1
|
Pettygrove BA, Nygaard TK, Borgogna TR, Malachowa N, Gaur G, Salo SE, Pallister KB, Burroughs O, Robinson C, Gao A, Sturdevant DE, Ricklefs S, DeLeo FR, Otto M, Stewart PS, Voyich JM. Staphylococcus aureus SaeR/S-regulated factors overcome human complement-mediated inhibition of aggregation to evade neutrophil killing. Proc Natl Acad Sci U S A 2025; 122:e2412447122. [PMID: 40359050 DOI: 10.1073/pnas.2412447122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 03/25/2025] [Indexed: 05/15/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a frequent culprit in implant-associated infections and employs many virulence factors to escape killing by the host immune system. The specific immune evasion strategies used by small aggregates of S. aureus on a surface, precursors to mature biofilm, are still relatively unknown. Time-lapse confocal microscopy was leveraged to quantify interactions between S. aureus aggregates and human neutrophils in vitro and identify specific mechanisms of resistance to neutrophil killing. Surface-associated wild-type S. aureus rapidly formed small biofilm aggregates when grown in human serum. Conversely, aggregation was inhibited when the SaeR/S two-component gene regulatory system was deleted. Wild-type aggregates began to show individual and population-level resistance to neutrophil killing upon reaching sizes of approximately 50 to 75 µm2, whereas Δsae clusters failed to reach these sizes and were readily cleared. Aggregation of Δsae strains was impaired by serum complement, and this inhibition required complement proteins C3 and factor B, but not C4 or C5, suggesting that this activity primarily occurs at the level of the alternative pathway. Several complement-inhibiting genes regulated by SaeR/S were identified that collectively facilitate biofilm aggregate formation in human, but not murine serum. Finally, aggregation of two related opportunistic pathogens, Staphylococcus epidermidis and Enterococcus faecalis, was inhibited by serum. These data demonstrate a function of serum complement, the ability to inhibit bacterial aggregation, that is potently blocked by S. aureus through the production of multiple complement-interfering proteins that are regulated by the SaeR/S system.
Collapse
Affiliation(s)
- Brian A Pettygrove
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infection Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Tyler K Nygaard
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Timothy R Borgogna
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Natalia Malachowa
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Gauri Gaur
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Shannon E Salo
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infection Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Kyler B Pallister
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Owen Burroughs
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Cassandra Robinson
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Annika Gao
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| | - Daniel E Sturdevant
- Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Stacy Ricklefs
- Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infection Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Philip S Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT 59717
| | - Jovanka M Voyich
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59717
| |
Collapse
|
2
|
Predtechenskaya M, Arbizzani CJ, Shomento SR, Borgogna TR, Voyich JM. Lung surfactant reduces Staphylococcus aureus cytotoxicity and protects host immune cells from membrane damage. Microbiol Spectr 2025:e0138624. [PMID: 40237467 DOI: 10.1128/spectrum.01386-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/20/2025] [Indexed: 04/18/2025] Open
Abstract
In this study, we identify that lung surfactant significantly reduces the cytotoxicity of Staphylococcus aureus (S. aureus) membrane-damaging toxins. Data demonstrate that natural surfactants from mice and rats and commercially available surfactant, Infasurf, protect human primary cells (neutrophils and peripheral blood mononuclear cells) from cytolytic activity caused by S. aureus supernatants. Supernatants from S. aureus grown in surfactant showed a significant reduction in plasma membrane damage against primary human cells as compared to supernatants grown without surfactant. This reduction was not due to a direct bactericidal effect of the surfactants on S. aureus growth. Rat and mouse surfactants downregulated the gene expression of saeR, the response regulator of the S. aureus two-component system SaeR/S that is responsible for the production of virulence factors which are important during lung infection and cause membrane damage in host cells. Rat and lung surfactants also reduced transcript abundance of SaeR/S-regulated genes lukF-PV, hla, and hlgA. Interestingly, the commercially available surfactant Infasurf did not recapitulate the effect of natural surfactants and did not decrease gene transcription of the virulence genes tested. These data suggest that components of natural surfactants protect lungs from S. aureus by suppressing S. aureus virulence factors and have implications for the role of surfactants in host defense against S. aureus.IMPORTANCEThis study explored the influence of lung surfactants on membrane-damaging Staphylococcus aureus (S. aureus) toxins. We demonstrate that natural and commercially available lung surfactants minimize the cytolytic capacity of S. aureus supernatants against primary human cells. Data indicate that cytolytic reduction by mouse and rat surfactants was partially due to surfactants reducing transcript abundance of virulence factors. This work identifies a novel role for surfactants and suggests their importance in modulating the severity of S. aureus lung infections.
Collapse
Affiliation(s)
- Maria Predtechenskaya
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Corbin J Arbizzani
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Sofia R Shomento
- University of Washington, School of Medicine, Seattle, Washington, USA
| | - Timothy R Borgogna
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Jovanka M Voyich
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
3
|
Piechowicz L, Kosznik-Kwaśnicka K, Kaźmierczak N, Grzenkowicz M, Stasiłojć M, Necel A, Werbowy O, Pałubicka A. Efficacy of Three Kayviruses Against Staphylococcus aureus Strains Isolated from COVID-19 Patients. Antibiotics (Basel) 2025; 14:257. [PMID: 40149068 PMCID: PMC11939781 DOI: 10.3390/antibiotics14030257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/21/2025] [Accepted: 03/01/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: The viral pandemic caused by the SARS-CoV-2 virus has affected millions of people. However, it was noticed that high mortality was often a result of bacterial co-infections. One of the main pathogens responsible for secondary infections in patients with viral respiratory tract infections, including COVID-19, is Staphylococcus aureus. In recent years, the number of infections caused by drug-resistant strains of S. aureus has been growing rapidly, often exceeding the number of infections caused by antibiotic-sensitive strains. In addition, biofilm-related infections are more difficult to treat due to the lower sensitivity of biofilm structure to antibiotics. Bacteriophages are seen as alternative treatment of bacterial infections. Therefore, in our work, we have analyzed the efficacy of three Kayviruses against S. aureus strains isolated from COVID-19 patients. Methods: We analyzed the ability of tested phages to remove S. aureus biofilm both from polystyrene plates as well as from the surface of pulmonary epithelial cells. Results: We have observed that tested Kayviruses had a broad host range. Furthermore, phages were able to effectively reduce biofilm biomass and number of viable cells in pure culture. During our research, none of the tested phages was shown to have a negative effect on cell viability and were able to inhibit the negative effect S. aureus had on cell condition. Conclusions: Our results show tested phages were effective in reducing the biofilm of S. aureus strains isolated from COVID-19 patients, had no adverse effect on lung epithelial cell viability. Therefore, it should be recognized that the properties of three studied Kayviruses give them an advantage in the selection of phages for treatment of staphylococcal infections.
Collapse
Affiliation(s)
- Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
| | - Katarzyna Kosznik-Kwaśnicka
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
| | - Natalia Kaźmierczak
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
| | - Milena Grzenkowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
- Department of Microbiology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdansk, Poland;
| | - Małgorzata Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, Dębinki 1, 80-211 Gdansk, Poland;
| | - Agnieszka Necel
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
| | - Olesia Werbowy
- Department of Microbiology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdansk, Poland;
| | - Anna Pałubicka
- Specialist Hospital in Kościerzyna Sp. z o.o., Department of Laboratory and Microbiological Diagnostics, Kościerzyna, Alojzego Piechowskiego 36, 83-400 Koscierzyna, Poland;
| |
Collapse
|
4
|
Piechowicz L, Kosznik-Kwaśnicka K, Jarzembowski T, Daca A, Necel A, Bonawenturczak A, Werbowy O, Stasiłojć M, Pałubicka A. Staphylococcus aureus Co-Infection in COVID-19 Patients: Virulence Genes and Their Influence on Respiratory Epithelial Cells in Light of Risk of Severe Secondary Infection. Int J Mol Sci 2024; 25:10050. [PMID: 39337536 PMCID: PMC11431965 DOI: 10.3390/ijms251810050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Pandemics from viral respiratory tract infections in the 20th and early 21st centuries were associated with high mortality, which was not always caused by a primary viral infection. It has been observed that severe course of infection, complications and mortality were often the result of co-infection with other pathogens, especially Staphylococcus aureus. During the COVID-19 pandemic, it was also noticed that patients infected with S. aureus had a significantly higher mortality rate (61.7%) compared to patients infected with SARS-CoV-2 alone. Our previous studies have shown that S. aureus strains isolated from patients with COVID-19 had a different protein profile than the strains in non-COVID-19 patients. Therefore, this study aims to analyze S. aureus strains isolated from COVID-19 patients in terms of their pathogenicity by analyzing their virulence genes, adhesion, cytotoxicity and penetration to the human pulmonary epithelial cell line A549. We have observed that half of the tested S. aureus strains isolated from patients with COVID-19 had a necrotizing effect on the A549 cells. The strains also showed greater variability in terms of their adhesion to the human cells than their non-COVID-19 counterparts.
Collapse
Affiliation(s)
- Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Katarzyna Kosznik-Kwaśnicka
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Tomasz Jarzembowski
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Agnieszka Daca
- Department of Physiopathology, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Agnieszka Necel
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Ada Bonawenturczak
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Olesia Werbowy
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Małgorzata Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland
| | - Anna Pałubicka
- Specialist Hospital in Koscierzyna Sp. z o.o., Department of Laboratory and Microbiological Diagnostics, Koscierzyna, Alojzego Piechowskiego 36, 83-400 Koscierzyna, Poland
| |
Collapse
|
5
|
Gao P, Wei Y, Hou S, Lai PM, Liu H, Tai SSC, Tang VYM, Prakash PH, Sze KH, Chen JHK, Sun H, Li X, Kao RYT. SaeR as a novel target for antivirulence therapy against Staphylococcus aureus. Emerg Microbes Infect 2023; 12:2254415. [PMID: 37671453 PMCID: PMC10494732 DOI: 10.1080/22221751.2023.2254415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/07/2023]
Abstract
Staphylococcus aureus is a major human pathogen responsible for a wide range of clinical infections. SaeRS is one of the two-component systems in S. aureus that modulate multiple virulence factors. Although SaeR is required for S. aureus to develop an infection, inhibitors have not been reported. Using an in vivo knockdown method, we demonstrated that SaeR is targetable for the discovery of antivirulence agent. HR3744 was discovered through a high-throughput screening utilizing a GFP-Lux dual reporter system driven by saeP1 promoter. The antivirulence efficacy of HR3744 was tested using Western blot, Quantitative Polymerase Chain Reaction, leucotoxicity, and haemolysis tests. In electrophoresis mobility shift assay, HR3744 inhibited SaeR-DNA probe binding. WaterLOGSY-NMR test showed HR3744 directly interacted with SaeR's DNA-binding domain. When SaeR was deleted, HR3744 lost its antivirulence property, validating the target specificity. Virtual docking and mutagenesis were used to confirm the target's specificity. When Glu159 was changed to Asn, the bacteria developed resistance to HR3744. A structure-activity relationship study revealed that a molecule with a slight modification did not inhibit SaeR, indicating the selectivity of HR3744. Interestingly, we found that SAV13, an analogue of HR3744, was four times more potent than HR3744 and demonstrated identical antivirulence properties and target specificity. In a mouse bacteraemia model, both HR3744 and SAV13 exhibited in vivo effectiveness. Collectively, we identified the first SaeR inhibitor, which exhibited in vitro and in vivo antivirulence properties, and proved that SaeR could be a novel target for developing antivirulence drugs against S. aureus infections.
Collapse
Affiliation(s)
- Peng Gao
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yuanxin Wei
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Suying Hou
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Pok-Man Lai
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Han Liu
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Sherlock Shing Chiu Tai
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Victor Yat Man Tang
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Pradeep Halebeedu Prakash
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kong-Hung Sze
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jonathan Hon Kwan Chen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Hongzhe Sun
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Xuechen Li
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Hong Kong, People’s Republic of China
| | - Richard Yi-Tsun Kao
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
6
|
Wei J, Zhang C, Ma W, Ma J, Liu Z, Ren F, Li N. Antibacterial Activity of Thesium chinense Turcz Extract Against Bacteria Associated with Upper Respiratory Tract Infections. Infect Drug Resist 2023; 16:5091-5105. [PMID: 37576521 PMCID: PMC10422991 DOI: 10.2147/idr.s425398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023] Open
Abstract
Purpose The drug resistance of Staphylococcus aureus, Streptococcus pneumoniae, Streptococcus pyogenes and Haemophilus influenzae has become more and more serious, and it is urgent to seek new antibacterial drugs. In this study, Thesium chinense Turcz. extracts were tested for its potential antibacterial activities. Methods T. chinense powder was extracted with 5 solvents of different polarity (ethyl alcohol, petroleum ether, ethyl acetate, n-butyl alcohol and double distilled water), and their antibacterial activities were tested. The Broth dilution method was used to evaluate the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of highly active plant extracts with a concentration of 1g/mL. The inhibitory activity of this extract on biofilm formation was investigated. Afterwards, we investigated its effect on the transcriptome of S. aureus. Results The ethanol extract coded as BRY, only inhibited S. aureus, whereas the ethyl acetate extract coded as BY2 showed inhibitory effect on all the tested bacteria. The MIC of BRY on S. aureus was 128 mg/mL, and the MBC was 512 mg/mL. The MIC of BY2 against S. aureus, S. pneumoniae, S. pyogenes and H. influenzae were 8 mg/mL, 4 mg/mL, 4 mg/mL, and 4 mg/mL, respectively. The MBC of BY2 for these four bacteria ranged from 4 to 256 mg/mL. Mechanism studies have shown that BRY and BY2 have an impact on anti-formation of biofilms at MIC concentrations. Transcriptome sequencing results showed that 531 genes were up-regulated and 340 genes showed down-regulated expression in S. aureus after BY2 treatment. Conclusion BY2 has a broader antibacterial spectrum than BRY. Meanwhile, the inhibitory effect of BY2 on S. aureus is better than BRY. The mechanism of BY2 against S. aureus may relate to its inhibition of ribosome synthesis, restriction of key enzymes of citric acid cycle, decrease of pathogenicity and influence on biofilm formation. The results confirmed that BY2 was the main antibacterial part of T. chinense, which can be used as a source of antibacterial agents.
Collapse
Affiliation(s)
- Juanru Wei
- School of Pharmacy, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Cong Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Wei Ma
- School of Pharmacy, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Juncheng Ma
- School of Pharmacy, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Zhenzhen Liu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Fucai Ren
- School of Pharmacy, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Ning Li
- School of Pharmacy, Anhui Medical University, Hefei, 230032, People’s Republic of China
| |
Collapse
|
7
|
Garvin KL, Kildow BJ, Hewlett AL, Hartman CW, Fey PD. The Challenge of Emerging Resistant Gram-Positive Pathogens in Hip and Knee Periprosthetic Joint Infections. J Bone Joint Surg Am 2023; 105:878-890. [PMID: 37053296 DOI: 10.2106/jbjs.22.00792] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
➤ An increase in resistant bacterial pathogens has occurred over the last 4 decades.➤ Careful patient selection and improving or correcting risk factors for periprosthetic joint infection (PJI) before elective surgical treatment are strongly recommended. ➤ Appropriate microbiological methods, including those used to detect and grow Cutibacterium acnes, are recommended. ➤ Antimicrobial agents used in the prevention or management of infection should be selected appropriately and the duration of therapy should be carefully considered in order to mitigate the risk of developing bacterial resistance.➤ Molecular methods including rapid polymerase chain reaction (PCR) diagnostics, 16S sequencing, and/or shotgun and/or targeted whole-genome sequencing are recommended in culture-negative cases of PJI.➤ Expert consultation with an infectious diseases specialist (if available) is recommended to assist with the appropriate antimicrobial management and monitoring of patients with PJI.
Collapse
Affiliation(s)
- Kevin L Garvin
- Department of Orthopaedic Surgery and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska
| | - Beau J Kildow
- Department of Orthopaedic Surgery and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska
| | - Angela L Hewlett
- Division of Infectious Diseases, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Curtis W Hartman
- Department of Orthopaedic Surgery and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska
| | - Paul D Fey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
8
|
Goncheva MI, Gibson RM, Shouldice AC, Dikeakos JD, Heinrichs DE. The Staphylococcus aureus protein IsdA increases SARS CoV-2 replication by modulating JAK-STAT signaling. iScience 2023; 26:105975. [PMID: 36687318 PMCID: PMC9838083 DOI: 10.1016/j.isci.2023.105975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/28/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (CoV-2) pandemic has affected millions globally. A significant complication of CoV-2 infection is secondary bacterial co-infection, as seen in approximately 25% of severe cases. The most common organism isolated during co-infection is Staphylococcus aureus. Here, we describe the development of an in vitro co-infection model where both viral and bacterial replication kinetics may be examined. We demonstrate CoV-2 infection does not alter bacterial interactions with host epithelial cells. In contrast, S. aureus enhances CoV-2 replication by 10- to 15-fold. We identify this pro-viral activity is due to the S. aureus iron-regulated surface determinant A (IsdA) protein and demonstrate IsdA modifies host transcription. We find that IsdA alters Janus Kinase - Signal Transducer and Activator of Transcription (JAK-STAT) signaling, by affecting JAK2-STAT3 levels, ultimately leading to increased viral replication. These findings provide key insight into the molecular interactions between host cells, CoV-2 and S. aureus during co-infection.
Collapse
Affiliation(s)
- Mariya I. Goncheva
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada,Corresponding author
| | - Richard M. Gibson
- ImPaKT Laboratory, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Ainslie C. Shouldice
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - David E. Heinrichs
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada,Corresponding author
| |
Collapse
|
9
|
Lane S, Hilliam Y, Bomberger JM. Microbial and Immune Regulation of the Gut-Lung Axis during Viral-Bacterial Coinfection. J Bacteriol 2023; 205:e0029522. [PMID: 36409130 PMCID: PMC9879096 DOI: 10.1128/jb.00295-22] [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] [Indexed: 11/23/2022] Open
Abstract
Viral-bacterial coinfections of the respiratory tract have long been associated with worsened disease outcomes. Clinical and basic research studies demonstrate that these infections are driven via complex interactions between the infecting pathogens, microbiome, and host immune response, although how these interactions contribute to disease progression is still not fully understood. Research over the last decade shows that the gut has a significant role in mediating respiratory outcomes, in a phenomenon known as the "gut-lung axis." Emerging literature demonstrates that acute respiratory viruses can modulate the gut-lung axis, suggesting that dysregulation of gut-lung cross talk may be a contributing factor during respiratory coinfection. This review will summarize the current literature regarding modulation of the gut-lung axis during acute respiratory infection, with a focus on the role of the microbiome, secondary infections, and the host immune response.
Collapse
Affiliation(s)
- Sidney Lane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yasmin Hilliam
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jennifer M. Bomberger
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
Langouët-Astrié C, Oshima K, McMurtry SA, Yang Y, Kwiecinski JM, LaRivière WB, Kavanaugh JS, Zakharevich I, Hansen KC, Shi D, Zhang F, Boguslawski KM, Perelman SS, Su G, Torres VJ, Liu J, Horswill AR, Schmidt EP. The influenza-injured lung microenvironment promotes MRSA virulence, contributing to severe secondary bacterial pneumonia. Cell Rep 2022; 41:111721. [PMID: 36450248 PMCID: PMC10082619 DOI: 10.1016/j.celrep.2022.111721] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 10/12/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
Influenza infection is substantially worsened by the onset of secondary pneumonia caused by bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA). The bidirectional interaction between the influenza-injured lung microenvironment and MRSA is poorly understood. By conditioning MRSA ex vivo in bronchoalveolar lavage fluid collected from mice at various time points of influenza infection, we found that the influenza-injured lung microenvironment dynamically induces MRSA to increase cytotoxin expression while decreasing metabolic pathways. LukAB, a SaeRS two-component system-dependent cytotoxin, is particularly important to the severity of post-influenza MRSA pneumonia. LukAB's activity is likely shaped by the post-influenza lung microenvironment, as LukAB binds to (and is activated by) heparan sulfate (HS) oligosaccharide sequences shed from the epithelial glycocalyx after influenza. Our findings indicate that post-influenza MRSA pneumonia is shaped by bidirectional host-pathogen interactions: host injury triggers changes in bacterial expression of toxins, the activity of which may be shaped by host-derived HS fragments.
Collapse
Affiliation(s)
| | - Kaori Oshima
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA
| | - Sarah A McMurtry
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA
| | - Yimu Yang
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA
| | - Jakub M Kwiecinski
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow 30387, Poland
| | - Wells B LaRivière
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA; Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jeffrey S Kavanaugh
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Igor Zakharevich
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045, USA
| | - Deling Shi
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Fuming Zhang
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Kristina M Boguslawski
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Sofya S Perelman
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Gouwei Su
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Jian Liu
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Alexander R Horswill
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Eric P Schmidt
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA 02115, USA
| |
Collapse
|
11
|
Song J, Zhao J, Cai X, Qin S, Chen Z, Huang X, Li R, Wang Y, Wang X. Lianhuaqingwen capsule inhibits non-lethal doses of influenza virus-induced secondary Staphylococcus aureus infection in mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115653. [PMID: 35995276 DOI: 10.1016/j.jep.2022.115653] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/07/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lianhuaqingwen capsule (LH-C) is a traditional Chinese medicine (TCM), consisting of two prescriptions, Ma-xing-shi-gan-tang (MXSGT) and Yinqiao San. It has been proven to have antiviral, antibacterial, and immunomodulatory effects in recent years. Clinically, it is commonly used in the treatment of respiratory tract infections. AIM OF THE STUDY It was demonstrated in our previous studies that LH-C has an effect of antivirus and inhibits influenza virus-induced bacterial adhesion to respiratory epithelial cells through down-regulation of cell adhesion molecules in vitro. However, LH-C's effect against influenza-induced secondary bacterial infection in animal studies remains unclear. Therefore, in the present study, we established a mouse model of infection with non-lethal doses of influenza virus(H1N1) and secondary infection of Staphylococcus aureus (S. aureus), to investigate the potential effects of LH-C. METHODS Experiments were carried out on BALB/c mice infecting non-lethal doses of H1N1 and non-lethal doses of S. aureus, and the viral, and bacterial doses were determined by observing and recording changes in the body weight, mortality, and pathological changes. Moreover, after LH-C treatment, the survival rate, body weight, lung index, viral titers, bacterial colonies, pathological changes, and the inflammatory cytokines in the mouse model have all been systematically determined. RESULTS In the superinfection models of H1N1 and S. aureus, the mortality rate was 100% in groups of mice infected with 20 PFU/50 μL of H1N1 and 105 CFU/mL of S. aureus, 20 PFU/50 μL of H1N1 and 106 CFU/mL of S. aureus, 4 PFU/50 μL of H1N1 and 106 CFU/mL of S. aureus. The mortality rate was 50% in the group of mice infected with 4 PFU/50 μL of H1N1 and 105 CFU/mL of S. aureus. The mortality rate was 37.5% in the group of mice infected with 20 PFU/50 μL of H1N1 alone and in the group of mice infected with 2 PFU/50 μL of H1N1 and 106 CFU/mL of S. aureus. The mortality rate in the group of mice infected with 2 PFU/50 μL of H1N1 and 106 CFU/mL of S. aureus was 30%. The infected mice of 2 PFU/50 μL of H1N1 and 106 CFU/mL of S. aureus had a weight loss of nearly 10%. About the histopathological changes in the lung tissue of infection mice, severe lung lesions were found in the superinfection models. LH-C improved survival in the superinfected mice, significantly reduced lung index, lowered viral titers and bacterial loads, and alleviated lung damage. It reduced lung inflammation by down-regulating mRNA expression levels of inflammatory mediators like IL-6, IL-1β, IL-10, TNF-α, IFN-β, MCP-1, and RANTES. CONCLUSIONS We found that superinfection from non-lethal doses of S. aureus following non-lethal doses of H1N1 was equally fatal in mice, confirming the severity of secondary infections. The ability of LH-C to alleviate lung injury resulting from secondary S. aureus infection induced by H1N1 was confirmed. These findings provided a further assessment of LH-C, suggesting that LH-C may have good therapeutic efficacy in influenza secondary bacterial infection disease.
Collapse
Affiliation(s)
- Jian Song
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jin Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xuejun Cai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shengle Qin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zexin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaodong Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yutao Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Xinhua Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China; Institute of Integration of Traditional and Western Medicine, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
12
|
R. Borgogna T, M. Voyich J. Examining the Executioners, Influenza Associated Secondary Bacterial Pneumonia. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.101666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Influenza infections typically present mild to moderate morbidities in immunocompetent host and are often resolved within 14 days of infection onset. Death from influenza infection alone is uncommon; however, antecedent influenza infection often leads to an increased susceptibility to secondary bacterial pneumonia. Bacterial pneumonia following viral infection exhibits mortality rates greater than 10-fold of those of influenza alone. Furthermore, bacterial pneumonia has been identified as the major contributor to mortality during each of the previous four influenza pandemics. Streptococcus pneumoniae, Staphylococcus aureus, Haemophilus influenzae, and Streptococcus pyogenes are the most prevalent participants in this pathology. Of note, these lung pathogens are frequently found as commensals of the upper respiratory tract. Herein we describe influenza-induced host-changes that lead to increased susceptibility to bacterial pneumonia, review virulence strategies employed by the most prevalent secondary bacterial pneumonia species, and highlight recent findings of bacterial sensing and responding to the influenza infected environment.
Collapse
|
13
|
Catabolic Ornithine Carbamoyltransferase Activity Facilitates Growth of Staphylococcus aureus in Defined Medium Lacking Glucose and Arginine. mBio 2022; 13:e0039522. [PMID: 35475645 PMCID: PMC9239276 DOI: 10.1128/mbio.00395-22] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Previous studies have found that arginine biosynthesis in Staphylococcus aureus is repressed via carbon catabolite repression (CcpA), and proline is used as a precursor. Unexpectedly, however, robust growth of S. aureus is not observed in complete defined medium lacking both glucose and arginine (CDM-R). Mutants able to grow on agar-containing defined medium lacking arginine (CDM-R) were selected and found to contain mutations within ahrC, encoding the canonical arginine biosynthesis pathway repressor (AhrC), or single nucleotide polymorphisms (SNPs) upstream of the native arginine deiminase (ADI) operon arcA1B1D1C1. Reverse transcription-PCR (RT-PCR) studies found that mutations within ccpA or ahrC or SNPs identified upstream of arcA1B1D1C1 increased the transcription of both arcB1 and argGH, encoding ornithine carbamoyltransferase and argininosuccinate synthase/lyase, respectively, facilitating arginine biosynthesis. Furthermore, mutations within the AhrC homologue argR2 facilitated robust growth within CDM-R. Complementation with arcB1 or arcA1B1D1C1, but not argGH, rescued growth in CDM-R. Finally, supplementation of CDM-R with ornithine stimulated growth, as did mutations in genes (proC and rocA) that presumably increased the pyrroline-5-carboxylate and ornithine pools. Collectively, these data suggest that the transcriptional regulation of ornithine carbamoyltransferase and, in addition, the availability of intracellular ornithine pools regulate arginine biosynthesis in S. aureus in the absence of glucose. Surprisingly, ~50% of clinical S. aureus isolates were able to grow in CDM-R. These data suggest that S. aureus is selected to repress arginine biosynthesis in environments with or without glucose; however, mutants may be readily selected that facilitate arginine biosynthesis and growth in specific environments lacking arginine.
Collapse
|
14
|
Sura T, Surabhi S, Maaß S, Hammerschmidt S, Siemens N, Becher D. The global proteome and ubiquitinome of bacterial and viral co-infected bronchial epithelial cells. J Proteomics 2022; 250:104387. [PMID: 34600154 DOI: 10.1016/j.jprot.2021.104387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/26/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022]
Abstract
Viral infections facilitate bacterial trafficking to the lower respiratory tract resulting in bacterial-viral co-infections. Bacterial dissemination to the lower respiratory tract is enhanced by influenza A virus induced epithelial cell damage and dysregulation of immune responses. Epithelial cells act as a line of defense and detect pathogens by a high variety of pattern recognition receptors. The post-translational modification ubiquitin is involved in almost every cellular process. Moreover, ubiquitination contributes to the regulation of host immune responses, influenza A virus uncoating and transport within host cells. We applied proteomics with a special focus on ubiquitination to assess the impact of single bacterial and viral as well as bacterial-viral co-infections on bronchial epithelial cells. We used Tandem Ubiquitin Binding Entities to enrich polyubiquitinated proteins and assess changes in the ubiquitinome. Infecting 16HBE cells with Streptococcus pyogenes led to an increased abundance of proteins related to mitochondrial translation and energy metabolism in proteome and ubiquitinome. In contrast, influenza A virus infection mainly altered the ubiquitinome. Co-infections had no additional impact on protein abundances or affected pathways. Changes in protein abundance and enriched pathways were assigned to imprints of both infecting pathogens. SIGNIFICANCE: Viral and bacterial co-infections of the lower respiratory tract are a burden for health systems worldwide. Therefore, it is necessary to elucidate the complex interplay between the host and the infecting pathogens. Thus, we analyzed the proteome and the ubiquitinome of co-infected bronchial epithelial cells to elaborate a potential synergism of the two infecting organisms. The results presented in this work can be used as a starting point for further analyses.
Collapse
Affiliation(s)
- Thomas Sura
- University of Greifswald, Center for Functional Genomics of Microbes, Institute of Microbiology, Department of Microbial Proteomics, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Surabhi Surabhi
- University of Greifswald, Center for Functional Genomics of Microbes, Interfaculty Institute for Genetics and Functional Genomics, Department of Molecular Genetics and Infection Biology, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Sandra Maaß
- University of Greifswald, Center for Functional Genomics of Microbes, Institute of Microbiology, Department of Microbial Proteomics, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Sven Hammerschmidt
- University of Greifswald, Center for Functional Genomics of Microbes, Interfaculty Institute for Genetics and Functional Genomics, Department of Molecular Genetics and Infection Biology, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Nikolai Siemens
- University of Greifswald, Center for Functional Genomics of Microbes, Interfaculty Institute for Genetics and Functional Genomics, Department of Molecular Genetics and Infection Biology, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Dörte Becher
- University of Greifswald, Center for Functional Genomics of Microbes, Institute of Microbiology, Department of Microbial Proteomics, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany.
| |
Collapse
|
15
|
Han C, Zhang T, Zhao Y, Dong L, Li X, Zheng J, Guo W, Xu Y, Cai C. Successful treatment of pleural empyema and necrotizing pneumonia caused by methicillin-resistant Staphylococcus aureus infection following influenza A virus infection: A case report and literature review. Front Pediatr 2022; 10:959419. [PMID: 36090578 PMCID: PMC9462453 DOI: 10.3389/fped.2022.959419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
With the rapid increase in the number of infections, children with Staphylococcus aureus (S. aureus) infection secondary to Influenza A virus (IAV), appear to have a great possibility of causing severe complications and illness. Despite some cases and research findings regarding the death of children with IAV and S. aureus, coinfection included, there were few details about successful treatment of pleural empyema and necrotizing pneumonia caused by methicillin-resistant Staphylococcus aureus (MRSA) infection following IAV. In this case report, we describe the clinical symptoms and treatment of a teenager with pleural empyema and necrotizing pneumonia related to S. aureus secondary infection who was initially infected by IAV. This case highlights the importance of early recognition and application of thoracoscopy for this potentially fatal pleural empyema caused by MRSA and IAV coinfection. We conclude that this is a significant case that contributes to raising awareness regarding rarely occurring severe respiratory infections by MRSA in a child with normal immune function after IAV. In addition, further studies are needed to explore risk factors for IAV coinfection with S. aureus.
Collapse
Affiliation(s)
- Chunjiao Han
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China.,Department of Pulmonology, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Tongqiang Zhang
- Department of Pulmonology, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Yidi Zhao
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China
| | - Lili Dong
- Department of Pulmonology, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Xiaole Li
- Department of Pulmonology, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Jiafeng Zheng
- Department of Pulmonology, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Wei Guo
- Department of Pulmonology, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Yongsheng Xu
- Department of Pulmonology, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Chunquan Cai
- Institute of Pediatrics, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| |
Collapse
|
16
|
Abu-Rub LI, Abdelrahman HA, Johar ARA, Alhussain HA, Hadi HA, Eltai NO. Antibiotics Prescribing in Intensive Care Settings during the COVID-19 Era: A Systematic Review. Antibiotics (Basel) 2021; 10:935. [PMID: 34438985 PMCID: PMC8389042 DOI: 10.3390/antibiotics10080935] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/18/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023] Open
Abstract
The prevalence of patients admitted to intensive care units (ICUs) with SARS-CoV-2 infection who were prescribed antibiotics is undetermined and might contribute to the increased global antibiotic resistance. This systematic review evaluates the prevalence of antibiotic prescribing in patients admitted to ICUs with SARS-CoV-2 infection using PRISMA guidelines. We searched and scrutinized results from PubMed and ScienceDirect databases for published literature restricted to the English language up to 11 May 2021. In addition, we included observational studies of humans with laboratory-confirmed SARS-CoV-2 infection, clinical characteristics, and antibiotics prescribed for ICU patients with SARS-CoV-2 infections. A total of 361 studies were identified, but only 38 were included in the final analysis. Antibiotic prescribing data were available from 2715 patients, of which prevalence of 71% was reported in old age patients with a mean age of 62.7 years. From the reported studies, third generation cephalosporin had the highest frequency amongst reviewed studies (36.8%) followed by azithromycin (34.2%). The estimated bacterial infection in 12 reported studies was 30.8% produced by 15 different bacterial species, and S. aureus recorded the highest bacterial infection (75%). The fundamental outcomes were the prevalence of ICU COVID-19 patients prescribed antibiotics stratified by age, type of antibiotics prescribed, and the presence of co-infections and comorbidities. In conclusion, more than half of ICU patients with SARS-CoV-2 infection received antibiotics, and prescribing is significantly higher than the estimated frequency of identified bacterial co-infection.
Collapse
Affiliation(s)
- Lubna I. Abu-Rub
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (H.A.A.); (H.A.A.)
| | - Hana A. Abdelrahman
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (H.A.A.); (H.A.A.)
| | | | - Hashim A. Alhussain
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (H.A.A.); (H.A.A.)
| | - Hamad Abdel Hadi
- Communicable Diseases Centre, Infectious Disease Division, Hamad Medical Corporation, Doha 3050, Qatar;
| | - Nahla O. Eltai
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (H.A.A.); (H.A.A.)
| |
Collapse
|
17
|
Feng J, Liu L, He Y, Wang M, Zhou D, Wang J. Novel insights into the pathogenesis of virus-induced ARDS: review on the central role of the epithelial-endothelial barrier. Expert Rev Clin Immunol 2021; 17:991-1001. [PMID: 34224287 DOI: 10.1080/1744666x.2021.1951233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Respiratory viruses can directly or indirectly damage the pulmonary defense barrier, potentially contributing to acute respiratory distress syndrome (ARDS). Despite developments in the understanding of the pathogenesis of ARDS, the underlying pathophysiology still needs to be elucidated.Areas covered: The PubMed database was reviewed for relevant papers published up to 2021. This review summarizes the currently immunological and clinical studies to provide a systemic overview of the epithelial-endothelial barrier, given the recently published immunological profiles upon viral pneumonia, and the potentially detrimental contribution to respiratory function caused by damage to this barrier.Expert opinion: The biophysical structure of host pulmonary defense is intrinsically linked with the ability of alveolar epithelial and capillary endothelial cells, known as the epithelial-endothelial barrier, to respond to, and instruct the delicate immune system to protect the lungs from infections and injuries. Recently published immunological profiles upon viral infection, and its contributions to the damage of respiratory function, suggest a central role for the pulmonary epithelial and endothelial barrier in the pathogenesis of ARDS. We suggest a central role and common pathways by which the epithelial-endothelial barrier contributes to the pathogenesis of ARDS.
Collapse
Affiliation(s)
- Jun Feng
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lina Liu
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang He
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Daixing Zhou
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junshuai Wang
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Wilden JJ, Jacob JC, Ehrhardt C, Ludwig S, Boergeling Y. Altered Signal Transduction in the Immune Response to Influenza Virus and S. pneumoniae or S. aureus Co-Infections. Int J Mol Sci 2021; 22:5486. [PMID: 34067487 PMCID: PMC8196994 DOI: 10.3390/ijms22115486] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/14/2021] [Accepted: 05/19/2021] [Indexed: 12/23/2022] Open
Abstract
Influenza virus is a well-known respiratory pathogen, which still leads to many severe pulmonary infections in the human population every year. Morbidity and mortality rates are further increased if virus infection coincides with co-infections or superinfections caused by bacteria such as Streptococcus pneumoniae (S. pneumoniae) and Staphylococcus aureus (S. aureus). This enhanced pathogenicity is due to complex interactions between the different pathogens and the host and its immune system and is mainly governed by altered intracellular signaling processes. In this review, we summarize the recent findings regarding the innate and adaptive immune responses during co-infection with influenza virus and S. pneumoniae or S. aureus, describing the signaling pathways involved and how these interactions influence disease outcomes.
Collapse
Affiliation(s)
- Janine J. Wilden
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, 48149 Muenster, Germany; (J.J.W.); (J.C.J.); (S.L.)
| | - Jasmin C. Jacob
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, 48149 Muenster, Germany; (J.J.W.); (J.C.J.); (S.L.)
- CiM-IMPRS, The Joined Graduate School of the Cells in Motion Interfaculty Centre, University of Muenster and the International Max Planck Research School—Molecular Biomedicine, 48149 Muenster, Germany
| | - Christina Ehrhardt
- Section of Experimental Virology, Center for Molecular Biomedicine (CMB), Institute of Medical Microbiology, Jena University Hospital, 07745 Jena, Germany;
| | - Stephan Ludwig
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, 48149 Muenster, Germany; (J.J.W.); (J.C.J.); (S.L.)
- “Cells in Motion Interfaculty Center (CIMIC)”, WWU Muenster, 48149 Muenster, Germany
| | - Yvonne Boergeling
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, 48149 Muenster, Germany; (J.J.W.); (J.C.J.); (S.L.)
| |
Collapse
|
19
|
Bacterial and fungal coinfections in COVID-19 patients hospitalized during the New York City pandemic surge. Infect Control Hosp Epidemiol 2020; 42:84-88. [PMID: 32703320 PMCID: PMC7417979 DOI: 10.1017/ice.2020.368] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We observed bacterial or fungal coinfections in COVID-19 patients admitted between March 1 and April 18, 2020 (152 of 4,267, 3.6%). Among these patients, mortality was 57%; 74% were intubated; 51% with bacteremia had central venous catheters. Time to culture positivity was 6–7 days, and 79% had received prior antibiotics. Metallo-β-lactamase–producing E. cloacae coinfections occurred in 5 patients.
Collapse
|
20
|
Abstract
Influenza A virus (IAV) causes annual epidemics and sporadic pandemics of respiratory disease. Secondary bacterial coinfection by organisms such as Staphylococcus aureus is the most common complication of primary IAV infection and is associated with high levels of morbidity and mortality. Here, we report the first identified S. aureus factor (lipase 1) that enhances IAV replication during infection via positive modulation of virus budding. The effect is observed in vivo in embryonated hen’s eggs and greatly enhances the yield of a vaccine strain, a finding that could be applied to address global shortages of influenza vaccines. Influenza A virus (IAV) causes annual epidemics of respiratory disease in humans, often complicated by secondary coinfection with bacterial pathogens such as Staphylococcus aureus. Here, we report that the S. aureus secreted protein lipase 1 enhances IAV replication in vitro in primary cells, including human lung fibroblasts. The proviral activity of lipase 1 is dependent on its enzymatic function, acts late in the viral life cycle, and results in increased infectivity through positive modulation of virus budding. Furthermore, the proviral effect of lipase 1 on IAV is exhibited during in vivo infection of embryonated hen’s eggs and, importantly, increases the yield of a vaccine strain of IAV by approximately 5-fold. Thus, we have identified the first S. aureus protein to enhance IAV replication, suggesting a potential role in coinfection. Importantly, this activity may be harnessed to address global shortages of influenza vaccines.
Collapse
|
21
|
Xiu L, Sheng S, Hu Z, Liu Y, Li J, Zhang H, Liang Y, Du R, Wang X. Exopolysaccharides from Lactobacillus kiferi as adjuvant enhanced the immuno-protective against Staphylococcus aureus infection. Int J Biol Macromol 2020; 161:10-23. [PMID: 32512102 DOI: 10.1016/j.ijbiomac.2020.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 01/17/2023]
Abstract
Exopolysaccharides from lactic acid bacteria (LAB) have gained more attention due to their health benefits. Most research on LAB EPS focuses on antitumor and antioxidant activities. To our knowledge, the immunoadjuvant activity of LAB EPS has not been thoroughly studied. In this study, the EPS produced by Lactobacillus kiferi WXD029 were purified by ethanol precipitation and column chromatography fractionation. The molecular weight of the EPS was 3.423 × 105 Da and was mainly composed of Glu, GlcN, and GalN in a molar ratio of 3.1:1:1. In vitro, EPS could significantly enhance the proliferation and phagocytic activity as well as induce the production of NO, TNF-α, IL-1β, and IL-6 in RAW264.7 cells. In vivo, the EPS adjuvant could increase the titers of S.aureus antigen-specific antibodies and markedly enhanced T cell proliferation. Notably, EPS adjuvant also induced a strong potential Th1, Th2 and Th17-cell mixture responses. Furthermore, immunization with S.aureus antigen plus EPS adjuvant induced a protective effect when compared with S.aureus antigen alone in murine bacteremia, pneumonia and mastitis model. Collectively, these results suggest that EPS derived from probiotic Lactobacillus kiferi strain is promising as an efficient adjuvant candidate for the prevention of S. aureus infections.
Collapse
Affiliation(s)
- Lei Xiu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot 010070, PR China
| | - Shouxin Sheng
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot 010070, PR China
| | - Zhongpeng Hu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot 010070, PR China
| | - Yang Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot 010070, PR China
| | - Jianwei Li
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot 010070, PR China
| | - Haochi Zhang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot 010070, PR China
| | - Yanchen Liang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot 010070, PR China
| | - Ruiping Du
- Animal Nutrition Institute, Agriculture and Animal Husbandry Academy of Inner Mongolia, Hohhot 010031, PR China.
| | - Xiao Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot 010070, PR China.
| |
Collapse
|
22
|
Jia L, Zhao J, Yang C, Liang Y, Long P, Liu X, Qiu S, Wang L, Xie J, Li H, Liu H, Guo W, Wang S, Li P, Zhu B, Hao R, Ma H, Jiang Y, Song H. Severe Pneumonia Caused by Coinfection With Influenza Virus Followed by Methicillin-Resistant Staphylococcus aureus Induces Higher Mortality in Mice. Front Immunol 2019; 9:3189. [PMID: 30761162 PMCID: PMC6364753 DOI: 10.3389/fimmu.2018.03189] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/31/2018] [Indexed: 01/20/2023] Open
Abstract
Background: Coinfection with influenza virus and bacteria is a major cause of high mortality during flu pandemics. Understanding the mechanisms behind such coinfections is of utmost importance both for the clinical treatment of influenza and the prevention and control of epidemics. Methods: To investigate the cause of high mortality during flu pandemics, we performed coinfection experiments with H1N1 influenza virus and Staphylococcus aureus in which mice were infected with bacteria at time points ranging from 0 to 7 days after infection with influenza virus. Results: The mortality rates of mice infected with bacteria were highest 0-3 days after infection with influenza virus; lung tissues extracted from these co-infected mice showed higher infiltrating cells and thicker lung parenchyma than lung samples from coinfected mice in which influenza virus was introduced at other times and sequences. The levels of interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-8, and IL-6 in the 0-3 day coinfected group were significantly higher than those in the other groups (p < 0.01), as were the mRNA levels of IFN-γ, IL-6, and TNF-α. Coinfection with influenza virus and S. aureus led to high mortality rates that are directly dependent on the sequence and timing of infection by both pathogens. Moreover, coinfection following this particular schedule induced severe pneumonia, leading to increased mortality. Conclusions: Our data suggest that prevention of bacterial co-infection in the early stage of influenza virus infection is critical to reducing the risk of clinical mortality.
Collapse
Affiliation(s)
- Leili Jia
- Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.,Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Jiangyun Zhao
- Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.,Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Chaojie Yang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Yuan Liang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Pengwei Long
- Chinese PLA Center for Disease Control and Prevention, Beijing, China.,Department of Health Care, Chinese PLA Joint Staff Headquarters Guard Bureau, Beijing, China
| | - Xiao Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Shaofu Qiu
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Ligui Wang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Jing Xie
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Hao Li
- Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Hongbo Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Weiguang Guo
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Shan Wang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Peng Li
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | | | - Rongzhang Hao
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Hui Ma
- The 6th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongbin Song
- Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.,Chinese PLA Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
23
|
Nygaard TK, Borgogna TR, Sward EW, Guerra FE, Dankoff JG, Collins MM, Pallister KB, Chen L, Kreiswirth BN, Voyich JM. Aspartic Acid Residue 51 of SaeR Is Essential for Staphylococcus aureus Virulence. Front Microbiol 2018; 9:3085. [PMID: 30619166 PMCID: PMC6302044 DOI: 10.3389/fmicb.2018.03085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/29/2018] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus is a common Gram-positive bacteria that is a major cause of human morbidity and mortality. The SaeR/S two-component sensory system of S. aureus is important for virulence gene transcription and pathogenesis. However, the influence of SaeR phosphorylation on virulence gene transcription is not clear. To determine the importance of potential SaeR phosphorylation sites for S. aureus virulence, we generated genomic alanine substitutions at conserved aspartic acid residues in the receiver domain of the SaeR response regulator in clinically significant S. aureus pulsed-field gel electrophoresis (PFGE) type USA300. Transcriptional analysis demonstrated a dramatic reduction in the transcript abundance of various toxins, adhesins, and immunomodulatory proteins for SaeR with an aspartic acid to alanine substitution at residue 51. These findings corresponded to a significant decrease in cytotoxicity against human erythrocytes and polymorphonuclear leukocytes, the ability to block human myeloperoxidase activity, and pathogenesis during murine soft-tissue infection. Analysis of SaeR sequences from over 8,000 draft S. aureus genomes revealed that aspartic acid residue 51 is 100% conserved. Collectively, these results demonstrate that aspartic acid residue 51 of SaeR is essential for S. aureus virulence and underscore a conserved target for novel antimicrobial strategies that treat infection caused by this pathogen.
Collapse
Affiliation(s)
- Tyler K Nygaard
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Timothy R Borgogna
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Eli W Sward
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Fermin E Guerra
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Jennifer G Dankoff
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Madison M Collins
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Kyler B Pallister
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Liang Chen
- Public Health Research Institute Tuberculosis Center, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Barry N Kreiswirth
- Public Health Research Institute Tuberculosis Center, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| |
Collapse
|