmental genomes were related with resistance to β-lactams (blaI, blaPCI), aminoglycosides (ant(6)-Ia), macrolides (mph(C), erm(B)), enterotoxins (seg, sei, sem, sen, seo, seu) and serine protease functions (splB), among others. Genes significantly more frequent among clinical genomes were associated with resistance to macrolides (erm(C)), phenicols (fexA), fosfomycin (murA), the leucocidin virulence gene (lukS-PV), and serine protease functions (splA, splE). It is suggested that mecA+S. aureus can be exchanged between clinical and environmental settings, with accessory traits (particularly antibiotic resistance, virulence and stress response) possibly being associated with the habitat. The interplay between phylogeny and accessory genome is an interesting contribution to better understanding the ecology and evolution of mecA+S. aureus.
Collapse
Affiliation(s)
- Jaqueline Rocha
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Vanessa Silva
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; CECAV - Veterinary and Animal Research Centre, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisboa, Portugal
| | - Patrícia Poeta
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisboa, Portugal
| | - João Botelho
- Centro de Biotecnología y Genómica de Plantas (CBGP), Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Célia M Manaia
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| |
Collapse
|
11
|
Paudel S, Guedry S, Obernuefemann CLP, Hultgren SJ, Walker JN, Kulkarni R. Defining the Roles of Pyruvate Oxidation, TCA Cycle, and Mannitol Metabolism in Methicillin-Resistant Staphylococcus aureus Catheter-Associated Urinary Tract Infection. Microbiol Spectr 2023; 11:e0536522. [PMID: 37378538 PMCID: PMC10433999 DOI: 10.1128/spectrum.05365-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is an important cause of complicated urinary tract infection (UTI) associated with the use of indwelling urinary catheters. Previous reports have revealed host and pathogen effectors critical for MRSA uropathogenesis. Here, we sought to determine the significance of specific metabolic pathways during MRSA UTI. First, we identified four mutants from the Nebraska transposon mutant library in the MRSA JE2 background that grew normally in rich medium but displayed significantly reduced growth in pooled human urine (HU). This prompted us to transduce the uropathogenic MRSA 1369 strain with the transposon mutants in sucD and fumC (tricarboxylic acid [TCA] cycle), mtlD (mannitol metabolism), and lpdA (pyruvate oxidation). Notably, sucD, fumC, and mtlD were also significantly upregulated in the MRSA 1369 strain upon exposure to HU. Compared to the WT, the MRSA 1369 lpdA mutant was significantly defective for (i) growth in HU, and (ii) colonization of the urinary tract and dissemination to the kidneys and the spleen in the mouse model of catheter-associated UTI (CAUTI), which may be attributed to its increased membrane hydrophobicity and higher susceptibility to killing by human blood. In contrast to their counterparts in the JE2 background, the sucD, fumC, and mtlD mutants in the MRSA 1369 background grew normally in HU; however, they displayed significant fitness defects in the CAUTI mouse model. Overall, identification of novel metabolic pathways important for the urinary fitness and survival of MRSA can be used for the development of novel therapeutics. IMPORTANCE While Staphylococcus aureus has historically not been considered a uropathogen, S. aureus urinary tract infection (UTI) is clinically significant in certain patient populations, including those with chronic indwelling urinary catheters. Moreover, most S. aureus strains causing catheter-associated UTI (CAUTI) are methicillin-resistant S. aureus (MRSA). MRSA is difficult to treat due to limited treatment options and the potential to deteriorate into life-threatening bacteremia, urosepsis, and shock. In this study, we found that pathways involved in pyruvate oxidation, TCA cycle, and mannitol metabolism are important for MRSA fitness and survival in the urinary tract. Improved understanding of the metabolic needs of MRSA in the urinary tract may help us develop novel inhibitors of MRSA metabolism that can be used to treat MRSA-CAUTI more effectively.
Collapse
Affiliation(s)
- Santosh Paudel
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Sarah Guedry
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Chloe L. P. Obernuefemann
- Center for Women’s Infectious Disease Research, Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Scott J. Hultgren
- Center for Women’s Infectious Disease Research, Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jennifer N. Walker
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
- Department of Epidemiology, Human Genetics, and Environmental Science, School of Public Health, University of Texas Health Science Center at Houston, Texas, USA
| | - Ritwij Kulkarni
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| |
Collapse
|
12
|
Zhang J, Tu J, Chen Y, Jin X. Clinical characteristics and homology analysis of Staphylococcus aureus from would infection at a tertiary hospital in southern Zhejiang, China. BMC Microbiol 2023; 23:217. [PMID: 37573311 PMCID: PMC10422768 DOI: 10.1186/s12866-023-02921-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 06/28/2023] [Indexed: 08/14/2023] Open
Abstract
OBJECTIVE Staphylococcus aureus (S. aureus), especially Methicillin resistant S. aureus (MRSA), has been disseminated across communities and hospitals, associated with severe infections and organ failure. In order to understand the clinical epidemiological characteristics of S. aureus stains in the First Affiliated Hospital of Wenzhou Medical University in 2018, the prevalence and the drug resistance of S. aureus stains were investigated, for improving the clinical effective prevention and control of S. aureus infection. METHODS A total of 105 S. aureus isolates were separated from wound infection of inpatients in the First Affiliated Hospital of Wenzhou Medical University in 2018, and the department distributions and drug resistance of the isolates were analyzed. The genotyping homology analysis was conducted through the random amplified polymorphic DNA typing (RAPD-PCR) coupled with NTSYS cluster analysis. RESULTS Among the 105 strains of S. aureus, 31 isolates were MRSA. The prevalence of MRSA among inpatients in the Departments of Burn, Trauma, Orthopedics, Nephrology and Neurosurgery were 35.48%, 19.35%, 9.68%, 6.45%, and 29.03%, respectively. Among the 105 strains, 35.24% strains were the hospital-acquired infections (HAI) and 64.76% strains were community-acquired infections (CAI). DNA genotyping of the 105 S. aureus strains showed seventeen different groups, most of which were type I, type VII, type IX, and type VII, the others were scattered. CONCLUSION This study highlights the prevalence of S. aureus strains in the First Affiliated Hospital of Wenzhou Medical University in 2018. The emergence and mutation of the strains should be closely monitored for the prevention and control of the S. aureus infection and transmission in the nosocomial settings.
Collapse
Affiliation(s)
- Jiarong Zhang
- Department of Nosocomial Infection Prevention and Control, the First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou, Zhejiang, 325000, China
| | - Jingjing Tu
- The Key Laboratory of Diagnosis and Controlment for the Development of Chronic Liver Disease of Zhejiang Province, Ouhai District, Wenzhou, Zhejiang, 325000, China
| | - Yongping Chen
- The Key Laboratory of Diagnosis and Controlment for the Development of Chronic Liver Disease of Zhejiang Province, Ouhai District, Wenzhou, Zhejiang, 325000, China.
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou, Zhejiang, 325000, China.
| | - Xiaoya Jin
- Department of Nosocomial Infection Prevention and Control, the First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou, Zhejiang, 325000, China.
- The Key Laboratory of Diagnosis and Controlment for the Development of Chronic Liver Disease of Zhejiang Province, Ouhai District, Wenzhou, Zhejiang, 325000, China.
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
13
|
Stone MC, Mychack A, Coe KA, Walker S. Combining Signal Peptidase and Lipoprotein Processing Inhibitors Overcomes Ayr Resistance in Staphylococcus aureus. Antimicrob Agents Chemother 2023; 67:e0011523. [PMID: 37097175 PMCID: PMC10190671 DOI: 10.1128/aac.00115-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/30/2023] [Indexed: 04/26/2023] Open
Abstract
Antibiotic resistance in bacterial pathogens is an ongoing public health concern. The arylomycins are a class of natural product antibiotics that target the type I signal peptidase, which carries out the terminal step in protein secretion. Here, we used transposon sequencing (Tn-Seq) to profile the effects of the optimized arylomycin derivative G0775 in Staphylococcus aureus. Our transposon libraries include both upregulation and inactivation mutants, allowing us to identify resistance mechanisms and targets for synergism. We identified several cell envelope pathways that, when inactivated, sensitize S. aureus to the arylomycin G0775. These pathways include the lipoprotein processing pathway, and we have shown that inhibitors of this pathway synergize with G0775 even though lipoprotein processing is nonessential in S. aureus. Moreover, we found that blocking this pathway completely reverses Ayr resistance, which is a major resistance mechanism to arylomycins, including G0775. Our Tn-Seq data also showed that upregulation of mprF and several other genes is protective against G0775. Because a subset of these genes was previously found in a Tn-Seq profile of the clinically important antibiotic daptomycin, we tested a set of daptomycin-nonsusceptible clinical isolates with gain-of-function mutations in mprF for susceptibility to arylomycin G0775. Despite structural and mechanistic differences between these antibiotics, we observed similar decreases in susceptibility. Taken together, our results highlight how Tn-Seq profiles that include both gene inactivation and upregulation can identify targets, antibiotic resistance mechanisms, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Madeleine C. Stone
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron Mychack
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Kathryn A. Coe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Suzanne Walker
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Qi X, Shen N, Al Othman A, Mezentsev A, Permyakova A, Yu Z, Lepoitevin M, Serre C, Durymanov M. Metal-Organic Framework-Based Nanomedicines for the Treatment of Intracellular Bacterial Infections. Pharmaceutics 2023; 15:1521. [PMID: 37242762 PMCID: PMC10220673 DOI: 10.3390/pharmaceutics15051521] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Metal-organic frameworks (MOFs) are a highly versatile class of ordered porous materials, which hold great promise for different biomedical applications, including antibacterial therapy. In light of the antibacterial effects, these nanomaterials can be attractive for several reasons. First, MOFs exhibit a high loading capacity for numerous antibacterial drugs, including antibiotics, photosensitizers, and/or photothermal molecules. The inherent micro- or meso-porosity of MOF structures enables their use as nanocarriers for simultaneous encapsulation of multiple drugs resulting in a combined therapeutic effect. In addition to being encapsulated into an MOF's pores, antibacterial agents can sometimes be directly incorporated into an MOF skeleton as organic linkers. Next, MOFs contain coordinated metal ions in their structure. Incorporation of Fe2/3+, Cu2+, Zn2+, Co2+, and Ag+ can significantly increase the innate cytotoxicity of these materials for bacteria and cause a synergistic effect. Finally, abundance of functional groups enables modifying the external surface of MOF particles with stealth coating and ligand moieties for improved drug delivery. To date, there are a number of MOF-based nanomedicines available for the treatment of bacterial infections. This review is focused on biomedical consideration of MOF nano-formulations designed for the therapy of intracellular infections such as Staphylococcus aureus, Mycobacterium tuberculosis, and Chlamydia trachomatis. Increasing knowledge about the ability of MOF nanoparticles to accumulate in a pathogen intracellular niche in the host cells provides an excellent opportunity to use MOF-based nanomedicines for the eradication of persistent infections. Here, we discuss advantages and current limitations of MOFs, their clinical significance, and their prospects for the treatment of the mentioned infections.
Collapse
Affiliation(s)
- Xiaoli Qi
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Ningfei Shen
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Aya Al Othman
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | | | | | - Zhihao Yu
- Institute of Porous Materials from Paris (IMAP), Ecole Normale Supérieure, ESPCI Paris, CNRS, PSL University, 75006 Paris, France
| | - Mathilde Lepoitevin
- Institute of Porous Materials from Paris (IMAP), Ecole Normale Supérieure, ESPCI Paris, CNRS, PSL University, 75006 Paris, France
| | - Christian Serre
- Institute of Porous Materials from Paris (IMAP), Ecole Normale Supérieure, ESPCI Paris, CNRS, PSL University, 75006 Paris, France
| | - Mikhail Durymanov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
- Faculty of Chemistry, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
15
|
Znaidia M, de Souza-Angelo Y, Létoffé S, Staropoli I, Grzelak L, Ghigo JM, Schwartz O, Casartelli N. Exposure to Secreted Bacterial Factors Promotes HIV-1 Replication in CD4 + T Cells. Microbiol Spectr 2023; 11:e0431322. [PMID: 36853052 PMCID: PMC10100953 DOI: 10.1128/spectrum.04313-22] [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: 10/21/2022] [Accepted: 02/08/2023] [Indexed: 03/01/2023] Open
Abstract
Microbial translocation is associated with systemic immune activation in HIV-1 disease. Circulating T cells can encounter microbial products in the bloodstream and lymph nodes, where viral replication takes place. The mechanisms by which bacteria contribute to HIV-associated pathogenesis are not completely deciphered. Here, we examined how bacteria may impact T cell function and viral replication. We established cocultures between a panel of live bacteria and uninfected or HIV-1-infected activated peripheral blood CD4-positive (CD4+) T cells. We show that some bacteria, such as Escherichia coli and Acinetobacter baumannii, sustain lymphocyte activation and enhance HIV-1 replication. Bacteria secrete soluble factors that upregulate CD25 and ICAM-1 cell surface levels and activate NF-κB nuclear translocation. Our data also demonstrate that CD25 polarizes at the virological synapse, suggesting a previously unappreciated role of CD25 during viral replication. These findings highlight how interactions between bacterial factors and T cells may promote T cell activation and HIV-1 replication. IMPORTANCE People living with HIV suffer from chronic immune activation despite effective antiretroviral therapy. Early after infection, HIV-1 actively replicates in the gut, causing the breakage of the intestinal epithelial barrier and microbial translocation. Microbial translocation and chronic immune activation have been proven linked; however, gaps in our knowledge on how bacteria contribute to the development of HIV-related diseases remain. Whether T cells in the peripheral blood react to bacterial products and how this affects viral replication are unknown. We show that some bacteria enriched in people living with HIV activate T cells and favor HIV-1's spread. Bacteria release soluble factors that cause the overexpression of cellular molecules related to their activation state. T cells overexpressing these molecules also replicate HIV-1 more efficiently. These results help us learn more about how HIV-1, T cells, and bacteria interact with each other, as well as the mechanisms behind chronic immune activation.
Collapse
Affiliation(s)
- M. Znaidia
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - Y. de Souza-Angelo
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - S. Létoffé
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Paris, France
| | - I. Staropoli
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - L. Grzelak
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - J. M. Ghigo
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Paris, France
| | - O. Schwartz
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
- Vaccine Research Institute, Créteil, France
| | - N. Casartelli
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| |
Collapse
|
16
|
Al-Trad EI, Chew CH, Che Hamzah AM, Suhaili Z, Rahman NIA, Ismail S, Puah SM, Chua KH, Kwong SM, Yeo CC. The Plasmidomic Landscape of Clinical Methicillin-Resistant Staphylococcus aureus Isolates from Malaysia. Antibiotics (Basel) 2023; 12:antibiotics12040733. [PMID: 37107095 PMCID: PMC10135026 DOI: 10.3390/antibiotics12040733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a priority nosocomial pathogen with plasmids playing a crucial role in its genetic adaptability, particularly in the acquisition and spread of antimicrobial resistance. In this study, the genome sequences of 79 MSRA clinical isolates from Terengganu, Malaysia, (obtained between 2016 and 2020) along with an additional 15 Malaysian MRSA genomes from GenBank were analyzed for their plasmid content. The majority (90%, 85/94) of the Malaysian MRSA isolates harbored 1-4 plasmids each. In total, 189 plasmid sequences were identified ranging in size from 2.3 kb to ca. 58 kb, spanning all seven distinctive plasmid replication initiator (replicase) types. Resistance genes (either to antimicrobials, heavy metals, and/or biocides) were found in 74% (140/189) of these plasmids. Small plasmids (<5 kb) were predominant (63.5%, 120/189) with a RepL replicase plasmid harboring the ermC gene that confers resistance to macrolides, lincosamides, and streptogramin B (MLSB) identified in 63 MRSA isolates. A low carriage of conjugative plasmids was observed (n = 2), but the majority (64.5%, 122/189) of the non-conjugative plasmids have mobilizable potential. The results obtained enabled us to gain a rare view of the plasmidomic landscape of Malaysian MRSA isolates and reinforces their importance in the evolution of this pathogen.
Collapse
Affiliation(s)
- Esra'a I Al-Trad
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| | - Ching Hoong Chew
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Kuala Nerus 21300, Malaysia
| | | | - Zarizal Suhaili
- Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, Besut 22200, Malaysia
| | - Nor Iza A Rahman
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| | - Salwani Ismail
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| | - Suat Moi Puah
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Stephen M Kwong
- Infectious Diseases & Microbiology, School of Medicine, Western Sydney University, Campbelltown 2560, Australia
| | - Chew Chieng Yeo
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| |
Collapse
|
17
|
Linz MS, Mattappallil A, Finkel D, Parker D. Clinical Impact of Staphylococcus aureus Skin and Soft Tissue Infections. Antibiotics (Basel) 2023; 12:557. [PMID: 36978425 PMCID: PMC10044708 DOI: 10.3390/antibiotics12030557] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
The pathogenic bacterium Staphylococcus aureus is the most common pathogen isolated in skin-and-soft-tissue infections (SSTIs) in the United States. Most S. aureus SSTIs are caused by the epidemic clone USA300 in the USA. These infections can be serious; in 2019, SSTIs with S. aureus were associated with an all-cause, age-standardized mortality rate of 0.5 globally. Clinical presentations of S. aureus SSTIs vary from superficial infections with local symptoms to monomicrobial necrotizing fasciitis, which can cause systemic manifestations and may lead to serious complications or death. In order to cause skin infections, S. aureus employs a host of virulence factors including cytolytic proteins, superantigenic factors, cell wall-anchored proteins, and molecules used for immune evasion. The immune response to S. aureus SSTIs involves initial responders such as keratinocytes and neutrophils, which are supported by dendritic cells and T-lymphocytes later during infection. Treatment for S. aureus SSTIs is usually oral therapy, with parenteral therapy reserved for severe presentations; it ranges from cephalosporins and penicillin agents such as oxacillin, which is generally used for methicillin-sensitive S. aureus (MSSA), to vancomycin for methicillin-resistant S. aureus (MRSA). Treatment challenges include adverse effects, risk for Clostridioides difficile infection, and potential for antibiotic resistance.
Collapse
Affiliation(s)
- Matthew S. Linz
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Arun Mattappallil
- Department of Pharmaceutical Services, University Hospital, Newark, NJ 07103, USA
| | - Diana Finkel
- Division of Infectious Diseases, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
18
|
Abramov VM, Kosarev IV, Machulin AV, Priputnevich TV, Deryusheva EI, Nemashkalova EL, Chikileva IO, Abashina TN, Panin AN, Melnikov VG, Suzina NE, Nikonov IN, Selina MV, Khlebnikov VS, Sakulin VK, Samoilenko VA, Gordeev AB, Sukhikh GT, Uversky VN, Karlyshev AV. Limosilactobacillus fermentum 3872 That Produces Class III Bacteriocin Forms Co-Aggregates with the Antibiotic-Resistant Staphylococcus aureus Strains and Induces Their Lethal Damage. Antibiotics (Basel) 2023; 12:antibiotics12030471. [PMID: 36978338 PMCID: PMC10044573 DOI: 10.3390/antibiotics12030471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
LF3872 was isolated from the milk of a healthy lactating and breastfeeding woman. Earlier, the genome of LF3872 was sequenced, and a gene encoding unique bacteriocin was discovered. We have shown here that the LF3872 strain produces a novel thermolabile class III bacteriolysin (BLF3872), exhibiting antimicrobial activity against antibiotic-resistant Staphylococcus aureus strains. Sequence analysis revealed the two-domain structural (lysozyme-like domain and peptidase M23 domain) organization of BLF3872. At least 25% residues of this protein are expected to be intrinsically disordered. Furthermore, BLF3872 is predicted to have a very high liquid-liquid phase separation. According to the electron microscopy data, the bacterial cells of LF3872 strain form co-aggregates with the S. aureus 8325-4 bacterial cells. LF3872 produced bacteriolysin BLF3872 that lyses the cells of the S. aureus 8325-4 mastitis-inducing strain. The sensitivity of the antibiotic-resistant S. aureus collection strains and freshly isolated antibiotic-resistant strains was tested using samples from women with lactation mastitis; the human nasopharynx and oral cavity; the oropharynx of pigs; and the cows with a diagnosis of clinical mastitis sensitive to the lytic action of the LF3872 strain producing BLF3872. The co-cultivation of LF3872 strain with various antibiotic-resistant S. aureus strains for 24 h reduced the level of living cells of these pathogens by six log. The LF3872 strain was found to be able to co-aggregate with all studied S. aureus strains. The cell-free culture supernatant of LF3872 (CSLF3872) induced S. aureus cell damage and ATP leakage. The effectiveness of the bacteriolytic action of LF3872 strain did not depend on the origin of the S. aureus strains. The results reported here are important for the creation of new effective drugs against antibiotic-resistant strains of S. aureus circulating in humans and animals.
Collapse
Affiliation(s)
- Vyacheslav M. Abramov
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
- Correspondence:
| | - Igor V. Kosarev
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Andrey V. Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Tatiana V. Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Ekaterina L. Nemashkalova
- Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Irina O. Chikileva
- Laboratory of Cell Immunity, Blokhin National Research Center of Oncology, Ministry of Health RF, 115478 Moscow, Russia
| | - Tatiana N. Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Alexander N. Panin
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Vyacheslav G. Melnikov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia
| | - Nataliya E. Suzina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Ilia N. Nikonov
- Federal State Educational Institution of Higher Professional Education, Moscow State Academy of Veterinary Medicine and Biotechnology named after K.I. Skryabin, 109472 Moscow, Russia
| | - Marina V. Selina
- Federal State Educational Institution of Higher Professional Education, Moscow State Academy of Veterinary Medicine and Biotechnology named after K.I. Skryabin, 109472 Moscow, Russia
| | | | - Vadim K. Sakulin
- Institute of Immunological Engineering, 142380 Lyubuchany, Russia
| | - Vladimir A. Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Alexey B. Gordeev
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Gennady T. Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Andrey V. Karlyshev
- Department of Biomolecular Sciences, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
19
|
Shearer HL, Loi VV, Weiland P, Bange G, Altegoer F, Hampton MB, Antelmann H, Dickerhof N. MerA functions as a hypothiocyanous acid reductase and defense mechanism in Staphylococcus aureus. Mol Microbiol 2023; 119:456-470. [PMID: 36779383 DOI: 10.1111/mmi.15035] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/14/2023]
Abstract
The major pathogen Staphylococcus aureus has to cope with host-derived oxidative stress to cause infections in humans. Here, we report that S. aureus tolerates high concentrations of hypothiocyanous acid (HOSCN), a key antimicrobial oxidant produced in the respiratory tract. We discovered that the flavoprotein disulfide reductase (FDR) MerA protects S. aureus from this oxidant by functioning as a HOSCN reductase, with its deletion sensitizing bacteria to HOSCN. Crystal structures of homodimeric MerA (2.4 Å) with a Cys43 -Cys48 intramolecular disulfide, and reduced MerACys43 S (1.6 Å) showed the FAD cofactor close to the active site, supporting that MerA functions as a group I FDR. MerA is controlled by the redox-sensitive repressor HypR, which we show to be oxidized to intermolecular disulfides under HOSCN stress, resulting in its inactivation and derepression of merA transcription to promote HOSCN tolerance. Our study highlights the HOSCN tolerance of S. aureus and characterizes the structure and function of MerA as a major HOSCN defense mechanism. Crippling the capacity to respond to HOSCN may be a novel strategy for treating S. aureus infections.
Collapse
Affiliation(s)
- Heather L Shearer
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Vu V Loi
- Freie Universität Berlin, Institute of Biology-Microbiology, Berlin, Germany
| | - Paul Weiland
- Center for Synthetic Microbiology (SYNMIKRO), Department of Chemistry, Philipps-University Marburg, Marburg, Germany.,Center for Tumor Biology and Immunology, Department of Medicine, Philipps-University Marburg, Marburg, Germany
| | - Gert Bange
- Center for Synthetic Microbiology (SYNMIKRO), Department of Chemistry, Philipps-University Marburg, Marburg, Germany.,Max-Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Florian Altegoer
- Center for Synthetic Microbiology (SYNMIKRO), Department of Chemistry, Philipps-University Marburg, Marburg, Germany.,Institute of Microbiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mark B Hampton
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Haike Antelmann
- Freie Universität Berlin, Institute of Biology-Microbiology, Berlin, Germany
| | - Nina Dickerhof
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
20
|
Kawakami T, Yokoyama K, Ikeda T, Kaneko H, Watanabe Y, Nakaminami H. Severity and intractableness of skin infections caused by
Panton–Valentine
leukocidin‐positive methicillin‐resistant
Staphylococcus aureus. JOURNAL OF CUTANEOUS IMMUNOLOGY AND ALLERGY 2023. [DOI: 10.1002/cia2.12294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Tamihiro Kawakami
- Division of Dermatology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Kae Yokoyama
- Division of Dermatology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Takaharu Ikeda
- Division of Dermatology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Hiroshi Kaneko
- Department of Clinical Microbiology, School of Pharmacy Tokyo University of Pharmacy and Life Sciences Tokyo Japan
| | - Yuji Watanabe
- Division of Clinical Laboratory Tohoku Medical and Pharmaceutical University Hospital Sendai Japan
| | - Hidemasa Nakaminami
- Department of Clinical Microbiology, School of Pharmacy Tokyo University of Pharmacy and Life Sciences Tokyo Japan
| |
Collapse
|
21
|
Wang WY, Chiu CF, Lee YT, Hsueh PR, Tsao SM. Molecular epidemiology and phenotypes of invasive methicillin-resistant vancomycin-intermediate Staphylococcus aureus in Taiwan. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:1203-1210. [PMID: 34635425 DOI: 10.1016/j.jmii.2021.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/25/2021] [Accepted: 09/04/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Patients with invasive infections caused by methicillin-resistant Staphylococcus aureus (MRSA), especially those with an elevated minimal inhibitory concentration (MIC) of vancomycin (VA), are likely to have treatment failure and poor outcomes. The aim of this study was to delineate and correlate the genotypes and phenotypes of clinical VA-intermediate S. aureus (VISA) from invasive infections in Taiwan. METHODS Between 2006 and 2010, a total of 670 non-duplicate MRSA isolates were collected from patients with invasive infections, mostly from blood, as part of a nationwide antimicrobial surveillance program named Tigecycline in vitro Surveillance in Taiwan. Among them, 10 (1.5%) VISA (VA MIC = 4 mg/L) isolates were identified. Molecular typing with staphylococcal cassette chromosome mec (SCCmec), multilocus sequence typing, staphylococcal protein A (spa), mec-associated hypervariable region (dru), accessory gene regulator (agr), and pulse-field gel electrophoresis, and phenotypic analysis including antibiotic susceptibility testing, gene encoding Panton-Valentine leukocidin (pvl), and superantigenic toxin profiles, were analyzed. RESULTS All but one isolate was defined as molecular health-care-associated MRSA: 6 as SCCmecIII-ST239-spa t037-agrI-dru7 (1 isolate) and dru14 (5 isolates), 2 as SCCmecII-ST5-spa t586-agrII-dru4, and one as SCCmecII-ST89-spa t3520-agrIII-dru7. One isolate was defined as SCCmecIV-ST59-spa t437-agrI-dru8, which was categorized as molecular community-associated MRSA. Five pulsotypes were identified; only one had a positive D-test and 3 were insusceptible to daptomycin (MIC ≧1 mg/L). Five isolates possessed sea-selk-selq, among them 4 belonged to SCCmecIII-ST239-spa t037-agrI. CONCLUSION In this study, VISA was rarely isolated from invasive MRSA infections, and most cases harbored limited genotypes and corresponding phenotypes.
Collapse
Affiliation(s)
- Wei-Yao Wang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Chen-Feng Chiu
- Department of Internal Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Taichung, Taiwan
| | - Yuan-Ti Lee
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Po-Ren Hsueh
- Departments of Laboratory Medicine and Internal Medicine, China Medical University Hospital, Taichung, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan; Ph.D Program for Aging, School of Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Ming Tsao
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
22
|
Popovich KJ, Weinstein RA. Questioning Old Staphylococcus aureus Beliefs With New Technology. J Infect Dis 2022; 227:1028-1030. [PMID: 36322783 DOI: 10.1093/infdis/jiac439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Kyle J Popovich
- Rush University Medical Center/Cook County Health, Section of Infectious Diseases, University Medical Center , Chicago, Illinois , USA
| | - Robert A Weinstein
- Rush University Medical Center/Cook County Health, Section of Infectious Diseases, University Medical Center , Chicago, Illinois , USA
| |
Collapse
|
23
|
Lade H, Joo HS, Kim JS. Molecular Basis of Non-β-Lactam Antibiotics Resistance in Staphylococcus aureus. Antibiotics (Basel) 2022; 11:1378. [PMID: 36290036 PMCID: PMC9598170 DOI: 10.3390/antibiotics11101378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the most successful human pathogens with the potential to cause significant morbidity and mortality. MRSA has acquired resistance to almost all β-lactam antibiotics, including the new-generation cephalosporins, and is often also resistant to multiple other antibiotic classes. The expression of penicillin-binding protein 2a (PBP2a) is the primary basis for β-lactams resistance by MRSA, but it is coupled with other resistance mechanisms, conferring resistance to non-β-lactam antibiotics. The multiplicity of resistance mechanisms includes target modification, enzymatic drug inactivation, and decreased antibiotic uptake or efflux. This review highlights the molecular basis of resistance to non-β-lactam antibiotics recommended to treat MRSA infections such as macrolides, lincosamides, aminoglycosides, glycopeptides, oxazolidinones, lipopeptides, and others. A thorough understanding of the molecular and biochemical basis of antibiotic resistance in clinical isolates could help in developing promising therapies and molecular detection methods of antibiotic resistance.
Collapse
Affiliation(s)
- Harshad Lade
- Department of Laboratory Medicine, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul 05355, Korea
| | - Hwang-Soo Joo
- Department of Biotechnology, College of Engineering, Duksung Women’s University, Seoul 01369, Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul 05355, Korea
| |
Collapse
|
24
|
Gu K, Ouyang P, Hong Y, Dai Y, Tang T, He C, Shu G, Liang X, Tang H, Zhu L, Xu Z, Yin L. Geraniol inhibits biofilm formation of methicillin-resistant Staphylococcus aureus and increase the therapeutic effect of vancomycin in vivo. Front Microbiol 2022; 13:960728. [PMID: 36147840 PMCID: PMC9485828 DOI: 10.3389/fmicb.2022.960728] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/16/2022] [Indexed: 12/04/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is among the common drug resistant bacteria, which has gained worldwide attention due to its high drug resistance and infection rates. Biofilms produced by S. aureus are known to increase antibiotic resistance, making the treatment of S. aureus infections even more challenging. Hence, inhibition of biofilm formation has become an alternative strategy for controlling persistent infections. In this study, we evaluated the efficacy of geraniol as a treatment for MRSA biofilm infection. The results of crystal violet staining indicated that 256 μg/mL concentration of geraniol inhibited USA300 biofilm formation by 86.13% and removed mature biofilms by 49.87%. Geraniol exerted its anti-biofilm effect by influencing the major components of the MRSA biofilm structure. We found that geraniol inhibited the synthesis of major virulence factors, including staphyloxanthin and autolysins. The colony count revealed that geraniol inhibited staphyloxanthin and sensitized USA300 cells to hydrogen peroxide. Interestingly, geraniol not only reduced the release of extracellular nucleic acids (eDNA) but also inhibited cell autolysis. Real-time polymerase chain reaction data revealed the downregulation of genes involved in biofilm formation, which verified the results of the phenotypic analysis. Geraniol increased the effect of vancomycin in eliminating USA300 biofilms in a mouse infection model. Our findings revealed that geraniol effectively inhibits biofilm formation in vitro. Furthermore, in combination with vancomycin, geraniol can reduce the biofilm adhesion to the implant in mice. This suggests the potential of geraniol as an anti-MRSA biofilm drug and can provide a solution for the clinical treatment of biofilm infection.
Collapse
|
25
|
Beavers WN, Munneke MJ, Stackhouse AR, Freiberg JA, Skaar EP. Host Polyunsaturated Fatty Acids Potentiate Aminoglycoside Killing of Staphylococcus aureus. Microbiol Spectr 2022; 10:e0276721. [PMID: 35377191 PMCID: PMC9045252 DOI: 10.1128/spectrum.02767-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
Aminoglycoside antibiotics rely on the proton motive force to enter the bacterial cell, and facultative anaerobes like Staphylococcus aureus can shift energy generation from respiration to fermentation, becoming tolerant of aminoglycosides. Following this metabolic shift, high concentrations of aminoglycosides are required to eradicate S. aureus infections, which endangers the host due to the toxicity of aminoglycosides. Membrane-disrupting molecules prevent aminoglycoside tolerance in S. aureus by facilitating passive entry of the drug through the membrane. Polyunsaturated fatty acids (PUFAs) increase membrane permeability when incorporated into S. aureus. Here, we report that the abundant host-derived PUFA arachidonic acid increases the susceptibility of S. aureus to aminoglycosides, decreasing the aminoglycoside concentration needed to kill S. aureus. We demonstrate that PUFAs and aminoglycosides synergize to kill multiple strains of S. aureus, including both methicillin-resistant and -susceptible S. aureus. We also present data showing that PUFAs and aminoglycosides effectively kill S. aureus small colony variants, strains that are particularly recalcitrant to killing by many antibiotics. We conclude that cotreatment with PUFAs, which are molecules with low host toxicity, and aminoglycosides decreases the aminoglycoside concentration necessary to kill S. aureus, lowering the toxic side effects to the host associated with prolonged aminoglycoside exposure. IMPORTANCE Staphylococcus aureus infects every niche of the human host, and these infections are the leading cause of Gram-positive sepsis. Aminoglycoside antibiotics are inexpensive, stable, and effective against many bacterial infections. However, S. aureus can shift its metabolism to become tolerant of aminoglycosides, requiring increased concentrations and/or longer courses of treatment, which can cause severe host toxicity. Here, we report that polyunsaturated fatty acids (PUFAs), which have low host toxicity, disrupt the S. aureus membrane, making the pathogen susceptible to aminoglycosides. Additionally, cotreatment with aminoglycosides is effective at killing S. aureus small colony variants, strains that are difficult to treat with antibiotics. Taken together, the data presented herein show the promise of PUFA cotreatment to increase the efficacy of aminoglycosides against S. aureus infections and decrease the risk to the human host of antibiotic-induced toxicity.
Collapse
Affiliation(s)
- William N. Beavers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathobiological Sciences, Louisiana State University and Agricultural and Mechanical College, School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Matthew J. Munneke
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alex R. Stackhouse
- Department of Pathobiological Sciences, Louisiana State University and Agricultural and Mechanical College, School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Jeffrey A. Freiberg
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
26
|
Comparative Genomic Analysis of a Panton–Valentine Leukocidin-Positive ST22 Community-Acquired Methicillin-Resistant Staphylococcus aureus from Pakistan. Antibiotics (Basel) 2022; 11:antibiotics11040496. [PMID: 35453247 PMCID: PMC9033113 DOI: 10.3390/antibiotics11040496] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/27/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Staphylococcus aureus (S. aureus) ST22 is considered a clinically important clone because an epidemic strain EMRSA-15 belongs to ST22, and several outbreaks of this clone have been documented worldwide. We performed genomic analysis of an S. aureus strain Lr2 ST22 from Pakistan and determined comparative analysis with other ST22 strains. The genomic data show that Lr2 belongs to spa-type t2986 and harbors staphylococcal cassette chromosome mec (SCCmec) type IVa(2B), one complete plasmid, and seven prophages or prophage-like elements. The strain harbors several prophage-associated virulence factors, including Panton–Valentine leukocidin (PVL) and toxic shock syndrome toxin (TSST). The single nucleotide polymorphism (SNPs)-based phylogenetic relationship inferred from whole genome and core genome revealed that strain Lr2 exhibits the nearest identities to a South African community-acquired methicillin-resistant S. aureus (CA-MRSA) ST22 strain and makes a separate clade with an Indian CA-MRSA ST22 strain. Although most ST22 strains carry blaZ, mecA, and mutations in gyrA, the Lr2 strain does not have the blaZ gene but, unlike other ST22 strains, carries the antibiotic resistance genes erm(C) and aac(6′)-Ie-aph(2″)-Ia. Among ST22 strains analyzed, only the strain Lr2 possesses both PVL and TSST genes. The functional annotation of genes unique to Lr2 revealed that mobilome is the third-largest Cluster of Orthologous Genes (COGs) category, which encodes genes associated with prophages and transposons. This possibly makes methicillin-resistant S. aureus (MRSA) Lr2 ST22 strain highly virulent, and this study would improve the knowledge of MRSA ST22 strains in Pakistan. However, further studies are needed on a large collection of MRSA to comprehend the genomic epidemiology and evolution of this clone in Pakistan.
Collapse
|
27
|
Forrester JD, Cao S, Schaps D, Liou R, Patil A, Stave C, Sokolow SH, Leo GD. Influence of Socioeconomic and Environmental Determinants of Health on Human Infection and Colonization with Antibiotic-Resistant and Antibiotic-Associated Pathogens: A Scoping Review. Surg Infect (Larchmt) 2022; 23:209-225. [PMID: 35100052 DOI: 10.1089/sur.2021.348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Antibiotic-resistant and antibiotic-associated pathogens are commonly encountered by surgeons. Pathogens such as methicillin-resistant Staphylococcus aureus (MRSA), Clostridioides difficile infection (CDI), and carbapenem-resistant Enterobacteriaceae (CRE) result in considerable human morbidity, mortality, and excess healthcare expenditure. Human colonization or infection can result from exposure to these pathogens across a range of domains both inside and outside of the built healthcare environment, exposure that may be influenced by socioeconomic and environmental determinants of health, the importance of which has not been investigated fully. Methods: We performed a scoping review of published literature describing potential socioeconomic and environmental variables that may increase the likelihood of human infection or colonization with common antibiotic-resistant or antibiotic-associated pathogens, using MRSA, CDI, and CRE as examples. Results: We identified 7,916 articles meeting initial search criteria. Of these, 101 provided supportive evidence of socioeconomic and environmental determinants of human infection or colonization and were included in the scoping review after abstract and full-text screening. Sixty-seven evaluated MRSA, nine evaluated CRE, and 29 evaluated CDI. Twenty-nine articles evaluated exposure to livestock or companion animals; 28, exposure to antibiotics; 20, impact of socioeconomic factors, education level, or race; 14, the influence of temperature, humidity, or season; 13, the effect of travel or human population migration; 11, exposure to built healthcare environments; and eight assessed impact of population density or urbanization. Conclusions: Although articles outlining socioeconomic and environmental drivers of antibiotic-resistant and antibiotic-associated infection are still disconcertedly few, evidence of such associations are overwhelming for MRSA and CDI and supportive for CRE. Additional research is needed to investigate the role and importance of different potential socioeconomic and environmental drivers of antibiotic-resistant and antibiotic-associated infections and colonization in humans.
Collapse
Affiliation(s)
- Joseph D Forrester
- Division of General Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Siqi Cao
- School of Medicine, Stanford University, Stanford, California, USA
| | - Diego Schaps
- School of Medicine, Duke University, Durham, North Carolina, USA
| | - Raymond Liou
- School of Medicine, Stanford University, Stanford, California, USA
| | | | - Christopher Stave
- School of Medicine, Stanford University, Stanford, California, USA
- Lane Medical Library, Stanford University, Stanford, California, USA
| | - Susanne H Sokolow
- Woods Institute for the Environment, Stanford University, Stanford, California, USA
- Marine Science Institute, University of California Santa Barbara, Santa Barbara, California, USA
| | - Giulio De Leo
- Woods Institute for the Environment, Stanford University, Stanford, California, USA
- Hopkins Marine Station, Stanford University, Stanford, California, USA
| |
Collapse
|
28
|
Monteith AJ, Miller JM, Beavers WN, Juttukonda LJ, Skaar EP. Increased Dietary Manganese Impairs Neutrophil Extracellular Trap Formation Rendering Neutrophils Ineffective at Combating Staphylococcus aureus. Infect Immun 2022; 90:e0068521. [PMID: 35191757 PMCID: PMC8929375 DOI: 10.1128/iai.00685-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 11/20/2022] Open
Abstract
Dietary metals can modify the risk to infection. Previously, we demonstrated that heightened dietary manganese (Mn) during systemic Staphylococcus aureus infection increases S. aureus virulence. However, immune cells also operate in these same environments and the effect of dietary Mn on neutrophil function in vivo has not been assessed. This study reveals that increased concentrations of Mn impairs mitochondrial respiration and superoxide production in neutrophils responding to S. aureus. As a result, high Mn accelerates primary degranulation, while impairing suicidal neutrophil extracellular trap (NET) formation, which decreases bactericidal activity. In vivo, elevated dietary Mn accumulated extracellularly in the heart, indicating that excess Mn may be more bioavailable in the heart. Coinciding with this phenotype, neutrophil function in the heart was most impacted by a high Mn diet, as neutrophils produced lower levels of mitochondrial superoxide and underwent less suicidal NET formation. Consistent with an ineffective neutrophil response when mice are on a high Mn diet, S. aureus burdens were increased in the heart and mice were more susceptible to systemic infection. Therefore, elevated dietary Mn not only affects S. aureus but also renders neutrophils less capable of restricting staphylococcal infection.
Collapse
Affiliation(s)
- Andrew J. Monteith
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeanette M. Miller
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - William N. Beavers
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Lillian J. Juttukonda
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, & Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
29
|
Maeda R, Kobayashi H, Higashidani M, Matsuhisa T, Sawa A, Miyake K, Tayama Y, Kimura K, Itoh H, Okano T, Seike S, Yamanaka H. Molecular epidemiological and pharmaceutical studies of methicillin-resistant Staphylococcus aureus isolated at hospitals in Kure City, Japan. Access Microbiol 2022; 4:000319. [PMID: 35355871 PMCID: PMC8941957 DOI: 10.1099/acmi.0.000319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/13/2021] [Indexed: 11/18/2022] Open
Abstract
Introduction Methicillin-resistant Staphylococcus aureus (MRSA) is one of the major pathogens of nosocomial infections throughout the world. In the medical field, it is extremely important to this pathogen’s trends when considering infection control. Hypothesis/Gap Statement We hypothesized that clarifying the characteristics of clinically isolated MRSA would contribute to infection control and proper use of antimicrobial agents against MRSA. Aim The purpose of this study is to elucidate the genetic and biological characteristics of the MRSA isolates found at our hospital and to reveal changes in the spread of this pathogen in the local area where we live. Methodology Pulse-field gel electrophoresis (PFGE) and polymerase chain reaction were used for the genetic analyses of MRSA isolates. Toxin production by each isolate was examined using toxin-specific detection systems. Results During the 3 years from 2017 through 2019, over 1000 MRSA strains were isolated at our hospital. Genomic analysis of 237 of these clinical isolates by PFGE revealed 12 PFGE types (types A to L), each consisting of five or more MRSA clinical strains with over 80% genetic similarity. Examination of the SCCmec genotypes found that 219 of 237 isolated MRSA strains (approximately 92%) were SCCmec genotype II or IV and that only four of the isolates carried the Panton−Valentine leukocidin (PVL) gene. Examination of the toxin production of the isolates using staphylococcal enterotoxin detection kits found that most isolates carrying the SCCmec genotype II produced enterotoxin B and/or C, and that most isolates carrying the SCCmec genotype IV produced enterotoxin A. Conclusion The present results revealed that MRSA isolates with common properties were isolated at certain rates throughout the 3 year study period, suggesting that relatively specific MRSA clones may have settled in the local area around our hospital. We also examine the relationship between antimicrobial usage over time and changes in MRSA isolation rates.
Collapse
Affiliation(s)
- Ryuto Maeda
- National Hospital Organization, Kure Medical Center and Chugoku Cancer Center, Aoyama-cho, Kure, Hiroshima 737-0023, Japan
- Research Center for Pharmaceutical Health Care and Sciences, Faculty of Pharmaceutical Sciences, Hiroshima International University, Hiro-Koshingai, Kure, Hiroshima 737-0112, Japan
| | - Hidetomo Kobayashi
- Laboratory of Molecular Microbiological Science, Faculty of Pharmaceutical Sciences, Hiroshima International University, Hiro-Koshingai, Kure, Hiroshima 737-0112, Japan
| | - Mami Higashidani
- National Hospital Organization, Kure Medical Center and Chugoku Cancer Center, Aoyama-cho, Kure, Hiroshima 737-0023, Japan
| | - Tetsuaki Matsuhisa
- National Hospital Organization, Kure Medical Center and Chugoku Cancer Center, Aoyama-cho, Kure, Hiroshima 737-0023, Japan
| | - Akihiro Sawa
- Research Center for Pharmaceutical Health Care and Sciences, Faculty of Pharmaceutical Sciences, Hiroshima International University, Hiro-Koshingai, Kure, Hiroshima 737-0112, Japan
| | - Katsushi Miyake
- Research Center for Pharmaceutical Health Care and Sciences, Faculty of Pharmaceutical Sciences, Hiroshima International University, Hiro-Koshingai, Kure, Hiroshima 737-0112, Japan
| | - Yoshitaka Tayama
- Research Center for Pharmaceutical Health Care and Sciences, Faculty of Pharmaceutical Sciences, Hiroshima International University, Hiro-Koshingai, Kure, Hiroshima 737-0112, Japan
| | - Kouji Kimura
- Research Center for Pharmaceutical Health Care and Sciences, Faculty of Pharmaceutical Sciences, Hiroshima International University, Hiro-Koshingai, Kure, Hiroshima 737-0112, Japan
| | - Hiroyuki Itoh
- Saiseikai Kure Hospital, Sanjo, Kure, Hiroshima 737-0821, Japan
| | - Taichi Okano
- Saiseikai Kure Hospital, Sanjo, Kure, Hiroshima 737-0821, Japan
| | - Soshi Seike
- Laboratory of Molecular Microbiological Science, Faculty of Pharmaceutical Sciences, Hiroshima International University, Hiro-Koshingai, Kure, Hiroshima 737-0112, Japan
| | - Hiroyasu Yamanaka
- Laboratory of Molecular Microbiological Science, Faculty of Pharmaceutical Sciences, Hiroshima International University, Hiro-Koshingai, Kure, Hiroshima 737-0112, Japan
| |
Collapse
|
30
|
Popovich KJ, Thiede SN, Zawitz C, Payne D, Aroutcheva A, Schoeny M, Green SJ, Snitkin ES, Weinstein RA. Genomic Analysis of Community Transmission Networks for MRSA among Females Entering a Large Inner-City Jail. Open Forum Infect Dis 2022; 9:ofac049. [PMID: 35211635 PMCID: PMC8863081 DOI: 10.1093/ofid/ofac049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/25/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Background
It is unclear if there are differences in MRSA risk between sexes in high-risk populations.
Methods
Females incarcerated at the Cook County Jail were enrolled within 72 hours of intake. Surveillance cultures (nares, throat, groin) were collected to determine prevalence of MRSA colonization. A survey was administered to identify colonization predictors. Univariate and multivariate analyses were performed to identify predictors of colonization at intake. Genomic sequencing was performed on MRSA colonization and archived clinical isolates.
Results
250 women were enrolled (70% AA, 15% Hispanic) with 70% previously in jail. The prevalence of MRSA colonization at intake was 20%, with 42% of those colonized solely in the throat or groin. Univariate predictors of MRSA colonization at entrance were illicit drug use, unstable housing, engaging in anal sex, recent exchange of sex for drugs/money, and a higher number of recent sexual partners. With multivariate adjustment for race/ethnicity, use of needles for illicit drugs was a significant predictor of MRSA. Use of illicit drugs was also associated with inclusion in a genomic cluster.
Nares colonization was significantly associated with not being in a genomic cluster (18.8% vs 78.6%, p<0.001), whereas exclusive extra-nasal colonization was associated (OR 15.89, p<0.001).
Conclusion
We found that a high proportion (20%) of females entered jail colonized with MRSA, suggesting that previously reported sex disparities of a lower risk in women may not apply to high-risk populations. Our findings suggest high-risk activities or venues in the community for MRSA, with potential for directing sex-specific interventions.
Collapse
Affiliation(s)
- Kyle J Popovich
- Rush University Medical Center/Cook County Health, Chicago, IL, USA
| | | | - Chad Zawitz
- Cermak Health Services, Cook County Health, Chicago, IL, USA
| | - Darjai Payne
- Rush University Medical Center, Chicago, IL, USA
| | - Alla Aroutcheva
- Rush University Medical Center/Cook County Health, Chicago, IL, USA
| | | | | | | | | |
Collapse
|
31
|
Fang HQ, Zeng J, Wang SK, Wang X, Chen F, Li B, Liu J, Jin Z, Liu YH, Tang YZ. Discovery of Novel Pleuromutilin Derivatives as Potent Antibacterial Agents for the Treatment of MRSA Infection. Molecules 2022; 27:931. [PMID: 35164203 PMCID: PMC8838415 DOI: 10.3390/molecules27030931] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
A series of novel pleuromutilin derivatives containing nitrogen groups on the side chain of C14 were synthesized under mild conditions. Most of the synthesized derivatives displayed potent antibacterial activities. Compound 9 was found to be the most active antibacterial derivative against MRSA (MIC = 0.06 μg/mL). Furthermore, the result of time-kill curves showed that compound 9 had a certain inhibitory effect against MRSA in vitro. Moreover, according to a surface plasmon resonance (SPR) study, compound 9 (KD = 1.77 × 10-8 M) showed stronger affinity to the 50S ribosome than tiamulin (KD = 2.50 × 10-8 M). The antibacterial activity of compound 9 was further evaluated in an MRSA-infected murine thigh model. Compared to the negative control group, tiamulin reduced MRSA load (~0.7 log10 CFU/mL), and compound 9 performed a treatment effect (~1.3 log10 CFU/mL). In addition, compound 9 was evaluated in CYP450 inhibition assay and showed only moderate in vitro CYP3A4 inhibition (IC50 = 2.92 μg/mL).
Collapse
Affiliation(s)
- Han-Qing Fang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
| | - Jie Zeng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
| | - Shou-Kai Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
| | - Xiao Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
| | - Fang Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
| | - Bo Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
| | - Jie Liu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
| | - Zhen Jin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Ya-Hong Liu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - You-Zhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.-Q.F.); (J.Z.); (S.-K.W.); (X.W.); (F.C.); (B.L.); (J.L.); (Z.J.); (Y.-H.L.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| |
Collapse
|
32
|
Monteith AJ, Miller JM, Williams JM, Voss K, Rathmell JC, Crofford LJ, Skaar EP. Altered Mitochondrial Homeostasis during Systemic Lupus Erythematosus Impairs Neutrophil Extracellular Trap Formation Rendering Neutrophils Ineffective at Combating Staphylococcus aureus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:454-463. [PMID: 34930781 PMCID: PMC8761356 DOI: 10.4049/jimmunol.2100752] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/09/2021] [Indexed: 01/17/2023]
Abstract
Inflammation involves a delicate balance between pathogen clearance and limiting host tissue damage, and perturbations in this equilibrium promote disease. Patients suffering from autoimmune diseases, such as systemic lupus erythematosus (SLE), have higher levels of serum S100A9 protein and increased risk for infection. S100A9 is highly abundant within neutrophils and modulates antimicrobial activity in response to bacterial pathogens. We reasoned that increased serum S100A9 in SLE patients reflects accumulation of S100A9 protein in neutrophils and may indicate altered neutrophil function. In this study, we demonstrate elevated S100A9 protein within neutrophils from SLE patients, and MRL/lpr mice associates with lower mitochondrial superoxide, decreased suicidal neutrophil extracellular trap formation, and increased susceptibility to Staphylococcus aureus infection. Furthermore, increasing mitochondrial superoxide production restored the antibacterial activity of MRL/lpr neutrophils in response to S. aureus These results demonstrate that accumulation of intracellular S100A9 associates with impaired mitochondrial homeostasis, thereby rendering SLE neutrophils inherently less bactericidal.
Collapse
Affiliation(s)
- Andrew J Monteith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Jeanette M Miller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Jonathan M Williams
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; and
| | - Kelsey Voss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; and
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN
| | - Leslie J Crofford
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; and
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN;
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; and
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
33
|
Mitochondrial calcium uniporter affects neutrophil bactericidal activity during Staphylococcus aureus infection. Infect Immun 2021; 90:e0055121. [PMID: 34871043 DOI: 10.1128/iai.00551-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neutrophils simultaneously restrict Staphylococcus aureus dissemination and facilitate bactericidal activity during infection through the formation of neutrophil extracellular traps (NETs). Neutrophils that produce higher levels of mitochondrial superoxide undergo enhanced terminal NET formation (suicidal NETosis) in response to S. aureus; however, mechanisms regulating mitochondrial homeostasis upstream of neutrophil antibacterial processes are not fully resolved. Here, we demonstrate that mitochondrial calcium uptake 1 (MICU1)-deficient (MICU1-/-) neutrophils accumulate higher levels of calcium and iron within the mitochondria in a mitochondrial calcium uniporter (MCU)-dependent manner. Corresponding with increased ion flux through the MCU, mitochondrial superoxide production is elevated, thereby increasing the propensity for MICU1-/- neutrophils to undergo suicidal NETosis rather than primary degranulation in response to S. aureus. Increased NET formation augments macrophage killing of bacterial pathogens. Similarly, MICU1-/- neutrophils alone are not more antibacterial towards S. aureus, but rather enhanced suicidal NETosis by MICU1-/- neutrophils facilitates increased bactericidal activity in the presence of macrophages. Similarly, mice with a deficiency in MICU1 restricted to cells expressing LysM exhibit lower bacterial burdens in the heart with increased survival during systemic S. aureus infection. Coinciding with the decrease in S. aureus burdens, MICU1-/- neutrophils in the heart produced higher levels of mitochondrial superoxide and undergo enhanced suicidal NETosis. These results demonstrate that ion flux by the MCU affects the antibacterial function of neutrophils during S. aureus infection.
Collapse
|
34
|
Further Insight into the Mechanism of Human PMN Lysis following Phagocytosis of Staphylococcus aureus. Microbiol Spectr 2021; 9:e0088821. [PMID: 34704790 PMCID: PMC8549732 DOI: 10.1128/spectrum.00888-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Staphylococcus aureus is an important human pathogen that can cause a variety of diseases ranging from mild superficial skin infections to life-threatening conditions like necrotizing pneumonia, endocarditis, and septicemia. Polymorphonuclear leukocytes (PMNs; neutrophils in particular herein) are essential for host defense against S. aureus infections, and the microbe is phagocytosed readily. Most ingested bacteria are killed, but some S. aureus strains—such as the epidemic USA300 strain—have an enhanced ability to cause PMN lysis after phagocytosis. Although progress has been made, the mechanism for lysis after phagocytosis of S. aureus remains incompletely determined. Here, we tested the hypothesis that disruption of phagosome integrity and escape of S. aureus from the PMN phagosome into the cytoplasm precedes PMN lysis. We used USA300 wild-type and isogenic deletion strains to evaluate and/or verify the role of selected S. aureus molecules in this cytolytic process. Compared to the wild-type USA300 strain, Δagr, Δhla, ΔlukGH, and Δpsm strains each caused significantly less lysis of human PMNs 3 h and/or 6 h after phagocytosis, consistent with previous studies. Most notably, confocal microscopy coupled with selective permeabilization assays demonstrated that phagosome membrane integrity is largely maintained prior to PMN lysis after S. aureus phagocytosis. We conclude that PMN lysis does not require escape of S. aureus from the phagosome to the cytoplasm and that these are independent phenomena. The findings are consistent with the ability of S. aureus (via selected molecules) to trigger lysis of human PMNs by an undetermined signaling mechanism. IMPORTANCES. aureus strain USA300 has the ability to cause rapid lysis of human neutrophils after phagocytosis. Although this phenomenon likely contributes to the success of USA300 as a human pathogen, our knowledge of the mechanism remains incomplete. Here, we used a selective permeabilization assay coupled with confocal microscopy to demonstrate that USA300 is contained within human neutrophil phagosomes until the point of host cell lysis. Thus, consistent with a process in macrophages, S. aureus fails to escape into the neutrophil cytoplasm prior to cytolysis.
Collapse
|
35
|
Long B, Gottlieb M. Diagnosis and Management of Cellulitis and Abscess in the Emergency Department Setting: An Evidence-Based Review. J Emerg Med 2021; 62:16-27. [PMID: 34657784 DOI: 10.1016/j.jemermed.2021.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/04/2021] [Accepted: 09/11/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cellulitis and abscess are a common reason for presentation to the emergency department, although there are several nuances to the care of these patients. OBJECTIVE The purpose of this narrative review article was to provide a summary of the background, pathophysiology, diagnosis, and management of cellulitis and abscesses with a focus on emergency clinicians. DISCUSSION The most common bacteria causing cellulitis are Staphylococcus aureus, Streptococcus pyogenes, and other β-hemolytic streptococci, and methicillin-resistant S. aureus is most common in abscesses. The history and physical examination are helpful in differentiating cellulitis and abscess in many cases, and point-of-care ultrasound can be a useful tool in unclear cases. Treatment for cellulitis typically involves a penicillin or cephalosporin, and treatment of abscesses is incision and drainage. Loop drainage is preferred over the traditional incision and drainage technique, and adjunctive antibiotics can be considered. Most patients can be managed as outpatient. CONCLUSIONS It is essential for emergency physicians to be aware of the current evidence regarding the diagnosis and management of patients with cellulitis and abscess.
Collapse
Affiliation(s)
- Brit Long
- Department of Emergency Medicine, Brooke Army Medical Center, Houston, Texas
| | - Michael Gottlieb
- Department of Emergency Medicine, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
36
|
Achek R, El-Adawy H, Hotzel H, Hendam A, Tomaso H, Ehricht R, Neubauer H, Nabi I, Hamdi TM, Monecke S. Molecular Characterization of Staphylococcus aureus Isolated from Human and Food Samples in Northern Algeria. Pathogens 2021; 10:pathogens10101276. [PMID: 34684225 PMCID: PMC8537606 DOI: 10.3390/pathogens10101276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 01/04/2023] Open
Abstract
Staphylococcus aureus is a commensal resident of the skin and nasal cavities of humans and can cause various infections. Some toxigenic strains can contaminate food matrices and cause foodborne intoxications. The present study aimed to provide relevant information (clonal complex lineages, agr types, virulence and antimicrobial resistance-associated genes) based on DNA microarray analyses as well as the origins and dissemination of several circulating clones of 60 Staphylococcus aureus isolated from food matrices (n = 24), clinical samples (n = 20), and nasal carriers (n = 16) in northern Algeria. Staphylococcus aureus were genotyped into 14 different clonal complexes. Out of 60 S. aureus, 13 and 10 isolates belonged to CC1-MSSA and CC97-MSSA, respectively. The CC 80-MRSA-IV was the predominant S. aureus strain in clinical isolates. The accessory gene regulator allele agr group III was mainly found among clinical isolates (70.4%). Panton–Valentine leukocidin genes lukF/lukS-PV were detected in 13.3% of isolates that all belonged to CC80-MRSA. The lukF/S-hlg, hlgA, and hla genes encoding for hemolysins and leucocidin components were detected in all Staphylococcusaureus isolates. Clinical and food isolates harbored more often the antibiotic resistance genes markers. Seventeen (28.3%) methicillin-resistant Staphylococcus aureus carrying the mecA gene localized on a SCCmec type IV element were identified. The penicillinase operon (blaZ/I/R) was found in 71.7% (43/60) of isolates. Food isolates belonging to CC97-MSSA carried several antibiotic resistance genes (blaZ, ermB, aphA3, sat, tetM, and tetK). The results of this study showed that all clones were found in their typical host, but interestingly, some nasal carriers had isolates assigned to CC705 thought to be absent in humans. The detection of MRSA strains among food isolates should be considered as a potential public health risk. Therefore, controlling the antibiotics prescription for a rational use in human and animal infections is mandatory.
Collapse
Affiliation(s)
- Rachid Achek
- Faculty of Nature and Life and Earth Sciences, Djilali-Bounaama University, Soufay, Khemis-Miliana 44225, Algeria;
- Laboratory of Food Hygiene and Quality Assurance System, High National Veterinary School, Oued Smar, Algiers 16059, Algeria;
| | - Hosny El-Adawy
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, 07743 Jena, Germany; (H.H.); (H.T.); (H.N.)
- Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 35516, Egypt
- Correspondence:
| | - Helmut Hotzel
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, 07743 Jena, Germany; (H.H.); (H.T.); (H.N.)
| | - Ashraf Hendam
- Climate Change Information Center, Renewable Energy and Expert Systems (CCICREES), Agricultural Research Center, 9 Algamaa Street, Giza 12619, Egypt;
| | - Herbert Tomaso
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, 07743 Jena, Germany; (H.H.); (H.T.); (H.N.)
| | - Ralf Ehricht
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany; (R.E.); (S.M.)
- InfectoGnostics Research Campus Jena e. V., 07743 Jena, Germany
- Institute of Physical Chemistry, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Heinrich Neubauer
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, 07743 Jena, Germany; (H.H.); (H.T.); (H.N.)
| | - Ibrahim Nabi
- Faculty of Sciences, Yahia Farès University, Urban Pole, Médéa 26000, Algeria;
| | - Taha Mossadak Hamdi
- Laboratory of Food Hygiene and Quality Assurance System, High National Veterinary School, Oued Smar, Algiers 16059, Algeria;
| | - Stefan Monecke
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany; (R.E.); (S.M.)
- InfectoGnostics Research Campus Jena e. V., 07743 Jena, Germany
- Institute for Medical Microbiology and Virology, Dresden University Hospital, 01307 Dresden, Germany
| |
Collapse
|
37
|
Hu H, Liu H, Kweon O, Hart ME. A naturally occurring point mutation in the hyaluronidase gene ( hysA1) of Staphylococcus aureus UAMS-1 results in reduced enzymatic activity. Can J Microbiol 2021; 68:1-13. [PMID: 34520677 DOI: 10.1139/cjm-2021-0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyaluronic acid is a high-molecular-weight polysaccharide that is widely distributed in animal tissues. Bacterial hyaluronidases degrade hyaluronic acid as secreted enzymes and have been shown to contribute to infection. Staphylococcus aureus UAMS-1 is a clinical isolate that codes for two hyaluronidases (hysA1 and hysA2). Previous research has shown the presence of a full-length HysA1 protein from the S. aureus UAMS-1 strain with no evidence of enzymatic activity. In this study, the coding and upstream promoter regions of hysA1 from the S. aureus UAMS-1 strain were cloned, sequenced, and compared to the hysA1 gene from the S. aureus Sanger 252 strain. A single base change resulting in an E480G amino acid change was identified in the hysA1 gene from the S. aureus UAMS-1 strain when compared to the hysA1 gene from S. aureus Sanger 252. A plasmid copy of hysA1 from S. aureus Sanger 252 transduced into an S. aureus UAMS-1 hysA2 deletion mutant strain restored near wild-type levels of enzymatic activity. Homology modeling of the HysA1 hyaluronidase was performed with SWISS-MODEL using hyaluronidase from Streptococcus pneumoniae as the template, followed by a series of structural analyses using PyMOL, PLIP, PDBsum, and HOPE servers. This glutamic acid is highly conserved among hyaluronidases from Staphylococcus and other gram-positive bacteria. A series of structural analyses suggested that Glu-480 in HysA1 is critically responsible for maintaining the structural and functional ensemble of the catalytic and tunnel-forming residues, which are essential for enzyme activity. The missense mutation of Glu-480 to Gly introduces a loss of side chain hydrogen bond interactions with key residues Arg-360 and Arg-364, which are responsible for the tunnel topology, resulting in displacement of the substrate from an ideal position for catalysis through a localized conformational change of the active site. There is a high degree of relatedness among several gram-positive bacterial hyaluronidases; the loss of enzymatic activity of HysA1 in the S. aureus UAMS-1 strain is most likely caused by the mutation identified in our study. The role of hyaluronidase in staphylococcal infection and the redundancy of this gene are yet to be determined.
Collapse
Affiliation(s)
- Haijing Hu
- Office of Dietary Supplement Programs, Center for Food Safety and Nutrition, U.S. Food and Drug Administration, College Park, MD 20740, USA
| | - Huanli Liu
- Branch of Microbiology, Arkansas Laboratory, Office of Regulatory Affairs, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Ohgew Kweon
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Mark E Hart
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
38
|
Monteith AJ, Miller JM, Maxwell CN, Chazin WJ, Skaar EP. Neutrophil extracellular traps enhance macrophage killing of bacterial pathogens. SCIENCE ADVANCES 2021; 7:eabj2101. [PMID: 34516771 PMCID: PMC8442908 DOI: 10.1126/sciadv.abj2101] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/20/2021] [Indexed: 05/16/2023]
Abstract
Neutrophils and macrophages are critical to the innate immune response, but cooperative mechanisms used by these cells to combat extracellular pathogens are not well understood. This study reveals that S100A9-deficient neutrophils produce higher levels of mitochondrial superoxide in response to Staphylococcus aureus and, as a result, form neutrophil extracellular traps (suicidal NETosis). Increased suicidal NETosis does not improve neutrophil killing of S. aureus in isolation but augments macrophage killing. NET formation enhances antibacterial activity by increasing phagocytosis by macrophages and by transferring neutrophil-specific antimicrobial peptides to them. Similar results were observed in response to other phylogenetically distinct bacterial pathogens including Streptococcus pneumoniae and Pseudomonas aeruginosa, implicating this as an immune defense mechanism that broadly enhances antibacterial activity. These results demonstrate that achieving maximal bactericidal activity through NET formation requires macrophages and that accelerated and more robust suicidal NETosis makes neutrophils adept at increasing antibacterial activity, especially when A9 deficient.
Collapse
Affiliation(s)
- Andrew J. Monteith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeanette M. Miller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - C. Noel Maxwell
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Walter J. Chazin
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
39
|
Iwabuchi C, Hino H, Shimada K, Kaneko H, Nakaminami H. Recurrent furunculosis by Panton-Valentine leukocidin-positive methicillin-resistant Staphylococcus aureus USA300 clone in Tokyo: A report of two cases. J Dermatol 2021; 48:E522-E523. [PMID: 34296468 DOI: 10.1111/1346-8138.16076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/12/2021] [Accepted: 07/06/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Chikako Iwabuchi
- Department of Dermatology, Nissan Tamagawa Hospital, Tokyo, Japan
| | - Haruko Hino
- Department of Dermatology, Nissan Tamagawa Hospital, Tokyo, Japan.,Department of Dermatology, Akasaka Toranomon Clinic, Tokyo, Japan
| | - Kyoka Shimada
- Department of Dermatology, Nissan Tamagawa Hospital, Tokyo, Japan
| | - Hiroshi Kaneko
- Department of Clinical Microbiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hidemasa Nakaminami
- Department of Clinical Microbiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
40
|
Beavers WN, DuMont AL, Monteith AJ, Maloney KN, Tallman KA, Weiss A, Christian AH, Toste FD, Chang CJ, Porter NA, Torres VJ, Skaar EP. Staphylococcus aureus Peptide Methionine Sulfoxide Reductases Protect from Human Whole-Blood Killing. Infect Immun 2021; 89:e0014621. [PMID: 34001560 PMCID: PMC8281210 DOI: 10.1128/iai.00146-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/07/2021] [Indexed: 11/20/2022] Open
Abstract
The generation of oxidative stress is a host strategy used to control Staphylococcus aureus infections. Sulfur-containing amino acids, cysteine and methionine, are particularly susceptible to oxidation because of the inherent reactivity of sulfur. Due to the constant threat of protein oxidation, many systems evolved to protect S. aureus from protein oxidation or to repair protein oxidation after it occurs. The S. aureus peptide methionine sulfoxide reductase (Msr) system reduces methionine sulfoxide to methionine. Staphylococci have four Msr enzymes, which all perform this reaction. Deleting all four msr genes in USA300 LAC (Δmsr) sensitizes S. aureus to hypochlorous acid (HOCl) killing; however, the Δmsr strain does not exhibit increased sensitivity to H2O2 stress or superoxide anion stress generated by paraquat or pyocyanin. Consistent with increased susceptibility to HOCl killing, the Δmsr strain is slower to recover following coculture with both murine and human neutrophils than USA300 wild type. The Δmsr strain is attenuated for dissemination to the spleen following murine intraperitoneal infection and exhibits reduced bacterial burdens in a murine skin infection model. Notably, no differences in bacterial burdens were observed in any organ following murine intravenous infection. Consistent with these observations, USA300 wild-type and Δmsr strains have similar survival phenotypes when incubated with murine whole blood. However, the Δmsr strain is killed more efficiently by human whole blood. These findings indicate that species-specific immune cell composition of the blood may influence the importance of Msr enzymes during S. aureus infection of the human host.
Collapse
Affiliation(s)
- William N. Beavers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ashley L. DuMont
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Andrew J. Monteith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - K. Nichole Maloney
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Keri A. Tallman
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Andy Weiss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alec H. Christian
- Department of Chemistry, University of California, Berkeley, Berkeley, California, USA
| | - F. Dean Toste
- Department of Chemistry, University of California, Berkeley, Berkeley, California, USA
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Ned A. Porter
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
41
|
Ziesmer J, Tajpara P, Hempel N, Ehrström M, Melican K, Eidsmo L, Sotiriou GA. Vancomycin-Loaded Microneedle Arrays against Methicillin-Resistant Staphylococcus Aureus Skin Infections. ADVANCED MATERIALS TECHNOLOGIES 2021; 6:2001307. [PMID: 34307835 PMCID: PMC8281827 DOI: 10.1002/admt.202001307] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/12/2021] [Indexed: 05/24/2023]
Abstract
Skin and soft tissue infections (SSTIs) caused by methicillin-resistant Staphylococcus aureus (MRSA) are a major healthcare burden, often treated with intravenous injection of the glycopeptide antibiotic vancomycin (VAN). However, low local drug concentration in the skin limits its treatment efficiency, while systemic exposure promotes the development of resistant bacterial strains. Topical administration of VAN on skin is ineffective as its high molecular weight prohibits transdermal penetration. In order to implement a local VAN delivery, microneedle (MN) arrays with a water-insoluble support layer for the controlled administration of VAN into the skin are developed. The utilization of such a support layer results in water-insoluble needle shafts surrounded by drug-loaded water-soluble tips with high drug encapsulation. The developed MN arrays can penetrate the dermal barriers of both porcine and fresh human skin. Permeation studies on porcine skin reveal that the majority of the delivered VAN is retained within the skin. It is shown that the VAN-MN array reduces MRSA growth both in vitro and ex vivo on skin. The developed VAN-MN arrays may be extended to several drugs and may facilitate localized treatment of MRSA-caused skin infections while minimizing adverse systemic effects.
Collapse
Affiliation(s)
- Jill Ziesmer
- Department of MicrobiologyTumour and Cell BiologyKarolinska InstitutetStockholmSE‐17177Sweden
| | - Poojabahen Tajpara
- Department of Medicine SolnaUnit of RheumatologyKarolinska InstitutetStockholmSE‐17177Sweden
| | | | - Marcus Ehrström
- Department of Reconstructive Plastic SurgeryKarolinska University Hospital SolnaStockholmSE‐17176Sweden
| | - Keira Melican
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES)Karolinska Institutet and KTH Royal Institute of TechnologyStockholmSE‐171 77Sweden
- Department of NeuroscienceKarolinska InstitutetStockholmSE‐171 77Sweden
| | - Liv Eidsmo
- Department of Medicine SolnaUnit of RheumatologyKarolinska InstitutetStockholmSE‐17177Sweden
- Diagnostiskt Centrum HudStockholmSE‐11137Sweden
- Leo Foundation Skin Immunology CenterUniversity of CopenhagenCopenhagenDK‐2100Denmark
| | - Georgios A. Sotiriou
- Department of MicrobiologyTumour and Cell BiologyKarolinska InstitutetStockholmSE‐17177Sweden
| |
Collapse
|
42
|
Sun Z, Peng Y, Wang M, Lin Y, Jalalah M, Alsareii SA, Harraz FA, Yang J, Li G. Electrochemical Deposition of Cu Metal-Organic Framework Films for the Dual Analysis of Pathogens. Anal Chem 2021; 93:8994-9001. [PMID: 34151551 DOI: 10.1021/acs.analchem.1c01763] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Metal-organic framework (MOF) thin films with flexible nature and prominent qualities have opened doors to new technological applications in different fields. Herein, we propose an electrochemical biosensor for the dual detection of Staphylococcus aureus based on the electrodeposition of Cu metal-organic framework (Cu-MOF) thin films. The promising sensing layer with features of good electronic conductivity and enhanced electron-transfer property can not only identify S. aureus through specific micrococcal nucleases in the supernatant but also detect the pathogen directly via aptamer recognition. The dual analysis design ensures the accuracy of this method for S. aureus detection in the range of 7-7 × 106 cfu/mL with the limits of detection of 1.9 and 5.2 cfu/mL. Moreover, the analytical method validation confirmed that the biosensor could efficiently work in complex biological samples, showing good selectivity and specificity and great potential for clinical diagnosis. More importantly, the current proposed strategy is simple and easy to implement without the need for extra signaling elements, which is convenient for timely clinical detection.
Collapse
Affiliation(s)
- Zhaowei Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Ying Peng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Minghui Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yuxin Lin
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Mohammed Jalalah
- Promising Centre for Sensors and Electronic Devices (PCSED), Advanced Materials and Nano-Research Centre, Najran University, P.O. Box 1988, Najran 11001, Saudi Arabia.,Department of Electrical Engineering, Faculty of Engineering, Najran University, Najran 11001, Saudi Arabia
| | - Saeed A Alsareii
- Department of Surgery, College of Medicine, Najran University, Najran 11001, Saudi Arabia
| | - Farid A Harraz
- Promising Centre for Sensors and Electronic Devices (PCSED), Advanced Materials and Nano-Research Centre, Najran University, P.O. Box 1988, Najran 11001, Saudi Arabia.,Nanomaterials and Nanotechnology Department, Central Metallurgical Research and Development Institute (CMRDI), P.O. 87 Helwan, Cairo 11421, Egypt
| | - Jie Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Genxi Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.,Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
43
|
Popovich KJ, Green SJ, Okamoto K, Rhee Y, Hayden MK, Schoeny M, Snitkin ES, Weinstein RA. MRSA Transmission in Intensive Care Units: Genomic Analysis of Patients, Their Environments, and Healthcare Workers. Clin Infect Dis 2021; 72:1879-1887. [PMID: 32505135 PMCID: PMC8315036 DOI: 10.1093/cid/ciaa731] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/03/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Methicillin-resistant Staphylococcus aureus (MRSA)-and now USA300 MRSA-is a significant intensive care unit (ICU) pathogen; healthcare worker (HCW) contamination may lead to patient cross-transmission. METHODS From September 2015 to February 2016, to study the spread of MRSA, we enrolled HCWs in 4 adult ICUs caring for patients on MRSA contact precautions. Samples were collected from patient body sites and high-touch surfaces in patient rooms. HCW hands, gloves, and personal protective equipment were sampled pre/post-patient encounter. Whole genome sequencing (WGS) was used to compare isolates from patients, HCWs, and environment. RESULTS There were 413 MRSA isolates sequenced (38% USA300, 52% USA100) from 66 patient encounters. Six of 66 HCWs were contaminated with MRSA prior to room entry. Isolates from a single patient encounter were typically either USA100 or USA300; in 8 (12%) encounters both USA300 and USA100 were isolated. WGS demonstrated that isolates from patients, HCWs, and environment often were genetically similar, although there was substantial between-encounter diversity. Strikingly, there were 5 USA100 and 1 USA300 clusters that contained similar strains (<22 single-nucleotide variants [SNVs], with most <10 SNVs) within the cluster despite coming from different encounters, suggesting intra- and inter-ICU spread of strains, that is, 4 of these genomic clusters were from encounters in the same ICU; 5 of 6 clusters occurred within 1 week. CONCLUSIONS We demonstrated frequent spread of MRSA USA300 and USA100 strains among patients, environment, and HCWs. WGS identified possible spread within and even between ICUs. Future analysis with detailed contact tracing in conjunction with genomic data may further elucidate pathways of MRSA spread and points for intervention.
Collapse
Affiliation(s)
- Kyle J Popovich
- Division of Infectious Disease, Rush University Medical Center/Cook County Health, Chicago, Illinois, USA
| | - Stefan J Green
- Genome Research Core, Research Resources Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Koh Okamoto
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Yoona Rhee
- Division of Infectious Disease, Rush University Medical Center/Cook County Health, Chicago, Illinois, USA
| | - Mary K Hayden
- Division of Infectious Disease, Rush University Medical Center, Chicago, Illinois, USA
| | - Michael Schoeny
- Community, Systems, and Mental Health Nursing Rush University Medical Center, Chicago, Illinois, USA
| | - Evan S Snitkin
- Department of Microbiology and Immunology and Division of Infectious Disease, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert A Weinstein
- Division of Infectious Disease, Rush University Medical Center/Cook County Health, Chicago, Illinois, USA
| |
Collapse
|
44
|
Huang SY, Wang X, Shen DY, Chen F, Zhang GY, Zhang Z, Li K, Jin Z, Du D, Tang YZ. Design, synthesis and biological evaluation of novel pleuromutilin derivatives as potent anti-MRSA agents targeting the 50S ribosome. Bioorg Med Chem 2021; 38:116138. [PMID: 33857737 DOI: 10.1016/j.bmc.2021.116138] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/13/2021] [Accepted: 03/23/2021] [Indexed: 10/21/2022]
Abstract
A series of novel pleuromutilin derivatives were designed and synthesized with 1,2,4-triazole as the linker connected to benzoyl chloride analogues under mild conditions. The in vitro antibacterial activities of the synthesized derivatives against four strains of Staphylococcus aureus (MRSA ATCC 43300, ATCC 29213, AD3 and 144) were tested by the broth dilution method. Most of the synthesized derivatives displayed potent activities, and 22-(3-amino-2-(4-methyl-benzoyl)-1,2,4-triazole-5-yl)-thioacetyl)-22-deoxypleuromutilin (compound 12) was found to be the most active antibacterial derivative against MRSA (MIC = 0.125 μg/mL). Furthermore, the time-kill curves showed compound 12 had a certain inhibitory effect against MRSA in vitro. The in vivo antibacterial activity of compound 12 was further evaluated using MRSA infected murine thigh model. Compound 12 exhibited superior antibacterial efficacy than tiamulin. It was also found that compound 12 had no significant inhibitory effect on the proliferation of RAW264.7 cells. Compound 12 was further evaluated in CYP450 inhibition assay and showed moderate inhibitory effect on CYP3A4 (IC50 = 3.95 μM). Moreover, seven candidate compounds showed different affinities with the 50S ribosome by SPR measurement. Subsequently, binding of compound 12 and 20 to the 50S ribosome was further investigated by molecular modeling. Three strong hydrogen bonds were formed through the interaction of compound 12 and 20 with 50S ribosome. The binding free energy of compound 12 and 20 with the ribosome was calculated to be -10.7 kcal/mol and -11.66 kcal/mol, respectively.
Collapse
Affiliation(s)
- Si-Yu Huang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiao Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ding-Yi Shen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Fang Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Guang-Yu Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhe Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Kang Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhen Jin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Dan Du
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - You-Zhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China.
| |
Collapse
|
45
|
Popovich KJ, Snitkin ES, Zawitz C, Aroutcheva A, Payne D, Thiede SN, Schoeny M, Green SJ, Hayden MK, Hota B, Weinstein RA. Frequent Methicillin-Resistant Staphylococcus aureus Introductions Into an Inner-city Jail: Indications of Community Transmission Networks. Clin Infect Dis 2021; 71:323-331. [PMID: 31425575 DOI: 10.1093/cid/ciz818] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Jails may facilitate spread of methicillin-resistant Staphylococcus aureus (MRSA) in urban areas. We examined MRSA colonization upon entrance to a large urban jail to determine if there are MRSA transmission networks preceding incarceration. METHODS Males incarcerated in Cook County Jail (Chicago) were enrolled, with enrichment for people living with human immunodeficiency virus (PLHIV), within 72 hours of intake. Surveillance cultures assessed prevalence of MRSA colonization. Whole-genome sequencing (WGS) identified preincarceration transmission networks.We examined methicillin-resistant Staphylococcus aureus (MRSA) isolates to determine if there are transmission networks that precede incarceration. A large proportion of individuals enter jail colonized with MRSA. Molecular epidemiology and colonization risk factors provide clues to community reservoirs for MRSA. RESULTS There were 718 individuals (800 incarcerations) enrolled; 58% were PLHIV. The prevalence of MRSA colonization at intake was 19%. In multivariate analysis, methamphetamine use, unstable housing, current/recent skin infection, and recent injection drug use were predictors of MRSA. Among PLHIV, recent injection drug use, current skin infection, and HIV care at outpatient clinic A that emphasizes comprehensive care to the lesbian, gay, bisexual, transgender community were predictors of MRSA. Fourteen (45%) of 31 detainees with care at clinic A had colonization. WGS revealed that this prevalence was not due to clonal spread in clinic but rather to an intermingling of distinct community transmission networks. In contrast, genomic analysis supported spread of USA500 strains within a network. Members of this USA500 network were more likely to be PLHIV (P < .01), men who have sex with men (P < .001), and methamphetamine users (P < .001). CONCLUSIONS A large proportion of individuals enter jail colonized with MRSA. Molecular epidemiology and colonization risk factors provide clues to identify colonized detainees entering jail and potential community reservoirs of MRSA.
Collapse
Affiliation(s)
- Kyle J Popovich
- Division of Infectious Diseases, Rush University Medical Center/Cook County Health, Chicago, Illinois, USA
| | - Evan S Snitkin
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Chicago, Illinois, USA
| | - Chad Zawitz
- Clinical Coordinator of HIV and Infectious Disease Services, Cermak Health Services, Cook County Health, Chicago, Illinois, USA
| | - Alla Aroutcheva
- Division of Infectious Diseases, Rush University Medical Center/Cook County Health, Chicago, Illinois, USA
| | - Darjai Payne
- Division of Infectious Diseases, Chicago, Illinois, USA
| | - Stephanie N Thiede
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Chicago, Illinois, USA
| | - Michael Schoeny
- Community, Systems, and Mental Health Nursing Rush University Medical Center, Chicago, Illinois, USA
| | - Stefan J Green
- Sequencing Core and Associate Director, Research Resources Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mary K Hayden
- Division of Infectious Diseases, Chicago, Illinois, USA
| | - Bala Hota
- Division of Infectious Diseases, Chicago, Illinois, USA
| | - Robert A Weinstein
- Division of Infectious Diseases, Rush University Medical Center/Cook County Health, Chicago, Illinois, USA
| |
Collapse
|
46
|
Popovich KJ. Using Genomic Sequencing to Delineate Community Networks of Methicillin-Resistant Staphylococcus aureus Transmission Among Men Who Have Sex With Men. J Infect Dis 2021; 223:543-546. [PMID: 33057661 PMCID: PMC7904285 DOI: 10.1093/infdis/jiaa652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Kyle J Popovich
- Rush University Medical Center, Chicago, Illinois, USA
- Cook County Health, Chicago, Illinois, USA
| |
Collapse
|
47
|
Sommers KJ, Bentley BS, Carden RG, Post SJ, Allen RA, Kontos RC, Black JW, Wuest WM, Minbiole KPC. Metallocene QACs: The Incorporation of Ferrocene Moieties into monoQAC and bisQAC Structures. ChemMedChem 2020; 16:467-471. [PMID: 33197298 DOI: 10.1002/cmdc.202000605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/12/2020] [Indexed: 01/12/2023]
Abstract
Inspired by the incorporation of metallocene functionalities into a variety of bioactive structures, particularly antimicrobial peptides, we endeavored to broaden the structural variety of quaternary ammonium compounds (QACs) by the incorporation of the ferrocene moiety. Accordingly, 23 ferrocene-containing mono- and bisQACs were prepared in high yields and tested for activity against a variety of bacteria, including Gram-negative strains and a panel of clinically isolated MRSA strains. Ferrocene QACs were shown to be effective antiseptics with some displaying single-digit micromolar activity against all bacteria tested, demonstrating yet another step in the expansion of structural variety of antiseptic QACs.
Collapse
Affiliation(s)
- Kyle J Sommers
- Department of Chemistry, Villanova University, Villanova, PA 19085, USA
| | - Brian S Bentley
- Department of Chemistry, Villanova University, Villanova, PA 19085, USA
| | - Robert G Carden
- Department of Chemistry, Villanova University, Villanova, PA 19085, USA
| | - Savannah J Post
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Ryan A Allen
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Renee C Kontos
- Department of Chemistry, Villanova University, Villanova, PA 19085, USA
| | - Jacob W Black
- Department of Chemistry, Villanova University, Villanova, PA 19085, USA
| | - William M Wuest
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | | |
Collapse
|
48
|
Abstract
Staphylococcus aureus is a leading agent of antibiotic-resistant bacterial infections in the world. S. aureus tightly controls metal homeostasis during infection, and disruption of metal uptake systems impairs staphylococcal virulence. We identified small molecules that interfere with metal handling in S. aureus to develop chemical probes to investigate metallobiology in this organism. Compound VU0026921 was identified as a small molecule that kills S. aureus both aerobically and anaerobically. The activity of VU0026921 is modulated by metal supplementation, is enhanced by genetic inactivation of Mn homeostasis genes, and correlates with increased cellular reactive oxygen species. Treatment with VU0026921 causes accumulation of multiple metals within S. aureus cells and concomitant upregulation of genes involved in metal detoxification. This work defines a small-molecule probe for further defining the role of metal toxicity in S. aureus and validates future antibiotic development targeting metal toxicity pathways. Metals are essential nutrients that all living organisms acquire from their environment. While metals are necessary for life, excess metal uptake can be toxic; therefore, intracellular metal levels are tightly regulated in bacterial cells. Staphylococcus aureus, a Gram-positive bacterium, relies on metal uptake and metabolism to colonize vertebrates. Thus, we hypothesized that an expanded understanding of metal homeostasis in S. aureus will lead to the discovery of pathways that can be targeted with future antimicrobials. We sought to identify small molecules that inhibit S. aureus growth in a metal-dependent manner as a strategy to uncover pathways that maintain metal homeostasis. Here, we demonstrate that VU0026921 kills S. aureus through disruption of metal homeostasis. VU0026921 activity was characterized through cell culture assays, transcriptional sequencing, compound structure-activity relationship, reactive oxygen species (ROS) generation assays, metal binding assays, and metal level analyses. VU0026921 disrupts metal homeostasis in S. aureus, increasing intracellular accumulation of metals and leading to toxicity through mismetalation of enzymes, generation of reactive oxygen species, or disruption of other cellular processes. Antioxidants partially protect S. aureus from VU0026921 killing, emphasizing the role of reactive oxygen species in the mechanism of killing, but VU0026921 also kills S. aureus anaerobically, indicating that the observed toxicity is not solely oxygen dependent. VU0026921 disrupts metal homeostasis in multiple Gram-positive bacteria, leading to increased reactive oxygen species and cell death, demonstrating the broad applicability of these findings. Further, this study validates VU0026921 as a probe to further decipher mechanisms required to maintain metal homeostasis in Gram-positive bacteria.
Collapse
|
49
|
Al-Tameemi H, Beavers WN, Norambuena J, Skaar EP, Boyd JM. Staphylococcus aureus lacking a functional MntABC manganese import system has increased resistance to copper. Mol Microbiol 2020; 115:554-573. [PMID: 33034093 DOI: 10.1111/mmi.14623] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 12/17/2022]
Abstract
S. aureus USA300 isolates utilize the copBL and copAZ gene products to prevent Cu intoxication. We created and examined a ΔcopAZ ΔcopBL mutant strain (cop-). The cop- strain was sensitive to Cu and accumulated intracellular Cu. We screened a transposon (Tn) mutant library in the cop- background and isolated strains with Tn insertions in the mntABC operon that permitted growth in the presence of Cu. The mutations were in mntA and they were recessive. Under the growth conditions utilized, MntABC functioned in manganese (Mn) import. When cultured with Cu, strains containing a mntA::Tn accumulated less Cu than the parent strain. Mn(II) supplementation improved growth when cop- was cultured with Cu and this phenotype was dependent upon the presence of MntR, which is a repressor of mntABC transcription. A ΔmntR strain had an increased Cu load and decreased growth in the presence of Cu, which was abrogated by the introduction of mntA::Tn. Over-expression of mntABC increased cellular Cu load and sensitivity to Cu. The presence of a mntA::Tn mutation protected iron-sulfur (FeS) enzymes from inactivation by Cu. The data presented are consistent with a model wherein defective MntABC results in decreased cellular Cu accumulation and protection to FeS enzymes from Cu poisoning.
Collapse
Affiliation(s)
- Hassan Al-Tameemi
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, New Brunswick, NJ, USA
| | - William N Beavers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Javiera Norambuena
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, New Brunswick, NJ, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey M Boyd
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
50
|
Ikeuchi K, Adachi E, Sasaki T, Suzuki M, Lim LA, Saito M, Koga M, Tsutsumi T, Kido Y, Uehara Y, Yotsuyanagi H. An Outbreak of USA300 Methicillin-Resistant Staphylococcus aureus Among People With HIV in Japan. J Infect Dis 2020; 223:610-620. [PMID: 33057717 DOI: 10.1093/infdis/jiaa651] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/09/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND USA300 produces Panton-Valentin leucocidin (PVL) and is known as a predominant community-associated methicillin-resistant Staphylococcus aureus (MRSA) strain in the United States, but it was extremely rare in Japan. We report here an outbreak of USA300 in people with HIV (PWH) in Tokyo, Japan. METHODS We analyzed the cases of PVL-MRSA infection between 2010 and 2020 and screened for nasal colonization of PVL-MRSA in PWH who visited an HIV/AIDS referral hospital from December 2019 to March 2020. Whole-genome sequencing-based single nucleotide polymorphism (SNP) analysis was performed on these isolates. RESULTS During the study period, a total of 21 PVL-MRSA infections in 14 patients were identified after 2014. The carriage prevalence was 4.3% (12/277) and PVL-MRSA carriers were more likely to have sexually transmitted infections (STIs) within a year compared with patients who had neither a history of PVL-MRSA infection nor colonization (33.3% [4/12] vs 10.1% [26/258]; P = .03). SNP analysis showed that all 26 isolates were ST8-SCCmecIVa-USA300. Twenty-four isolates were closely related (≤100 SNP differences) and had the nonsynonymous SNPs associated with carbohydrate metabolism and antimicrobial tolerance. CONCLUSIONS An outbreak of USA300 has been occurring among PWH in Tokyo and a history of STI was a risk of colonization.
Collapse
Affiliation(s)
- Kazuhiko Ikeuchi
- IMSUT Hospital, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Eisuke Adachi
- IMSUT Hospital, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Takashi Sasaki
- Animal Research Center, Sapporo Medical University School of Medicine, Sapporo City, Hokkaido, Japan
| | - Masato Suzuki
- IMSUT Hospital, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Lay Ahyoung Lim
- IMSUT Hospital, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Makoto Saito
- IMSUT Hospital, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Michiko Koga
- IMSUT Hospital, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Takeya Tsutsumi
- IMSUT Hospital, The University of Tokyo, Minato-ku, Tokyo, Japan
| | | | - Yuki Uehara
- St Luke's International Hospital, Chuo-ku, Tokyo, Japan
| | | |
Collapse
|