1
|
Evans DCS, Mitkin AA, Rohde H, Meyer RL. Extracellular DNA and polysaccharide intercellular adhesin protect Staphylococcus epidermidis biofilms from phagocytosis by polymorphonuclear neutrophils. Microbiol Res 2025; 297:128176. [PMID: 40239428 DOI: 10.1016/j.micres.2025.128176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/20/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025]
Abstract
Staphylococcus epidermidis is the leading cause of implant-associated infections, where it forms biofilms that are highly effective at evading the immune system. Here we investigate to what extent the biofilm extracellular matrix components extracellular DNA (eDNA) and polysaccharide intercellular adhesin (PIA) protect S. epidermidis from phagocytosis by polymorphonuclear neutrophils (PMN). We visualised phagocytosis using time-lapse confocal laser scanning microscopy of PMN interacting with planktonic S. epidermidis and 24 h old biofilms formed by the wildtype strain or mutant strains lacking either eDNA or PIA. We also compared phagocytosis of 24 h vs. 6 h old biofilms. PMN easily moved around and phagocytised S. epidermidis that were adhered to a surface from a planktonic culture. In contrast, PMN quickly became immobilised when interacting with biofilms. Very few PMN were able to phagocytise young (6 h) and mature (24 h) biofilms, suggesting that the accumulation of matrix components quickly provides a protective effect. Biofilms lacking either eDNA or PIA were much less dense, and many more PMN were able to phagocytise bacteria in these biofilms. Our findings suggest that both eDNA and PIA contribute to the ability of S. epidermidis biofilms to resist phagocytosis.
Collapse
Affiliation(s)
| | | | - Holger Rohde
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
2
|
Zhong J, Bæk O, Doughty R, Jørgensen BM, Jensen HE, Thymann T, Sangild PT, Brunse A, Nguyen DN. Reduced parenteral glucose supply during neonatal infection attenuates neurological and renal pathology associated with modulation of innate and Th1 immunity. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167723. [PMID: 39978441 DOI: 10.1016/j.bbadis.2025.167723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/17/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Premature infants are highly susceptible to infections that can lead to sepsis with life-threatening organ dysfunctions. The clinical practice of high parenteral glucose supply in preterm infants can exacerbate infection outcomes through excessive glycolysis-induced inflammatory response. This in turn can affect the health of vital preterm organs, including the brain and kidneys. We hypothesized that reduced parenteral glucose supply to infected preterm newborns may help protect against pathology in these two key organs. METHODS Cesarean-delivered preterm pigs were nourished with high or low parenteral glucose levels (21 % vs. 5 %), infused with Staphylococcus epidermidis or saline, and monitored in heated, oxygenated incubators until 22 h. Blood, brain, and kidney samples were collected for histological, immunohistological, q-PCR, ELISA, and biochemical analyses. RESULTS Infection led to multiple pathological changes (e.g. edema), increased inflammation and tissue injury (indicated by gene expression data) in both brain and kidneys of preterm piglets. Reduced glucose supply in infected animals alleviated histopathological manifestations in the brain, and reduced neuroinflammation with enhanced M2 microglial phenotype. Reduced glucose supply also decreased plasma creatinine, and the severity of renal edema, tubular vacuolization and dilatation. Multiple genes related to innate and Th1 immunity in both organs were dampened by reduced glucose supply. Correlation analysis showed that renal inflammation was more closely connected to systemic inflammation compared to neuroinflammation. CONCLUSION Reduced glucose supply can reduce renal and neuro-inflammation during neonatal infection, thereby protecting brain and kidney health in infected preterm neonates.
Collapse
Affiliation(s)
- Jingren Zhong
- Comparative Pediatrics, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Ole Bæk
- Comparative Pediatrics, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Richard Doughty
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Benjamin Meyer Jørgensen
- Section of Pathological Sciences, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Henrik Elvang Jensen
- Section of Pathological Sciences, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Thomas Thymann
- Comparative Pediatrics, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Per Torp Sangild
- Comparative Pediatrics, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark; Department of Pediatrics, Odense University Hospital, Odense, Denmark; Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
| | - Anders Brunse
- Comparative Pediatrics, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark.
| | - Duc Ninh Nguyen
- Comparative Pediatrics, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
3
|
Shirmohammadpour M, Mehrasbi MR, Noshiranzadeh N, Afshar D, Mansori K, Mirzaei B. Investigation of the effect of anti-PIA/PNAG antibodies on biofilm formation in Escherichia coli. Front Microbiol 2025; 16:1552670. [PMID: 40115191 PMCID: PMC11922938 DOI: 10.3389/fmicb.2025.1552670] [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: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Polysaccharide Intercellular Adhesin (PIA), a surface polysaccharide produced by Staphylococcus aureus and Staphylococcus epidermidis, is a compelling target for opsonic and protective antibodies against these bacteria. Escherichia coli has recently made an exopolysaccharide called poly-β(1,6)-N-acetylglucosamine (PNAG), biochemically indistinguishable from PIA. This study investigated the effect of antibodies generated against PNAG on biofilm formation and the opsonization activity of secreted antibodies in Escherichia coli. Following purification and structural confirmation of PIA polysaccharide from producing Staphylococcus epidermidis, the ability to inhibit biofilm and the function of secreted antibodies for the mentioned polysaccharide were evaluated using semi-quantitative methods in a mouse model. Subsequently, the opsonic activity of antibodies targeting Escherichia coli strain ATCC 25922 was evaluated. The extracted polysaccharide was confirmed using FTIR, NMR, and colorimetric methods, and the results showed that the purified PIA induced protective antibodies with 40.48% opsonization properties in E. coli. The sera of the PIA-immunized groups showed a significant increase in antibody production and protective IgG titer levels compared to the control group. Also, the antibodies produced showed a substantial difference in inhibiting biofilm production in vitro compared to non-immunized serum. Antibodies directed against PIA with a lethality of 40.48% showed a significant effect on the absence of biofilm formation in E. coli. Despite the opsonic properties of the antibodies for E. coli, the simultaneous impact of these antibodies on infections caused by S. epidermidis and E. coli may have a role that requires further investigation and studies in animal models.
Collapse
Affiliation(s)
- Mina Shirmohammadpour
- Department of Microbiology and Virology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Student Research Committee, Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohammad Reza Mehrasbi
- Department of Environmental Health Engineering, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Nader Noshiranzadeh
- Department of Chemistry, Faculty of Sciences, University of Zanjan, Zanjan, Iran
| | - Davoud Afshar
- Department of Microbiology and Virology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kamyar Mansori
- Department of Biostatistics and Epidemiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Bahman Mirzaei
- Department of Microbiology and Virology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
4
|
Zhang T, Luo R, Ehrström M, Melican K. Staphylococcus lugdunensis does not exert competitive exclusion on human corneocytes. MICROBIOLOGY (READING, ENGLAND) 2025; 171. [PMID: 39888662 DOI: 10.1099/mic.0.001522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Human skin is our primary physical barrier and largest immune organ, and it also hosts a protective microbiota. Staphylococci are prominent members of the skin microbiota, including the ubiquitous coagulase-negative staphylococci (CoNS). The coagulase-positive Staphylococcus aureus is found as part of the microbiota, but it poses clinical concern due to its potential pathogenicity and antibiotic resistance. Recently, a CoNS, Staphylococcus lugdunensis, has been shown to inhibit S. aureus growth via the production of a novel antibiotic, lugdunin. In this study, we use human skin models to understand the spatial relationships between the CoNS Staphylococcus epidermidis and S. lugdunensis with S. aureus during colonization of human skin. We investigated the attachment patterns of the bacteria, both individually and in competition. Surprisingly, we found that attachment did not always correlate with colonization ability. S. lugdunensis exhibited significantly reduced attachment to human skin stratum corneum but was an efficient longer-term colonizer. S. lugdunensis had a distinct attachment pattern on human corneocytes, with no significant overlap, or competitive exclusion, with the other strains. S. lugdunensis is a potential probiotic strain, with a proven ability to suppress S. aureus. Before this potential can be realized, however, further research is needed to understand how this strain adheres and interacts with other bacteria in the human skin microenvironment.
Collapse
Affiliation(s)
- Tianqi Zhang
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ran Luo
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | - Keira Melican
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
5
|
Kaplan JB, Assa M, Mruwat N, Sailer M, Regmi S, Kridin K. Facultatively Anaerobic Staphylococci Enable Anaerobic Cutibacterium Species to Grow and Form Biofilms Under Aerobic Conditions. Microorganisms 2024; 12:2601. [PMID: 39770803 PMCID: PMC11678309 DOI: 10.3390/microorganisms12122601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Facultatively anaerobic Staphylococcus spp. and anaerobic Cutibacterium spp. are among the most prominent bacteria on human skin. Although skin microbes generally grow as multispecies biofilms, few studies have investigated the interaction between staphylococci and Cutibacterium spp. in dual-species biofilms. Here, we measured the mono- and dual-species biofilm formation of four staphylococcal species (S. epidermidis, S. hominis, S. capitis, and S. aureus) and two Cutibacterium spp. (C. acnes and C. avidum) cultured in vitro under both aerobic and anaerobic conditions. The biofilms were quantitated by rinsing them to remove planktonic cells, detaching the biofilm bacteria via sonication, and enumerating the cells by dilution plating. When cultured alone, staphylococci formed biofilms under both aerobic and anaerobic conditions, whereas Cutibacterium spp. formed biofilms only under anaerobic conditions. In co-culture, staphylococcal biofilm formation was unaffected by the presence of Cutibacterium spp., regardless of oxygen availability. However, Cutibacterium spp. biofilm formation was significantly enhanced in the presence of staphylococci, enabling robust growth under both anaerobic and aerobic conditions. Fluorescence confocal microscopy of the aerobic dual-species biofilms suggested that staphylococci create anaerobic niches at the base of the biofilm where C. acnes can grow. These findings demonstrate that staphylococci facilitate the colonization of Cutibacterium spp. in oxygen-rich environments, potentially explaining their presence in high numbers on the oxygen-exposed stratum corneum.
Collapse
Affiliation(s)
- Jeffrey B. Kaplan
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel; (N.M.); (K.K.)
| | - Michael Assa
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
| | - Noor Mruwat
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel; (N.M.); (K.K.)
| | - Miloslav Sailer
- Kane Biotech Inc., Winnipeg, MB R3T 6G2, Canada; (M.S.); (S.R.)
| | - Suresh Regmi
- Kane Biotech Inc., Winnipeg, MB R3T 6G2, Canada; (M.S.); (S.R.)
| | - Khalaf Kridin
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel; (N.M.); (K.K.)
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
| |
Collapse
|
6
|
Weißelberg S, Both A, Failla AV, Huang J, Linder S, Ohnezeit D, Bartsch P, Aepfelbacher M, Rohde H. Staphylococcus epidermidis alters macrophage polarization and phagocytic uptake by extracellular DNA release in vitro. NPJ Biofilms Microbiomes 2024; 10:131. [PMID: 39567551 PMCID: PMC11579364 DOI: 10.1038/s41522-024-00604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
Biofilm formation shields Staphylococcus epidermidis from host defense mechanisms, contributing to chronic implant infections. Using wild-type S. epidermidis 1457, a PIA-negative mutant (1457-M10), and an eDNA-negative mutant (1457ΔatlE), this study examined the influence of biofilm matrix components on human monocyte-derived macrophage (hMDM) interactions. The wild-type strain was resistant to phagocytosis and induced an anti-inflammatory response in hMDMs, while both mutants were more susceptible to phagocytosis and triggered a pro-inflammatory response. Removing eDNA from the 1457 biofilm matrix increased hMDM uptake and a pro-inflammatory reaction, whereas adding eDNA to the 1457ΔatlE mutant reduced phagocytosis and promoted an anti-inflammatory response. Inhibiting TLR9 enhanced bacterial uptake and induced a pro-inflammatory response in hMDMs exposed to wild-type S. epidermidis. This study highlights the critical role of eDNA in immune evasion and the central role of TLR9 in modulating macrophage responses, advancing the understanding of implant infections.
Collapse
Affiliation(s)
- Samira Weißelberg
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Anna Both
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Antonio Virgilio Failla
- UKE Microscopy Imaging Facility (Umif), Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jiabin Huang
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Stefan Linder
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Denise Ohnezeit
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Patricia Bartsch
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Martin Aepfelbacher
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Holger Rohde
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
7
|
Cruikshank MJ, Pitzer JM, Ameri K, Rother CV, Cooper K, Nuxoll AS. Characterization of Staphylococcus lugdunensis biofilms through ethyl methanesulfonate mutagenesis. AIMS Microbiol 2024; 10:880-893. [PMID: 39628716 PMCID: PMC11609425 DOI: 10.3934/microbiol.2024038] [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: 07/27/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 12/06/2024] Open
Abstract
Staphylococcus lugdunensis is a coagulase-negative species responsible for a multitude of infections. These infections often resemble those caused by the more pathogenic staphylococcal species, Staphylococcus aureus, such as skin and soft tissue infections, prosthetic joint infections, and infective endocarditis. Despite a high mortality rate and infections that differ from other coagulase-negative species, little is known regarding S. lugdunensis pathogenesis. The objective of this study is to identify the essential factors for biofilm formation in S. lugdunensis. S. lugdunensis was mutagenized through ethyl methanesulfonate (EMS) exposure, and the individual cells were separated using a cell sorter and examined for biofilm formation at 8 hr and 24 hr timepoints. Mutations that resulted in either increased or decreased biofilm formation were sequenced to identify the genes responsible for the respective phenotypes. A mutation within the S. lugdunensis surface protein A (slsA) gene was common among all of the low biofilm formers, thus suggesting that high expression of this protein is important in biofilm formation. However, other mutations common among the mutants with decreased biofilm formation were in the putative divalent cation transport gene, mgtE. Conversely, a mutation in the gene that codes for the von Willebrand factor binding protein, vwbl, was common among the mutants with increased biofilm formation. Following proteinase K treatment, a significant dispersal of the S. lugdunensis biofilm matrix occurred, thus confirming the presence of primarily protein-mediated biofilms; this is in agreement with previous S. lugdunensis studies. Additionally, all low biofilm formers exhibited decreased protein levels (1.95-2.77 fold change) within the biofilm matrix, while no difference was observed with extracellular DNA (eDNA) or polysaccharides. This study presents a unique methodology to identify genes that affect biofilm formation and sheds light on S. lugdunensis pathogenesis.
Collapse
Affiliation(s)
- McKenna J. Cruikshank
- Department of Biology, University of Nebraska at Kearney, Kearney, NE, United States
| | - Justine M. Pitzer
- Department of Biology, University of Nebraska at Kearney, Kearney, NE, United States
| | - Kimia Ameri
- School of Interdisciplinary Informatics, College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Caleb V. Rother
- Department of Biology, University of Nebraska at Kearney, Kearney, NE, United States
| | - Kathryn Cooper
- School of Interdisciplinary Informatics, College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Austin S. Nuxoll
- Department of Biology, University of Nebraska at Kearney, Kearney, NE, United States
| |
Collapse
|
8
|
Lang JC, Brutscher A, Ehrström M, Melican K. Tissue resident cells differentiate S. aureus from S. epidermidis via IL-1β following barrier disruption in healthy human skin. PLoS Pathog 2024; 20:e1012056. [PMID: 39208402 PMCID: PMC11389914 DOI: 10.1371/journal.ppat.1012056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 09/11/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024] Open
Abstract
The Staphylococcus sp. are a dominant part of the human skin microbiome and present across the body. Staphylococcus epidermidis is a ubiquitous skin commensal, while S. aureus is thought to colonize at least 30% of the population. S. aureus are not only colonizers but a leading cause of skin and soft tissue infections and a critical healthcare concern. To understand how healthy human skin may differentiate commensal bacteria, such as S. epidermidis, from the potential pathogen methicillin-resistant S. aureus (MRSA), we use ex vivo human skin models that allow us to study this host-bacterial interaction in the most clinically relevant environment. Our work highlights the role of the outer stratum corneum as a protective physical barrier against invasion by colonizing Staphylococci. We show how the structural cells of the skin can internalize and respond to different Staphylococci with increasing sensitivity. In intact human skin, a discriminatory IL-1β response was identified, while disruption of the protective stratum corneum triggered an increased and more diverse immune response. We identified and localized tissue resident Langerhans cells (LCs) as a potential source of IL-1β and go on to show a dose-dependent response of MUTZ-LCs to S. aureus but not S. epidermidis. This suggests an important role of LCs in sensing and discriminating between bacteria in healthy human skin, particularly in intact skin and provides a detailed snapshot of how human skin differentiates between friend and potential foe. With the rise in antibiotic resistance, understanding the innate immune response of healthy skin may help us find ways to enhance or manipulate these natural defenses to prevent invasive infection.
Collapse
Affiliation(s)
- Julia C Lang
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Brutscher
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Keira Melican
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Beck C, Krusche J, Notaro A, Walter A, Kränkel L, Vollert A, Stemmler R, Wittmann J, Schaller M, Slavetinsky C, Mayer C, De Castro C, Peschel A. Wall teichoic acid substitution with glucose governs phage susceptibility of Staphylococcus epidermidis. mBio 2024; 15:e0199023. [PMID: 38470054 PMCID: PMC11005348 DOI: 10.1128/mbio.01990-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 02/20/2024] [Indexed: 03/13/2024] Open
Abstract
The species- and clone-specific susceptibility of Staphylococcus cells for bacteriophages is governed by the structures and glycosylation patterns of wall teichoic acid (WTA) glycopolymers. The glycosylation-dependent phage-WTA interactions in the opportunistic pathogen Staphylococcus epidermidis and in other coagulase-negative staphylococci (CoNS) have remained unknown. We report a new S. epidermidis WTA glycosyltransferase TagE whose deletion confers resistance to siphoviruses such as ΦE72 but enables binding of otherwise unbound podoviruses. S. epidermidis glycerolphosphate WTA was found to be modified with glucose in a tagE-dependent manner. TagE is encoded together with the enzymes PgcA and GtaB providing uridine diphosphate-activated glucose. ΦE72 transduced several other CoNS species encoding TagE homologs, suggesting that WTA glycosylation via TagE is a frequent trait among CoNS that permits interspecies horizontal gene transfer. Our study unravels a crucial mechanism of phage-Staphylococcus interaction and horizontal gene transfer, and it will help in the design of anti-staphylococcal phage therapies.IMPORTANCEPhages are highly specific for certain bacterial hosts, and some can transduce DNA even across species boundaries. How phages recognize cognate host cells remains incompletely understood. Phages infecting members of the genus Staphylococcus bind to wall teichoic acid (WTA) glycopolymers with highly variable structures and glycosylation patterns. How WTA is glycosylated in the opportunistic pathogen Staphylococcus epidermidis and in other coagulase-negative staphylococci (CoNS) species has remained unknown. We describe that S. epidermidis glycosylates its WTA backbone with glucose, and we identify a cluster of three genes responsible for glucose activation and transfer to WTA. Their inactivation strongly alters phage susceptibility patterns, yielding resistance to siphoviruses but susceptibility to podoviruses. Many different CoNS species with related glycosylation genes can exchange DNA via siphovirus ΦE72, suggesting that glucose-modified WTA is crucial for interspecies horizontal gene transfer. Our finding will help to develop antibacterial phage therapies and unravel routes of genetic exchange.
Collapse
Affiliation(s)
- Christian Beck
- Cluster of Excellence “Controlling Microbes to Fight Infections (CMFI)”, University of Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Infection Biology, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Janes Krusche
- Cluster of Excellence “Controlling Microbes to Fight Infections (CMFI)”, University of Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Infection Biology, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Anna Notaro
- Department of Agricultural Sciences, University of Naples, Naples, Italy
| | - Axel Walter
- Cluster of Excellence “Controlling Microbes to Fight Infections (CMFI)”, University of Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Organismic Interactions/Glycobiology, University of Tübingen, Tübingen, Germany
| | - Lara Kränkel
- Cluster of Excellence “Controlling Microbes to Fight Infections (CMFI)”, University of Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Infection Biology, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Anneli Vollert
- Electron-Microscopy, Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Regine Stemmler
- Cluster of Excellence “Controlling Microbes to Fight Infections (CMFI)”, University of Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Infection Biology, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Johannes Wittmann
- Leibniz Institute, DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Martin Schaller
- Electron-Microscopy, Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Slavetinsky
- Cluster of Excellence “Controlling Microbes to Fight Infections (CMFI)”, University of Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Infection Biology, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
- Pediatric Surgery and Urology, University Children's Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Christoph Mayer
- Cluster of Excellence “Controlling Microbes to Fight Infections (CMFI)”, University of Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Organismic Interactions/Glycobiology, University of Tübingen, Tübingen, Germany
| | | | - Andreas Peschel
- Cluster of Excellence “Controlling Microbes to Fight Infections (CMFI)”, University of Tübingen, Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Infection Biology, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| |
Collapse
|
10
|
Dombach JL, Christensen GL, Allgood SC, Quintana JLJ, Detweiler CS. Inhibition of multiple staphylococcal growth states by a small molecule that disrupts membrane fluidity and voltage. mSphere 2024; 9:e0077223. [PMID: 38445864 PMCID: PMC10964410 DOI: 10.1128/msphere.00772-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/26/2024] [Indexed: 03/07/2024] Open
Abstract
New molecular approaches to disrupting bacterial infections are needed. The bacterial cell membrane is an essential structure with diverse potential lipid and protein targets for antimicrobials. While rapid lysis of the bacterial cell membrane kills bacteria, lytic compounds are generally toxic to whole animals. In contrast, compounds that subtly damage the bacterial cell membrane could disable a microbe, facilitating pathogen clearance by the immune system with limited compound toxicity. A previously described small molecule, D66, terminates Salmonella enterica serotype Typhimurium (S. Typhimurium) infection of macrophages and reduces tissue colonization in mice. The compound dissipates bacterial inner membrane voltage without rapid cell lysis under broth conditions that permeabilize the outer membrane or disable efflux pumps. In standard media, the cell envelope protects Gram-negative bacteria from D66. We evaluated the activity of D66 in Gram-positive bacteria because their distinct envelope structure, specifically the absence of an outer membrane, could facilitate mechanism of action studies. We observed that D66 inhibited Gram-positive bacterial cell growth, rapidly increased Staphylococcus aureus membrane fluidity, and disrupted membrane voltage while barrier function remained intact. The compound also prevented planktonic staphylococcus from forming biofilms and a disturbed three-dimensional structure in 1-day-old biofilms. D66 furthermore reduced the survival of staphylococcal persister cells and of intracellular S. aureus. These data indicate that staphylococcal cells in multiple growth states germane to infection are susceptible to changes in lipid packing and membrane conductivity. Thus, agents that subtly damage bacterial cell membranes could have utility in preventing or treating disease.IMPORTANCEAn underutilized potential antibacterial target is the cell membrane, which supports or associates with approximately half of bacterial proteins and has a phospholipid makeup distinct from mammalian cell membranes. Previously, an experimental small molecule, D66, was shown to subtly damage Gram-negative bacterial cell membranes and to disrupt infection of mammalian cells. Here, we show that D66 increases the fluidity of Gram-positive bacterial cell membranes, dissipates membrane voltage, and inhibits the human pathogen Staphylococcus aureus in several infection-relevant growth states. Thus, compounds that cause membrane damage without lysing cells could be useful for mitigating infections caused by S. aureus.
Collapse
Affiliation(s)
- Jamie L. Dombach
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Grace L. Christensen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Samual C. Allgood
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Joaquin L. J. Quintana
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Corrella S. Detweiler
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
11
|
Chick HM, Rees ME, Lewis ML, Williams LK, Bodger O, Harris LG, Rushton S, Wilkinson TS. Using the Traditional Ex Vivo Whole Blood Model to Discriminate Bacteria by Their Inducible Host Responses. Biomedicines 2024; 12:724. [PMID: 38672079 PMCID: PMC11047930 DOI: 10.3390/biomedicines12040724] [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: 02/28/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Whole blood models are rapid and versatile for determining immune responses to inflammatory and infectious stimuli, but they have not been used for bacterial discrimination. Staphylococcus aureus, S. epidermidis and Escherichia coli are the most common causes of invasive disease, and rapid testing strategies utilising host responses remain elusive. Currently, immune responses can only discriminate between bacterial 'domains' (fungi, bacteria and viruses), and very few studies can use immune responses to discriminate bacteria at the species and strain level. Here, whole blood was used to investigate the relationship between host responses and bacterial strains. Results confirmed unique temporal profiles for the 10 parameters studied: IL-6, MIP-1α, MIP-3α, IL-10, resistin, phagocytosis, S100A8, S100A8/A9, C5a and TF3. Pairwise analysis confirmed that IL-6, resistin, phagocytosis, C5a and S100A8/A9 could be used in a discrimination scheme to identify to the strain level. Linear discriminant analysis (LDA) confirmed that (i) IL-6, MIP-3α and TF3 could predict genera with 95% accuracy; (ii) IL-6, phagocytosis, resistin and TF3 could predict species at 90% accuracy and (iii) phagocytosis, S100A8 and IL-10 predicted strain at 40% accuracy. These data are important because they confirm the proof of concept that host biomarker panels could be used to identify bacterial pathogens.
Collapse
Affiliation(s)
- Heather M. Chick
- Microbiology and Infectious Disease, Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK; (H.M.C.); (M.E.R.); (M.L.L.); (L.K.W.); (L.G.H.)
| | - Megan E. Rees
- Microbiology and Infectious Disease, Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK; (H.M.C.); (M.E.R.); (M.L.L.); (L.K.W.); (L.G.H.)
| | - Matthew L. Lewis
- Microbiology and Infectious Disease, Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK; (H.M.C.); (M.E.R.); (M.L.L.); (L.K.W.); (L.G.H.)
| | - Lisa K. Williams
- Microbiology and Infectious Disease, Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK; (H.M.C.); (M.E.R.); (M.L.L.); (L.K.W.); (L.G.H.)
- Department of Animal and Agriculture, Hartpury University, Hartpury, Gloucestershire GL19 3BE, UK
| | - Owen Bodger
- Patient and Population Health an Informatics Research, Swansea University Medical School, Swansea SA2 8PP, UK;
| | - Llinos G. Harris
- Microbiology and Infectious Disease, Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK; (H.M.C.); (M.E.R.); (M.L.L.); (L.K.W.); (L.G.H.)
| | - Steven Rushton
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Thomas S. Wilkinson
- Microbiology and Infectious Disease, Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK; (H.M.C.); (M.E.R.); (M.L.L.); (L.K.W.); (L.G.H.)
| |
Collapse
|
12
|
Chen Q, Ye J, Li XB, Zeng K, Zeng S. Drug resistance and influencing factors of biofilm bacteria in upper urinary calculi patients with double J stent indwelling. BMC Urol 2023; 23:165. [PMID: 37838674 PMCID: PMC10576326 DOI: 10.1186/s12894-023-01339-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023] Open
Abstract
OBJECTIVE To analyze the distribution and drug resistance of biofilm bacteria infected with upper urinary calculi patients with double J stent indwelling, and to explore the influencing factors of Biofilm Bacteria Infections. METHODS A total of 400 patients with upper urinary calculi who adopted double J stent inserting in our hospital from January 2019 to January 2022 were included. Urine and double J stent samples were collected, pathogen cultures were performed, and then drug sensitivity test analysis was carried out for isolates. Univariate and multivariate logistic regression analyzes were used to analyze the influencing factors of patients with double J stent associated biofilm bacteria infections. RESULTS A total of 13 strains (3.2%) of biofilm bacteria were detected in urine samples and 168 strains (42%) in double J stent samples (P < 0.05), 95 strains (23.7%) of pathogenic bacteria were separated from urine samples and 117 strains (29.2%) from double J-stent samples (P > 0.05). Escherichia coli were the most common bacteria. There was significantly higher drug resistance observed in biofilm bacteria versus urine-cultured pathogens (P < 0.05). Advanced age, long-term catheterization, inadequate water intake, hypoproteinemia, abnormal renal function, and diabetes mellitus were independent risk factors for biofilm bacteria infection associated with double J stent(P < 0.05). CONCLUSION Among the upper urinary calculi patients with double J stent indwelling, the positive rate and drug resistance of biofilm bacteria obtained from double J stent were significantly higher than that from urine. More attention should be paid to the factors that influence biofilm bacteria infections.
Collapse
Affiliation(s)
- Qian Chen
- Department of Nephrology, First People's Hospital of Zigong City, Zigong, China
| | - JunBing Ye
- Department of Urology, First People's Hospital of Zigong City, Zigong, China.
| | - Xiao Bin Li
- Department of Urology, First People's Hospital of Zigong City, Zigong, China
| | - Ke Zeng
- Department of Urology, First People's Hospital of Zigong City, Zigong, China
| | - Shiping Zeng
- Department of Urology, First People's Hospital of Zigong City, Zigong, China
| |
Collapse
|
13
|
Afshar M, Møllebjerg A, Minero GA, Hollensteiner J, Poehlein A, Himmelbach A, Lange J, Meyer RL, Brüggemann H. Biofilm formation and inflammatory potential of Staphylococcus saccharolyticus: A possible cause of orthopedic implant-associated infections. Front Microbiol 2022; 13:1070201. [DOI: 10.3389/fmicb.2022.1070201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/09/2022] [Indexed: 11/29/2022] Open
Abstract
Staphylococcus saccharolyticus, a coagulase-negative staphylococcal species, has some unusual characteristics for human-associated staphylococci, such as slow growth and its preference for anoxic culture conditions. This species is a relatively abundant member of the human skin microbiota, but its microbiological properties, as well as the pathogenic potential, have scarcely been investigated so far, despite being occasionally isolated from different types of infections including orthopedic implant-associated infections. Here, we investigated the growth and biofilm properties of clinical isolates of S. saccharolyticus and determined host cell responses. Growth assessments in anoxic and oxic conditions revealed strain-dependent outcomes, as some strains can also grow aerobically. All tested strains of S. saccharolyticus were able to form biofilm in a microtiter plate assay. Strain-dependent differences were determined by optical coherence tomography, revealing that medium supplementation with glucose and sodium chloride enhanced biofilm formation. Visualization of the biofilm by confocal laser scanning microscopy revealed the role of extracellular DNA in the biofilm structure. In addition to attached biofilms, S. saccharolyticus also formed bacterial aggregates at an early stage of growth. Transcriptome analysis of biofilm-grown versus planktonic cells revealed a set of upregulated genes in biofilm-embedded cells, including factors involved in adhesion, colonization, and competition such as epidermin, type I toxin-antitoxin system, and phenol-soluble modulins (beta and epsilon). To investigate consequences for the host after encountering S. saccharolyticus, cytokine profiling and host cell viability were assessed by infection experiments with differentiated THP-1 cells. The microorganism strongly triggered the secretion of the tested pro-inflammatory cyto- and chemokines IL-6, IL-8, and TNF-alpha, determined at 24 h post-infection. S. saccharolyticus was less cytotoxic than Staphylococcus aureus. Taken together, the results indicate that S. saccharolyticus has substantial pathogenic potential. Thus, it can be a potential cause of orthopedic implant-associated infections and other types of deep-seated infections.
Collapse
|
14
|
Brás S, França A. Transcriptome Mining to Identify Molecular Markers for the Diagnosis of Staphylococcus epidermidis Bloodstream Infections. Antibiotics (Basel) 2022; 11:1596. [PMID: 36421239 PMCID: PMC9687011 DOI: 10.3390/antibiotics11111596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/08/2022] [Accepted: 11/09/2022] [Indexed: 12/01/2023] Open
Abstract
Bloodstream infections caused by Staphylococcus epidermidis are often misdiagnosed since no diagnostic marker found so far can unequivocally discriminate "true" infection from sample contamination. While attempts have been made to find genomic and/or phenotypic differences between invasive and commensal isolates, possible changes in the transcriptome of these isolates under in vivo-mimicking conditions have not been investigated. Herein, we characterized the transcriptome, by RNA sequencing, of three clinical and three commensal isolates after 2 h of exposure to whole human blood. Bioinformatics analysis was used to rank the genes with the highest potential to distinguish invasive from commensal isolates and among the ten genes identified as candidates, the gene SERP2441 showed the highest potential. A collection of 56 clinical and commensal isolates was then used to validate, by quantitative PCR, the discriminative power of the selected genes. A significant variation was observed among isolates, and the discriminative power of the selected genes was lost, undermining their potential use as markers. Nevertheless, future studies should include an RNA sequencing characterization of a larger collection of isolates, as well as a wider range of conditions to increase the chances of finding further candidate markers for the diagnosis of bloodstream infections caused by S. epidermidis.
Collapse
Affiliation(s)
- Susana Brás
- LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Angela França
- LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- LABBELS–Associate Laboratory, Braga and Guimarães, Portugal
| |
Collapse
|
15
|
Zeng K, Huang J, Li X, Ye J, Yang J, Zhang S. Distribution and Drug Resistance of Pathogenic Bacteria in Diabetic Patients with Double J-Stent Associated Infections. Infect Drug Resist 2022; 15:2671-2678. [PMID: 35652084 PMCID: PMC9148917 DOI: 10.2147/idr.s360086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/29/2022] [Indexed: 11/29/2022] Open
Abstract
Objective To analyze the distribution and drug resistance of pathogenic bacteria in diabetic patients with double J-stent associated infections, and to explore the strategies for prevention and treatment of the infections. Methods From January 2019 to December 2021, 266 diabetic patients treated with double J-stent placement in our hospital assessed for eligibility were recruited. Urine and double J-stent samples were collected for pathogenicity assay and screened for biofilm bacteria. Pathogenic bacteria distribution and drug resistance were examined. Results A total of 97 strains (36.5%) of pathogenic bacteria were isolated from urine samples and 129 strains (48.5%) from double J-stent samples (P > 0.05). 3 strains (1.1%) of biofilm bacteria were separated from urine samples and 106 strains (39.8%) from double J-stent samples (P < 0.05). In the double J-stent samples, there were significantly higher ratios of Gram-positive bacteria separated from biofilm bacteria versus the urine-cultured pathogens (44.3%/61.3%, P < 0.05), and higher drug resistance was observed in biofilm bacteria versus urine-cultured pathogens (P < 0.05). Fosfomycin tromethamine showed remarkable susceptibility to both urinary cultured pathogens and double J-stent biofilm bacteria. Conclusion Diabetic patients with double J-stent biofilm-positive bacteria are mainly Gram-positive bacteria, which are prone to biofilm formation and show strong drug resistance.
Collapse
Affiliation(s)
- Ke Zeng
- Department of Basic Medicine, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, People’s Republic of China
| | - Jun Huang
- Department of Urology, First People’s Hospital of Zigong City, Zigong, Sichuan, People’s Republic of China
| | - Xiaobin Li
- Department of Urology, First People’s Hospital of Zigong City, Zigong, Sichuan, People’s Republic of China
| | - Junbing Ye
- Department of Urology, First People’s Hospital of Zigong City, Zigong, Sichuan, People’s Republic of China
| | - Jian Yang
- Department of Urology, First People’s Hospital of Zigong City, Zigong, Sichuan, People’s Republic of China
| | - Suchuan Zhang
- Laboratory Department, First People’s Hospital of Zigong City, Zigong, Sichuan, People’s Republic of China
| |
Collapse
|
16
|
Kaur H, Kaur A, Soni SK, Rishi P. Microbially-derived cocktail of carbohydrases as an anti-biofouling agents: a 'green approach'. BIOFOULING 2022; 38:455-481. [PMID: 35673761 DOI: 10.1080/08927014.2022.2085566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/12/2022] [Accepted: 05/30/2022] [Indexed: 06/15/2023]
Abstract
Enzymes, also known as biocatalysts, display vital properties like high substrate specificity, an eco-friendly nature, low energy inputs, and cost-effectiveness. Among their numerous known applications, enzymes that can target biofilms or their components are increasingly being investigated for their anti-biofouling action, particularly in healthcare, food manufacturing units and environmental applications. Enzymes can target biofilms at different levels like during the attachment of microorganisms, formation of exopolymeric substances (EPS), and their disruption thereafter. In this regard, a consortium of carbohydrases that can target heterogeneous polysaccharides present in the EPS matrix may provide an effective alternative to conventional chemical anti-biofouling methods. Further, for complete annihilation of biofilms, enzymes can be used alone or in conjunction with other antimicrobial agents. Enzymes hold the promise to replace the conventional methods with greener, more economical, and more efficient alternatives. The present article explores the potential and future perspectives of using carbohydrases as effective anti-biofilm agents.
Collapse
Affiliation(s)
- Harmanpreet Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Arashdeep Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | | | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
17
|
Gaio V, Lima T, Vilanova M, Cerca N, França A. mazEF Homologue Has a Minor Role in Staphylococcus epidermidis 1457 Virulence Potential. Front Cell Infect Microbiol 2022; 11:803134. [PMID: 35096651 PMCID: PMC8792614 DOI: 10.3389/fcimb.2021.803134] [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: 10/27/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcus epidermidis biofilm cells are characterized by increased antimicrobial tolerance and improved ability to evade host immune system defenses. These features are, in part, due to the presence of viable but non-culturable (VBNC) cells. A previous study identified genes potentially involved in VBNC cells formation in S. epidermidis biofilms, among which SERP1682/1681 raised special interest due to their putative role as a toxin–antitoxin system of the mazEF family. Herein, we constructed an S. epidermidis mutant lacking the mazEF genes homologues and determined their role in (i) VBNC state induction during biofilm formation, (ii) antimicrobial susceptibility, (iii) survival in human blood and plasma, and (iv) activation of immune cells. Our results revealed that mazEF homologue did not affect the proportion of VBNC cells in S. epidermidis 1457, refuting the previous hypothesis that mazEF homologue could be linked with the emergence of VBNC cells in S. epidermidis biofilms. Additionally, mazEF homologue did not seem to influence key virulence factors on this strain, since its deletion did not significantly affect the mutant biofilm formation capacity, antimicrobial tolerance or the response by immune cells. Surprisingly, our data suggest that mazEF does not behave as a toxin–antitoxin system in S. epidermidis strain 1457, since no decrease in the viability and culturability of bacteria was found when only the mazF toxin homologue was being expressed.
Collapse
Affiliation(s)
- Vânia Gaio
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Tânia Lima
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Manuel Vilanova
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Nuno Cerca
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Angela França
- Laboratory of Research in Biofilms Rosário Oliveira, Centre of Biological Engineering, University of Minho, Braga, Portugal
| |
Collapse
|
18
|
The Transcription Factor SpoVG Is of Major Importance for Biofilm Formation of Staphylococcus epidermidis under In Vitro Conditions, but Dispensable for In Vivo Biofilm Formation. Int J Mol Sci 2022; 23:ijms23063255. [PMID: 35328675 PMCID: PMC8949118 DOI: 10.3390/ijms23063255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus epidermidis is a common cause of device related infections on which pathogens form biofilms (i.e., multilayered cell populations embedded in an extracellular matrix). Here, we report that the transcription factor SpoVG is essential for the capacity of S. epidermidis to form such biofilms on artificial surfaces under in vitro conditions. Inactivation of spoVG in the polysaccharide intercellular adhesin (PIA) producing S. epidermidis strain 1457 yielded a mutant that, unlike its parental strain, failed to produce a clear biofilm in a microtiter plate-based static biofilm assay. A decreased biofilm formation capacity was also observed when 1457 ΔspoVG cells were co-cultured with polyurethane-based peripheral venous catheter fragments under dynamic conditions, while the cis-complemented 1457 ΔspoVG::spoVG derivative formed biofilms comparable to the levels seen with the wild-type. Transcriptional studies demonstrated that the deletion of spoVG significantly altered the expression of the intercellular adhesion (ica) locus by upregulating the transcription of the ica operon repressor icaR and down-regulating the transcription of icaADBC. Electrophoretic mobility shift assays (EMSA) revealed an interaction between SpoVG and the icaA-icaR intergenic region, suggesting SpoVG to promote biofilm formation of S. epidermidis by modulating ica expression. However, when mice were challenged with the 1457 ΔspoVG mutant in a foreign body infection model, only marginal differences in biomasses produced on the infected catheter fragments between the mutant and the parental strain were observed. These findings suggest that SpoVG is critical for the PIA-dependent biofilm formation of S. epidermis under in vitro conditions, but is largely dispensable for biofilm formation of this skin commensal under in vivo conditions.
Collapse
|
19
|
Look Who's Talking: Host and Pathogen Drivers of Staphylococcus epidermidis Virulence in Neonatal Sepsis. Int J Mol Sci 2022; 23:ijms23020860. [PMID: 35055041 PMCID: PMC8775791 DOI: 10.3390/ijms23020860] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
Preterm infants are at increased risk for invasive neonatal bacterial infections. S. epidermidis, a ubiquitous skin commensal, is a major cause of late-onset neonatal sepsis, particularly in high-resource settings. The vulnerability of preterm infants to serious bacterial infections is commonly attributed to their distinct and developing immune system. While developmentally immature immune defences play a large role in facilitating bacterial invasion, this fails to explain why only a subset of infants develop infections with low-virulence organisms when exposed to similar risk factors in the neonatal ICU. Experimental research has explored potential virulence mechanisms contributing to the pathogenic shift of commensal S. epidermidis strains. Furthermore, comparative genomics studies have yielded insights into the emergence and spread of nosocomial S. epidermidis strains, and their genetic and functional characteristics implicated in invasive disease in neonates. These studies have highlighted the multifactorial nature of S. epidermidis traits relating to pathogenicity and commensalism. In this review, we discuss the known host and pathogen drivers of S. epidermidis virulence in neonatal sepsis and provide future perspectives to close the gap in our understanding of S. epidermidis as a cause of neonatal morbidity and mortality.
Collapse
|
20
|
Gaio V, Lopes N, Cerca N, França A. codY and pdhA Expression Is Induced in Staphylococcus epidermidis Biofilm and Planktonic Populations With Higher Proportions of Viable but Non-Culturable Cells. Front Cell Infect Microbiol 2021; 11:771666. [PMID: 34869073 PMCID: PMC8634161 DOI: 10.3389/fcimb.2021.771666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/27/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus epidermidis biofilm cells can enter a physiological state known as viable but non-culturable (VBNC), where, despite being alive, they do not grow in conventional laboratory media. As such, the presence of VBNC cells impacts the diagnosis of S. epidermidis biofilm-associated infections. Previous transcriptomics analysis of S. epidermidis strain 9142 biofilms with higher proportions of VBNC cells suggested that the genes pdhA, codY and mazEF could be involved in the induction of the VBNC state. However, it was previously demonstrated that VBNC induction is strain-dependent. To properly assess the role of these genes in VBNC induction, the construction of mutant strains is necessary. Thus, herein, we assessed if VBNC cells could be induced in strain 1457, a strain amenable to genetic manipulation, and if the previously identified genes were involved in the modulation of the VBNC state in this strain. Furthermore, we evaluated the formation of VBNC cells on planktonic cultures. Our results showed that despite being commonly associated with biofilms, the proportion of VBNC cells can be modulated in both biofilm and planktonic cultures and that the expression of codY and pdhA was upregulated under VBNC inducing conditions in both phenotypes. Overall, our study revealed that the formation of VBNC cells in S. epidermidis is independent of the mode of growth and that the genes codY and pdhA seem to be relevant for the regulation of this physiological condition.
Collapse
Affiliation(s)
- Vânia Gaio
- Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Nathalie Lopes
- Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Nuno Cerca
- Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Angela França
- Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| |
Collapse
|
21
|
Glycan-Dependent Corneocyte Adherence of Staphylococcus epidermidis Mediated by the Lectin Subdomain of Aap. mBio 2021; 12:e0290820. [PMID: 34253065 PMCID: PMC8406310 DOI: 10.1128/mbio.02908-20] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Staphylococcus epidermidis and other coagulase-negative staphylococci (CoNS) that colonize skin are known to promote skin immunity and inhibit colonization of pathogens that cause skin and soft tissue infections, including Staphylococcus aureus. However, S. epidermidis adherence to corneocytes, the cells that constitute the uppermost layer of the skin epidermis, remains poorly understood. Our study documents that S. epidermidis corneocyte adherence is dependent upon the accumulation-associated protein (Aap). Aap is composed of two distinct A and B domains. The A domain is comprised of a repeat region and a conserved L-type lectin domain, whereas the fibrillar B domain, which is comprised of G5 and E repeats, is linked to the cell wall in a sortase-dependent manner. Our studies revealed that adherence to corneocytes is dependent upon the lectin subdomain within the A domain. However, significant adherence was only observed when the lectin domain was expressed with both the A repeat and the B domain, suggesting further interactions between these three domains. Our data also suggest that the A repeat domain is important for stability or expression of Aap. Deglycosylation treatment suggested that glycans expressed in the host stratum corneum serve as potential binding partners for Aap-mediated corneocyte adherence. Last, bioinformatic analyses of the predominant commensal species of CoNS identified open reading frames (ORFs) homologous to aap, thus suggesting that Aap orthologues containing lectin-like domains may provide the basis for staphylococcal colonization of skin. Corroborating these observations, adherence to corneocytes in an S. aureus mgrA mutant was dependent upon SasG, the Aap orthologue in S. aureus. IMPORTANCE Staphylococcus aureus is the most significant cause of skin and soft tissue infections yet it rarely colonizes the skin of healthy individuals. This is believed to be due, in part, to inhibition of colonization via toxic substances produced by normal skin flora, including by S. epidermidis. Furthermore, we surmise that S. aureus colonization inhibition may also be due to competition for binding sites on host corneocytes. To understand these potential interactions between S. aureus and S. epidermidis and, potentially, other coagulase-negative staphylococci, we must first understand how staphylococci adhere to corneocytes. This work documents that S. epidermidis adherence to corneocytes is dependent upon the fibrillar cell wall-associated protein Aap. Our work further documents that Aap binds to glycans exposed on the corneocyte surface, which are commonly exploited by bacteria to facilitate adherence to host cells. Furthermore, we find that Aap orthologues may be responsible for corneocyte adherence in other staphylococci, including in S. aureus.
Collapse
|
22
|
Frequency of cbrA, cbrB, ndvB, and phoBR Genes in Relation to Biofilm Formation in Pseudomonas aeruginosa Clinical Isolates. JOURNAL OF MEDICAL MICROBIOLOGY AND INFECTIOUS DISEASES 2021. [DOI: 10.52547/jommid.9.2.97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
23
|
Staphylococcus epidermidis clones express Staphylococcus aureus-type wall teichoic acid to shift from a commensal to pathogen lifestyle. Nat Microbiol 2021; 6:757-768. [PMID: 34031577 DOI: 10.1038/s41564-021-00913-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
Most clonal lineages of Staphylococcus epidermidis are commensals present on human skin and in the nose. However, some globally spreading healthcare-associated and methicillin-resistant S. epidermidis (HA-MRSE) clones are major causes of difficult-to-treat implant or bloodstream infections. The molecular determinants that alter the lifestyle of S. epidermidis have remained elusive, and their identification might provide therapeutic targets. We reasoned that changes in surface-exposed wall teichoic acid (WTA) polymers of S. epidermidis, which potentially shape host interactions, may be linked to differences between colonization and infection abilities of different clones. We used a combined epidemiological and functional approach to show that while commensal clones express poly-glycerolphosphate WTA, S. epidermidis multilocus sequence type 23, which emerged in the past 15 years and is one of the main infection-causing HA-MRSE clones, contains an accessory genetic element, tarIJLM, that leads to the production of a second, Staphylococcus aureus-type WTA (poly-ribitolphosphate (RboP)). Production of RboP-WTA by S. epidermidis impaired in vivo colonization but augmented endothelial attachment and host mortality in a mouse sepsis model. tarIJLM was absent from commensal human sequence types but was found in several other HA-MRSE clones. Moreover, RboP-WTA enabled S. epidermidis to exchange DNA with S. aureus via siphovirus bacteriophages, thereby creating a possible route for the inter-species exchange of methicillin resistance, virulence and colonization factors. We conclude that tarIJLM alters the lifestyle of S. epidermidis from commensal to pathogenic and propose that RboP-WTA might be a robust target for preventive and therapeutic interventions against MRSE infections.
Collapse
|
24
|
Mu Y, Zeng H, Chen W. Quercetin Inhibits Biofilm Formation by Decreasing the Production of EPS and Altering the Composition of EPS in Staphylococcus epidermidis. Front Microbiol 2021; 12:631058. [PMID: 33763049 PMCID: PMC7982815 DOI: 10.3389/fmicb.2021.631058] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/26/2021] [Indexed: 11/25/2022] Open
Abstract
Staphylococcus epidermidis is an opportunistic pathogen, and its biofilm formation ability is an important virulent factor. Quercetin, a typical flavonoid ubiquitously used in dietary supplementation, is known for its antioxidant property, but its anti-biofilm activity against S. epidermidis remains unknown. In this study, the anti-biofilm activity of quercetin was investigated using S. epidermidis ATCC35984, a strong biofilm-positive strain. An attempt was made to disclose the mechanisms of the anti-biofilm activity of quercetin. S. epidermidis exhibited a less cell surface hydrophobicity after quercetin treatment. Also, quercetin effectively inhibited S. epidermidis cells from adhering to the glass slides. Quercetin downregulated the intercellular adhesion (ica) locus and then polysaccharide intercellular adhesin (PIA) production was reduced. Therefore, S. epidermidis cells became less hydrophobic, which supported quercetin’s anti-biofilm effect. Our study suggests that quercetin from plants be given further attention as a potential anti-biofilm agent against the biofilm formation of S. epidermidis, even biofilm infections of other bacteria.
Collapse
Affiliation(s)
- Yongqi Mu
- Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, China.,College of Life Sciences, Tarim University, Alar, China
| | - Hong Zeng
- Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, China.,College of Life Sciences, Tarim University, Alar, China
| | - Wei Chen
- Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, China.,College of Life Sciences, Tarim University, Alar, China.,College of Animal Sciences Tarim University, Alar, China.,Key Laboratory of Tarim Animal Husbandry and Science Technology of Xinjiang Production and Construction Corps, Tarim University, Alar, China.,Engineering Laboratory for Tarim Animal Diseases Diagnosis and Control of Xinjiang Production and Construction Corps, Tarim University, Alar, China
| |
Collapse
|
25
|
Hendriks A, Mnich ME, Clemente B, Cruz AR, Tavarini S, Bagnoli F, Soldaini E. Staphylococcus aureus-Specific Tissue-Resident Memory CD4 + T Cells Are Abundant in Healthy Human Skin. Front Immunol 2021; 12:642711. [PMID: 33796109 PMCID: PMC8008074 DOI: 10.3389/fimmu.2021.642711] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/18/2021] [Indexed: 12/12/2022] Open
Abstract
The skin is an immunocompetent tissue that harbors several kinds of immune cells and a plethora of commensal microbes constituting the skin microbiome. Staphylococcus aureus is a prominent skin pathogen that colonizes a large proportion of the human population. We currently have an incomplete understanding of the correlates of protection against S. aureus infection, however genetic and experimental evidence has shown that CD4+ T cells play a key role in orchestrating a protective anti-S. aureus immune response. A high S. aureus-specific memory CD4+ T cell response has been reported in the blood of healthy subjects. Since T cells are more abundant in the skin than in blood, we hypothesized that S. aureus-specific CD4+ T cells could be present in the skin of healthy individuals. Indeed, we observed proliferation of tissue-resident memory CD4+ T cells and production of IL-17A, IL-22, IFN-γ and TNF-β by cells isolated from abdominal skin explants in response to heat-killed S. aureus. Remarkably, these cytokines were produced also during an ex vivo epicutaneous S. aureus infection of human skin explants. These findings highlight the importance of tissue-resident memory CD4+ T cells present at barrier sites such as the skin, a primary entry site for S. aureus. Further phenotypical and functional characterization of these cells will ultimately aid in the development of novel vaccine strategies against this elusive pathogen.
Collapse
Affiliation(s)
- Astrid Hendriks
- GSK, Siena, Italy.,Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Malgorzata Ewa Mnich
- GSK, Siena, Italy.,Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | - Ana Rita Cruz
- GSK, Siena, Italy.,Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | | | | |
Collapse
|
26
|
Rifampin, Rifapentine, and Rifabutin Are Active against Intracellular Periprosthetic Joint Infection-Associated Staphylococcus epidermidis. Antimicrob Agents Chemother 2021; 65:AAC.01275-20. [PMID: 33199387 DOI: 10.1128/aac.01275-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/10/2020] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus epidermidis is a major cause of periprosthetic joint infection (PJI); its intracellular persistence within osteoblasts may compromise therapy if that therapy is not intracellularly active. The intracellular activity of rifampin, rifapentine, and rifabutin was assessed against five rifampin-susceptible and two rifampin-resistant S. epidermidis isolates. Compared to no treatment, treatment resulted in a ≥2-fold log10 reduction of intracellular rifampin-susceptible, but not rifampin-resistant, S. epidermidis These findings show activity of rifampin, rifapentine, and rifabutin against intraosteoblast PJI-associated S. epidermidis.
Collapse
|
27
|
Breslawec AP, Wang S, Li C, Poulin MB. Anionic amino acids support hydrolysis of poly-β-(1,6)-N-acetylglucosamine exopolysaccharides by the biofilm dispersing glycosidase Dispersin B. J Biol Chem 2020; 296:100203. [PMID: 33334876 PMCID: PMC7949127 DOI: 10.1074/jbc.ra120.015524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
The exopolysaccharide poly-β-(1→6)-N-acetylglucosamine (PNAG) is a major structural determinant of bacterial biofilms responsible for persistent and nosocomial infections. The enzymatic dispersal of biofilms by PNAG-hydrolyzing glycosidase enzymes, such as Dispersin B (DspB), is a possible approach to treat biofilm-dependent bacterial infections. The cationic charge resulting from partial de-N-acetylation of native PNAG is critical for PNAG-dependent biofilm formation. We recently demonstrated that DspB has increased catalytic activity on de-N-acetylated PNAG oligosaccharides, but the molecular basis for this increased activity is not known. Here, we analyze the role of anionic amino acids surrounding the catalytic pocket of DspB in PNAG substrate recognition and hydrolysis using a combination of site-directed mutagenesis, activity measurements using synthetic PNAG oligosaccharide analogs, and in vitro biofilm dispersal assays. The results of these studies support a model in which bound PNAG is weakly associated with a shallow anionic groove on the DspB protein surface with recognition driven by interactions with the -1 GlcNAc residue in the catalytic pocket. An increased rate of hydrolysis for cationic PNAG was driven, in part, by interaction with D147 on the anionic surface. Moreover, we identified that a DspB mutant with improved hydrolysis of fully acetylated PNAG oligosaccharides correlates with improved in vitro dispersal of PNAG-dependent Staphylococcus epidermidis biofilms. These results provide insight into the mechanism of substrate recognition by DspB and suggest a method to improve DspB biofilm dispersal activity by mutation of the amino acids within the anionic binding surface.
Collapse
Affiliation(s)
- Alexandra P Breslawec
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Shaochi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Crystal Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Myles B Poulin
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA.
| |
Collapse
|
28
|
Nakamura K, O'Neill AM, Williams MR, Cau L, Nakatsuji T, Horswill AR, Gallo RL. Short chain fatty acids produced by Cutibacterium acnes inhibit biofilm formation by Staphylococcus epidermidis. Sci Rep 2020; 10:21237. [PMID: 33277548 PMCID: PMC7718897 DOI: 10.1038/s41598-020-77790-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Biofilm formation by bacterial pathogens is associated with numerous human diseases and can confer resistance to both antibiotics and host defenses. Many strains of Staphylococcus epidermidis are capable of forming biofilms and are important human pathogens. Since S. epidermidis coexists with abundant Cutibacteria acnes on healthy human skin and does not typically form a biofilm in this environment, we hypothesized that C. acnes may influence biofilm formation of S. epidermidis. Culture supernatants from C. acnes and other species of Cutibacteria inhibited S. epidermidis but did not inhibit biofilms by Pseudomonas aeruginosa or Bacillus subtilis, and inhibited biofilms by S. aureus to a lesser extent. Biofilm inhibitory activity exhibited chemical properties of short chain fatty acids known to be produced from C. acnes. The addition of the pure short chain fatty acids propionic, isobutyric or isovaleric acid to S. epidermidis inhibited biofilm formation and, similarly to C. acnes supernatant, reduced polysaccharide synthesis by S. epidermidis. Both short chain fatty acids and C. acnes culture supernatant also increased sensitivity of S. epidermidis to antibiotic killing under biofilm-forming conditions. These observations suggest the presence of C. acnes in a diverse microbial community with S. epidermidis can be beneficial to the host and demonstrates that short chain fatty acids may be useful to limit formation of a biofilm by S. epidermidis.
Collapse
Affiliation(s)
- Kouki Nakamura
- Department of Dermatology, University of California San Diego, 9500 Gillman Dr. #0869, La Jolla, CA, 92093, USA
| | - Alan M O'Neill
- Department of Dermatology, University of California San Diego, 9500 Gillman Dr. #0869, La Jolla, CA, 92093, USA
| | - Michael R Williams
- Department of Dermatology, University of California San Diego, 9500 Gillman Dr. #0869, La Jolla, CA, 92093, USA
| | - Laura Cau
- Department of Dermatology, University of California San Diego, 9500 Gillman Dr. #0869, La Jolla, CA, 92093, USA
- SILAB, R&D Department, Brive, France
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California San Diego, 9500 Gillman Dr. #0869, La Jolla, CA, 92093, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, USA
| | - Richard L Gallo
- Department of Dermatology, University of California San Diego, 9500 Gillman Dr. #0869, La Jolla, CA, 92093, USA.
| |
Collapse
|
29
|
Meyer KJ, Taylor HB, Seidel J, Gates MF, Lewis K. Pulse Dosing of Antibiotic Enhances Killing of a Staphylococcus aureus Biofilm. Front Microbiol 2020; 11:596227. [PMID: 33240251 PMCID: PMC7680849 DOI: 10.3389/fmicb.2020.596227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022] Open
Abstract
Biofilms are highly tolerant to antibiotics and underlie the recalcitrance of many chronic infections. We demonstrate that mature Staphylococcus aureus biofilms can be substantially sensitized to the treatment by pulse dosing of an antibiotic – in this case, oxacillin. Pulse (periodic) dosing was compared to continuous application of antibiotic and was studied in a novel in vitro flow system which allowed for robust biofilm growth and tractable pharmacokinetics of dosing regimens. Our results highlight that a subpopulation of the biofilm survives antibiotic without becoming resistant, a population we refer to as persister bacteria. When oxacillin was continuously present the persister level did not decline, but, importantly, providing correctly timed periodic breaks decreased the surviving population. We found that the length of the periodic break impacted efficacy, and there was an optimal length that sensitized the biofilm to repeat treatment without allowing resistance expansion. Periodic dosing provides a potential simple solution to a complicated problem.
Collapse
Affiliation(s)
- Kirsten J Meyer
- Department of Biology, Antimicrobial Discovery Center, Northeastern University, Boston, MA, United States
| | - Hannah B Taylor
- Department of Biology, Antimicrobial Discovery Center, Northeastern University, Boston, MA, United States
| | - Jazlyn Seidel
- Department of Biology, Antimicrobial Discovery Center, Northeastern University, Boston, MA, United States
| | - Michael F Gates
- Department of Biology, Antimicrobial Discovery Center, Northeastern University, Boston, MA, United States
| | - Kim Lewis
- Department of Biology, Antimicrobial Discovery Center, Northeastern University, Boston, MA, United States
| |
Collapse
|
30
|
A Giant Extracellular Matrix Binding Protein of Staphylococcus epidermidis Binds Surface-Immobilized Fibronectin via a Novel Mechanism. mBio 2020; 11:mBio.01612-20. [PMID: 33082256 PMCID: PMC7587433 DOI: 10.1128/mbio.01612-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Although it is normally an innocuous part of the human skin microbiota, Staphylococcus epidermidis has emerged as a major nosocomial pathogen, and implanted foreign materials are an essential risk factor for the development of an infection. The extraordinary efficiency of S. epidermidis to colonize artificial surfaces is particularly related to the ability to form biofilms. Biofilm formation itself critically depends on stable pathogen binding to extracellular host matrix components, e.g. fibronectin (Fn), covering inserted devices in vast amounts. Extracellular matrix binding protein (Embp) and its subdomains referred to as the F-repeat and the FG-repeat are critical for adherence of S. epidermidis to surface-immobilized Fn. Embp-Fn interactions preferentially occur with surface-bound, but not folded, globular Fn via binding to the F3 domain. High-resolution structure analysis of F- and FG-repeats revealed that both repeats are composed of two tightly connected triple α-helix bundles, exhibiting an elongated but rather rigid structural organization in solution. Both F- and FG-repeat possess Fn-binding capacity via interactions with type III subdomain FN12, involving residues within the C and F β-sheet. FN12 essentially supports stability of the globular Fn state, and thus these findings reasonably explain why Embp-mediated interaction of S. epidermidis necessitates Fn surface immobilization. Thus, Embp employs an uncharacterized bacterial Fn-binding mechanism to promote staphylococcal adherence.IMPORTANCE Staphylococcus epidermidis is a leading pathogen in implant-associated hospital infections. The pathogenesis critically depends on bacterial binding to ECM components, specifically fibronectin (Fn). The cell surface-localized, 1-MDa extracellular matrix binding protein (Embp) is essentially characterized by 10 F- and 40 FG-repeats. These repetitive units, each characterized by two α-helical bundles, organize themselves in a rigid, elongated form. Embp binds preferentially to surface-localized but not soluble Fn, with both F- and FG-repeats being sufficient for Fn binding and resulting bacterial adherence. Binding preferentially involves Fn type III domain, specifically residues of FN12 β-sheets C and F. Both play key role in stabilizing the globular Fn conformation, explaining the necessity of Fn surface immobilization for a subsequent interaction with Embp. In comparison to many other bacterial Fn-binding proteins using the Fn N terminus, Embp employs a previously undescribed mechanism supporting the adhesion of S. epidermidis to surface-immobilized Fn.
Collapse
|
31
|
Zhao Z, Lin Q, Feng Y, Zhou Y, Wang X. Determination of monosaccharides hydrolyzed from polysaccharides in activated sludge by ion chromatography-mass spectrometry with online pretreatment of column switching technique. Anal Bioanal Chem 2020; 412:8061-8071. [PMID: 32975656 DOI: 10.1007/s00216-020-02955-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/28/2020] [Accepted: 09/14/2020] [Indexed: 10/23/2022]
Abstract
The simultaneous determination of monosaccharides present in the activated sludge would be crucial to understand the water treatment mechanism. Herein, an ion chromatography-mass spectrometry (IC-MS) with online pretreatment of column switching technique was proposed to analyze monosaccharides hydrolyzed from extracellular polysaccharides in the activated sludge. When the matrix was eliminated in the first dimension, monosaccharides were immediately identified by IC-MS. The improved ionization efficiency was achieved with the addition of T-joint prior to MS. During the performance test, our established method showed excellent detection limits ranging from 0.34 to 2.15 μg/L for all sugar targets. Great linearity (R ≥ 0.9955) was also achieved using this method in the range from 0.01 to 5 mg/L. Furthermore, the average recoveries were obtained between 84.82 and 113.46%. RSDs for peak areas and retention times were determined as 3.76% and 0.27%, respectively. Finally, this approach provided a rapid, convenient, and practical determination of monosaccharides in the activated sludge, which would be helpful for the analysis of monosaccharides derived from other biological samples.
Collapse
Affiliation(s)
- Zhendong Zhao
- Key Laboratory of Water Pollution Treatment and Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, Hainan, China.,Analytical and Testing Center, Hainan University, Haikou, 570228, Hainan, China
| | - Qiang Lin
- Key Laboratory of Water Pollution Treatment and Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, Hainan, China.
| | - Yuhong Feng
- Analytical and Testing Center, Hainan University, Haikou, 570228, Hainan, China. .,School of Chemical Engineering and Technology, Hainan University, Haikou, 570228, Hainan, China.
| | - Yang Zhou
- School of Chemical Engineering and Technology, Hainan University, Haikou, 570228, Hainan, China
| | - Xianghui Wang
- Key Laboratory of Water Pollution Treatment and Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, Hainan, China
| |
Collapse
|
32
|
Weiß E, Schlatterer K, Beck C, Peschel A, Kretschmer D. Formyl-Peptide Receptor Activation Enhances Phagocytosis of Community-Acquired Methicillin-Resistant Staphylococcus aureus. J Infect Dis 2020; 221:668-678. [PMID: 31573600 DOI: 10.1093/infdis/jiz498] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/27/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Formyl-peptide receptors (FPRs) are important pattern recognition receptors that sense specific bacterial peptides. Formyl-peptide receptors are highly expressed on neutrophils and monocytes, and their activation promotes the migration of phagocytes to sites of infection. It is currently unknown whether FPRs may also influence subsequent processes such as bacterial phagocytosis and killing. Staphylococcus aureus, especially highly pathogenic community-acquired methicillin-resistant S aureus strains, release high amounts of FPR2 ligands, the phenol-soluble modulins. METHODS We demonstrate that FPR activation leads to upregulation of complement receptors 1 and 3 as well as FCγ receptor I on neutrophils and, consequently, increased opsonic phagocytosis of S aureus and other pathogens. RESULTS Increased phagocytosis promotes killing of S aureus and interleukin-8 release by neutrophils. CONCLUSIONS We show here for the first time that FPRs govern opsonic phagocytosis. Manipulation of FPR2 activation could open new therapeutic opportunities against bacterial pathogens.
Collapse
Affiliation(s)
- Elisabeth Weiß
- Infection Biology, Interfaculty Institute for Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
| | - Katja Schlatterer
- Infection Biology, Interfaculty Institute for Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
| | - Christian Beck
- Infection Biology, Interfaculty Institute for Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
| | - Andreas Peschel
- Infection Biology, Interfaculty Institute for Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
| | - Dorothee Kretschmer
- Infection Biology, Interfaculty Institute for Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
| |
Collapse
|
33
|
Gaio V, Cerca N. Biofilm released cells can easily be obtained in a fed-batch system using ica+ but not with ica- isolates. PeerJ 2020; 8:e9549. [PMID: 32742809 PMCID: PMC7368429 DOI: 10.7717/peerj.9549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/24/2020] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus epidermidis is one of the major opportunistic bacterial pathogens in healthcare facilities, mainly due to its strong ability to form biofilms in the surface of indwelling medical devices. To study biofilms under in vitro conditions, both fed-batch and flow systems are widely used, with the first being the most frequent due to their low cost and ease of use. Aim To assess if a fed-batch system previously developed to obtain biofilm released cells (Brc) from strong biofilm producing S. epidermidis isolates could also be used to obtain and characterize Brc from isolates with lower abilities to form biofilms. Methodology The applicability of a fed-batch system to obtain Brc from biofilms of 3 ica + and 3 ica - isolates was assessed by quantifying the biofilm and Brc biomass by optical density (OD) and colony-forming units (CFU) measurements. The effect of media replacement procedures of fed-batch systems on the amount of biofilm was determined by quantifying the biofilm and biofilm bulk fluid, by CFU, after consecutive washing steps. Results The fed-batch model was appropriate to obtain Brc from ica+ isolates, that presented a greater ability to form biofilms and release cells. However, the same was not true for ica - isolates, mainly because the washing procedure would physically remove a significant number of cells from the biofilm. Conclusions This study demonstrates that a fed-batch system is only feasible to be used to obtain Brc from S. epidermidis when studying strong and cohesive biofilm-forming isolates.
Collapse
Affiliation(s)
- Vânia Gaio
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| | - Nuno Cerca
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| |
Collapse
|
34
|
Abstract
The skin microbiome is rich in opportunities for novel therapeutics for skin diseases, and synthetic biology offers the advantage of providing novel functionality or therapeutic benefit to live biotherapeutic products. The development of novel bacterial strains whose growth can be controlled without the use of antibiotics or genetic elements conferring antibiotic resistance enables modulation of therapeutic exposure and improves safety. This study presents the design and in vitro evidence of a skin commensal whose growth can be controlled through d-alanine. The basis of this strain will support future clinical studies of this strain in humans. Using live microbes as therapeutic candidates is a strategy that has gained traction across multiple therapeutic areas. In the skin, commensal microorganisms play a crucial role in maintaining skin barrier function, homeostasis, and cutaneous immunity. Alterations of the homeostatic skin microbiome are associated with a number of skin diseases. Here, we present the design of an engineered commensal organism, Staphylococcus epidermidis, for use as a live biotherapeutic product (LBP) candidate for skin diseases. The development of novel bacterial strains whose growth can be controlled without the use of antibiotics or genetic elements conferring antibiotic resistance enables modulation of therapeutic exposure and improves safety. We therefore constructed an auxotrophic strain of S. epidermidis that requires exogenously supplied d-alanine. The S. epidermidis NRRL B-4268 Δalr1 Δalr2 Δdat strain (SEΔΔΔ) contains deletions of three biosynthetic genes: two alanine racemase genes, alr1 and alr2 (SE1674 and SE1079), and the d-alanine aminotransferase gene, dat (SE1423). These three deletions restricted growth in d-alanine-deficient medium, pooled human blood, and skin. In the presence of d-alanine, SEΔΔΔ colonized and increased expression of human β-defensin 2 in cultured human skin models in vitro. SEΔΔΔ showed a low propensity to revert to d-alanine prototrophy and did not form biofilms on plastic in vitro. These studies support the potential safety and utility of SEΔΔΔ as a live biotherapeutic strain whose growth can be controlled by d-alanine. IMPORTANCE The skin microbiome is rich in opportunities for novel therapeutics for skin diseases, and synthetic biology offers the advantage of providing novel functionality or therapeutic benefit to live biotherapeutic products. The development of novel bacterial strains whose growth can be controlled without the use of antibiotics or genetic elements conferring antibiotic resistance enables modulation of therapeutic exposure and improves safety. This study presents the design and in vitro evidence of a skin commensal whose growth can be controlled through d-alanine. The basis of this strain will support future clinical studies of this strain in humans.
Collapse
|
35
|
Lauten A, Martinović M, Kursawe L, Kikhney J, Affeld K, Kertzscher U, Falk V, Moter A. Bacterial biofilms in infective endocarditis: an in vitro model to investigate emerging technologies of antimicrobial cardiovascular device coatings. Clin Res Cardiol 2020; 110:323-331. [PMID: 32444905 PMCID: PMC7907033 DOI: 10.1007/s00392-020-01669-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 05/08/2020] [Indexed: 12/01/2022]
Abstract
Objective In spite of the progress in antimicrobial and surgical therapy, infective endocarditis (IE) is still associated with a high morbidity and mortality. IE is characterized by bacterial biofilms of the endocardium, especially of the aortic and mitral valve leading to their destruction. About one quarter of patients with formal surgery indication cannot undergo surgery. This group of patients needs further options of therapy, but due to a lack of models for IE prospects of research are low. Therefore, the purpose of this project was to establish an in vitro model of infective endocarditis to allow growth of bacterial biofilms on porcine aortic valves, serving as baseline for further research. Methods and results A pulsatile two-chamber circulation model was constructed that kept native porcine aortic valves under sterile, physiologic hemodynamic and temperature conditions. To create biofilms on porcine aortic valves the system was inoculated with Staphylococcus epidermidis PIA 8400. Aortic roots were incubated in the model for increasing periods of time (24 h and 40 h) and bacterial titration (1.5 × 104 CFU/mL and 1.5 × 105 CFU/mL) with 5 L cardiac output per minute. After incubation, tissue sections were analysed by fluorescence in situ hybridization (FISH) for direct visualization of the biofilms. Pilot tests for biofilm growth showed monospecies colonization consisting of cocci with time- and inocula-dependent increase after 24 h and 40 h (n = 4). In n = 3 experiments for 24 h, with the same inocula, FISH visualized biofilms with ribosome-containing, and thus metabolic active cocci, tissue infiltration and similar colonization pattern as observed by the FISH in human IE heart valves infected by S. epidermidis. Conclusion These results demonstrate the establishment of a novel in vitro model for bacterial biofilm growth on porcine aortic roots mimicking IE. The model will allow to identify predilection sites of valves for bacterial adhesion and biofilm growth and it may serve as baseline for further research on IE therapy and prevention, e.g. the development of antimicrobial transcatheter approaches to IE. Graphic abstract ![]()
Collapse
Affiliation(s)
- Alexander Lauten
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Standort Berlin, Berlin, Germany. .,Department of Cardiology, Charité, Universitaetsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany. .,Department of General and Interventional Cardiology and Rhythmology, HELIOS Klinikum Erfurt, Erfurt, Germany. .,Devie Medical GmbH, Bachstr. 18, 7743, Jena, Deutschland.
| | - Marc Martinović
- Department of Cardiology, Charité, Universitaetsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.
| | - Laura Kursawe
- Biofilmcenter, Department of Microbiology, Infectious Diseases and Immunology, Charité, Universitaetsmedizin Berlin, Berlin, Germany
| | - Judith Kikhney
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Standort Berlin, Berlin, Germany.,Biofilmcenter, Department of Microbiology, Infectious Diseases and Immunology, Charité, Universitaetsmedizin Berlin, Berlin, Germany
| | - Klaus Affeld
- Institute for Cardiovascular Computer-Assisted Medicine, Labor für Biofluidmechanik, Charité, Universitaetsmedizin Berlin, Berlin, Germany
| | - Ulrich Kertzscher
- Institute for Cardiovascular Computer-Assisted Medicine, Labor für Biofluidmechanik, Charité, Universitaetsmedizin Berlin, Berlin, Germany
| | - Volkmar Falk
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Standort Berlin, Berlin, Germany.,Department of Cardiology, Charité, Universitaetsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.,Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
| | - Annette Moter
- Biofilmcenter, Department of Microbiology, Infectious Diseases and Immunology, Charité, Universitaetsmedizin Berlin, Berlin, Germany
| |
Collapse
|
36
|
Walker JN, Poppler LH, Pinkner CL, Hultgren SJ, Myckatyn TM. Establishment and Characterization of Bacterial Infection of Breast Implants in a Murine Model. Aesthet Surg J 2020; 40:516-528. [PMID: 31259380 DOI: 10.1093/asj/sjz190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Staphylococcus epidermidis and Pseudomonas aeruginosa are the most common causes of Gram-positive and Gram-negative breast implant-associated infection. Little is known about how these bacteria infect breast implants as a function of implant surface characteristics and timing of infection. OBJECTIVES The aim of this work was to establish a mouse model for studying the impact of various conditions on breast implant infection. METHODS Ninety-one mice were implanted with 273 breast implant shells and infected with S. epidermidis or P. aeruginosa. Smooth, microtextured, and macrotextured breast implant shells were implanted in each mouse. Bacterial inoculation occurred during implantation or 1 day later. Implants were retrieved 1 or 7 days later. Explanted breast implant shells were sonicated, cultured, and colony-forming units determined or analyzed with scanning electron microscopy. RESULTS P. aeruginosa could be detected on all device surfaces at 1- and 7- days post infection (dpi), when mice were implanted and infected concurrently or when they were infected 1- day after implantation. However, P. aeruginosa infection was more robust on implant shells retrieved at 7 dpi and particularly on the macrotextured devices that were infected 1 day post implantation. S. epidermidis was mostly cleared from implants when mice were infected and implanted concurrently. Other the other hand, S. epidermidis could be detected on all device surfaces at 1 dpi and 2 days post implantation. However, S. epidermdis infection was suppressed by 7 dpi and 8 days post implantation. CONCLUSIONS S. epidermidis required higher inoculating doses to cause infection and was cleared within 7 days. P. aeruginosa infected at lower inoculating doses, with robust biofilms noted 7 days later.
Collapse
|
37
|
Walker JN, Pinkner CL, Lynch AJL, Ortbal S, Pinkner JS, Hultgren SJ, Myckatyn TM. Deposition of Host Matrix Proteins on Breast Implant Surfaces Facilitates Staphylococcus Epidermidis Biofilm Formation: In Vitro Analysis. Aesthet Surg J 2020; 40:281-295. [PMID: 30953053 DOI: 10.1093/asj/sjz099] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Staphylococcus epidermidis is a primary cause of breast implant-associated infection. S epidermidis possesses several virulence factors that enable it to bind both abiotic surfaces and host factors to form a biofilm. In addition S epidermidis colocalizes with matrix proteins coating explanted human breast implants. OBJECTIVES The authors sought to identify matrix proteins that S epidermidis may exploit to infect various breast implant surfaces in vitro. METHODS A combination of in vitro assays was used to characterize S epidermidis strains isolated from human breast implants to gain a better understanding of how these bacteria colonize breast implant surfaces. These included determining the (1) minimum inhibitory and bactericidal concentrations for irrigation solutions commonly used to prevent breast implant contamination; (2) expression and carriage of polysaccharide intercellular adhesin and serine-aspartate repeat proteins, which bind fibrinogen (SdrG) and collagen (SdrF), respectively; and (3) biofilm formation on varying implant surface characteristics, in different growth media, and supplemented with fibrinogen and Types I and III collagen. Scanning electron microscopy and immunofluorescence staining analyses were performed to corroborate findings from these assays. RESULTS Textured breast implant surfaces support greater bacterial biofilm formation at baseline, and the addition of collagen significantly increases biomass on all surfaces tested. We found that S epidermidis isolated from breast implants all encoded SdrF. Consistent with this finding, these strains had a clear affinity for Type I collagen, forming dense, highly structured biofilms in its presence. CONCLUSIONS The authors found that S epidermidis may utilize SdrF to interact with Type I collagen to form biofilm on breast implant surfaces. LEVEL OF EVIDENCE: 5
Collapse
Affiliation(s)
| | | | | | - Sarah Ortbal
- Washington University School of Medicine, St. Louis, MO
| | | | | | | |
Collapse
|
38
|
da Silva Filho RG, Campos AC, Souza IDS, Saramago CSDM, de Lima e Silva AA. Production of Poly-γ-Glutamic Acid (γ-PGA) by Clinical Isolates of Staphylococcus Epidermidis. Open Microbiol J 2020. [DOI: 10.2174/1874285802014010030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background and Objective:Poly-γ-glutamic acid (γ-PGA) is a constituent of theBacillus anthraciscapsule and a potential virulence factor ofS. epidermidis. In this study, a methodology for the isolation, purification and quantification of γ-PGA in the isolates was adapted. In addition, the fate of the produced γ-PGA and its antiphagocytic activity were investigated.Methods:ThecapBgene was investigated by the PCR method in 50 isolates ofS. epidermidis. A modified methodology was used for the extraction, purification, and quantification of γ-PGA using Cetyltrimethylammonium Bromide (CTAB) solution. The fate of γ-PGA was determined in Tryptic Soy Broth (TSB) medium, as well as the effect of ethanol, NaCl and KCl on the induction of the polymer production. The ability of neutrophils to phagocyte both FITC-labeled latex particles in the presence of free γ-PGA andS. epidermidiswith and without anchored γ-PGA was evaluated by cytometry.Results:The production of γ-PGA was detected in 40 isolates; all of them werecapBgene carriers. Free γ-PGA was detected and in the strain, the amount of released γ-PGA in the supernatant was 67% greater than the cell anchored γ-PGA. Phagocytosis tests performed with one γ-PGA producer isolate showed a significant reduction in neutrophil internalization.Conclusion:The adapted methodology was able to detect γ-PGA in the isolates studied. In addition to being found attached to the cell wall, it was demonstrated in this study that γ-PGA can also be found in the culture supernatant. Free γ-PGA did not determine a reduction in the internalization of latex by neutrophils, but cells with anchored γ-PGA showed significant protection against phagocytosis.
Collapse
|
39
|
Kipanga PN, Liu M, Panda SK, Mai AH, Veryser C, Van Puyvelde L, De Borggraeve WM, Van Dijck P, Matasyoh J, Luyten W. Biofilm inhibiting properties of compounds from the leaves of Warburgia ugandensis Sprague subsp ugandensis against Candida and staphylococcal biofilms. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112352. [PMID: 31676401 DOI: 10.1016/j.jep.2019.112352] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/17/2019] [Accepted: 10/24/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Warburgia ugandensis Sprague subspecies ugandensis is a plant widely distributed in Eastern, Central and Southern Africa. In humans, it is used to treat respiratory infections, tooth aches, malaria, skin infections, venereal diseases, diarrhea, fevers and aches. AIM OF THE STUDY This study aims to identify the bioactive compounds against clinically important biofilm-forming strains of Candida and staphylococci that are responsible for tissue and implanted device-related infections. METHODS Using a bioassay-guided fractionation approach, hexane -, ethanol -, acetone - and water extracts from the leaves of W. ugandensis, their subsequent fractions and isolated compounds were tested against both developing and preformed 24 h-biofilms of Candida albicans SC5314, Candida glabrata BG2, Candida glabrata ATCC 2001, Staphylococcus epidermidis 1457 and Staphylococcus aureus USA 300 using microtiter susceptibility tests. Planktonic cells were also tested in parallel for comparison purposes. Confocal scanning laser microscopy was also used to visualize effects of isolated compounds on biofilm formation. RESULTS Warburganal, polygodial and alpha-linolenic acid (ALA) were the major bioactive compounds isolated from the acetone extract of W. ugandensis. For both warburganal and polygodial, the biofilm inhibitory concentration that inhibits 50% of C. albicans developing biofilms (BIC50) was 4.5 ± 1 and 10.8 ± 5 μg/mL respectively. Against S. aureus developing biofilms, this value was 37.9 ± 8 μg/mL and 25 μg/mL with warburganal and ALA respectively. Eradication of preformed 24 h biofilms was also observed. Interestingly, synergy between the sesquiterpenoids and azoles against developing C. albicans biofilms resulted in an approximately ten-fold decrease of the effective concentration required to completely inhibit growth of the biofilms by individual compounds. The hydroxyl group in position C-9 in warburganal was identified as essential for activity against staphylococcal biofilms. We also identified additional promising bioactive sesquiterpenoids; drimenol and drimendiol from the structure-activity relationship (SAR) studies. CONCLUSIONS ALA and four sesquiterpenoids: polygodial, warburganal, drimenol and drimendiol, have shown biofilm-inhibitory activity that has not been reported before and is worth following up. These compounds are potential drug candidates to manage biofilm-based infections, possibly in combination with azoles.
Collapse
Affiliation(s)
- Purity N Kipanga
- Faculty of Pharmaceutical Sciences, KU Leuven, Belgium; Department of Biology, Animal Physiology and Neurobiology Division, KU Leuven, Belgium
| | - Maoxuan Liu
- Faculty of Pharmaceutical Sciences, KU Leuven, Belgium
| | - Sujogya K Panda
- Department of Biology, Animal Physiology and Neurobiology Division, KU Leuven, Belgium
| | - Anh Hung Mai
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven, Belgium
| | - Cedrick Veryser
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven, Belgium
| | - Luc Van Puyvelde
- Department of Biology, Animal Physiology and Neurobiology Division, KU Leuven, Belgium
| | | | - Patrick Van Dijck
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium; Laboratory of Molecular Cell Biology, KU Leuven, Belgium.
| | | | - Walter Luyten
- Department of Biology, Animal Physiology and Neurobiology Division, KU Leuven, Belgium
| |
Collapse
|
40
|
Feng G, Cheng Y, Worobo R, Borca‐Tasciuc D, Moraru C. Nanoporous anodic alumina reduces
Staphylococcus
biofilm formation. Lett Appl Microbiol 2019; 69:246-251. [DOI: 10.1111/lam.13201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 11/30/2022]
Affiliation(s)
- G. Feng
- Department of Food Science Cornell University Ithaca NY USA
| | - Y. Cheng
- Department of Food Science Cornell University Ithaca NY USA
| | - R.W. Worobo
- Department of Food Science Cornell University Ithaca NY USA
| | - D.A. Borca‐Tasciuc
- Department of Mechanical, Aerospace and Nuclear Engineering Rensselaer Polytechnic Institute Troy NY USA
| | - C.I. Moraru
- Department of Food Science Cornell University Ithaca NY USA
| |
Collapse
|
41
|
Effect of daptomycin and vancomycin on Staphylococcus epidermidis biofilms: An in vitro assessment using fluorescence in situ hybridization. PLoS One 2019; 14:e0221786. [PMID: 31454398 PMCID: PMC6711592 DOI: 10.1371/journal.pone.0221786] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/14/2019] [Indexed: 11/24/2022] Open
Abstract
Colonization of in-dwelling catheters by microbial biofilms is a major concern in patient health eventually leading to catheter-related blood stream infections. Biofilms are less susceptible to standard antibiotic therapies that are effective against planktonic bacteria. Standard procedure for the detection of microorganisms on the catheter tip is culture. However, viable but non-culturable cells (VBNCs) may be missed. The aim of this study was to evaluate the use of fluorescence in situ hybridization (FISH) as an indicator to visualize and quantify the effect of the antibiotics daptomycin and vancomycin on biofilms in situ. We established an in vitro catheter biofilm model of Staphylococcus epidermidis biofilms on polyurethane catheters. Biofilm activity was measured by FISH and correlated to colony forming units (CFU) data. Digital image analysis was used for quantification of total biofilm mass and the area of the FISH positive biofilm cells. FISH showed a pronounced effect of both antibiotics on the biofilms, with daptomycin having a significantly stronger effect in terms of both reduction of biofilm mass and number of FISH-positive cells. This supports the anti-biofilm capacity of daptomycin. Interestingly, neither antibiotic was able to eradicate all of the FISH-positive cells. In summary, FISH succeeded in visualization, quantification, and localization of antibiotic activity on biofilms. This technique adds a new tool to the arsenal of test systems for anti-biofilm compounds. FISH is a valuable complementary technique to CFU since it can be highly standardized and provides information on biofilm architecture and quantity and localization of survivor cells.
Collapse
|
42
|
Le KY, Villaruz AE, Zheng Y, He L, Fisher EL, Nguyen TH, Ho TV, Yeh AJ, Joo HS, Cheung GYC, Otto M. Role of Phenol-Soluble Modulins in Staphylococcus epidermidis Biofilm Formation and Infection of Indwelling Medical Devices. J Mol Biol 2019; 431:3015-3027. [PMID: 30954574 PMCID: PMC10999989 DOI: 10.1016/j.jmb.2019.03.030] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/10/2019] [Accepted: 03/22/2019] [Indexed: 12/20/2022]
Abstract
Phenol-soluble modulins (PSMs) are amphipathic, alpha-helical peptides that are secreted by staphylococci in high amounts in a quorum-sensing-controlled fashion. Studies performed predominantly in Staphylococcus aureus showed that PSMs structure biofilms, which results in reduced biofilm mass, while it has also been reported that S. aureus PSMs stabilize biofilms due to amyloid formation. We here analyzed the roles of PSMs in in vitro and in vivo biofilms of Staphylococcus epidermidis, the leading cause of indwelling device-associated biofilm infection. We produced isogenic deletion mutants for every S. epidermidis psm locus and a sequential deletion mutant in which production of all PSMs was abolished. In vitro analysis substantiated the role of all PSMs in biofilm structuring. PSM-dependent biofilm expansion was not observed, in accordance with our finding that no S. epidermidis PSM produced amyloids. In a mouse model of indwelling device-associated infection, the total psm deletion mutant had a significant defect in dissemination. Notably, the total psm mutant produced a significantly more substantial biofilm on the implanted catheter than the wild-type strain. Our study, which for the first time directly quantified the impact of PSMs on biofilm expansion on an implanted device, shows that the in vivo biofilm infection phenotype in S. epidermidis is in accordance with the PSM biofilm structuring and detachment model, which has important implications for the potential therapeutic application of quorum-sensing blockers.
Collapse
Affiliation(s)
- Katherine Y Le
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA; Division of Hospital Internal Medicine, Department of Medicine, Mayo Clinic College of Medicine, 200 1st Street SW, Rochester, MN 55902, USA
| | - Amer E Villaruz
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Yue Zheng
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Lei He
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA; Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No. 160 Pujian Road, Shanghai 200127, China
| | - Emilie L Fisher
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Thuan H Nguyen
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Trung V Ho
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Anthony J Yeh
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Hwang-Soo Joo
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Gordon Y C Cheung
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA.
| |
Collapse
|
43
|
Shehatou C, Logunov SL, Dunman PM, Haidaris CG, Klubben WS. Characterizing the Antimicrobial Properties of 405 nm Light and the Corning® Light-Diffusing Fiber Delivery System. Lasers Surg Med 2019; 51:887-896. [PMID: 31302937 PMCID: PMC6916415 DOI: 10.1002/lsm.23132] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2019] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND OBJECTIVES Hospital-acquired infections (HAIs) and multidrug resistant bacteria pose a significant threat to the U.S. healthcare system. With a dearth of new antibiotic approvals, novel antimicrobial strategies are required to help solve this problem. Violet-blue visible light (400-470 nm) has been shown to elicit strong antimicrobial effects toward many pathogens, including representatives of the ESKAPE bacterial pathogens, which have a high propensity to cause HAIs. However, phototherapeutic solutions to prevention or treating infections are currently limited by efficient and nonobtrusive light-delivery mechanisms. STUDY DESIGN/MATERIALS AND METHODS Here, we investigate the in vitro antimicrobial properties of flexible Corning® light-diffusing fiber (LDF) toward members of the ESKAPE pathogens in a variety of growth states and in the context of biological materials. Bacteria were grown on agar surfaces, in liquid culture and on abiotic surfaces. We also explored the effects of 405 nm light within the presence of lung surfactant, human serum, and on eukaryotic cells. Pathogens tested include Enterococcus spp, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp., Staphylococcus epidermidis, Streptococcus pyogenes, Candida albicans, and Escherichia coli. RESULTS Overall, the LDF delivery of 405 nm violet-blue light exerted a significant degree of microbicidal activity against a wide range of pathogens under diverse experimental conditions. CONCLUSIONS The results exemplify the fiber's promise as a non-traditional approach for the prevention and/or therapeutic intervention of HAIs. Lasers Surg. Med. © 2019 The Authors. Lasers in Surgery and Medicine Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cindy Shehatou
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642
| | - Stephan L Logunov
- Division of Science and Technology, Corning Research & Development Corporation, Corning, New York, 14831
| | - Paul M Dunman
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642
| | - Constantine G Haidaris
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642
| | - W Spencer Klubben
- Division of Science and Technology, Corning Research & Development Corporation, Corning, New York, 14831
| |
Collapse
|
44
|
Acosta S, Quintanilla L, Alonso M, Aparicio C, Rodríguez-Cabello JC. Recombinant AMP/Polypeptide Self-Assembled Monolayers with Synergistic Antimicrobial Properties for Bacterial Strains of Medical Relevance. ACS Biomater Sci Eng 2019; 5:4708-4716. [DOI: 10.1021/acsbiomaterials.9b00247] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sergio Acosta
- Bioforge lab, CIBER-BBN, Edificio LUCIA, University of Valladolid, Paseo Belén 19, Valladolid 47011, Spain
| | - Luis Quintanilla
- Bioforge lab, CIBER-BBN, Edificio LUCIA, University of Valladolid, Paseo Belén 19, Valladolid 47011, Spain
| | - Matilde Alonso
- Bioforge lab, CIBER-BBN, Edificio LUCIA, University of Valladolid, Paseo Belén 19, Valladolid 47011, Spain
| | - Conrado Aparicio
- MDRCBB, Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-250A Moos Tower, 515 Delaware Street Southeast, Minneapolis, Minnesota 55455, United States
| | | |
Collapse
|
45
|
Schiffer C, Hilgarth M, Ehrmann M, Vogel RF. Bap and Cell Surface Hydrophobicity Are Important Factors in Staphylococcus xylosus Biofilm Formation. Front Microbiol 2019; 10:1387. [PMID: 31293539 PMCID: PMC6603148 DOI: 10.3389/fmicb.2019.01387] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/03/2019] [Indexed: 01/04/2023] Open
Abstract
Staphylococcus (S.) xylosus is a coagulase-negative Staphylococcus species naturally present in food of animal origin with a previously described potential for biofilm formation. In this study we characterized biofilm formation of five selected strains isolated from raw fermented dry sausages, upon different growth conditions. Four strains exhibited a biofilm positive phenotype with strain-dependent intensities. Biofilm formation of S. xylosus was influenced by the addition of glucose, sodium chloride and lactate to the growth medium, respectively. It was further dependent on strain-specific cell surface properties. Three strains exhibited hydrophobic and two hydrophilic cell surface properties. The biofilm positive hydrophilic strain TMW 2.1523 adhered significantly better to hydrophilic than to hydrophobic supports, whereas the differences in adherence to hydrophobic versus hydrophilic supports were not as distinct for the hydrophobic strains TMW 2.1023, TMW 2.1323, and TMW 2.1521. Comparative genomics enabled prediction of functional biofilm-related genes and link these to phenotypic variations. While a wide range of biofilm associated factors/genes previously described for S. aureus and S. epidermidis were absent in the genomes of the five strains analyzed, they all possess the gene encoding biofilm associated protein Bap. The only biofilm negative strain TMW 2.1602 showed a mutation in the bap sequence. This study demonstrates that Bap and surface hydrophobicity are important factors in S. xylosus biofilm formation with potential impact on the assertiveness of a starter strain against autochthonous staphylococci by competitive exclusion during raw sausage fermentation.
Collapse
Affiliation(s)
- Carolin Schiffer
- Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Freising, Germany
| | - Maik Hilgarth
- Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Freising, Germany
| | - Matthias Ehrmann
- Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Freising, Germany
| | - Rudi F Vogel
- Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Freising, Germany
| |
Collapse
|
46
|
Booker AE, Hoyt DW, Meulia T, Eder E, Nicora CD, Purvine SO, Daly RA, Moore JD, Wunch K, Pfiffner SM, Lipton MS, Mouser PJ, Wrighton KC, Wilkins MJ. Deep-Subsurface Pressure Stimulates Metabolic Plasticity in Shale-Colonizing Halanaerobium spp. Appl Environ Microbiol 2019; 85:e00018-19. [PMID: 30979840 PMCID: PMC6544827 DOI: 10.1128/aem.00018-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/10/2019] [Indexed: 01/12/2023] Open
Abstract
Bacterial Halanaerobium strains become the dominant persisting microbial community member in produced fluids across geographically distinct hydraulically fractured shales. Halanaerobium is believed to be inadvertently introduced into this environment during the drilling and fracturing process and must therefore tolerate large changes in pressure, temperature, and salinity. Here, we used a Halanaerobium strain isolated from a natural gas well in the Utica Point Pleasant formation to investigate metabolic and physiological responses to growth under high-pressure subsurface conditions. Laboratory incubations confirmed the ability of Halanaerobium congolense strain WG8 to grow under pressures representative of deep shale formations (21 to 48 MPa). Under these conditions, broad metabolic and physiological shifts were identified, including higher abundances of proteins associated with the production of extracellular polymeric substances. Confocal laser scanning microscopy indicated that extracellular polymeric substance (EPS) production was associated with greater cell aggregation when biomass was cultured at high pressure. Changes in Halanaerobium central carbon metabolism under the same conditions were inferred from nuclear magnetic resonance (NMR) and gas chromatography measurements, revealing large per-cell increases in production of ethanol, acetate, and propanol and cessation of hydrogen production. These metabolic shifts were associated with carbon flux through 1,2-propanediol in response to slower fluxes of carbon through stage 3 of glycolysis. Together, these results reveal the potential for bioclogging and corrosion (via organic acid fermentation products) associated with persistent Halanaerobium growth in deep, hydraulically fractured shale ecosystems, and offer new insights into cellular mechanisms that enable these strains to dominate deep-shale microbiomes.IMPORTANCE The hydraulic fracturing of deep-shale formations for hydrocarbon recovery accounts for approximately 60% of U.S. natural gas production. Microbial activity associated with this process is generally considered deleterious due to issues associated with sulfide production, microbially induced corrosion, and bioclogging in the subsurface. Here we demonstrate that a representative Halanaerobium species, frequently the dominant microbial taxon in hydraulically fractured shales, responds to pressures characteristic of the deep subsurface by shifting its metabolism to generate more corrosive organic acids and produce more polymeric substances that cause "clumping" of biomass. While the potential for increased corrosion of steel infrastructure and clogging of pores and fractures in the subsurface may significantly impact hydrocarbon recovery, these data also offer new insights for microbial control in these ecosystems.
Collapse
Affiliation(s)
- Anne E Booker
- Department of Microbiology, Ohio State University, Columbus, Ohio, USA
| | - David W Hoyt
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Tea Meulia
- College of Food, Agricultural, and Environmental Sciences, Ohio State University, Columbus, Ohio, USA
| | - Elizabeth Eder
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Carrie D Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Samuel O Purvine
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Rebecca A Daly
- Department of Microbiology, Ohio State University, Columbus, Ohio, USA
| | - Joseph D Moore
- DowDuPont Industrial Biosciences, Wilmington, Delaware, USA
| | - Kenneth Wunch
- DowDuPont Industrial Biosciences, Wilmington, Delaware, USA
| | - Susan M Pfiffner
- Center for Environmental Biotechnology, University of Tennessee, Knoxville, Tennessee, USA
| | - Mary S Lipton
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Paula J Mouser
- Department of Civil and Environmental Engineering, University of New Hampshire, Durham, New Hampshire, USA
| | - Kelly C Wrighton
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Michael J Wilkins
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
47
|
Gaio V, Cerca N. Cells released from S. epidermidis biofilms present increased antibiotic tolerance to multiple antibiotics. PeerJ 2019; 7:e6884. [PMID: 31143534 PMCID: PMC6525591 DOI: 10.7717/peerj.6884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022] Open
Abstract
Biofilm released cells (Brc) are thought to present an intermediary phenotype between biofilm and planktonic cells and this has the potential of affecting their antimicrobial tolerance. AIM Compare the antimicrobial tolerance profiles of Brc, planktonic or biofilm cultures of S. epidermidis. METHODOLOGY Planktonic, biofilm cultures or Brc from 11 isolates were exposed to peak serum concentrations (PSC) of antibiotics. The antimicrobial killing effect in the three populations was determined by CFU. RESULTS Increased Brc tolerance to vancomycin, teicoplanin, rifampicin, erythromycin, and tetracycline was confirmed in model strain 9142. Furthermore, significant differences in the susceptibility of Brc to vancomycin were further found in 10 other clinical isolates. CONCLUSIONS Brc from distinct clinical isolates presented a decreased susceptibility to most antibiotics tested and maintained that enhanced tolerance despite growing planktonically for up to 6 h. Our data suggest that Brc maintain the typical enhanced antibiotic tolerance of biofilm populations, further suggesting that addressing antimicrobial susceptibility in planktonic cultures might not reflect the full potential of biofilm-associated bacteria to survive therapy.
Collapse
Affiliation(s)
- Vânia Gaio
- Laboratory of Research in Biofilms Rosário Oliveira-Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Nuno Cerca
- Laboratory of Research in Biofilms Rosário Oliveira-Centre of Biological Engineering, University of Minho, Braga, Portugal
| |
Collapse
|
48
|
Langerhans Cells Sense Staphylococcus aureus Wall Teichoic Acid through Langerin To Induce Inflammatory Responses. mBio 2019; 10:mBio.00330-19. [PMID: 31088921 PMCID: PMC6520447 DOI: 10.1128/mbio.00330-19] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The bacterium Staphylococcus aureus is an important cause of skin infections and is also associated with the occurrence and severity of eczema. Langerhans cells (LCs), a specific subset of skin immune cells, participate in the immune response to S. aureus, but it is yet unclear how LCs recognize S. aureus. Therefore, we investigated the molecular mechanism underlying the interaction between LCs and S. aureus. We identified that wall teichoic acid, an abundant polymer on the S. aureus surface, is recognized by langerin, a receptor unique to LCs. This interaction allows LCs to discriminate S. aureus from other related staphylococcal species and initiates a proinflammatory response similar to that observed in patients with eczema. Our data therefore provide important new insights into the relationship between S. aureus, LCs, and eczema. Staphylococcus aureus is a major cause of skin and soft tissue infections and aggravator of the inflammatory skin disease atopic dermatitis (AD [eczema]). Epicutaneous exposure to S. aureus induces Th17 responses through skin Langerhans cells (LCs), which paradoxically contribute to host defense but also to AD pathogenesis. The molecular mechanisms underlying the interaction between S. aureus and LCs are poorly understood. Here we demonstrate that human LCs directly interact with S. aureus through the pattern recognition receptor langerin (CD207). Human, but not mouse, langerin interacts with S. aureus through the conserved β-N-acetylglucosamine (GlcNAc) modifications on wall teichoic acid (WTA), thereby discriminating S. aureus from other staphylococcal species. Importantly, the specific S. aureus WTA glycoprofile strongly influences the level of proinflammatory cytokines that are produced by in vitro-generated LCs. Finally, in a murine epicutaneous infection model, S. aureus strongly upregulated transcripts of Cxcl1, Il6, and Il17, which required the presence of both human langerin and WTA β-GlcNAc. Our findings provide molecular insight into the unique proinflammatory capacities of S. aureus in relation to skin inflammation.
Collapse
|
49
|
Transcriptional Regulation of icaADBC by both IcaR and TcaR in Staphylococcus epidermidis. J Bacteriol 2019; 201:JB.00524-18. [PMID: 30602488 DOI: 10.1128/jb.00524-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/17/2018] [Indexed: 12/17/2022] Open
Abstract
S. epidermidis is a primary cause of biofilm-mediated infections in humans due to adherence to foreign bodies. A major staphylococcal biofilm accumulation molecule is polysaccharide intracellular adhesin (PIA), which is synthesized by enzymes encoded by the icaADBC operon. Expression of PIA is highly variable among clinical isolates, suggesting that PIA expression levels are selected in certain niches of the host. However, the mechanisms that govern enhanced icaADBC transcription and PIA synthesis in these isolates are not known. We hypothesized that enhanced PIA synthesis in these isolates was due to function of IcaR and/or TcaR. Thus, two S. epidermidis isolates (1457 and CSF41498) with different icaADBC transcription and PIA expression levels were studied. Constitutive expression of both icaR and tcaR demonstrated that both repressors are functional and can completely repress icaADBC transcription in both 1457 and CSF41498. However, it was found that IcaR was the primary repressor for CSF41498 and TcaR was the primary repressor for 1457. Further analysis demonstrated that icaR transcription was repressed in 1457 in comparison to CSF41498, suggesting that TcaR functions as a repressor only in the absence of IcaR. Indeed, DNase I footprinting suggests IcaR and TcaR may bind to the same site within the icaR-icaA intergenic region. Lastly, we found mutants expressing variable amounts of PIA could rapidly be selected from both 1457 and CSF41498. Collectively, we propose that strains producing enhanced PIA synthesis are selected within certain niches of the host through several genetic mechanisms that function to repress icaR transcription, thus increasing PIA synthesis.IMPORTANCE Staphylococcus epidermidis is a commensal bacterium that resides on our skin. As a commensal, it protects humans from bacterial pathogens through a variety of mechanisms. However, it is also a significant cause of biofilm infections due to its ability to bind to plastic. Polysaccharide intercellular adhesin is a significant component of biofilm, and we propose that the expression of this polysaccharide is beneficial in certain host niches, such as providing extra strength when the bacterium is colonizing the lumen of a catheter, and detrimental in others, such as colonization of the skin surface. We show here that fine-tuning of icaADBC transcription, and thus PIA synthesis, is mediated via two transcriptional repressors, IcaR and TcaR.
Collapse
|
50
|
Cooke AC, Nello AV, Ernst RK, Schertzer JW. Analysis of Pseudomonas aeruginosa biofilm membrane vesicles supports multiple mechanisms of biogenesis. PLoS One 2019; 14:e0212275. [PMID: 30763382 PMCID: PMC6375607 DOI: 10.1371/journal.pone.0212275] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 01/30/2019] [Indexed: 01/07/2023] Open
Abstract
Outer Membrane Vesicles (OMVs) are ubiquitous in bacterial environments and enable interactions within and between species. OMVs are observed in lab-grown and environmental biofilms, but our understanding of their function comes primarily from planktonic studies. Planktonic OMVs assist in toxin delivery, cell-cell communication, horizontal gene transfer, small RNA trafficking, and immune system evasion. Previous studies reported differences in size and proteomic cargo between planktonic and agar plate biofilm OMVs, suggesting possible differences in function between OMV types. In Pseudomonas aeruginosa interstitial biofilms, extracellular vesicles were reported to arise through cell lysis, in contrast to planktonic OMV biogenesis that involves the Pseudomonas Quinolone Signal (PQS) without appreciable autolysis. Differences in biogenesis mechanism could provide a rationale for observed differences in OMV characteristics between systems. Using nanoparticle tracking, we found that P. aeruginosa PAO1 planktonic and biofilm OMVs had similar characteristics. However, P. aeruginosa PA14 OMVs were smaller, with planktonic OMVs also being smaller than their biofilm counterparts. Large differences in Staphylococcus killing ability were measured between OMVs from different strains, and a smaller within-strain difference was recorded between PA14 planktonic and biofilm OMVs. Across all conditions, the predatory ability of OMVs negatively correlated with their size. To address biogenesis mechanism, we analyzed vesicles from wild type and pqsA mutant biofilms. This showed that PQS is required for physiological-scale production of biofilm OMVs, and time-course analysis confirmed that PQS production precedes OMV production as it does in planktonic cultures. However, a small sub-population of vesicles was detected in pqsA mutant biofilms whose size distribution more resembled sonicated cell debris than wild type OMVs. These results support the idea that, while a small and unique population of vesicles in P. aeruginosa biofilms may result from cell lysis, the PQS-induced mechanism is required to generate the majority of OMVs produced by wild type communities.
Collapse
Affiliation(s)
- Adam C. Cooke
- Department of Biological Sciences, Binghamton University, Binghamton, New York, United States of America
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, United States of America
| | - Alexander V. Nello
- Department of Biological Sciences, Binghamton University, Binghamton, New York, United States of America
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, United States of America
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, United States of America
| | - Jeffrey W. Schertzer
- Department of Biological Sciences, Binghamton University, Binghamton, New York, United States of America
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, United States of America
| |
Collapse
|